Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Mol Cell Cardiol ; 189: 12-24, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38401179

ABSTRACT

Cardiomyocytes activate the unfolded protein response (UPR) transcription factor ATF6 during pressure overload-induced hypertrophic growth. The UPR is thought to increase ER protein folding capacity and maintain proteostasis. ATF6 deficiency during pressure overload leads to heart failure, suggesting that ATF6 protects against myocardial dysfunction by preventing protein misfolding. However, conclusive evidence that ATF6 prevents toxic protein misfolding during cardiac hypertrophy is still pending. Here, we found that activation of the UPR, including ATF6, is a common response to pathological cardiac hypertrophy in mice. ATF6 KO mice failed to induce sufficient levels of UPR target genes in response to chronic isoproterenol infusion or transverse aortic constriction (TAC), resulting in impaired cardiac growth. To investigate the effects of ATF6 on protein folding, the accumulation of poly-ubiquitinated proteins as well as soluble amyloid oligomers were directly quantified in hypertrophied hearts of WT and ATF6 KO mice. Whereas only low levels of protein misfolding was observed in WT hearts after TAC, ATF6 KO mice accumulated increased quantities of misfolded protein, which was associated with impaired myocardial function. Collectively, the data suggest that ATF6 plays a critical adaptive role during cardiac hypertrophy by protecting against protein misfolding.


Subject(s)
Aortic Valve Stenosis , Cardiomegaly , Animals , Mice , Cardiomegaly/pathology , Myocytes, Cardiac/metabolism , Myocardium/metabolism , Transcription Factors/metabolism , Gene Expression Regulation , Aortic Valve Stenosis/metabolism , Mice, Knockout
2.
Circulation ; 147(1): 66-82, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36317534

ABSTRACT

BACKGROUND: Cardiac hypertrophy increases demands on protein folding, which causes an accumulation of misfolded proteins in the endoplasmic reticulum (ER). These misfolded proteins can be removed by the adaptive retrotranslocation, polyubiquitylation, and a proteasome-mediated degradation process, ER-associated degradation (ERAD), which, as a biological process and rate, has not been studied in vivo. To investigate a role for ERAD in a pathophysiological model, we examined the function of the functional initiator of ERAD, valosin-containing protein-interacting membrane protein (VIMP), positing that VIMP would be adaptive in pathological cardiac hypertrophy in mice. METHODS: We developed a new method involving cardiac myocyte-specific adeno-associated virus serovar 9-mediated expression of the canonical ERAD substrate, TCRα, to measure the rate of ERAD, ie, ERAD flux, in the heart in vivo. Adeno-associated virus serovar 9 was also used to either knock down or overexpress VIMP in the heart. Then mice were subjected to transverse aortic constriction to induce pressure overload-induced cardiac hypertrophy. RESULTS: ERAD flux was slowed in both human heart failure and mice after transverse aortic constriction. Surprisingly, although VIMP adaptively contributes to ERAD in model cell lines, in the heart, VIMP knockdown increased ERAD and ameliorated transverse aortic constriction-induced cardiac hypertrophy. Coordinately, VIMP overexpression exacerbated cardiac hypertrophy, which was dependent on VIMP engaging in ERAD. Mechanistically, we found that the cytosolic protein kinase SGK1 (serum/glucocorticoid regulated kinase 1) is a major driver of pathological cardiac hypertrophy in mice subjected to transverse aortic constriction, and that VIMP knockdown decreased the levels of SGK1, which subsequently decreased cardiac pathology. We went on to show that although it is not an ER protein, and resides outside of the ER, SGK1 is degraded by ERAD in a noncanonical process we call ERAD-Out. Despite never having been in the ER, SGK1 is recognized as an ERAD substrate by the ERAD component DERLIN1, and uniquely in cardiac myocytes, VIMP displaces DERLIN1 from initiating ERAD, which decreased SGK1 degradation and promoted cardiac hypertrophy. CONCLUSIONS: ERAD-Out is a new preferentially favored noncanonical form of ERAD that mediates the degradation of SGK1 in cardiac myocytes, and in so doing is therefore an important determinant of how the heart responds to pathological stimuli, such as pressure overload.


Subject(s)
Cardiomegaly , Endoplasmic Reticulum-Associated Degradation , Animals , Humans , Mice , Cardiomegaly/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum-Associated Degradation/physiology , Myocytes, Cardiac/metabolism , Unfolded Protein Response/physiology
3.
Am J Physiol Heart Circ Physiol ; 325(2): H311-H320, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37294892

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) is a central regulator of protein synthesis that senses and responds to a variety of stimuli to coordinate cellular metabolism with environmental conditions. To ensure that protein synthesis is inhibited during unfavorable conditions, translation is directly coupled to the sensing of cellular protein homeostasis. Thus, translation is attenuated during endoplasmic reticulum (ER) stress by direct inhibition of the mTORC1 pathway. However, residual mTORC1 activity is maintained during prolonged ER stress, which is thought to be involved in translational reprogramming and adaption to ER stress. By analyzing the dynamics of mTORC1 regulation during ER stress, we unexpectedly found that mTORC1 is transiently activated in cardiomyocytes within minutes at the onset of ER stress before being inhibited during chronic ER stress. This dynamic regulation of mTORC1 appears to be mediated, at least in part, by ATF6, as its activation was sufficient to induce the biphasic control of mTORC1. We further showed that protein synthesis remains dependent on mTORC1 throughout the ER stress response and that mTORC1 activity is essential for posttranscriptional induction of several unfolded protein response genes. Pharmacological inhibition of mTORC1 increased cell death during ER stress, indicating that the mTORC1 pathway serves adaptive functions during ER stress in cardiomyocytes potentially by controlling the expression of protective unfolded protein response genes.NEW & NOTEWORTHY Cells coordinate translation rates with protein quality control to ensure that protein synthesis is initiated primarily when proper protein folding can be achieved. Long-term activity of the unfolded protein response is therefore associated with an inhibition of mTORC1, a central regulator of protein synthesis. Here, we found that mTORC1 is transiently activated early in response to ER stress before it is inhibited. Importantly, partial mTORC1 activity remained essential for the upregulation of adaptive unfolded protein response genes and cell survival in response to ER stress. Our data reveal a complex regulation of mTORC1 during ER stress and its involvement in the adaptive unfolded protein response.


Subject(s)
Myocytes, Cardiac , Signal Transduction , Mechanistic Target of Rapamycin Complex 1/metabolism , Myocytes, Cardiac/metabolism , Endoplasmic Reticulum Stress , Unfolded Protein Response , Cell Death , Proteins/metabolism
4.
J Biol Chem ; 295(22): 7566-7583, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32327487

ABSTRACT

We have previously demonstrated that ischemia/reperfusion (I/R) impairs endoplasmic reticulum (ER)-based protein folding in the heart and thereby activates an unfolded protein response sensor and effector, activated transcription factor 6α (ATF6). ATF6 then induces mesencephalic astrocyte-derived neurotrophic factor (MANF), an ER-resident protein with no known structural homologs and unclear ER function. To determine MANF's function in the heart in vivo, here we developed a cardiomyocyte-specific MANF-knockdown mouse model. MANF knockdown increased cardiac damage after I/R, which was reversed by AAV9-mediated ectopic MANF expression. Mechanistically, MANF knockdown in cultured neonatal rat ventricular myocytes (NRVMs) impaired protein folding in the ER and cardiomyocyte viability during simulated I/R. However, this was not due to MANF-mediated protection from reactive oxygen species generated during reperfusion. Because I/R impairs oxygen-dependent ER protein disulfide formation and such impairment can be caused by reductive stress in the ER, we examined the effects of the reductive ER stressor DTT. MANF knockdown in NRVMs increased cell death from DTT-mediated reductive ER stress, but not from nonreductive ER stresses caused by thapsigargin-mediated ER Ca2+ depletion or tunicamycin-mediated inhibition of ER protein glycosylation. In vitro, recombinant MANF exhibited chaperone activity that depended on its conserved cysteine residues. Moreover, in cells, MANF bound to a model ER protein exhibiting improper disulfide bond formation during reductive ER stress but did not bind to this protein during nonreductive ER stress. We conclude that MANF is an ER chaperone that enhances protein folding and myocyte viability during reductive ER stress.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Molecular Chaperones/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Nerve Growth Factors/metabolism , Animals , Cell Survival , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Glycosylation , HeLa Cells , Humans , Mice , Mice, Knockout , Molecular Chaperones/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Myocytes, Cardiac/pathology , Nerve Growth Factors/genetics , Reactive Oxygen Species
5.
Am J Physiol Heart Circ Physiol ; 320(5): H1813-H1821, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33666503

ABSTRACT

Although peroxisomes have been extensively studied in other cell types, their presence and function have gone virtually unexamined in cardiac myocytes. Here, in neonatal rat ventricular myocytes (NRVM) we showed that several known peroxisomal proteins co-localize to punctate structures with a morphology typical of peroxisomes. Surprisingly, we found that the peroxisomal protein, fatty acyl-CoA reductase 1 (FAR1), was upregulated by pharmacological and pathophysiological ER stress induced by tunicamycin (TM) and simulated ischemia-reperfusion (sI/R), respectively. Moreover, FAR1 induction in NRVM was mediated by the ER stress sensor, activating transcription factor 6 (ATF6). Functionally, FAR1 knockdown reduced myocyte death during oxidative stress induced by either sI/R or hydrogen peroxide (H2O2). Thus, Far1 is an ER stress-inducible gene, which encodes a protein that localizes to peroxisomes of cardiac myocytes, where it reduces myocyte viability during oxidative stress. Since FAR1 is critical for plasmalogen synthesis, these results imply that plasmalogens may exert maladaptive effects on the viability of myocytes exposed to oxidative stress.NEW & NOTEWORTHY The peroxisomal enzyme, FAR1, was shown to be an ER stress- and ATF6-inducible protein that localizes to peroxisomes in cardiac myocytes. FAR1 decreases myocyte viability during oxidative stress.


Subject(s)
Activating Transcription Factor 6/metabolism , Aldehyde Oxidoreductases/biosynthesis , Endoplasmic Reticulum Stress , Myocardial Reperfusion Injury/enzymology , Myocytes, Cardiac/enzymology , Peroxisomes/enzymology , Activating Transcription Factor 6/genetics , Aldehyde Oxidoreductases/genetics , Animals , Animals, Newborn , Cell Hypoxia , Cell Survival , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Enzyme Induction , Hydrogen Peroxide/toxicity , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidative Stress , Peroxisomes/drug effects , Peroxisomes/metabolism , Rats , Tunicamycin/toxicity
6.
Circ Res ; 124(1): 79-93, 2019 01 04.
Article in English | MEDLINE | ID: mdl-30582446

ABSTRACT

RATIONALE: Endoplasmic reticulum (ER) stress dysregulates ER proteostasis, which activates the transcription factor, ATF6 (activating transcription factor 6α), an inducer of genes that enhance protein folding and restore ER proteostasis. Because of increased protein synthesis, it is possible that protein folding and ER proteostasis are challenged during cardiac myocyte growth. However, it is not known whether ATF6 is activated, and if so, what its function is during hypertrophic growth of cardiac myocytes. OBJECTIVE: To examine the activity and function of ATF6 during cardiac hypertrophy. METHODS AND RESULTS: We found that ER stress and ATF6 were activated and ATF6 target genes were induced in mice subjected to an acute model of transverse aortic constriction, or to free-wheel exercise, both of which promote adaptive cardiac myocyte hypertrophy with preserved cardiac function. Cardiac myocyte-specific deletion of Atf6 (ATF6 cKO [conditional knockout]) blunted transverse aortic constriction and exercise-induced cardiac myocyte hypertrophy and impaired cardiac function, demonstrating a role for ATF6 in compensatory myocyte growth. Transcript profiling and chromatin immunoprecipitation identified RHEB (Ras homologue enriched in brain) as an ATF6 target gene in the heart. RHEB is an activator of mTORC1 (mammalian/mechanistic target of rapamycin complex 1), a major inducer of protein synthesis and subsequent cell growth. Both transverse aortic constriction and exercise upregulated RHEB, activated mTORC1, and induced cardiac hypertrophy in wild type mouse hearts but not in ATF6 cKO hearts. Mechanistically, knockdown of ATF6 in neonatal rat ventricular myocytes blocked phenylephrine- and IGF1 (insulin-like growth factor 1)-mediated RHEB induction, mTORC1 activation, and myocyte growth, all of which were restored by ectopic RHEB expression. Moreover, adeno-associated virus 9- RHEB restored cardiac growth to ATF6 cKO mice subjected to transverse aortic constriction. Finally, ATF6 induced RHEB in response to growth factors, but not in response to other activators of ATF6 that do not induce growth, indicating that ATF6 target gene induction is stress specific. CONCLUSIONS: Compensatory cardiac hypertrophy activates ER stress and ATF6, which induces RHEB and activates mTORC1. Thus, ATF6 is a previously unrecognized link between growth stimuli and mTORC1-mediated cardiac growth.


Subject(s)
Activating Transcription Factor 6/metabolism , Hypertrophy, Left Ventricular/enzymology , Mechanistic Target of Rapamycin Complex 1/metabolism , Myocytes, Cardiac/enzymology , Ras Homolog Enriched in Brain Protein/metabolism , Transcriptional Activation , Ventricular Function, Left , Ventricular Remodeling , Activating Transcription Factor 6/deficiency , Activating Transcription Factor 6/genetics , Animals , Animals, Newborn , Disease Models, Animal , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum Stress , Genetic Predisposition to Disease , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Phenotype , Protein Folding , Proteostasis , Ras Homolog Enriched in Brain Protein/genetics , Signal Transduction
7.
J Mol Cell Cardiol ; 143: 132-144, 2020 06.
Article in English | MEDLINE | ID: mdl-32339566

ABSTRACT

The effects of ER stress on protein secretion by cardiac myocytes are not well understood. In this study, the ER stressor thapsigargin (TG), which depletes ER calcium, induced death of cultured neonatal rat ventricular myocytes (NRVMs) in high media volume but fostered protection in low media volume. In contrast, another ER stressor, tunicamycin (TM), a protein glycosylation inhibitor, induced NRVM death in all media volumes, suggesting that protective proteins were secreted in response to TG but not TM. Proteomic analyses of TG- and TM-conditioned media showed that the secretion of most proteins was inhibited by TG and TM; however, secretion of several ER-resident proteins, including GRP78 was increased by TG but not TM. Simulated ischemia, which decreases ER/SR calcium also increased secretion of these proteins. Mechanistically, secreted GRP78 was shown to enhance survival of NRVMs by collaborating with a cell-surface protein, CRIPTO, to activate protective AKT signaling and to inhibit death-promoting SMAD2 signaling. Thus, proteins secreted during ER stress mediated by ER calcium depletion can enhance cardiac myocyte viability.


Subject(s)
Endoplasmic Reticulum Stress , Myocytes, Cardiac/metabolism , Proteome , Proteomics , Animals , Apoptosis , Autocrine Communication , Biomarkers , Calcium/metabolism , Calcium Signaling/drug effects , Cell Survival , Cells, Cultured , Disease Susceptibility , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , Mice , Myocytes, Cardiac/drug effects , Neoplasm Proteins/metabolism , Paracrine Communication , Proteomics/methods , Rats , Sarcoplasmic Reticulum/metabolism , Signal Transduction/drug effects , Thapsigargin/pharmacology
8.
Int J Mol Sci ; 21(3)2020 Feb 08.
Article in English | MEDLINE | ID: mdl-32046286

ABSTRACT

There are more than 2000 transcription factors in eukaryotes, many of which are subject to complex mechanisms fine-tuning their activity and their transcriptional programs to meet the vast array of conditions under which cells must adapt to thrive and survive. For example, conditions that impair protein folding in the endoplasmic reticulum (ER), sometimes called ER stress, elicit the relocation of the ER-transmembrane protein, activating transcription factor 6α (ATF6α), to the Golgi, where it is proteolytically cleaved. This generates a fragment of ATF6α that translocates to the nucleus, where it regulates numerous genes that restore ER protein-folding capacity but is degraded soon after. Thus, upon ER stress, ATF6α is converted from a stable, transmembrane protein, to a rapidly degraded, nuclear protein that is a potent transcription factor. This review focuses on the molecular mechanisms governing ATF6α location, activity, and stability, as well as the transcriptional programs ATF6α regulates, whether canonical genes that restore ER protein-folding or unexpected, non-canonical genes affecting cellular functions beyond the ER. Moreover, we will review fascinating roles for an ATF6α isoform, ATF6ß, which has a similar mode of activation but, unlike ATF6α, is a long-lived, weak transcription factor that may moderate the genetic effects of ATF6α.


Subject(s)
Activating Transcription Factor 6/metabolism , Endoplasmic Reticulum Stress , Transcription, Genetic , Animals , Gene Expression Regulation , Humans , Myocardium/metabolism , Protein Isoforms/metabolism , Transcription Factors/metabolism
9.
Int J Mol Sci ; 21(4)2020 Feb 18.
Article in English | MEDLINE | ID: mdl-32085622

ABSTRACT

Activating transcription factor-6 α (ATF6) is one of the three main sensors and effectors of the endoplasmic reticulum (ER) stress response and, as such, it is critical for protecting the heart and other tissues from a variety of environmental insults and disease states. In the heart, ATF6 has been shown to protect cardiac myocytes. However, its roles in other cell types in the heart are unknown. Here we show that ATF6 decreases the activation of cardiac fibroblasts in response to the cytokine, transforming growth factor ß (TGFß), which can induce fibroblast trans-differentiation into a myofibroblast phenotype through signaling via the TGFß-Smad pathway. ATF6 activation suppressed fibroblast contraction and the induction of α smooth muscle actin (αSMA). Conversely, fibroblasts were hyperactivated when ATF6 was silenced or deleted. ATF6 thus represents a novel inhibitor of the TGFß-Smad axis of cardiac fibroblast activation.


Subject(s)
Activating Transcription Factor 6/metabolism , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Myocardium/pathology , Unfolded Protein Response , Animals , Biomarkers/metabolism , Endoplasmic Reticulum/drug effects , Fibroblasts/drug effects , Fibrosis , Gene Expression Regulation/drug effects , Heart Ventricles/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Phosphorylation/drug effects , Signal Transduction/drug effects , Smad2 Protein/metabolism , Stress Fibers/drug effects , Stress Fibers/metabolism , Transforming Growth Factor beta/pharmacology
10.
Circ Res ; 120(5): 862-875, 2017 Mar 03.
Article in English | MEDLINE | ID: mdl-27932512

ABSTRACT

RATIONALE: Endoplasmic reticulum (ER) stress causes the accumulation of misfolded proteins in the ER, activating the transcription factor, ATF6 (activating transcription factor 6 alpha), which induces ER stress response genes. Myocardial ischemia induces the ER stress response; however, neither the function of this response nor whether it is mediated by ATF6 is known. OBJECTIVE: Here, we examined the effects of blocking the ATF6-mediated ER stress response on ischemia/reperfusion (I/R) in cardiac myocytes and mouse hearts. METHODS AND RESULTS: Knockdown of ATF6 in cardiac myocytes subjected to I/R increased reactive oxygen species and necrotic cell death, both of which were mitigated by ATF6 overexpression. Under nonstressed conditions, wild-type and ATF6 knockout mouse hearts were similar. However, compared with wild-type, ATF6 knockout hearts showed increased damage and decreased function after I/R. Mechanistically, gene array analysis showed that ATF6, which is known to induce genes encoding ER proteins that augment ER protein folding, induced numerous oxidative stress response genes not previously known to be ATF6-inducible. Many of the proteins encoded by the ATF6-induced oxidative stress genes identified here reside outside the ER, including catalase, which is known to decrease damaging reactive oxygen species in the heart. Catalase was induced by the canonical ER stressor, tunicamycin, and by I/R in cardiac myocytes from wild-type but not in cardiac myocytes from ATF6 knockout mice. ER stress response elements were identified in the catalase gene and were shown to bind ATF6 in cardiac myocytes, which increased catalase promoter activity. Overexpression of catalase, in vivo, restored ATF6 knockout mouse heart function to wild-type levels in a mouse model of I/R, as did adeno-associated virus 9-mediated ATF6 overexpression. CONCLUSIONS: ATF6 serves an important role as a previously unappreciated link between the ER stress and oxidative stress gene programs, supporting a novel mechanism by which ATF6 decreases myocardial I/R damage.


Subject(s)
Activating Transcription Factor 6/biosynthesis , Endoplasmic Reticulum Stress/physiology , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Oxidative Stress/physiology , Activating Transcription Factor 6/deficiency , Animals , Animals, Newborn , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Myocytes, Cardiac , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
12.
J Mol Cell Cardiol ; 103: 48-55, 2017 02.
Article in English | MEDLINE | ID: mdl-28077321

ABSTRACT

Deletion of Ca2+/calmodulin-dependent protein kinase II delta (CaMKIIδ) has been shown to protect against in vivo ischemia/reperfusion (I/R) injury. It remains unclear which CaMKIIδ isoforms and downstream mechanisms are responsible for the salutary effects of CaMKIIδ gene deletion. In this study we sought to compare the roles of the CaMKIIδB and CaMKIIδC subtypes and the mechanisms by which they contribute to ex vivo I/R damage. WT, CaMKIIδKO, and mice expressing only CaMKIIδB or δC were subjected to ex vivo global ischemia for 25min followed by reperfusion. Infarct formation was assessed at 60min reperfusion by triphenyl tetrazolium chloride (TTC) staining. Deletion of CaMKIIδ conferred significant protection from ex vivo I/R. Re-expression of CaMKIIδC in the CaMKIIδKO background reversed this effect and exacerbated myocardial damage and dysfunction following I/R, while re-expression of CaMKIIδB was protective. Selective activation of CaMKIIδC in response to I/R was evident in a subcellular fraction enriched for cytosolic/membrane proteins. Further studies demonstrated differential regulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling and tumor necrosis factor alpha (TNF-α) expression by CaMKIIδB and CaMKIIδC. Selective activation of CaMKIIδC was also observed and associated with NF-κB activation in neonatal rat ventricular myocytes (NRVMs) subjected to oxidative stress. Pharmacological inhibition of NF-κB or TNF-α significantly ameliorated infarct formation in WT mice and those that re-express CaMKIIδC, demonstrating distinct roles for CaMKIIδ subtypes in I/R and implicating acute activation of CaMKIIδC and NF-κB in the pathogenesis of reperfusion injury.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Animals , Biopsy , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Disease Models, Animal , Echocardiography , Gene Knockout Techniques , Mice , Mice, Transgenic , Myocardial Infarction/diagnosis , Myocardial Infarction/etiology , Myocardial Infarction/mortality , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/mortality , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Phosphorylation , Rats , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Ventricular Dysfunction
14.
Circ Res ; 117(6): 536-46, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26137860

ABSTRACT

RATIONALE: Hydroxymethyl glutaryl-coenzyme A reductase degradation protein 1 (Hrd1) is an endoplasmic reticulum (ER)-transmembrane E3 ubiquitin ligase that has been studied in yeast, where it contributes to ER protein quality control by ER-associated degradation (ERAD) of misfolded proteins that accumulate during ER stress. Neither Hrd1 nor ERAD has been studied in the heart, or in cardiac myocytes, where protein quality control is critical for proper heart function. OBJECTIVE: The objective of this study were to elucidate roles for Hrd1 in ER stress, ERAD, and viability in cultured cardiac myocytes and in the mouse heart, in vivo. METHODS AND RESULTS: The effects of small interfering RNA-mediated Hrd1 knockdown were examined in cultured neonatal rat ventricular myocytes. The effects of adeno-associated virus-mediated Hrd1 knockdown and overexpression were examined in the hearts of mice subjected to pressure overload-induced pathological cardiac hypertrophy, which challenges protein-folding capacity. In cardiac myocytes, the ER stressors, thapsigargin and tunicamycin increased ERAD, as well as adaptive ER stress proteins, and minimally affected cell death. However, when Hrd1 was knocked down, thapsigargin and tunicamycin dramatically decreased ERAD, while increasing maladaptive ER stress proteins and cell death. In vivo, Hrd1 knockdown exacerbated cardiac dysfunction and increased apoptosis and cardiac hypertrophy, whereas Hrd1 overexpression preserved cardiac function and decreased apoptosis and attenuated cardiac hypertrophy in the hearts of mice subjected to pressure overload. CONCLUSIONS: Hrd1 and ERAD are essential components of the adaptive ER stress response in cardiac myocytes. Hrd1 contributes to preserving heart structure and function in a mouse model of pathological cardiac hypertrophy.


Subject(s)
Adaptation, Physiological/physiology , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum-Associated Degradation/physiology , Myocytes, Cardiac/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Animals , Animals, Newborn , Cells, Cultured , Endoplasmic Reticulum/metabolism , Gene Knockdown Techniques , Mice , Rats , Rats, Sprague-Dawley
15.
Proc Natl Acad Sci U S A ; 116(40): 19774-19776, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31515450

Subject(s)
Amides , Peptides , Animals , Mice
16.
J Mol Cell Cardiol ; 100: 54-63, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27721024

ABSTRACT

BACKGROUND: Myocardial infarction is followed by cardiac dysfunction, cellular death, and ventricular remodeling, including tissue fibrosis. S100A4 protein plays multiple roles in cellular survival, and tissue fibrosis, but the relative role of the S100A4 in the myocardium after myocardial infarction is unknown. This study aims to investigate the role of S100A4 in myocardial remodeling and cardiac function following infarct damage. METHODS AND RESULTS: S100A4 expression is low in the adult myocardium, but significantly increased following myocardial infarction. Deletion of S100A4 increased cardiac damage after myocardial infarction, whereas cardiac myocyte-specific overexpression of S100A4 protected the infarcted myocardium. Decreased cardiac function in S100A4 Knockout mice was accompanied with increased cardiac remodeling, fibrosis, and diminished capillary density in the remote myocardium. Loss of S100A4 caused increased apoptotic cell death both in vitro and in vivo in part mediated by decreased VEGF expression. Conversely, S100A4 overexpression protected cells against apoptosis in vitro and in vivo. Increased pro-survival AKT-signaling explained reduced apoptosis in S100A4 overexpressing cells. CONCLUSION: S100A4 expression protects cardiac myocytes against myocardial ischemia and is required for stabilization of cardiac function after MI.


Subject(s)
Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardium/metabolism , S100 Calcium-Binding Protein A4/genetics , Stress, Physiological/genetics , Animals , Cell Death/genetics , Disease Models, Animal , Echocardiography , Gene Expression , Hemodynamics , Mice , Mice, Knockout , Myocardial Infarction/diagnosis , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardial Ischemia/diagnosis , Myocardial Ischemia/physiopathology , Myocardium/pathology , S100 Calcium-Binding Protein A4/metabolism , Ventricular Remodeling
17.
Proc Natl Acad Sci U S A ; 110(31): 12661-6, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23842089

ABSTRACT

Mechanistic target of rapamycin complex 1 (mTORC1), necessary for cellular growth, is regulated by intracellular signaling mediating inhibition of mTORC1 activation. Among mTORC1 regulatory binding partners, the role of Proline Rich AKT Substrate of 40 kDa (PRAS40) in controlling mTORC1 activity and cellular growth in response to pathological and physiological stress in the heart has never been addressed. This report shows PRAS40 is regulated by AKT in cardiomyocytes and that AKT-driven phosphorylation relieves the inhibitory function of PRAS40. PRAS40 overexpression in vitro blocks mTORC1 in cardiomyocytes and decreases pathological growth. Cardiomyocyte-specific overexpression in vivo blunts pathological remodeling after pressure overload and preserves cardiac function. Inhibition of mTORC1 by PRAS40 preferentially promotes protective mTORC2 signaling in chronic diseased myocardium. In contrast, strong PRAS40 phosphorylation by AKT allows for physiological hypertrophy both in vitro and in vivo, whereas cardiomyocyte-specific overexpression of a PRAS40 mutant lacking capacity for AKT-phosphorylation inhibits physiological growth in vivo, demonstrating that AKT-mediated PRAS40 phosphorylation is necessary for induction of physiological hypertrophy. Therefore, PRAS40 phosphorylation acts as a molecular switch allowing mTORC1 activation during physiological growth, opening up unique possibilities for therapeutic regulation of the mTORC1 complex to mitigate pathologic myocardial hypertrophy by PRAS40.


Subject(s)
Cardiomegaly/metabolism , Multiprotein Complexes/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Phosphoproteins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/therapy , Male , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Multiprotein Complexes/genetics , Muscle Proteins/genetics , Mutation , Myocytes, Cardiac/pathology , Phosphoproteins/genetics , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics
18.
Circ Res ; 112(9): 1244-52, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23487407

ABSTRACT

RATIONALE: Cardiac hypertrophy results from the complex interplay of differentially regulated cascades based on the phosphorylation status of involved signaling molecules. Although numerous critical regulatory kinases and phosphatases have been identified in the myocardium, the intracellular mechanism for temporal regulation of signaling duration and intensity remains obscure. In the nonmyocyte context, control of folding, activity, and stability of proteins is mediated by the prolyl isomerase Pin1, but the role of Pin1 in the heart is unknown. OBJECTIVE: To establish the role of Pin1 in the heart. METHODS AND RESULTS: Here, we show that either genetic deletion or cardiac overexpression of Pin1 blunts hypertrophic responses induced by transaortic constriction and consequent cardiac failure in vivo. Mechanistically, we find that Pin1 directly binds to Akt, mitogen activated protein kinase (MEK), and Raf-1 in cultured cardiomyocytes after hypertrophic stimulation. Furthermore, loss of Pin1 leads to diminished hypertrophic signaling of Akt and MEK, whereas overexpression of Pin1 increases Raf-1 phosphorylation on the autoinhibitory site Ser259, leading to reduced MEK activation. CONCLUSIONS: Collectively, these data support a role for Pin1 as a central modulator of the intensity and duration of 2 major hypertrophic signaling pathways, thereby providing a novel target for regulation and control of cardiac hypertrophy.


Subject(s)
Cardiomegaly/enzymology , Myocytes, Cardiac/enzymology , Peptidylprolyl Isomerase/metabolism , Signal Transduction , Animals , Cardiomegaly/diagnostic imaging , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Dependovirus/genetics , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/metabolism , Myocytes, Cardiac/pathology , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/deficiency , Peptidylprolyl Isomerase/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Rats , Time Factors , Transduction, Genetic , Transfection , Ultrasonography , raf Kinases/metabolism
19.
J Mol Cell Cardiol ; 71: 11-5, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24140798

ABSTRACT

The hypertrophic growth of cardiac myocytes is a highly dynamic process that underlies physiological and pathological adaptation of the heart. Accordingly, a better understanding of the molecular underpinnings of cardiac myocyte hypertrophy is required in order to fully appreciate the causes and functional consequences of the changes in the size of the healthy and diseased heart. Hypertrophy is driven by increases in cardiac myocyte protein, which must be balanced by cellular ability to maintain protein quality in order to avoid maladaptive accumulation of toxic misfolded proteins. Recent studies have shown that the endoplasmic reticulum (ER), which, in cardiac myocytes, comprises the sarco/endoplasmic reticulum (SR/ER), is the site of most protein synthesis. Thus, the protein quality control machinery located at the SR/ER is likely to be an important determinant of whether the heart responds adaptively to hypertrophic growth stimuli. The SR/ER-transmembrane protein, ATF6, serves a critical protein quality control function as a first responder to the accumulation of potentially toxic, misfolded proteins. Misfolded proteins transform ATF6 into a transcription factor that regulates a gene program that is partly responsible for enhancing protein quality control. Two ATF6-inducible genes that have been studied in the heart and shown to be adaptive are RCAN1 and Derl3, which encode proteins that decrease protein-folding demand, and enhance degradation of misfolded proteins, respectively. Thus, the ATF6-regulated SR/ER protein quality control system is important for maintaining protein quality during growth, making ATF6, and other components of the system, potentially attractive targets for the therapeutic management pathological cardiac hypertrophy. This article is part of a Special Issue entitled "Protein Quality Control, the Ubiquitin Proteasome System, and Autophagy".


Subject(s)
Activating Transcription Factor 6/metabolism , Endoplasmic Reticulum/metabolism , Heart/physiology , Muscle Proteins/biosynthesis , Myocytes, Cardiac/metabolism , Animals , Humans , Protein Biosynthesis , Protein Folding , Transcription Factors/metabolism , Ubiquitin/metabolism
20.
Circulation ; 128(19): 2132-44, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24008870

ABSTRACT

BACKGROUND: The mechanistic target of rapamycin (mTOR) comprises 2 structurally distinct multiprotein complexes, mTOR complexes 1 and 2 (mTORC1 and mTORC2). Deregulation of mTOR signaling occurs during and contributes to the severity of myocardial damage from ischemic heart disease. However, the relative roles of mTORC1 versus mTORC2 in the pathogenesis of ischemic damage are unknown. METHODS AND RESULTS: Combined pharmacological and molecular approaches were used to alter the balance of mTORC1 and mTORC2 signaling in cultured cardiac myocytes and in mouse hearts subjected to conditions that mimic ischemic heart disease. The importance of mTOR signaling in cardiac protection was demonstrated by pharmacological inhibition of both mTORC1 and mTORC2 with Torin1, which led to increased cardiomyocyte apoptosis and tissue damage after myocardial infarction. Predominant mTORC1 signaling mediated by suppression of mTORC2 with Rictor similarly increased cardiomyocyte apoptosis and tissue damage after myocardial infarction. In comparison, preferentially shifting toward mTORC2 signaling by inhibition of mTORC1 with PRAS40 led to decreased cardiomyocyte apoptosis and tissue damage after myocardial infarction. CONCLUSIONS: These results suggest that selectively increasing mTORC2 while concurrently inhibiting mTORC1 signaling is a novel therapeutic approach for the treatment of ischemic heart disease.


Subject(s)
Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/metabolism , Myocardial Infarction/metabolism , Myocardial Ischemia/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis/physiology , Carrier Proteins/metabolism , Humans , Male , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/genetics , Myocardial Infarction/pathology , Myocardial Ischemia/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Naphthyridines/pharmacology , Primary Cell Culture , Rapamycin-Insensitive Companion of mTOR Protein , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL