54 results on '"Donna J. Thuerauf"'
Search Results
2. Noncanonical Form of ERAD Regulates Cardiac Hypertrophy
- Author
-
Erik A. Blackwood, Lauren F. MacDonnell, Donna J. Thuerauf, Alina S. Bilal, Victoria B. Murray, Kenneth C. Bedi, Kenneth B. Margulies, and Christopher C. Glembotski
- Subjects
Physiology (medical) ,Cardiology and Cardiovascular Medicine - 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.
- Published
- 2023
- Full Text
- View/download PDF
3. Optimization of Large‐Scale Adeno‐Associated Virus (AAV) Production
- Author
-
Alina S. Bilal, Sarah N. Parker, Victoria B. Murray, Lauren F. MacDonnell, Donna J. Thuerauf, Christopher C. Glembotski, and Erik A. Blackwood
- Subjects
Medical Laboratory Technology ,General Immunology and Microbiology ,General Neuroscience ,Health Informatics ,General Pharmacology, Toxicology and Pharmaceutics ,General Biochemistry, Genetics and Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
4. Design and Production of Heart Chamber-Specific AAV9 Vectors
- Author
-
Alina S, Bilal, Donna J, Thuerauf, Erik A, Blackwood, and Christopher C, Glembotski
- Subjects
Mice ,Genetic Vectors ,Gene Transfer Techniques ,Animals ,Heart Atria ,Dependovirus ,Serogroup - Abstract
Adeno-associated virus serotype 9 (AAV9) is often used in heart research involving gene delivery due to its cardiotropism, high transduction efficiency, and little to no pathogenicity, making it highly applicable for gene manipulation, in vivo. However, current AAV9 technology is limited by the lack of strains that can selectively express and elucidate gene function in an atrial- and ventricular-specific manner. In fact, study of gene function in cardiac atria has been limited due to the lack of an appropriate tool to study atrial gene expression in vivo, hindering progress in the study of atrial-specific diseases such as atrial fibrillation, the most common cardiac arrhythmia in the USA.This chapter describes the method for the design and production of such chamber-specific AAV9 vectors, with the use of Nppa and Myl2 promoters to enhance atrial- and ventricular-specific expression. While several gene promoter candidates were considered and tested, Nppa and Myl2 were selected for use here because of their clearly defined regulatory elements that confer cardiac chamber-specific expression. Accordingly, Nppa (-425/+25) and Myl2 (-226/+36) promoter fragments are inserted into AAV9 vectors. The atrial- and ventricular-specific expression conferred by these new recombinant AAV9 was confirmed in a double-fluorescent Cre-dependent reporter mouse model. At only 450 and 262 base pairs of Nppa and Myl2 promoters, respectively, these AAV9 that drive chamber-specific AAV9 transgene expression address two major limitations of AAV9 technology, i.e., achieving chamber-specificity while maximizing space in the AAV genome for insertion of larger transgenes.
- Published
- 2022
5. The peroxisomal enzyme, FAR1, is induced during ER stress in an ATF6-dependent manner in cardiac myocytes
- Author
-
Adrian Arrieta, Erik A Blackwood, Christopher C Glembotski, Lauren MacDonnell, Kayleigh G Marsh, and Donna J. Thuerauf
- Subjects
Cell Survival ,Physiology ,Myocardial Reperfusion Injury ,Oxidative phosphorylation ,medicine.disease_cause ,chemistry.chemical_compound ,Physiology (medical) ,Peroxisomes ,medicine ,Animals ,Myocyte ,Myocytes, Cardiac ,Cells, Cultured ,Rapid Report ,ATF6 ,Tunicamycin ,Cardiac myocyte ,Hydrogen Peroxide ,Peroxisome ,Endoplasmic Reticulum Stress ,Aldehyde Oxidoreductases ,Cell Hypoxia ,Activating Transcription Factor 6 ,Rats ,Cell biology ,Oxidative Stress ,Animals, Newborn ,chemistry ,Enzyme Induction ,Unfolded protein response ,Cardiology and Cardiovascular Medicine ,Oxidative stress - 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 (H(2)O(2)). 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.
- Published
- 2021
- Full Text
- View/download PDF
6. PRAS40 prevents development of diabetic cardiomyopathy and improves hepatic insulin sensitivity in obesity
- Author
-
Mirko Völkers, Shirin Doroudgar, Nathalie Nguyen, Mathias H Konstandin, Pearl Quijada, Shabana Din, Luis Ornelas, Donna J Thuerauf, Natalie Gude, Kilian Friedrich, Stephan Herzig, Christopher C Glembotski, and Mark A Sussman
- Subjects
diabetes ,PRAS40 ,mTOR ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Diabetes is a multi‐organ disease and diabetic cardiomyopathy can result in heart failure, which is a leading cause of morbidity and mortality in diabetic patients. In the liver, insulin resistance contributes to hyperglycaemia and hyperlipidaemia, which further worsens the metabolic profile. Defects in mTOR signalling are believed to contribute to metabolic dysfunctions in diabetic liver and hearts, but evidence is missing that mTOR activation is causal to the development of diabetic cardiomyopathy. This study shows that specific mTORC1 inhibition by PRAS40 prevents the development of diabetic cardiomyopathy. This phenotype was associated with improved metabolic function, blunted hypertrophic growth and preserved cardiac function. In addition PRAS40 treatment improves hepatic insulin sensitivity and reduces systemic hyperglycaemia in obese mice. Thus, unlike rapamycin, mTORC1 inhibition with PRAS40 improves metabolic profile in diabetic mice. These findings may open novel avenues for therapeutic strategies using PRAS40 directed against diabetic‐related diseases.
- Published
- 2013
- Full Text
- View/download PDF
7. Proteomic analysis of the cardiac myocyte secretome reveals extracellular protective functions for the ER stress response
- Author
-
Erik A Blackwood, Shirin Doroudgar, Khalid Azizi, Jennifer E. Van Eyk, Miroslava Stastna, Christopher C. Glembotski, Zoe Sand, Donna J. Thuerauf, Amber N Pentoney, Haley N Stephens, Hugo A. Katus, and Tobias Jakobi
- Subjects
Proteomics ,0301 basic medicine ,Glycosylation ,Thapsigargin ,Proteome ,Cell Survival ,Apoptosis ,Heart failure ,Cardioprotection ,030204 cardiovascular system & hematology ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Paracrine Communication ,Animals ,Myocytes, Cardiac ,Secretion ,Calcium Signaling ,Endoplasmic Reticulum Chaperone BiP ,Molecular Biology ,Protein kinase B ,Cells, Cultured ,Membrane Glycoproteins ,Epidermal Growth Factor ,Cardiac myocyte death ,Chemistry ,Endoplasmic reticulum ,Cardiokine ,Tunicamycin ,Endoplasmic Reticulum Stress ,Neoplasm Proteins ,Rats ,Cell biology ,Autocrine Communication ,Sarcoplasmic Reticulum ,030104 developmental biology ,Secretory protein ,Proteostasis ,Unfolded protein response ,Calcium ,Disease Susceptibility ,ER stress ,Cardiology and Cardiovascular Medicine ,Biomarkers ,Signal Transduction - 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.
- Published
- 2020
- Full Text
- View/download PDF
8. Design and Production of Heart Chamber-Specific AAV9 Vectors
- Author
-
Alina S. Bilal, Donna J. Thuerauf, Erik A. Blackwood, and Christopher C. Glembotski
- Published
- 2022
- Full Text
- View/download PDF
9. Pharmacologic ATF6 activation confers global protection in widespread disease models by reprograming cellular proteostasis
- Author
-
Khalid Azizi, Rockland Wiseman, Ryan J Paxman, Erik A Blackwood, Jeffery W. Kelly, Donna J. Thuerauf, Christopher C. Glembotski, and Lars Plate
- Subjects
Male ,0301 basic medicine ,Myocardial Infarction ,General Physics and Astronomy ,02 engineering and technology ,Endoplasmic Reticulum ,Kidney ,Mice ,Medicine ,Myocytes, Cardiac ,Myocardial infarction ,lcsh:Science ,Cells, Cultured ,Mice, Knockout ,Multidisciplinary ,Widespread Disease ,Cerebral Infarction ,021001 nanoscience & nanotechnology ,Cell biology ,3. Good health ,Treatment Outcome ,medicine.anatomical_structure ,Reperfusion Injury ,Female ,Kidney Diseases ,Signal transduction ,Cardiology and Cardiovascular Medicine ,0210 nano-technology ,Reprogramming ,Heart Ventricles ,Science ,Primary Cell Culture ,Ischemia ,Biology ,Protective Agents ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Animals ,Humans ,Molecular Biology ,ATF6 ,business.industry ,General Chemistry ,medicine.disease ,Activating Transcription Factor 6 ,Rats ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Proteostasis ,Animals, Newborn ,Unfolded Protein Response ,Unfolded protein response ,lcsh:Q ,business ,Neuroscience ,Reperfusion injury - Abstract
Pharmacologic activation of stress-responsive signaling pathways provides a promising approach for ameliorating imbalances in proteostasis associated with diverse diseases. However, this approach has not been employed in vivo. Here we show, using a mouse model of myocardial ischemia/reperfusion, that selective pharmacologic activation of the ATF6 arm of the unfolded protein response (UPR) during reperfusion, a typical clinical intervention point after myocardial infarction, transcriptionally reprograms proteostasis, ameliorates damage and preserves heart function. These effects were lost upon cardiac myocyte-specific Atf6 deletion in the heart, demonstrating the critical role played by ATF6 in mediating pharmacologically activated proteostasis-based protection of the heart. Pharmacological activation of ATF6 is also protective in renal and cerebral ischemia/reperfusion models, demonstrating its widespread utility. Thus, pharmacologic activation of ATF6 represents a proteostasis-based therapeutic strategy for ameliorating ischemia/reperfusion damage, underscoring its unique translational potential for treating a wide range of pathologies caused by imbalanced proteostasis., Imbalanced proteostasis is associated with diverse diseases, including ischemia/reperfusion injury in the heart. Here the authors show that the ATF6 arm of the unfolded protein response can be pharmacologically activated with a small molecule in vivo, providing protection from ischemia/reperfusion injury in the heart, the brain, and the kidney.
- Published
- 2019
10. Optimizing Adeno-Associated Virus Serotype 9 for Studies of Cardiac Chamber-Specific Gene Regulation
- Author
-
Erik A Blackwood, Donna J. Thuerauf, Christopher C Glembotski, and Alina S Bilal
- Subjects
Serotype ,Regulation of gene expression ,business.industry ,Heart ,medicine.disease_cause ,Virology ,Article ,Adenoviridae ,Tissue specificity ,Mice ,Physiology (medical) ,Cardiac chamber ,medicine ,Myocyte ,Animals ,Humans ,Gene Regulatory Networks ,Serotyping ,Cardiology and Cardiovascular Medicine ,business ,Adeno-associated virus ,Heart atrium - Published
- 2021
11. Mesencephalic astrocyte–derived neurotrophic factor is an ER‐resident chaperone that protects against reductive stress in the heart
- Author
-
Donna J. Thuerauf, Cathrine Aivati, Alina S. Bilal, Adrian Arrieta, Christopher C. Glembotski, Michelle Santo Domingo, Shirin Doroudgar, Amber N Pentoney, Winston T Stauffer, Erik A Blackwood, and Anup Sarakki
- Subjects
Programmed cell death ,Gene knockdown ,biology ,Chemistry ,ATF6 ,Endoplasmic reticulum ,Biochemistry ,Cell biology ,Neurotrophic factors ,Chaperone (protein) ,Genetics ,biology.protein ,Unfolded protein response ,Molecular Biology ,Transcription factor ,Biotechnology - 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.
- Published
- 2021
- Full Text
- View/download PDF
12. Mesencephalic astrocyte-derived neurotrophic factor is an ER-resident chaperone that protects against reductive stress in the heart
- Author
-
Anup Sarakki, Cathrine Aivati, Erik A Blackwood, Shirin Doroudgar, Christopher C. Glembotski, Winston T Stauffer, Donna J. Thuerauf, Michelle Santo Domingo, Amber N Pentoney, Adrian Arrieta, and Alina S. Bilal
- Subjects
0301 basic medicine ,Programmed cell death ,Glycosylation ,Cell Survival ,Myocardial Reperfusion Injury ,Endoplasmic Reticulum ,Biochemistry ,03 medical and health sciences ,Mice ,Neurotrophic factors ,Animals ,Humans ,Myocytes, Cardiac ,Editors' Picks ,Nerve Growth Factors ,Molecular Biology ,Transcription factor ,Mice, Knockout ,Gene knockdown ,030102 biochemistry & molecular biology ,biology ,Chemistry ,ATF6 ,Endoplasmic reticulum ,Myocardium ,Cell Biology ,Endoplasmic Reticulum Stress ,Cell biology ,030104 developmental biology ,Chaperone (protein) ,Unfolded protein response ,biology.protein ,Reactive Oxygen Species ,HeLa Cells ,Molecular Chaperones - 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 Ca(2+) 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.
- Published
- 2020
13. ATF6 Decreases Myocardial Ischemia/Reperfusion Damage and Links ER Stress and Oxidative Stress Signaling Pathways in the Heart
- Author
-
Christopher C. Glembotski, Donna J. Thuerauf, Erik A Blackwood, Asal G Fahem, Randal J. Kaufman, Christoph Hofmann, Shirin Doroudgar, Khalid Azizi, and Jung-Kang Jin
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Physiology ,Activating transcription factor ,Myocardial Reperfusion Injury ,Biology ,medicine.disease_cause ,Article ,Antioxidants ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,chemistry.chemical_classification ,Reactive oxygen species ,ATF6 ,Endoplasmic reticulum ,Activating Transcription Factor 6 ,Cell biology ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,chemistry ,Catalase ,Knockout mouse ,Unfolded Protein Response ,biology.protein ,Unfolded protein response ,Cardiology and Cardiovascular Medicine ,Oxidative stress ,Signal Transduction - 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.
- Published
- 2017
- Full Text
- View/download PDF
14. Junctophilin-2 gene therapy rescues heart failure by normalizing RyR2-mediated Ca2+ release
- Author
-
David L. Beavers, Ann P. Quick, Giselle Barreto-Torres, Qiongling Wang, Xander H.T. Wehrens, Christopher C. Glembotski, Jordan Showell, Leonne E Philippen, Shirin Doroudgar, Donna J. Thuerauf, and Julia O. Reynolds
- Subjects
0301 basic medicine ,Cardiac function curve ,medicine.medical_specialty ,Ejection fraction ,Ryanodine receptor ,business.industry ,030204 cardiovascular system & hematology ,medicine.disease ,Ryanodine receptor 2 ,T-tubule ,Contractility ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,medicine.anatomical_structure ,JPH2 ,Heart failure ,Internal medicine ,medicine ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background Junctophilin-2 (JPH2) is the primary structural protein for the coupling of transverse (T)-tubule associated cardiac L-type Ca channels and type-2 ryanodine receptors on the sarcoplasmic reticulum within junctional membrane complexes (JMCs) in cardiomyocytes. Effective signaling between these channels ensures adequate Ca-induced Ca release required for normal cardiac contractility. Disruption of JMC subcellular domains, a common feature of failing hearts, has been attributed to JPH2 downregulation. Here, we tested the hypothesis that adeno-associated virus type 9 (AAV9) mediated overexpression of JPH2 could halt the development of heart failure in a mouse model of transverse aortic constriction (TAC). Methods and results Following TAC, a progressive decrease in ejection fraction was paralleled by a progressive decrease of cardiac JPH2 levels. AAV9-mediated expression of JPH2 rescued cardiac contractility in mice subjected to TAC. AAV9-JPH2 also preserved T-tubule structure. Moreover, the Ca 2+ spark frequency was reduced and the Ca 2+ transient amplitude was increased in AAV9-JPH2 mice following TAC, consistent with JPH2-mediated normalization of SR Ca 2+ handling. Conclusions This study demonstrates that AAV9-mediated JPH2 gene therapy maintained cardiac function in mice with early stage heart failure. Moreover, restoration of JPH2 levels prevented loss of T-tubules and suppressed abnormal SR Ca 2+ leak associated with contractile failure following TAC. These findings suggest that targeting JPH2 might be an attractive therapeutic approach for treating pathological cardiac remodeling during heart failure.
- Published
- 2016
- Full Text
- View/download PDF
15. Small Nppa and Myl2 Promoters Are Sufficient to Maintain Chamber‐specific Expression on an AAV9 Platform
- Author
-
Erik A Blackwood, Alina S. Bilal, Donna J. Thuerauf, and Christopher C. Glembotski
- Subjects
MYL2 ,Expression (architecture) ,Genetics ,Promoter ,Biology ,Molecular Biology ,Biochemistry ,Biotechnology ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
16. ATF6β is an Adaptive Transcription Factor in Cardiac Myocyte
- Author
-
Donna J. Thuerauf, Christopher C. Glembotski, Erik A Blackwood, Tak Ki Dicky Cheung, and Alina S. Bilal
- Subjects
Cardiac myocyte ,Genetics ,Biology ,Molecular Biology ,Biochemistry ,Transcription factor ,Biotechnology ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
17. ATF6 Regulates Cardiac Hypertrophy by Transcriptional Induction of the mTORC1 Activator, Rheb
- Author
-
Winston T Stauffer, Christoph Dieterich, Shirin Doroudgar, Donna J. Thuerauf, Adrian Arrieta, Christopher C. Glembotski, Hugo A. Katus, Michelle Santo Domingo, Fred W. Kolkhorst, Anup Sarakki, Erik A Blackwood, Alina S. Bilal, Christoph Hofmann, Tobias Jakobi, and Oliver J. Müller
- Subjects
Male ,Transcriptional Activation ,Protein Folding ,Physiology ,Activating transcription factor ,Mechanistic Target of Rapamycin Complex 1 ,Endoplasmic Reticulum ,Ventricular Function, Left ,Animals ,Genetic Predisposition to Disease ,Myocytes, Cardiac ,Transcription factor ,Mice, Knockout ,biology ,Ventricular Remodeling ,ATF6 ,Activator (genetics) ,Chemistry ,Endoplasmic reticulum ,Endoplasmic Reticulum Stress ,Cell biology ,Activating Transcription Factor 6 ,Mice, Inbred C57BL ,Disease Models, Animal ,Proteostasis ,Phenotype ,Animals, Newborn ,Unfolded protein response ,biology.protein ,Hypertrophy, Left Ventricular ,Ras Homolog Enriched in Brain Protein ,Cardiology and Cardiovascular Medicine ,RHEB ,Signal Transduction - 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.
- Published
- 2018
18. Abstract 352: Manf, a Structurally Unique Redox-sensitive Chaperone, Restores Er-protein Folding in the Ischemic Heart
- Author
-
Adrian Arrieta, Erik A Blackwood, Cathrine Aivati, Winston T Stauffer, Michelle Santo Domingo, Alina S Bilal, Anup V Sarakki, Donna J Thuerauf, Shirin Doroudgar, and Christopher C Glembotski
- Subjects
biology ,Physiology ,Chemistry ,Endoplasmic reticulum ,Genetic enhancement ,Sarcoplasm ,Redox sensitive ,Cell biology ,Membrane protein ,Chaperone (protein) ,biology.protein ,Protein folding ,Cardiology and Cardiovascular Medicine ,Ischemic heart - Abstract
Rationale: In cardiomyocytes, secreted and membrane proteins critical for heart function are synthesized and folded in the sarcoplasmic/endoplasmic reticulum (SR/ER). We previously showed that myocardial ischemia decreases oxygen required for disulfide bond formation in nascent proteins, causing ER stress, i.e. the toxic accumulation of unfolded proteins, which contributes to cardiomyocyte death. In response to ER stress, the transcription factor ATF6 induces various ER-resident proteins that restore SR/ER protein folding, including ER chaperones. We found that ATF6 induces mesencephalic astrocyte-derived neurotrophic factor (MANF), a recently identified protein of unknown function. MANF is structurally unique, so its function could not be inferred by analogy to other proteins. Since we found that MANF is an ATF6-inducible ER-resident protein we hypothesized that it functions as a chaperone, and since MANF has 8 cysteine residues that are conserved in a wide range of species, that its chaperone function is redox-regulated and protective in the ischemic heart. Methods: The ability of recombinant MANF (rMANF) to suppress misfolded protein aggregation was examined in an in vitro chaperone assay. The effect of MANF knockdown on cell viability during simulated ischemia (sI) was determined in neonatal rat ventricular myocytes (NRVM). The effect of MANF loss-of-function in the ischemic heart, in vivo , was determined in a novel mouse model in which MANF is knocked down in cardiomyocytes. Results: rMANF formed disulfide-dependent complexes with and suppressed aggregation of model misfolded proteins in vitro , and these effects were lost when the cysteines in rMANF were mutated to alanine. In NRVM, MANF knockdown decreased viability during simulated ischemia; this viability deficit was restored upon ectopic expression of wild type, but not mutant MANF. MANF knockdown in the heart, in vivo , increased ischemia/reperfusion damage, and this damage was mitigated using an AAV9-based gene therapy approach to restore MANF expression. Conclusions: MANF is a novel redox-sensitive SR/ER-resident chaperone that is a critical contributor to SR/ER protein folding during the adaptive ER stress response and mitigates ischemia/reperfusion damage in the heart.
- Published
- 2018
- Full Text
- View/download PDF
19. Abstract 547: Pharmacologic ATF6 Activation Confers Global Protection in Widespread Disease Models by Reprogramming Cellular Proteostasis
- Author
-
Erik A Blackwood, Khalid Azizi, Lars Plate, Jeffery W. Kelly, Donna J. Thuerauf, Ryan J Paxman, Rockland Wiseman, and Christopher C. Glembotski
- Subjects
Proteostasis ,Physiology ,ATF6 ,Widespread Disease ,Biology ,Cardiology and Cardiovascular Medicine ,Reprogramming ,Cell biology - Abstract
Pharmacologic activation of stress-responsive signaling pathways provides a promising approach for ameliorating imbalances in proteostasis associated with diverse diseases. However, this approach has not been employed in vivo . Here, using a mouse model of myocardial ischemia/reperfusion, we showed that selective pharmacologic activation of the ATF6 arm of the unfolded protein response (UPR) during reperfusion, a typical clinical intervention point after myocardial infarction, transcriptionally reprograms proteostasis, ameliorates damage and preserves heart function. These effects were lost upon cardiac myocyte-specific Atf6 deletion in the heart, demonstrating the critical role played by ATF6 in mediating pharmacologically activated proteostasis-based protection of the heart. Pharmacological activation of ATF6 was also protective in renal and cerebral ischemia/reperfusion models, demonstrating its widespread utility. Thus, pharmacologic activation of ATF6 represents a first-in-class proteostasis-based therapeutic strategy for ameliorating ischemia/reperfusion damage, underscoring its unique translational potential for treating a wide range of pathologies caused by imbalanced proteostasis.
- Published
- 2018
- Full Text
- View/download PDF
20. ATF6 ubiquitylation is required for its transcriptional activity and degradation
- Author
-
Cathrine Aivati, Donna J. Thuerauf, and Christopher C. Glembotski
- Subjects
Transcriptional activity ,Ubiquitin ,biology ,ATF6 ,Chemistry ,Genetics ,biology.protein ,Degradation (geology) ,Molecular Biology ,Biochemistry ,Biotechnology ,Cell biology - Published
- 2018
- Full Text
- View/download PDF
21. Hrd1 and ER-Associated Protein Degradation, ERAD, Are Critical Elements of the Adaptive ER Stress Response in Cardiac Myocytes
- Author
-
Shirin Doroudgar, Mark A. Sussman, Oliver J. Müller, Mohsin Khan, Mirko Völkers, Christopher C. Glembotski, Wei Wang, Sadia Mohsin, Xander H.T. Wehrens, Jonathan L. Respress, Donna J. Thuerauf, and Natalie Gude
- Subjects
medicine.medical_specialty ,biology ,medicine.diagnostic_test ,Physiology ,Ubiquitin-Protein Ligases ,Proteolysis ,Endoplasmic reticulum ,Endoplasmic Reticulum-Associated Degradation ,Protein degradation ,Endoplasmic-reticulum-associated protein degradation ,Reductase ,Endoplasmic Reticulum Stress ,Adaptation, Physiological ,Article ,Ubiquitin ligase ,Cell biology ,Endocrinology ,Internal medicine ,biology.protein ,medicine ,Animals ,Myocyte ,Myocytes, Cardiac ,Protein folding ,Cardiology and Cardiovascular Medicine - 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.
- Published
- 2015
- Full Text
- View/download PDF
22. Abstract 21206: Manf, a Structurally Unique Redox-Sensitive Chaperone, Restores ER-Protein Folding in the Ischemic Heart
- Author
-
Adrian Arrieta, Erik A Blackwood, Winston T Stauffer, Michelle Santo Domingo, Donna J Thuerauf, Shirin Doroudgar, and Christopher C Glembotski
- Subjects
Physiology (medical) ,Cardiology and Cardiovascular Medicine - Abstract
Rationale: In cardiomyocytes, most secreted and membrane proteins are synthesized and folded in the sarcoplasmic/endoplasmic reticulum (SR/ER). We previously showed that during myocardial ischemia, decreased oxygen creates a reducing environment in the SR/ER, preventing protein disulfide isomerases (PDIs) from forming disulfide bonds in nascent proteins, causing ER stress, i.e. the toxic accumulation of unfolded proteins which contributes to cardiomyocyte death. In response to ER stress, the transcription factor, ATF6 induces chaperones that restore SR/ER protein folding. We found that ATF6 also induces mesencephalic astrocyte-derived neurotrophic factor (MANF), a recently identified protein of unknown function. MANF is structurally unique, so its function cannot be inferred from other proteins. Since MANF is induced by ATF6, is ER-localized, and possesses a conserved pattern of cysteines found in all known species of MANF, we hypothesized that MANF is a redox-regulated chaperone that optimizes cardiomyocyte viability during ischemia. Methods: The ability of MANF to bind misfolded proteins during reductive ER stress or ischemia were assessed in neonatal rat ventricular myocytes (NRVM). The ability of recombinant MANF (rMANF) to suppress aggregation of misfolded proteins was examined in an in vitro chaperone assay. Finally, the effects of MANF loss-of-function in the ischemic heart, in vivo , were determined by generating a transgenic mouse model that expresses a cardiomyocyte-specific MANF-targeted microRNA. Results: In NRVM subjected to reductive ER stress or simulated ischemia, MANF formed disulfide-linked complexes with misfolded proteins. Under reducing conditions, rMANF suppressed aggregation of model misfolded proteins in vitro , and mutant rMANF in which the cysteine residues were mutated to alanine did not suppress misfolded protein aggregation. MANF knockdown in the heart, in vivo , increased damage from myocardial infarction, and an AAV9-based gene therapy approach rescued the effects of MANF deficiency, in vivo . Conclusions: MANF is a redox-sensitive SR/ER-resident chaperone that is a critical contributor to SR/ER protein folding during the adaptive ER stress response and decreases tissue damage in the ischemic heart.
- Published
- 2017
- Full Text
- View/download PDF
23. Mechanistic Target of Rapamycin Complex 2 Protects the Heart From Ischemic Damage
- Author
-
Christopher C. Glembotski, Donna J. Thuerauf, Shabana Din, Kaitleen Samse, Shirin Doroudgar, Mark A. Sussman, Natalie Gude, Anya Y. Joyo, Pearl Quijada, Mathias H. Konstandin, Haruhiro Toko, Luis Ornelas, and Mirko Völkers
- Subjects
Male ,Primary Cell Culture ,Myocardial Infarction ,Myocardial Ischemia ,Apoptosis ,Mechanistic Target of Rapamycin Complex 2 ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,mTORC2 ,Article ,Mice ,Physiology (medical) ,medicine ,Animals ,Humans ,Myocytes, Cardiac ,Myocardial infarction ,Naphthyridines ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Mice, Knockout ,biology ,business.industry ,TOR Serine-Threonine Kinases ,RPTOR ,medicine.disease ,Recombinant Proteins ,Cell biology ,Mice, Inbred C57BL ,Rapamycin-Insensitive Companion of mTOR Protein ,Multiprotein Complexes ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Carrier Proteins ,Cardiology and Cardiovascular Medicine ,business ,Signal Transduction - 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.
- Published
- 2013
- Full Text
- View/download PDF
24. PRAS40 prevents development of diabetic cardiomyopathy and improves hepatic insulin sensitivity in obesity
- Author
-
Shirin Doroudgar, Natalie Gude, Pearl Quijada, Kilian Friedrich, Mark A. Sussman, Mirko Völkers, Stephan Herzig, Shabana Din, Christopher C. Glembotski, Mathias H. Konstandin, Luis Ornelas, Nathalie Nguyen, and Donna J. Thuerauf
- Subjects
Male ,Cardiac function curve ,medicine.medical_specialty ,Diabetic Cardiomyopathies ,PRAS40 ,medicine.medical_treatment ,Genetic Vectors ,Mice, Obese ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Diet, High-Fat ,Adenoviridae ,Diabetes Mellitus, Experimental ,Mice ,Insulin resistance ,Internal medicine ,Diabetic cardiomyopathy ,Diabetes mellitus ,medicine ,Animals ,Insulin ,Myocytes, Cardiac ,Obesity ,Cells, Cultured ,Research Articles ,PI3K/AKT/mTOR pathway ,diabetes ,TOR Serine-Threonine Kinases ,Phosphoproteins ,medicine.disease ,3. Good health ,Mice, Inbred C57BL ,Phenotype ,Endocrinology ,Multiprotein Complexes ,Heart failure ,Metabolome ,mTOR ,Molecular Medicine - Abstract
Diabetes is a multi-organ disease and diabetic cardiomyopathy can result in heart failure, which is a leading cause of morbidity and mortality in diabetic patients. In the liver, insulin resistance contributes to hyperglycaemia and hyperlipidaemia, which further worsens the metabolic profile. Defects in mTOR signalling are believed to contribute to metabolic dysfunctions in diabetic liver and hearts, but evidence is missing that mTOR activation is causal to the development of diabetic cardiomyopathy. This study shows that specific mTORC1 inhibition by PRAS40 prevents the development of diabetic cardiomyopathy. This phenotype was associated with improved metabolic function, blunted hypertrophic growth and preserved cardiac function. In addition PRAS40 treatment improves hepatic insulin sensitivity and reduces systemic hyperglycaemia in obese mice. Thus, unlike rapamycin, mTORC1 inhibition with PRAS40 improves metabolic profile in diabetic mice. These findings may open novel avenues for therapeutic strategies using PRAS40 directed against diabetic-related diseases.
- Published
- 2013
- Full Text
- View/download PDF
25. Pathological hypertrophy amelioration by PRAS40-mediated inhibition of mTORC1
- Author
-
Mark A. Sussman, Anya Y. Joyo, Donna J. Thuerauf, Mathias H. Konstandin, Shirin Doroudgar, Haruhiro Toko, Christopher C. Glembotski, Natalie Gude, Mirko Völkers, Luis Ornelas, Shabana Din, Eri Joyo, and Pearl Quijada
- Subjects
Male ,Proto-Oncogene Proteins c-akt ,Muscle Proteins ,Cardiomegaly ,Mechanistic Target of Rapamycin Complex 2 ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,mTORC2 ,Muscle hypertrophy ,Mice ,Animals ,Myocytes, Cardiac ,Phosphorylation ,Protein kinase B ,Pressure overload ,Multidisciplinary ,TOR Serine-Threonine Kinases ,Biological Sciences ,Phosphoproteins ,Molecular biology ,Cell biology ,Multiprotein Complexes ,Mutation ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Signal Transduction - 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.
- Published
- 2013
- Full Text
- View/download PDF
26. Junctophilin-2 gene therapy rescues heart failure by normalizing RyR2-mediated Ca
- Author
-
Julia O, Reynolds, Ann P, Quick, Qiongling, Wang, David L, Beavers, Leonne E, Philippen, Jordan, Showell, Giselle, Barreto-Torres, Donna J, Thuerauf, Shirin, Doroudgar, Christopher C, Glembotski, and Xander H T, Wehrens
- Subjects
Heart Failure ,Male ,Genetic Vectors ,Membrane Proteins ,Muscle Proteins ,Ryanodine Receptor Calcium Release Channel ,Genetic Therapy ,Article ,Adenoviridae ,Mice, Inbred C57BL ,Mice ,Animals ,Myocytes, Cardiac ,Calcium Signaling ,Cells, Cultured - Abstract
Junctophilin-2 (JPH2) is the primary structural protein for the coupling of transverse (T)-tubule associated cardiac L-type Ca channels and type-2 ryanodine receptors on the sarcoplasmic reticulum within junctional membrane complexes (JMCs) in cardiomyocytes. Effective signaling between these channels ensures adequate Ca-induced Ca release required for normal cardiac contractility. Disruption of JMC subcellular domains, a common feature of failing hearts, has been attributed to JPH2 downregulation. Here, we tested the hypothesis that adeno-associated virus type 9 (AAV9) mediated overexpression of JPH2 could halt the development of heart failure in a mouse model of transverse aortic constriction (TAC).Following TAC, a progressive decrease in ejection fraction was paralleled by a progressive decrease of cardiac JPH2 levels. AAV9-mediated expression of JPH2 rescued cardiac contractility in mice subjected to TAC. AAV9-JPH2 also preserved T-tubule structure. Moreover, the CaThis study demonstrates that AAV9-mediated JPH2 gene therapy maintained cardiac function in mice with early stage heart failure. Moreover, restoration of JPH2 levels prevented loss of T-tubules and suppressed abnormal SR Ca
- Published
- 2016
27. S100A4 protects the myocardium against ischemic stress
- Author
-
Kelli Ilves, Luis Ornelas, Mirko Völkers, Mark A. Sussman, Christopher C. Glembotski, Farid G. Khalafalla, Donna J. Thuerauf, Alexandria Casillas, Kathleen M. Broughton, Natalie Gude, Mathias H. Konstandin, Pearl Quijada, Haruhiro Toko, Shirin Doroudgar, and Kristine Nguyen
- Subjects
0301 basic medicine ,Cardiac function curve ,medicine.medical_specialty ,Myocardial Infarction ,Myocardial Ischemia ,Gene Expression ,030204 cardiovascular system & hematology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Fibrosis ,In vivo ,Stress, Physiological ,Internal medicine ,medicine ,Myocyte ,Animals ,S100 Calcium-Binding Protein A4 ,cardiovascular diseases ,Myocardial infarction ,Ventricular remodeling ,Molecular Biology ,Mice, Knockout ,Cell Death ,Ventricular Remodeling ,business.industry ,Myocardium ,Hemodynamics ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,Apoptosis ,Echocardiography ,Knockout mouse ,cardiovascular system ,Cardiology ,Cardiology and Cardiovascular Medicine ,business - 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.
- Published
- 2016
28. Regulation of microRNA expression in the heart by the ATF6 branch of the ER stress response
- Author
-
Christopher C. Glembotski, Peter J. Belmont, Wenqiong J. Chen, and Donna J. Thuerauf
- Subjects
Male ,Transcription, Genetic ,Primary Cell Culture ,Mice, Transgenic ,Article ,Rats, Sprague-Dawley ,Mice ,Downregulation and upregulation ,RNA interference ,microRNA ,Gene expression ,Animals ,Endoplasmic Reticulum Chaperone BiP ,Molecular Biology ,Cells, Cultured ,Heat-Shock Proteins ,Oligonucleotide Array Sequence Analysis ,Membrane Glycoproteins ,biology ,Microarray analysis techniques ,ATF6 ,Gene Expression Profiling ,Myocardium ,Endoplasmic Reticulum Stress ,Molecular biology ,Activating Transcription Factor 6 ,Rats ,Up-Regulation ,Cell biology ,Mice, Inbred C57BL ,MicroRNAs ,Unfolded protein response ,biology.protein ,RNA Interference ,Calreticulin ,Cardiology and Cardiovascular Medicine - Abstract
A nodal regulator of endoplasmic reticulum stress is the transcription factor, ATF6, which is activated by ischemia and protects the heart from ischemic damage, in vivo. To explore mechanisms of ATF6-mediated protection in the heart, a whole-genome microRNA (miRNA) array analysis of RNA from the hearts of ATF6 transgenic (TG) mice was performed. The array identified 13 ATF6-regulated miRNAs, eight of which were downregulated, suggesting that they could contribute to increasing levels of their mRNAs. The down-regulated miRNAs, including miR-455, were predicted to target 45 mRNAs that we had previously shown by microarray analysis to be up-regulated by ATF6 in the heart. One of the miR-455 targets was calreticulin (Calr), which is up-regulated in the pathologic heart, where it modulates hypertrophic growth, potentially reducing the impact of the pathology. To validate the effects of miR-455, we showed that Calr protein was increased by ATF6 in mouse hearts, in vivo. In cultured cardiac myocytes, treatment with the ER stressor, tunicamycin, or with adenovirus encoding activated ATF6 decreased miR-455 and increased Calr levels, consistent with the effects of ATF6 on miR-455 and Calr, in vivo. Moreover, transfection of cultured cardiac myocytes with a synthetic precursor, premiR-455, decreased Calr levels, while transfection with an antisense, antimiR-455, increased Calr levels. The results of this study suggest that ER stress can regulate gene expression via ATF6-mediated changes in micro-RNA levels. Moreover, these findings support the hypothesis that ATF6-mediated down-regulation of miR-455 augments Calr expression, which may contribute to the protective effects of ATF6 in the heart.
- Published
- 2012
- Full Text
- View/download PDF
29. Roles for Endoplasmic Reticulum–Associated Degradation and the Novel Endoplasmic Reticulum Stress Response Gene Derlin-3 in the Ischemic Heart
- Author
-
Matthew N. San Pedro, Mark A. Sussman, Brett J. Hilton, Peter J. Belmont, Natalie Gude, Roland Wolkowicz, Donna J. Thuerauf, Nicole Gellings Lowe, Wenqiong J. Chen, and Christopher C. Glembotski
- Subjects
Programmed cell death ,Physiology ,ATF6 ,Endoplasmic reticulum ,Unfolded protein response ,Activating transcription factor ,Myocyte ,Context (language use) ,Endoplasmic-reticulum-associated protein degradation ,Biology ,Cardiology and Cardiovascular Medicine ,Molecular biology ,Cell biology - Abstract
Rationale : Stresses, such as ischemia, impair folding of nascent proteins in the rough endoplasmic reticulum (ER), activating the unfolded protein response, which restores efficient ER protein folding, thus leading to protection from stress. In part, the unfolded protein response alleviates ER stress and cell death by increasing the degradation of terminally misfolded ER proteins via ER-associated degradation (ERAD). ERAD is increased by the ER stress modulator, activating transcription factor (ATF)6, which can induce genes that encode components of the ERAD machinery. Objective : Recently, it was shown that the mouse heart is protected from ischemic damage by ATF6; however, ERAD has not been studied in the cardiac context. A recent microarray study showed that the Derlin-3 (Derl3) gene, which encodes an important component of the ERAD machinery, is robustly induced by ATF6 in the mouse heart. Methods and Results : In the present study, activated ATF6 induced Derl3 in cultured cardiomyocytes, and in the heart, in vivo. Simulated ischemia (sI), which activates ER stress, induced Derl3 in cultured myocytes, and in an in vivo mouse model of myocardial infarction, Derl3 was also induced. Derl3 overexpression enhanced ERAD and protected cardiomyocytes from simulated ischemia–induced cell death, whereas dominant-negative Derl3 decreased ERAD and increased simulated ischemia–induced cardiomyocyte death. Conclusions : This study describes a potentially protective role for Derl3 in the heart, and is the first to investigate the functional consequences of enhancing ERAD in the cardiac context.
- Published
- 2010
- Full Text
- View/download PDF
30. Ischemia Activates the ATF6 Branch of the Endoplasmic Reticulum Stress Response
- Author
-
Donna J. Thuerauf, Marie Marcinko, Peter J. Belmont, Shirin Doroudgar, and Christopher C. Glembotski
- Subjects
Cell Survival ,Active Transport, Cell Nucleus ,Ischemia ,Myocardial Reperfusion Injury ,Biology ,Endoplasmic Reticulum ,Biochemistry ,Stress, Physiological ,Heat shock protein ,medicine ,Animals ,Humans ,Myocyte ,Myocytes, Cardiac ,RNA, Messenger ,Endoplasmic Reticulum Chaperone BiP ,Molecular Biology ,Heat-Shock Proteins ,Cell Nucleus ,ATF6 ,Protein Synthesis, Post-Translational Modification, and Degradation ,Endoplasmic reticulum ,Cell Biology ,medicine.disease ,Molecular biology ,Cell Hypoxia ,Activating Transcription Factor 6 ,Rats ,Cell biology ,Cytosol ,Gene Expression Regulation ,Chaperone (protein) ,Unfolded protein response ,biology.protein - Abstract
Stresses that perturb the folding of nascent endoplasmic reticulum (ER) proteins activate the ER stress response. Upon ER stress, ER-associated ATF6 is cleaved; the resulting active cytosolic fragment of ATF6 translocates to the nucleus, binds to ER stress response elements (ERSEs), and induces genes, including the ER-targeted chaperone, GRP78. Recent studies showed that nutrient and oxygen starvation during tissue ischemia induce certain ER stress response genes, including GRP78; however, the role of ATF6 in mediating this induction has not been examined. In the current study, simulating ischemia (sI) in a primary cardiac myocyte model system caused a reduction in the level of ER-associated ATF6 with a coordinate increase of ATF6 in nuclear fractions. An ERSE in the GRP78 gene not previously shown to be required for induction by other ER stresses was found to bind ATF6 and to be critical for maximal ischemia-mediated GRP78 promoter induction. Activation of ATF6 and the GRP78 promoter, as well as grp78 mRNA accumulation during sI, were reversed upon simulated reperfusion (sI/R). Moreover, dominant-negative ATF6, or ATF6-targeted miRNA blocked sI-mediated grp78 induction, and the latter increased cardiac myocyte death upon simulated reperfusion, demonstrating critical roles for endogenous ATF6 in ischemia-mediated ER stress activation and cell survival. This is the first study to show that ATF6 is activated by ischemia but inactivated upon reperfusion, suggesting that it may play a role in the induction of ER stress response genes during ischemia that could have a preconditioning effect on cell survival during reperfusion.
- Published
- 2009
- Full Text
- View/download PDF
31. Coordination of Growth and Endoplasmic Reticulum Stress Signaling by Regulator of Calcineurin 1 (RCAN1), a Novel ATF6-inducible Gene
- Author
-
Wenqiong J. Chen, Donna J. Thuerauf, Joshua J. Martindale, Marie Marcinko, Natalie Gude, Archana Tadimalla, Peter J. Belmont, Mark A. Sussman, and Christopher C. Glembotski
- Subjects
Small interfering RNA ,Muscle Proteins ,Mice, Transgenic ,Biology ,Endoplasmic Reticulum ,Models, Biological ,Biochemistry ,Mice ,Animals ,RNA, Small Interfering ,Molecular Biology ,Transcription factor ,Cells, Cultured ,Oligonucleotide Array Sequence Analysis ,Models, Statistical ,ATF6 ,Myocardium ,Protein Synthesis, Post-Translational Modification, and Degradation ,Endoplasmic reticulum ,Calcium-Binding Proteins ,Intracellular Signaling Peptides and Proteins ,NFAT ,Cell Biology ,Activating Transcription Factor 6 ,Rats ,Cell biology ,Calcineurin ,Unfolded protein response ,RNA ,Signal transduction ,Signal Transduction - Abstract
Exposing cells to conditions that modulate growth can impair endoplasmic reticulum (ER) protein folding, leading to ER stress and activation of the transcription factor, ATF6. ATF6 binds to ER stress response elements in target genes, inducing expression of proteins that enhance the ER protein folding capacity, which helps overcome the stress and foster survival. To examine the mechanism of ATF6-mediated survival in vivo, we developed a transgenic mouse model that expresses a novel conditionally activated form of ATF6. We previously showed that activating ATF6 protected the hearts of ATF6 transgenic mice from ER stresses. In the present study, transcript profiling identified modulatory calcineurin interacting protein-1 (MCIP1), also known as regulator of calcineurin 1 (RCAN1), as a novel ATF6-inducible gene that encodes a known regulator of calcineurin/nuclear factor of activated T cells (NFAT)-mediated growth and development in many tissues. The ability of ATF6 to induce RCAN1 in vivo was replicated in cultured cardiac myocytes, where adenoviral (AdV)-mediated overexpression of activated ATF6 induced the RCAN1 promoter, up-regulated RCAN1 mRNA, inhibited calcineurin phosphatase activity, and exerted a striking growth modulating effect that was inhibited by RCAN1-targeted small interfering RNA. These results demonstrate that RCAN1 is a novel ATF6 target gene that may coordinate growth and ER stress signaling pathways. By modulating growth, RCAN1 may reduce the need for ER protein folding, thus helping to overcome the stress and enhance survival. Moreover, these results suggest that RCAN1 may also be a novel integrator of growth and ER stress signaling in many other tissues that depend on calcineurin/NFAT signaling for optimal growth and development.
- Published
- 2008
- Full Text
- View/download PDF
32. Abstract 402: Selective Protein Secretion Upon Endoplasmic Reticulum Stress Protects Cardiac Myocytes
- Author
-
Donna J. Thuerauf, Mirko Voelkers, Shirin Doroudgar, Christopher C. Glembotski, Mirka Stastna, and Jennifer E. Van Eyk
- Subjects
Secretory protein ,Biochemistry ,Physiology ,Chemistry ,Endoplasmic reticulum ,Cardiac myocyte ,Myocyte ,Cardiology and Cardiovascular Medicine ,Reticulum ,Function (biology) - Abstract
Protein secretion is important for proper cardiac myocyte function. Many secreted proteins are synthesized and folded in the sarco- endo-plasmic reticulum (SR/ER). A number of diseases, including heart disease, alter the ER in ways that impair ER protein folding, causing ER stress, which can result in cardiac myocyte dysfunction and decreased viability. In studies aimed at assessing the effects of ER stress on cardiac myocyte viability, heart disease-related ER stress was mimicked by treating neonatal rat ventricular myocytes (NRVM) with either tunicamycin (TM) or thapsigargin (TG), which inhibit SR/ER protein glycosylation or decrease SR/ER calcium, respectively. When treated in high culture media volumes, both TM and TG caused cardiac myocyte death; however, in low culture media volumes, while TM still caused death, remarkably, TG was protective, suggesting that potentially protective factors were secreted in response to TG but not TM. To characterize these factors, the identities of proteins in control-, TM-, and TG-conditioned medium from NRVM were determined by proteomic approaches using high performance liquid chromatography and mass spectrometry. Twenty-four different proteins, known to be synthesized in the ER, were identified in control-conditioned medium. The levels of eighteen of these proteins, including extracellular matrix proteins, hormones, and growth factors were decreased in TM- and TG-conditioned medium. However, the levels of three SR/ER-resident, calcium-binding chaperones, glucose regulated protein 78 (GRP78), glucose regulated protein 94 (GRP94), and calreticulin were increased in TG-conditioned medium but not in TM-conditioned medium. Furthermore, we found that ischemia/reperfusion, which decreases SR/ER calcium, upregulated secretion of the proteins selectively secreted in response to TG. Thus, while ER stress mediated by TM or TG decreases the movement of most proteins through the secretory pathway, TG, which mimics the effects of heart disease on SR/ER calcium in cardiac myocytes, selectively enhances the secretion of a subset of proteins, which confer protection. Therefore, proteins once thought to be permanent residents of the SR/ER may have novel, extracellular, protective roles in the diseased heart.
- Published
- 2015
- Full Text
- View/download PDF
33. Activation of the Unfolded Protein Response in Infarcted Mouse Heart and Hypoxic Cultured Cardiac Myocytes
- Author
-
Natalie Gude, Marta Rubio, Christopher C. Glembotski, Mark A. Sussman, Donna J. Thuerauf, and Marie Marcinko
- Subjects
X-Box Binding Protein 1 ,Protein Folding ,medicine.medical_specialty ,XBP1 ,Physiology ,RNA Splicing ,Myocardial Infarction ,Regulatory Factor X Transcription Factors ,Biology ,Mice ,Internal medicine ,medicine ,Animals ,Myocyte ,Myocytes, Cardiac ,Hypoxia ,Endoplasmic Reticulum Chaperone BiP ,Transcription factor ,Cells, Cultured ,Heat-Shock Proteins ,Messenger RNA ,Endoplasmic reticulum ,Hypoxia (medical) ,Neoplasm Proteins ,Rats ,Cell biology ,DNA-Binding Proteins ,Basic-Leucine Zipper Transcription Factors ,Endocrinology ,Animals, Newborn ,Gene Expression Regulation ,Apoptosis ,Unfolded protein response ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Molecular Chaperones ,Transcription Factors - Abstract
Endoplasmic reticulum (ER) stresses that reduce ER protein folding activate the unfolded protein response (UPR). One effector of the UPR is the transcription factor X-box binding protein-1 (XBP1), which is expressed on ER stress-mediated splicing of the XBP1 mRNA. XBP1 induces certain ER-targeted proteins, eg, glucose-regulated protein 78 (GRP78), that help resolve the ER stress and foster cell survival. In this study, we determined whether hypoxia can activate the UPR in the cardiac context. Neonatal rat ventricular myocyte cultures subjected to hypoxia (16 hours) exhibited increased XBP1 mRNA splicing, XBP1 protein expression, GRP78 promoter activation, and GRP78 protein levels; however, the levels of these UPR markers declined during reoxygenation, suggesting that the UPR is activated during hypoxia but not during reoxygenation. When cells were infected with a recombinant adenovirus (AdV) encoding dominant-negative XBP1 (AdV-XBP1dn), UPR markers were reduced; however, hypoxia/reoxygenation-induced apoptosis increased. Confocal immunocytofluorescence demonstrated that hypoxia induced GRP78 in neonatal rat and isolated adult mouse ventricular myocytes. Moreover, mouse hearts subjected to in vivo myocardial infarction exhibited increased GRP78 expression in cardiac myocytes near the infarct, but not in healthy cells distal to the infarct. These results indicate that hypoxia activates the UPR in cardiac myocytes and that XBP1-inducible proteins may contribute to protecting the myocardium during hypoxic stress.
- Published
- 2006
- Full Text
- View/download PDF
34. Opposing Roles for ATF6α and ATF6β in Endoplasmic Reticulum Stress Response Gene Induction
- Author
-
Donna J. Thuerauf, Christopher C. Glembotski, and Lisa E. Morrison
- Subjects
Transcriptional Activation ,Gene isoform ,Cytomegalovirus ,Biology ,Endoplasmic Reticulum ,Biochemistry ,chemistry.chemical_compound ,Genes, Reporter ,Humans ,Protein Isoforms ,RNA, Small Interfering ,Endoplasmic Reticulum Chaperone BiP ,Molecular Biology ,Leucine Zippers ,Activator (genetics) ,ATF6 ,Endoplasmic reticulum ,Cell Biology ,DNA-binding domain ,Tunicamycin ,beta-Galactosidase ,Molecular biology ,Transmembrane protein ,Activating Transcription Factor 6 ,DNA-Binding Proteins ,Kinetics ,Basic-Leucine Zipper Transcription Factors ,Gene Expression Regulation ,chemistry ,Unfolded protein response ,HeLa Cells ,Transcription Factors - Abstract
The endoplasmic reticulum (ER) transmembrane proteins, ATF6alpha and ATF6beta, are cleaved in response to ER stress, which can be induced by tunicamycin. The resulting N-terminal fragments of both ATF6 isoforms, which have conserved basic leucine-zipper and DNA binding domains but divergent transcriptional activation domains, translocate to the nucleus where they bind to ER stress-response elements (ERSE) in ER stress-response genes (ERSRG), such as GRP78. Although it is known that ATF6alpha is a potent activator of ERSRGs, the transcriptional potency and functions of ATF6beta remain to be explored. Accordingly, N-terminal fragments of each ATF6 isoform (N-ATF6alpha and N-ATF6beta) were overexpressed in HeLa cells and the effects on GRP78 induction were assessed. When expressed at similar levels, N-ATF6alpha conferred approximately 200-fold greater GRP78 promoter activation than N-ATF6beta. Because ER stress activates nuclear translocation of both ATF6alpha and beta and because both bind to ERSEs, the effect of co-expressing them on GRP78 induction was assessed. Surprisingly, N-ATF6beta inhibited N-ATF6alpha-mediated GRP78 promoter activation in a dominant-negative manner. Moreover, N-ATF6beta inhibited TN-mediated GRP78 promoter activation, which requires endogenous ATF6alpha. ATF6 isoform-specific small inhibitory RNAs were used to show that, as expected, endogenous ATF6alpha was required for maximal ERSRG induction; however, endogenous ATF6beta moderated ERSRG induction. These results indicate that compared with ATF6alpha, ATF6beta is a very poor activator of ERSRG induction and it represses ATF6alpha-mediated ERSRG induction. Thus, ATF6beta may serve as a transcriptional repressor functioning in part to regulate the strength and duration of ATF6alpha-mediated ERSRG activation during the ER stress response.
- Published
- 2004
- Full Text
- View/download PDF
35. Mimicking Phosphorylation of αB-Crystallin on Serine-59 Is Necessary and Sufficient to Provide Maximal Protection of Cardiac Myocytes From Apoptosis
- Author
-
Donna J. Thuerauf, Holly Hoover, Christopher C. Glembotski, and Lisa E. Morrison
- Subjects
Programmed cell death ,Cell Survival ,Physiology ,Molecular Sequence Data ,Apoptosis ,Caspase 3 ,Biology ,Transfection ,Rats, Sprague-Dawley ,Structure-Activity Relationship ,Heat shock protein ,In Situ Nick-End Labeling ,Animals ,Sorbitol ,Myocyte ,Myocytes, Cardiac ,Phosphorylation ,Cells, Cultured ,Base Sequence ,Molecular Mimicry ,Osmolar Concentration ,alpha-Crystallin B Chain ,Cell Hypoxia ,Rats ,Cell biology ,Amino Acid Substitution ,Biochemistry ,Cell culture ,Caspases ,Mutagenesis, Site-Directed ,Cardiology and Cardiovascular Medicine - Abstract
AlphaB-crystallin (alphaBC), a small heat shock protein expressed in high levels in the heart, is phosphorylated on Ser-19, 45, and 59 after stress. However, it is not known whether alphaBC phosphorylation directly affects cell survival. In the present study, constructs were prepared that encode forms of alphaBC harboring Ser to Ala (blocks phosphorylation) or Ser to Glu (mimics phosphorylation) mutations at positions 19, 45, and 59. The effects of each form on apoptosis of cultured cardiac myocytes after hyperosmotic or hypoxic stress were assessed. Compared with controls, cells that expressed alphaBC with Ser to Ala substitutions at all three positions, alphaBC(AAA), exhibited more stress-induced apoptosis. Cells expressing either alphaBC(AAE) or (EEE) exhibited 3-fold less apoptosis than cells expressing alphaBC(AAA), indicating that phosphorylation of Ser-59 confers protection. alphaBC is known to bind to procaspase-3 and to decrease caspase-3 activation. Compared with cells expressing alphaBC(AAA), the activation of caspase-3 was decreased by 3-fold in cells expressing alphaBC(AAE). These results demonstrate that mimicking the phosphorylation of alphaBC on Ser-59 is necessary and sufficient to confer caspase-3 inhibition and protection of cardiac myocytes against hyperosmotic or hypoxic stress. These findings provide direct evidence that alphaBC(S59P) contributes to the cardioprotection observed after physiologically relevant stresses, such as transient hypoxia. Identifying the targets of alphaBC(S59P) will reveal important details about the mechanism underlying the cytoprotective effects of this small heat shock protein.
- Published
- 2003
- Full Text
- View/download PDF
36. MANF, a structurally unique redox-sensitive chaperone, restores ER-protein folding in the ischemic heart
- Author
-
Amber N Pentoney, Shirin Doroudgar, Michelle Santo Domingo, Christopher C. Glembotski, Donna J. Thuerauf, Adrian Arrieta, Erik A Blackwood, and Winston T Stauffer
- Subjects
0301 basic medicine ,biology ,Chemistry ,Redox sensitive ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Chaperone (protein) ,biology.protein ,Protein folding ,Cardiology and Cardiovascular Medicine ,Ischemic heart ,Molecular Biology ,030217 neurology & neurosurgery - Published
- 2017
- Full Text
- View/download PDF
37. Sarco/endoplasmic Reticulum Calcium ATPase-2 Expression Is Regulated by ATF6 during the Endoplasmic Reticulum Stress Response
- Author
-
Catherine Andrews, Leo Y. Su, Holly Hoover, Christopher C. Glembotski, Patrick M. McDonough, Jessica Hernandez, Wolfgang H. Dillmann, Donna J. Thuerauf, and Julia Meller
- Subjects
medicine.medical_specialty ,ATF6 ,Endoplasmic reticulum ,T-type calcium channel ,chemistry.chemical_element ,Cell Biology ,Calcium ,Biology ,Biochemistry ,Cell biology ,Calcium ATPase ,Endocrinology ,chemistry ,Cytoplasm ,Internal medicine ,medicine ,Myocyte ,Molecular Biology ,Calcium signaling - Abstract
The recently described transcription factor, ATF6, mediates the expression of proteins that compensate for potentially stressful changes in the endoplasmic reticulum (ER), such as reduced ER calcium. In cardiac myocytes the maintenance of optimal calcium levels in the sarcoplasmic reticulum (SR), a specialized form of the ER, is required for proper contractility. The present study investigated the hypothesis that ATF6 serves as a regulator of the expression of sarco/endoplasmic reticulum calcium ATPase-2 (SERCA2), a protein that transports calcium into the SR from the cytoplasm. Depletion of SR calcium in cultured cardiac myocytes fostered the translocation of ATF6 from the ER to the nucleus, activated the promoter for rat SERCA2, and led to increased levels of SERCA2 protein. SERCA2 promoter induction by calcium depletion was partially blocked by dominant-negative ATF6, whereas constitutively activated ATF6 led to SERCA2 promoter activation. Mutation analyses identified a promoter-proximal ER stress-response element in the rat SERCA2 gene that was required for maximal induction by ATF6 and calcium depletion. Although this element was shown to be responsible for all of the effects of ATF6 on SERCA2 promoter activation, it was responsible for only a portion of the effects of calcium depletion. Thus, SERCA2 induction in response to calcium depletion appears to be a potentially physiologically important compensatory response to this stress that involves intracellular signaling pathways that are both dependent and independent of ATF6.
- Published
- 2001
- Full Text
- View/download PDF
38. αB-crystallin Gene Induction and Phosphorylation by MKK6-activated p38
- Author
-
Holly Hoover, Christopher C. Glembotski, Donna J. Thuerauf, and Joshua J. Martindale
- Subjects
Regulation of gene expression ,p38 mitogen-activated protein kinases ,Cell Biology ,Biology ,Biochemistry ,Cytoprotection ,Molecular biology ,Heat shock protein ,Serum response factor ,Phosphorylation ,sense organs ,Signal transduction ,Protein kinase A ,Molecular Biology - Abstract
The MAPK kinase MKK6 selectively stimulates p38 MAPK and confers protection against stress-induced apoptosis in cardiac myocytes. However, the events lying downstream of p38 that mediate this protection are unknown. The small heat shock protein, alphaB-crystallin, which is expressed in only a few cell types, including cardiac myocytes, may participate in MKK6-mediated cytoprotection. In the present study, we showed that, in cultured cardiac myocytes, expression of MKK6(Glu), an active form of MKK6, led to p38-dependent increases in alphaB-crystallin mRNA, protein, and transcription. MKK6(Glu) also induced p38-dependent activation of the downstream MAPK-activated protein kinase, MAPKAP-K2, and the phosphorylation of alphaB-crystallin on serine-59. Initially, exposure of cells to the hyperosmotic stressor, sorbitol, stimulated MKK6, p38, and MAPKAP-K2 and increased phosphorylation of alphaB-crystallin on serine 59. However, after longer times of exposure to sorbitol, the cells began to undergo apoptosis. This sorbitol-induced apoptosis was increased when p38 was inhibited in a manner that would block alphaB-crystallin induction and phosphorylation. Thus, under these conditions, the activation of MKK6, p38, and MAPKAP-K2 by sorbitol can provide a degree of protection against stress-induced apoptosis. Supporting this view was the finding that sorbitol-induced apoptosis was nearly completely blocked in cells expressing MKK6(Glu). Therefore, the cytoprotective effects of MKK6 in cardiac myocytes are due, in part, to phosphorylation of alphaB-crystallin on serine 59 and to the induction of alphaB-crystallin gene expression.
- Published
- 2000
- Full Text
- View/download PDF
39. p38 Mitogen-activated Protein Kinase Mediates the Transcriptional Induction of the Atrial Natriuretic Factor Gene through a Serum Response Element
- Author
-
Kelli M. DeMartin, Dietmar Zechner, Patrick M. McDonough, Christopher C. Glembotski, Deanna S. Hanford, Ron Prywes, Nichole D. Arnold, and Donna J. Thuerauf
- Subjects
MAPK/ERK pathway ,ATF6 ,Activator (genetics) ,Promoter ,Cell Biology ,Biology ,Serum Response Element ,Biochemistry ,Molecular biology ,Serum response factor ,cardiovascular system ,Protein kinase A ,Molecular Biology ,Transcription factor - Abstract
In various cell types certain stresses can stimulate p38 mitogen-activated protein kinase (p38 MAPK), leading to the transcriptional activation of genes that contribute to appropriate compensatory responses. In this report the mechanism of p38-activated transcription was studied in cardiac myocytes where this MAPK is a key regulator of the cell growth and the cardiac-specific gene induction that occurs in response to potentially stressful stimuli. In the cardiac atrial natriuretic factor (ANF) gene, a promoter-proximal serum response element (SRE), which binds serum response factor (SRF), was shown to be critical for ANF induction in primary cardiac myocytes transfected with the selective p38 MAPK activator, MKK6 (Glu). This ANF SRE does not possess sequences typically required for the binding of the Ets-related ternary complex factors (TCFs), such as Elk-1, indicating that p38-mediated induction through this element may take place independently of such TCFs. Although p38 did not phosphorylate SRF in vitro, it efficiently phosphorylated ATF6, a newly discovered SRF-binding protein that is believed to serve as a co-activator of SRF-inducible transcription at SREs. Expression of an ATF6 antisense RNA blocked p38-mediated ANF induction through the ANF SRE. Moreover, when fused to the Gal4 DNA-binding domain, an N-terminal 273-amino acid fragment of ATF6 was sufficient to support trans-activation of Gal4/luciferase expression in response to p38 but not the other stress kinase, N-terminal Jun kinase (JNK); p38-activating cardiac growth promoters also stimulated ATF6 trans-activation. These results indicate that through ATF6, p38 can augment SRE-mediated transcription independently of Ets-related TCFs, representing a novel mechanism of SRF-dependent transcription by MAP kinases.
- Published
- 1998
- Full Text
- View/download PDF
40. A Role for the p38 Mitogen-activated Protein Kinase Pathway in Myocardial Cell Growth, Sarcomeric Organization, and Cardiac-specific Gene Expression
- Author
-
Dietmar Zechner, Christopher C. Glembotski, Deanna S. Hanford, Patrick M. McDonough, and Donna J. Thuerauf
- Subjects
Sarcomeres ,MAPK/ERK pathway ,Pyridines ,SB 203580 ,medicine.medical_treatment ,p38 mitogen-activated protein kinases ,Cardiomegaly ,MAP Kinase Kinase 6 ,Biology ,p38 Mitogen-Activated Protein Kinases ,Article ,Phenylephrine ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Animals ,Enzyme Inhibitors ,Cells, Cultured ,Cell Size ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Kinase ,Myocardium ,Growth factor ,Imidazoles ,Cell Biology ,Transfection ,Molecular biology ,Rats ,Cell biology ,Gene Expression Regulation ,chemistry ,Mitogen-activated protein kinase ,Calcium-Calmodulin-Dependent Protein Kinases ,biology.protein ,Mitogen-Activated Protein Kinases ,Cell Division ,030217 neurology & neurosurgery - Abstract
Three hallmark features of the cardiac hypertrophic growth program are increases in cell size, sarcomeric organization, and the induction of certain cardiac-specific genes. All three features of hypertrophy are induced in cultured myocardial cells by alpha1- adrenergic receptor agonists, such as phenylephrine (PE) and other growth factors that activate mitogen- activated protein kinases (MAPKs). In this study the MAPK family members extracellular signal-regulated kinase (ERK), c-jun NH2-terminal kinase (JNK), and p38 were activated by transfecting cultured cardiac myocytes with constructs encoding the appropriate kinases possessing gain-of-function mutations. Transfected cells were then analyzed for changes in cell size, sarcomeric organization, and induction of the genes for the A- and B-type natriuretic peptides (NPs), as well as the alpha-skeletal actin (alpha-SkA) gene. While activation of JNK and/or ERK with MEKK1COOH or Raf-1 BXB, respectively, augmented cell size and effected relatively modest increases in NP and alpha-SkA promoter activities, neither upstream kinase conferred sarcomeric organization. However, transfection with MKK6 (Glu), which specifically activated p38, augmented cell size, induced NP and alpha-Ska promoter activities by up to 130-fold, and elicited sarcomeric organization in a manner similar to PE. Moreover, all three growth features induced by MKK6 (Glu) or PE were blocked with the p38-specific inhibitor, SB 203580. These results demonstrate novel and potentially central roles for MKK6 and p38 in the regulation of myocardial cell hypertrophy.
- Published
- 1997
- Full Text
- View/download PDF
41. Differential Effects of Protein Kinase C, Ras, and Raf-1 Kinase on the Induction of the Cardiac B-type Natriuretic Peptide Gene through a Critical Promoter-proximal M-CAT Element
- Author
-
Christopher C. Glembotski and Donna J. Thuerauf
- Subjects
Heart Ventricles ,Molecular Sequence Data ,Oligonucleotides ,Protein Serine-Threonine Kinases ,Mitogen-activated protein kinase kinase ,Biochemistry ,MAP2K7 ,Proto-Oncogene Proteins ,Animals ,ASK1 ,c-Raf ,Promoter Regions, Genetic ,Molecular Biology ,Cells, Cultured ,Protein Kinase C ,Base Sequence ,biology ,MAP kinase kinase kinase ,Cyclin-dependent kinase 4 ,Cyclin-dependent kinase 2 ,Cell Biology ,Molecular biology ,Rats ,Proto-Oncogene Proteins c-raf ,Gene Expression Regulation ,ras Proteins ,cardiovascular system ,biology.protein ,Cyclin-dependent kinase 9 ,Atrial Natriuretic Factor ,hormones, hormone substitutes, and hormone antagonists - Abstract
The cardiac genes for the A- and B-type natriuretic peptides (ANP and BNP) are coordinately induced by growth promoters, such as alpha1-adrenergic receptor agonists (e.g. phenylephrine (PE)). Although inducible elements in the ANP gene have been identified, responsible elements in the BNP gene are unknown. In this study, reporter constructs transfected into neonatal rat ventricular myocytes showed that in the context of 2.5 kilobase pairs of native BNP 5'-flanking sequences, a 2-base pair mutation in a promoter-proximal M-CAT site (CATTCT) disrupted basal and PE-inducible transcription by more than 98%. Expression of constitutively active forms of Ras, Raf-1 kinase, and protein kinase C, all of which are activated by PE in cardiac myocytes, strongly stimulated BNP reporter expression. Isolated M-CAT elements conferred PE, protein kinase C, and Ras inducibility to a minimal BNP promoter, however, they did not confer Raf-1 inducibility. These results show that M-CAT elements can serve as targets for Ras-dependent, Raf-1-independent pathways, implying the involvement of c-Jun N-terminal kinase and/or p38 mitogen-activated protein kinases, but not extracellular signal-regulated protein kinase/mitogen-activated protein kinase. Moreover, the essential M-CAT element distinguishes the BNP gene from the ANP gene, which utilizes serum response elements and an Sp1-like sequence.
- Published
- 1997
- Full Text
- View/download PDF
42. Regulation of cardiac hypertrophic signaling by prolyl isomerase Pin1
- Author
-
Shirin Doroudgar, Kun Ping Lu, Natalie Gude, Lucia Ormachea, Donna J. Thuerauf, Anya Y. Joyo, Christopher C. Glembotski, Oliver J. Müller, Mark A. Sussman, Shabana Din, Haruhiro Toko, Chun-Hau Chen, Mirko Völkers, Mathias H. Konstandin, Brett Collins, Eri Joyo, and Takafumi Uchida
- Subjects
Male ,medicine.medical_specialty ,Cell signaling ,Time Factors ,Physiology ,Cardiomegaly ,Transfection ,Article ,Mice ,Transduction, Genetic ,Internal medicine ,medicine ,Prolyl isomerase ,Animals ,Humans ,Myocytes, Cardiac ,Extracellular Signal-Regulated MAP Kinases ,Protein kinase B ,Ultrasonography ,Peptidylprolyl isomerase ,Mice, Knockout ,Mitogen-Activated Protein Kinase Kinases ,biology ,Dependovirus ,Peptidylprolyl Isomerase ,Cell biology ,Rats ,Mice, Inbred C57BL ,NIMA-Interacting Peptidylprolyl Isomerase ,Disease Models, Animal ,Endocrinology ,HEK293 Cells ,Mitogen-activated protein kinase ,PIN1 ,biology.protein ,Phosphorylation ,RNA Interference ,raf Kinases ,Signal transduction ,Cardiology and Cardiovascular Medicine ,Proto-Oncogene Proteins c-akt ,Signal Transduction - 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.
- Published
- 2013
43. Abstract 18: Synoviolin, an E3 Ubiquitin Ligase, Modulates Cardiac Myocyte Size and Restores Heart Function in Hypertrophic Cardiomyopathy
- Author
-
Shirin Doroudgar, Mirko Völkers, Donna J Thuerauf, Ashley Bumbar, Mohsin Khan, Sadia Mohsin, Wei Wang, Jonathan L Respress, Natalie Gude, Oliver J Müller, Xander H Wehrens, Mark A Sussman, and Christopher C Glembotski
- Subjects
Physiology ,Cardiology and Cardiovascular Medicine - Abstract
The endoplasmic reticulum (ER) is essential for protein homeostasis, or proteostasis, which governs the balance of the proteome. In addition to secreted and membrane proteins, proteins bound for many other cellular locations are also made on ER-bound ribosomes, emphasizing the importance of protein quality and quantity control in the ER. Unlike cytosolic E3 ubiquitin ligases studied in the heart, synoviolin/Hrd1, which has not been studied in the heart, is an ER transmembrane E3 ubiquitin ligase, which we found to be upregulated upon protein misfolding in cardiac myocytes. Given the strategic location of synoviolin in the ER membrane, we addressed the hypothesis that synoviolin is critical for regulating the balance of the proteome, and accordingly, myocyte size. We showed that in vitro, adenovirus-mediated overexpression of synoviolin decreased cardiac myocyte size and protein synthesis, but unlike atrophy-related ubiquitin ligases, synoviolin did not increase global protein degradation. Furthermore, targeted gene therapy using adeno-associated virus 9 (AAV9) showed that overexpression of synoviolin in the left ventricle attenuated maladaptive cardiac hypertrophy and preserved cardiac function in mice subjected to trans-aortic constriction (AAV9-control TAC = 22.5 ± 6.2% decrease in EF vs. AAV9-synoviolin TAC at 6 weeks post TAC; P
- Published
- 2012
- Full Text
- View/download PDF
44. Protein disulfide isomerase-associated 6 is an ATF6-inducible ER stress response protein that protects cardiac myocytes from ischemia/reperfusion-mediated cell death
- Author
-
Donna J. Thuerauf, Christopher C. Glembotski, John A. Vekich, and Peter J. Belmont
- Subjects
Male ,Chromatin Immunoprecipitation ,Immunoblotting ,CAAT box ,Activating transcription factor ,Protein Disulfide-Isomerases ,Electrophoretic Mobility Shift Assay ,Mice, Transgenic ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,Mice ,Tandem Mass Spectrometry ,Animals ,Myocytes, Cardiac ,Protein disulfide-isomerase ,Molecular Biology ,Transcription factor ,Cell Death ,ATF6 ,Endoplasmic reticulum ,Endoplasmic Reticulum Stress ,Molecular biology ,Transmembrane protein ,Activating Transcription Factor 6 ,Mice, Inbred C57BL ,MicroRNAs ,Reperfusion Injury ,Unfolded protein response ,Cardiology and Cardiovascular Medicine ,Chromatography, Liquid - Abstract
Proper folding of secreted and transmembrane proteins made in the rough endoplasmic reticulum (ER) requires oxygen for disulfide bond formation. Accordingly, ischemia can impair ER protein folding and initiate the ER stress response, which we previously showed is activated in the ischemic heart and in culture cardiac myocytes subjected to simulated ischemia. ER stress and ischemia activate the transcription factor, activating transcription factor 6 (ATF6), which induces numerous genes, many of which have not been identified, or examined in the heart. Using an ATF6 transgenic mouse model, we previously showed that ATF6 protected the heart from ischemic damage; however, the mechanism of this protection remains to be determined. In this study, we showed that, in the mouse heart, and in cultured cardiac myocytes, ATF6 induced the protein disulfide isomerase associated 6 (PDIA6) gene, which encodes an ER enzyme that catalyzes protein disulfide bond formation. Moreover, in cultured cardiac myocytes, ER stress-mediated PDIA6 promoter activation was ATF6-dependent, and required an ER stress response element (ERSE) and a nearby CCAAT box element. Electromobility shift assays and chromatin immunoprecipitation showed that ATF6 bound to the ERSE in the PDIA6 promoter, in vitro, and in the mouse heart, in vivo. Gain- and loss-of-function studies showed that PDIA6 protected cardiac myocytes against simulated ischemia/reperfusion-induced death in a manner that was dependent on the catalytic activity of PDIA6. Thus, by facilitating disulfide bond formation, and enhanced ER protein folding, PDIA6 may contribute to the protective effects of ATF6 in the ischemic mouse heart.
- Published
- 2012
45. Brain natriuretic peptide is induced by alpha 1-adrenergic agonists as a primary response gene in cultured rat cardiac myocytes
- Author
-
Deanna S. Hanford, S F Murray, Donna J. Thuerauf, and Christopher C. Glembotski
- Subjects
Regulation of gene expression ,Agonist ,medicine.medical_specialty ,Messenger RNA ,medicine.drug_class ,Stimulation ,Cell Biology ,Biology ,Brain natriuretic peptide ,Biochemistry ,Endocrinology ,Transcription (biology) ,Internal medicine ,cardiovascular system ,medicine ,Protein biosynthesis ,Molecular Biology ,Phenylephrine ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
To better understand the molecular basis for increased atrial natriuretic factor (ANF) and brain natriuretic peptide (BNP) expression during overload-induced cardiac hypertrophy, we studied the induction of the genes in primary myocardial cells by the alpha 1-adrenergic agonist, phenylephrine (PE), a potent hypertrophic agent. PE augmented the transcription of both genes to similar extents, although the time course of mRNA accumulation differed. Increases in ANF mRNA were evident only after 6-8 h of PE exposure, when transcript levels were 2-4-fold over control. However, similar increases in BNP mRNA were observed as soon as 1 h of PE exposure. Moreover, while ANF mRNA levels continued to increase through 24 h of PE treatment, maximal levels of BNP mRNA (8-10-fold over control) were observed at 4 h, after which transcript levels declined to about 3-fold over control. The early induction of the BNP mRNA by PE was independent of protein synthesis, whereas the late induction of both genes required protein synthesis. Interestingly, the early BNP induction was only partially blocked by the transcription inhibitor, actinomycin D, indicating that, in part, the inductive effects of PE might be the result of transcript stabilization. Indeed, the BNP transcript, which was shown to possess a half-life of less than 1 h in control cells, was stabilized by the addition of PE, while the ANF transcript possessed a half-life of at least 24 h under all conditions. These data indicate that the induction of BNP by alpha 1-adrenergic agonists has characteristics of both a primary and secondary response gene, while ANF is a typical secondary response gene. Moreover, alpha 1-adrenergic stimulation enhances BNP expression through both transcriptional activation and transcript stabilization, while ANF expression is enhanced primarily transcriptionally.
- Published
- 1994
- Full Text
- View/download PDF
46. Regulation of rat brain natriuretic peptide transcription. A potential role for GATA-related transcription factors in myocardial cell gene expression
- Author
-
Christopher C. Glembotski, Donna J. Thuerauf, and Deanna S. Hanford
- Subjects
TATA box ,GATA2 ,Cell Biology ,Biology ,Biochemistry ,Molecular biology ,Fusion gene ,Transcription (biology) ,embryonic structures ,Gene expression ,GATA transcription factor ,Enhancer ,Molecular Biology ,Transcription factor - Abstract
GATA-binding proteins are transcription factors that regulate the stage- and tissue-specific expression of globin genes in cells of the erythroid lineage. Recently, a cardiac GATA-binding protein was found to be the earliest gene expressed during cardiogenesis; however, the target genes of this transcription factor in the heart are unknown. Since brain natriuretic peptide (BNP) is activated early in cardiac growth and development, we evaluated whether it could serve as a target gene for GATA-binding protein-mediated induction. Upon isolating and sequencing 2.5 kilobases of the rat BNP 5'-flanking sequence (FS), a variety of putative transcriptional enhancer sites were identified, including several GATA consensus sequences (WGATAR), one of which apparently serves as the major promoter site. Primary myocardial cells were transfected with BNP/luciferase fusion genes; reporter expression was strongly induced by typical growth factors such as phorbol esters, serum, or alpha 1-adrenergic agonists, as well as by GATA-4 overexpression. Truncation analyses showed that inducibility mapped primarily to the proximal -116 base pair of the rat BNP 5'-FS, where there are two consensus GATA sites in addition to the GATA sequence at the TATA box. Point mutation analyses showed that at least one of the GATA sites was required to confer full GATA-4-inducible transcription. These results demonstrate that a proximal region of the rat BNP 5'-FS is required for growth factor- and GATA-inducible transcription, supporting the view that the BNP gene could serve as a target for GATA-binding proteins during early cardiac development.
- Published
- 1994
- Full Text
- View/download PDF
47. Abstract P134: Synoviolin Is a Stress-Inducible Endoplasmic/Sarcoplasmic Reticulum E3 Ubiquitin Ligase that Preserves Cardiac Function
- Author
-
Shirin Doroudgar, Donna J Thuerauf, Mohsin Khan, Sadia Mohsin, Mirko Völkers, Natalie Gude, Oliver J Múller, Mark A Sussman, and Christopher C Glembotski
- Subjects
Physiology ,Cardiology and Cardiovascular Medicine - Abstract
We recently reported that synoviolin1 (syvn1) is a novel endoplasmic reticulum stress response (ERSR) protein that is up-regulated in the mouse heart by ATF6, a cardioprotective, nodal transcription factor of the ERSR. Syvn1 is a unique E3 ubiquitin ligase that retrotranslocates misfolded proteins from endoplasmic/sarcoplasmic reticulum (ER/SR) to the cytosol and subsequently polyubiquitinates them, targeting them for degradation. We now report that syvn1 expression is induced with tunicamycin, thapsigargin, and dithiothreitol, ER stressors that activate ATF6. Moreover, adenovirus-mediated syvn1 overexpression in neonatal rat ventricular cardiac myocytes (NRVCMs) increases contractility. Consistent with this finding, syvn1 overexpression increases calcium transient amplitude as well as diastolic calcium. We also find that syvn1 overexpression decreases secretion of MANF, a protective, anti-hypertrophic, ER/SR protein which we find is conditionally secreted when ER/SR calcium is depleted. We also report MANF as the first example of a is protein whose secretion from ventricular myocytes is conditionally dependent on ER/SR calcium. Furthermore, syvn1 reduces the growth of NRVCMs treated with the α-adrenergic agonist, phenylephrine. Moreover, while knockdown of syvn1 in NRVCMs using syvn1-targeted siRNA increases cell death, overexpression of syvn1 promotes cell survival. To investigate the roles of syvn1, in vivo , we examined the effects of syvn1 overexpression in cardiac myocytes in a mouse model of trans-aortic banding. Syvn1 overexpression, in vivo, was achieved by intravenous delivery of recombinant AAV serotype 9 (AAV9; cardiac specific serotype) with MLC2v promoter driving syvn1 expression. Baseline cardiac function, as measured by echocardiography six weeks after gene delivery, shows no difference between AAV9-control and AAV9-syvn1 treated mice. Trans-aortic banding decreases cardiac function in mice injected with AAV9-control. In contrast, cardiac function is preserved in mice injected with AAV9-syvn1. The findings in this study suggest that syvn1 is a novel stress-inducible cardiac E3 ligase with unique functions in regulating protein secretion, maintaining cell viability, and preserving cardiac function.
- Published
- 2011
- Full Text
- View/download PDF
48. Effects of the isoform-specific characteristics of ATF6 alpha and ATF6 beta on endoplasmic reticulum stress response gene expression and cell viability
- Author
-
Peter J. Belmont, Donna J. Thuerauf, Christopher C. Glembotski, and Marie Marcinko
- Subjects
Gene isoform ,Transcriptional Activation ,Cell Survival ,Mutant ,Cytomegalovirus ,Biology ,Endoplasmic Reticulum ,Biochemistry ,Gene expression ,Humans ,Protein Isoforms ,Viability assay ,RNA, Small Interfering ,Molecular Biology ,Endoplasmic Reticulum Chaperone BiP ,Heat-Shock Proteins ,Regulation of gene expression ,Activator (genetics) ,ATF6 ,Endoplasmic reticulum ,Cell Biology ,DNA ,beta-Galactosidase ,Molecular biology ,Cell biology ,Activating Transcription Factor 6 ,Protein Structure, Tertiary ,Basic-Leucine Zipper Transcription Factors ,Gene Expression Regulation ,HeLa Cells ,Molecular Chaperones ,Protein Binding - Abstract
The endoplasmic reticulum (ER)-transmembrane proteins, ATF6 alpha and ATF6 beta, are cleaved during the ER stress response (ERSR). The resulting N-terminal fragments (N-ATF6 alpha and N-ATF6 beta) have conserved DNA-binding domains and divergent transcriptional activation domains. N-ATF6 alpha and N-ATF6 beta translocate to the nucleus, bind to specific regulatory elements, and influence expression of ERSR genes, such as glucose-regulated protein 78 (GRP78), that contribute to resolving the ERSR, thus, enhancing cell viability. We previously showed that N-ATF6 alpha is a rapidly degraded, strong transcriptional activator, whereas beta is a slowly degraded, weak activator. In this study we explored the molecular basis and functional impact of these isoform-specific characteristics in HeLa cells. Mutants in the transcriptional activation domain or DNA-binding domain of N-ATF6 alpha exhibited loss of function and increased expression, the latter of which suggested decreased rates of degradation. Fusing N-ATF6 alpha to the mutant estrogen receptor generated N-ATF6 alpha-MER, which, without tamoxifen exhibited loss-of-function and high expression, but in the presence of tamoxifen N-ATF6 alpha-MER exhibited gain-of-function and low expression. N-ATF6 beta conferred loss-of-function and high expression to N-ATF6 alpha, suggesting that ATF6 beta is an endogenous inhibitor of ATF6 alpha. In vitro DNA binding experiments showed that recombinant N-ATF6 beta inhibited the binding of recombinant N-ATF6 alpha to an ERSR element from the GRP78 promoter. Moreover, siRNA-mediated knock-down of endogenous ATF6 beta increased GRP78 promoter activity and GRP78 gene expression, as well as augmenting cell viability. Thus, the relative levels of ATF6 alpha and -beta, may contribute to regulating the strength and duration of ATF6-dependent ERSR gene induction and cell viability.
- Published
- 2007
49. Endoplasmic reticulum stress gene induction and protection from ischemia/reperfusion injury in the hearts of transgenic mice with a tamoxifen-regulated form of ATF6
- Author
-
Mark A. Sussman, Ross Whittaker, Rayne Fernandez, Natalie Gude, Christopher C. Glembotski, Joshua J. Martindale, and Donna J. Thuerauf
- Subjects
Genetically modified mouse ,Male ,Transcriptional Activation ,medicine.medical_specialty ,Protein Folding ,Necrosis ,Cardiotonic Agents ,Physiology ,Transgene ,Mice, Transgenic ,Myocardial Reperfusion Injury ,Biology ,Endoplasmic Reticulum ,Mice ,Stress, Physiological ,Internal medicine ,medicine ,Animals ,RNA, Messenger ,Endoplasmic Reticulum Chaperone BiP ,Cells, Cultured ,Heat-Shock Proteins ,Membrane Glycoproteins ,ATF6 ,Endoplasmic reticulum ,Myocardium ,Recovery of Function ,medicine.disease ,Cell biology ,Activating Transcription Factor 6 ,Mice, Inbred C57BL ,Tamoxifen ,Endocrinology ,Gene Expression Regulation ,Echocardiography ,Unfolded protein response ,Female ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Reperfusion injury ,Ex vivo ,Molecular Chaperones - Abstract
Ischemia/reperfusion (I/R) affects the integrity of the endoplasmic reticulum (ER), the site of synthesis and folding of numerous proteins. Therefore, I/R may activate the unfolded protein response (UPR), resulting in the induction of a collection of ER stress proteins, many of which are protective and function to resolve the ER stress. In this study, we showed that when mouse hearts were subjected to ex vivo I/R, the levels of 2 ER stress-inducible markers of the UPR, the ER-targeted cytoprotective chaperones glucose-regulated proteins 78 and 94 (GRP78 and GRP94), were increased, consistent with I/R-mediated UPR activation in the heart. The UPR-mediated activation of ATF6 ( A ctivation of T ranscription F actor 6 ) induces cytoprotective ER stress proteins, including GRP78 and GRP94. To examine whether ATF6 protects the myocardium from I/R injury in the heart, we generated transgenic (TG) mice featuring cardiac-restricted expression of a novel tamoxifen-activated form of ATF6, ATF6-MER. When NTG and ATF6-MER TG mice were treated with or without tamoxifen for 5 days, only the hearts from the tamoxifen-treated TG mice exhibited increased levels of many ER stress–inducible mRNAs and proteins; for example, GRP78 and GRP94 transcript levels were increased by 8- and 15-fold, respectively. The tamoxifen-treated TG mouse hearts also exhibited better functional recovery from ex vivo I/R, as well as significantly reduced necrosis and apoptosis. These results suggest that the UPR is activated in the heart during I/R and that, as a result, the ATF6 branch of the UPR may induce expression of proteins that can function to reduce I/R injury.
- Published
- 2006
50. MAP kinase kinase 6-p38 MAP kinase signaling cascade regulates cyclooxygenase-2 expression in cardiac myocytes in vitro and in vivo
- Author
-
Donna J. Thuerauf, Norbert Degousee, Eva Stefanski, Joshua J. Martindale, Thomas F. Lindsay, Jason E. Fish, Philip A. Marsden, Martin Cieslak, Barry B. Rubin, and Christopher C. Glembotski
- Subjects
MAPK/ERK pathway ,Physiology ,MAP Kinase Signaling System ,Pyridines ,Heart Ventricles ,RNA polymerase II ,MAP Kinase Kinase 6 ,p38 Mitogen-Activated Protein Kinases ,Dinoprostone ,Gene Expression Regulation, Enzymologic ,Animals, Genetically Modified ,Rats, Sprague-Dawley ,Tubulin ,Gene expression ,Animals ,Myocytes, Cardiac ,RNA, Messenger ,Enzyme Inhibitors ,Phosphorylation ,Cells, Cultured ,Regulation of gene expression ,Messenger RNA ,biology ,Kinase ,Imidazoles ,Molecular biology ,Rats ,Isoenzymes ,Animals, Newborn ,Cyclooxygenase 2 ,Prostaglandin-Endoperoxide Synthases ,Mitogen-activated protein kinase ,Calcium-Calmodulin-Dependent Protein Kinases ,biology.protein ,Signal transduction ,Mitogen-Activated Protein Kinases ,Cardiology and Cardiovascular Medicine ,Interleukin-1 - Abstract
Cyclooxygenase-2 (COX-2) catalyzes the rate-limiting step in delayed prostaglandin biosynthesis. The purpose of this study was to evaluate the role of the MAP kinase kinase 6 (MKK6)–p38 MAPK signaling cascade in the regulation of myocardial COX-2 gene expression, in vitro and in vivo. RT-PCR analysis identified p38α and p38β2 MAPK mRNA in rat cardiac myocytes. Interleukin-1β induced the phosphorylation of p38α and p38β2 MAPK in cardiomyocytes and stimulated RNA polymerase II binding to the COX-2 promoter, COX-2 transcription, COX-2 protein synthesis, and prostaglandin E 2 (PGE 2 ) release. Infecting cardiomyocytes with adenoviruses that encode mutant, phosphorylation-resistant MKK6 or p38β2 MAPK inhibited interleukin-1β–induced p38 MAPK activation, COX-2 gene expression, and PGE 2 release. Treatment with the p38α and p38β2 MAPK inhibitor, SB202190, attenuated interleukin-1β–induced COX-2 transcription and accelerated the degradation of COX-2 mRNA. Cells infected with adenoviruses encoding wild-type or constitutively activated MKK6 or p38β2 MAPK, in the absence of interleukin-1β, exhibited increased cellular p38 MAPK activity, COX-2 mRNA expression, and COX-2 protein synthesis, which was blocked by SB202190. In addition, elevated levels of COX-2 protein were identified in the hearts of transgenic mice with cardiac-restricted expression of wild-type or constitutively activated MKK6, in comparison with nontransgenic littermates. These results provide direct evidence that MKK6 mediated p38 MAPK activation is necessary for interleukin-1β–induced cardiac myocyte COX-2 gene expression and PGE 2 biosynthesis in vitro and is sufficient for COX-2 gene expression by cardiac myocytes in vitro and in vivo.
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.