201
|
Sampieri L, Di Giusto P, Alvarez C. CREB3 Transcription Factors: ER-Golgi Stress Transducers as Hubs for Cellular Homeostasis. Front Cell Dev Biol 2019; 7:123. [PMID: 31334233 PMCID: PMC6616197 DOI: 10.3389/fcell.2019.00123] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022] Open
Abstract
CREB3 family of transcription factors are ER localized proteins that belong to the bZIP family. They are transported from the ER to the Golgi, cleaved by S1P and S2P proteases and the released N-terminal domains act as transcription factors. CREB3 family members regulate the expression of a large variety of genes and according to their tissue-specific expression profiles they play, among others, roles in acute phase response, lipid metabolism, development, survival, differentiation, organelle autoregulation, and protein secretion. They have been implicated in the ER and Golgi stress responses as regulators of the cell secretory capacity and cell specific cargos. In this review we provide an overview of the diverse functions of each member of the family (CREB3, CREB3L1, CREB3L2, CREB3L3, CREB3L4) with special focus on their role in the central nervous system.
Collapse
Affiliation(s)
- Luciana Sampieri
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Pablo Di Giusto
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Cecilia Alvarez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
202
|
Wan YJ, Guo Q, Liu D, Jiang Y, Zeng KW, Tu PF. Protocatechualdehyde reduces myocardial fibrosis by directly targeting conformational dynamics of collagen. Eur J Pharmacol 2019; 855:183-191. [DOI: 10.1016/j.ejphar.2019.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 04/17/2019] [Accepted: 05/02/2019] [Indexed: 10/26/2022]
|
203
|
|
204
|
Belmadani S, Matrougui K. Broken heart: A matter of the endoplasmic reticulum stress bad management? World J Cardiol 2019; 11:159-170. [PMID: 31367278 PMCID: PMC6658386 DOI: 10.4330/wjc.v11.i6.159] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/29/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases are the number one cause of morbidity and mortality in the United States and worldwide. The induction of the endoplasmic reticulum (ER) stress, a result of a disruption in the ER homeostasis, was found to be highly associated with cardiovascular diseases such as hypertension, diabetes, ischemic heart diseases and heart failure. This review will discuss the latest literature on the different aspects of the involvement of the ER stress in cardiovascular complications and the potential of targeting the ER stress pathways as a new therapeutic approach for cardiovascular complications.
Collapse
Affiliation(s)
- Souad Belmadani
- Department of Physiological Science, Eastern Virginia Medical School, Norfolk, VA 23501, United States
| | - Khalid Matrougui
- Department of Physiological Science, Eastern Virginia Medical School, Norfolk, VA 23501, United States
| |
Collapse
|
205
|
Irisin Protects Heart Against Ischemia-Reperfusion Injury Through a SOD2-Dependent Mitochondria Mechanism. J Cardiovasc Pharmacol 2019; 72:259-269. [PMID: 29979350 DOI: 10.1097/fjc.0000000000000608] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Irisin, a muscle-origin protein derived from the extracellular domain of the fibronectin domain-containing 5 protein (FNDC5), has been shown to modulate mitochondria welfare through paracrine action. Here, we test the hypothesis that irisin contributes to cardioprotection after myocardial infarction by preserving mitochondrial function in cardiomyocytes. Animal model studies show that intravenous administration of exogenous irisin produces dose-dependent protection against ischemia/reperfusion (I/R)-induced injury to the heart as reflected by the improvement of left ventricular ejection fraction and the reduction in serum level of cTnI (n = 15, P < 0.05). I/R-induced apoptosis of cardiomyocytes is reduced after irisin treatment. The irisin-mediated protection has, at least in part, an effect on mitochondrial function because administration of irisin increases irisin staining in the mitochondria of the infarct area. Irisin also reduces I/R-induced oxidative stress as determined by mitochondrial membrane potential evaluation and superoxide FLASH event recording (n = 4, P < 0.05). The interaction between irisin and superoxide dismutase2 (SOD2) plays a key role in the protective process because irisin treatment increases SOD activity (n = 10, P < 0.05) and restores the mitochondria localization of SOD2 in cardiomyocytes (n = 5, P < 0.05). These results demonstrate that irisin plays a protective role against I/R injury to the heart. Targeting the action of irisin in mitochondria presents a novel therapeutic intervention for myocardial infarction.
Collapse
|
206
|
Glembotski CC, Rosarda JD, Wiseman RL. Proteostasis and Beyond: ATF6 in Ischemic Disease. Trends Mol Med 2019; 25:538-550. [PMID: 31078432 DOI: 10.1016/j.molmed.2019.03.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/16/2019] [Accepted: 03/25/2019] [Indexed: 12/28/2022]
Abstract
Endoplasmic reticulum (ER) stress is a pathological hallmark of numerous ischemic diseases, including stroke and myocardial infarction (MI). In these diseases, ER stress leads to activation of the unfolded protein response (UPR) and subsequent adaptation of cellular physiology in ways that dictate cellular fate following ischemia. Recent evidence highlights a protective role for the activating transcription factor 6 (ATF6) arm of the UPR in mitigating adverse outcomes associated with ischemia/reperfusion (I/R) injury in multiple disease models. This suggests ATF6 as a potential therapeutic target for intervening in diverse ischemia-related disorders. Here, we discuss the evidence demonstrating the importance of ATF6 signaling in protecting different tissues against ischemic damage and discuss preclinical results focused on defining the potential for pharmacologically targeting ATF6 to intervene in such diseases.
Collapse
Affiliation(s)
- Christopher C Glembotski
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Jessica D Rosarda
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
207
|
Yarapureddy S, Abril J, Foote J, Kumar S, Asad O, Sharath V, Faraj J, Daniel D, Dickman P, White-Collins A, Hingorani P, Sertil AR. ATF6α Activation Enhances Survival against Chemotherapy and Serves as a Prognostic Indicator in Osteosarcoma. Neoplasia 2019; 21:516-532. [PMID: 31029032 PMCID: PMC6484364 DOI: 10.1016/j.neo.2019.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 02/25/2019] [Accepted: 02/26/2019] [Indexed: 01/31/2023] Open
Abstract
Patients with metastatic or relapsed/refractory osteosarcoma (OS) have a 5-year survival rate of <30%. This has remained unchanged over several decades. One of the factors contributing to lack of improvement in survival is the development of chemoresistance. Hence, elucidating and targeting the mechanisms that promote survival against chemotherapy and lead to chemoresistance is pivotal to improving outcomes for these patients. We identified that endoplasmic reticulum (ER) stress-activated transcription factor, ATF6α, is essential for the survival of OS cells against chemotherapy induced cell death. ATF6α cleavage and activity were enhanced in OS cells compared to normal osteoblasts and knockdown of ATF6α expression enhanced sensitivity of OS cells against chemotherapy induced cell death. This was in part due to increased Bax activation. Pharmacologic inhibition or knock-down of downstream targets of ATF6α, protein disulfide isomerases (PDI) and ERO1β, a thiol oxidase that is involved in the re-oxidation of PDIs also independently induced pronounced killing of OS cells following chemotherapy. Analysis of primary tumors from OS patients reveals that patients with high levels of nuclear ATF6α: (1) also had increased expression of its downstream targets the chaperone BiP and enzyme PDI, (2) had a significant likelihood of developing metastasis at diagnosis, (3) had significantly poorer overall and progression free survival, and (4) had poorer response to chemotherapy. These findings suggest that targeting survival signaling by the ATF6α pathway in OS cells may favor eradication of refractory OS tumor cells and ATF6α could be a useful predictor for chemo-responsiveness and prognosis.
Collapse
Affiliation(s)
- Suma Yarapureddy
- Department of Basic Medical Sciences, University of Arizona, College of Medicine, Phoenix, AZ
| | - Jazmine Abril
- Department of Basic Medical Sciences, University of Arizona, College of Medicine, Phoenix, AZ
| | - Janet Foote
- Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ
| | - Saravana Kumar
- Department of Basic Medical Sciences, University of Arizona, College of Medicine, Phoenix, AZ
| | - Omar Asad
- Department of Basic Medical Sciences, University of Arizona, College of Medicine, Phoenix, AZ
| | - Veena Sharath
- Department of Basic Medical Sciences, University of Arizona, College of Medicine, Phoenix, AZ
| | - Janine Faraj
- Department of Basic Medical Sciences, University of Arizona, College of Medicine, Phoenix, AZ
| | - Dustin Daniel
- Department of Basic Medical Sciences, University of Arizona, College of Medicine, Phoenix, AZ
| | - Paul Dickman
- Department of Hematology and Oncology, Phoenix Children's Hospital, Phoenix, Arizona
| | - Andrea White-Collins
- Department of Hematology and Oncology, Phoenix Children's Hospital, Phoenix, Arizona
| | - Pooja Hingorani
- Department of Hematology and Oncology, Phoenix Children's Hospital, Phoenix, Arizona.
| | - Aparna R Sertil
- Department of Basic Medical Sciences, University of Arizona, College of Medicine, Phoenix, AZ.
| |
Collapse
|
208
|
Wang F, Pulinilkunnil T, Flibotte S, Nislow C, Vlodavsky I, Hussein B, Rodrigues B. Heparanase protects the heart against chemical or ischemia/reperfusion injury. J Mol Cell Cardiol 2019; 131:29-40. [PMID: 31004678 DOI: 10.1016/j.yjmcc.2019.04.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/14/2019] [Accepted: 04/10/2019] [Indexed: 12/14/2022]
Abstract
Although cancer cells use heparanase for tumor metastasis, favourable effects of heparanase have been reported in the management of Alzheimer's disease and diabetes. Indeed, we previously established a protective function for heparanase in the acutely diabetic heart, where it conferred cardiomyocyte resistance to oxidative stress and apoptosis by provoking changes in gene expression. In this study, we tested if overexpression of heparanase can protect the heart against chemically induced or ischemia/reperfusion (I/R) injury. Transcriptomic analysis of Hep-tg hearts reveal that 240 genes related to the stress response, immune response, cell death, and development were altered in a pro-survival direction encompassing genes promoting the unfolded protein response (UPR) and autophagy, as well as those protecting against oxidative stress. The observed UPR activation was adaptive and not apoptotic, was mediated by activation of ATF6α, and when combined with mTOR inhibition, induced autophagy. Subjecting wild type (WT) mice to increasing concentrations of the ER stress inducer thapsigargin evoked a transition from adaptive to apoptotic UPR, an effect that was attenuated in Hep-tg mouse hearts. Consistent with these observations, when exposed to I/R, the infarct size and markers of apoptosis were significantly lower in the Hep-tg heart compared to WT. Finally, UPR and autophagy inhibitors reduced the protective effects of heparanase overexpression during I/R. Our data suggest that the mechanisms that underlie the role of heparanase in promoting cell survival could be uniquely beneficial to the heart by providing protection against cellular stresses, and could be useful for exploitation as a therapeutic target for the treatment of heart disease.
Collapse
Affiliation(s)
- Fulong Wang
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, 100 Tucker Park Road, Saint John, NB E2L 4L5, Canada
| | | | - Corey Nislow
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Bahira Hussein
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Brian Rodrigues
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
209
|
Endoplasmic Reticulum Stress-Induced NLRP1 Inflammasome Activation Contributes to Myocardial Ischemia/Reperfusion Injury. Shock 2019; 51:511-518. [DOI: 10.1097/shk.0000000000001175] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
210
|
Deng T, Wang Y, Wang C, Yan H. FABP4 silencing ameliorates hypoxia reoxygenation injury through the attenuation of endoplasmic reticulum stress-mediated apoptosis by activating PI3K/Akt pathway. Life Sci 2019; 224:149-156. [PMID: 30904493 DOI: 10.1016/j.lfs.2019.03.046] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/19/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Endoplasmic reticulum (ER) stress and subsequent apoptosis play a vital role in myocardial ischemia reperfusion (IR) injury. Fatty acid binding protein 4 (FABP4) may induce ER stress. The aim of this study was to investigate the mechanism and effect of FABP4 on IR injury in vitro. Rat H9c2 cells were exposed to hypoxia reoxygenation (HR) to create an IR model in vitro. FABP4 was overexpressed in HR-injured H9c2 cells. Transfection with FABP4 siRNA increased cell viability and decreased LDH upon HR stimulation. FABP4 cessation also suppressed apoptotic cells and caspase-3 activity after HR. Downregulation of FABP4 significantly inhibited ER stress by decreasing the protein expression of p-PERK, GRP78, and ATF6. FABP4 silencing also restrained the ER stress-mediated apoptotic pathway, as indicated by decreased pro-apoptotic proteins p-JNK, CHOP, Bax, and caspase-12, as well as upregulation of Bcl-2 during HR. Furthermore, FABP4 silencing activated the PI3K/Akt pathway. Blocking this pathway by the specific PI3K inhibitor-LY294002 restored HR-induced ER stress and subsequently reversed the protective effect of FABP4 silencing on HR injury. Taken together, our findings revealed that FABP4 silencing exerts protective effects against HR injury in H9c2 cells through inhibiting ER stress-induced cell apoptosis via activation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Tianming Deng
- Department of Cardiology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China.
| | - Yanhong Wang
- Department of Geriatrcs, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Chongchong Wang
- Department of Cardiology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Hua Yan
- Department of Cardiology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| |
Collapse
|
211
|
McKimpson WM, Kitsis RN. A New Role for the ER Unfolded Protein Response Mediator ATF6: Induction of a Generalized Antioxidant Program. Circ Res 2019; 120:759-761. [PMID: 28254796 DOI: 10.1161/circresaha.117.310577] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Wendy M McKimpson
- From the Department of Medicine (W.M.M., R.N.K.), Department of Cell Biology (W.M.M., R.N.K.), Wilf Family Cardiovascular Research Center (W.M.M., R.N.K.), Einstein-Mount Sinai Diabetes Research Center (R.N.K.), and Albert Einstein Cancer Center (R.N.K.), Albert Einstein College of Medicine, Bronx, NY
| | - Richard N Kitsis
- From the Department of Medicine (W.M.M., R.N.K.), Department of Cell Biology (W.M.M., R.N.K.), Wilf Family Cardiovascular Research Center (W.M.M., R.N.K.), Einstein-Mount Sinai Diabetes Research Center (R.N.K.), and Albert Einstein Cancer Center (R.N.K.), Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
212
|
Correll RN, Grimes KM, Prasad V, Lynch JM, Khalil H, Molkentin JD. Overlapping and differential functions of ATF6α versus ATF6β in the mouse heart. Sci Rep 2019; 9:2059. [PMID: 30765833 PMCID: PMC6375966 DOI: 10.1038/s41598-019-39515-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/02/2019] [Indexed: 01/22/2023] Open
Abstract
Hemodynamic stress on the mammalian heart results in compensatory hypertrophy and activation of the unfolded protein response through activating transcription factor 6α (ATF6α) in cardiac myocytes, but the roles of ATF6α or the related transcription factor ATF6β in regulating this hypertrophic response are not well-understood. Here we examined the effects of loss of ATF6α or ATF6β on the cardiac response to pressure overload. Mice gene-deleted for Atf6 or Atf6b were subjected to 2 weeks of transverse aortic constriction, and each showed a significant reduction in hypertrophy with reduced expression of endoplasmic reticulum (ER) stress-associated proteins compared with controls. However, with long-term pressure overload both Atf6 and Atf6b null mice showed enhanced decompensation typified by increased heart weight, pulmonary edema and reduced function compared to control mice. Our subsequent studies using cardiac-specific transgenic mice expressing the transcriptionally active N-terminus of ATF6α or ATF6β revealed that these factors control overlapping gene expression networks that include numerous ER protein chaperones and ER associated degradation components. This work reveals previously unappreciated roles for ATF6α and ATF6β in regulating the pressure overload induced cardiac hypertrophic response and in controlling the expression of genes that condition the ER during hemodynamic stress.
Collapse
Affiliation(s)
- Robert N Correll
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, 35487, USA.,Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Kelly M Grimes
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Vikram Prasad
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Jeffrey M Lynch
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Hadi Khalil
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA. .,Howard Hughes Medical Institute, Cincinnati, Ohio, 45229, USA.
| |
Collapse
|
213
|
Blackwood EA, Azizi K, Thuerauf DJ, Paxman RJ, Plate L, Kelly JW, Wiseman RL, Glembotski CC. Pharmacologic ATF6 activation confers global protection in widespread disease models by reprograming cellular proteostasis. Nat Commun 2019; 10:187. [PMID: 30643122 PMCID: PMC6331617 DOI: 10.1038/s41467-018-08129-2] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 12/11/2018] [Indexed: 01/05/2023] Open
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.
Collapse
Affiliation(s)
- Erik A Blackwood
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, CA, 92182, USA
| | - Khalid Azizi
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, CA, 92182, USA
| | - Donna J Thuerauf
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, CA, 92182, USA
| | - Ryan J Paxman
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Lars Plate
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - R Luke Wiseman
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Christopher C Glembotski
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, CA, 92182, USA.
| |
Collapse
|
214
|
Affiliation(s)
- Ellen Malovrh
- From the Department of Cardiology, Angiology, and Pneumology, University Hospital Heidelberg, University of Heidelberg, Germany (E.M., M.V.).,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (E.M., M.V.)
| | - Mirko Völkers
- From the Department of Cardiology, Angiology, and Pneumology, University Hospital Heidelberg, University of Heidelberg, Germany (E.M., M.V.).,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (E.M., M.V.)
| |
Collapse
|
215
|
Zhang G, Wang X, Gillette TG, Deng Y, Wang ZV. Unfolded Protein Response as a Therapeutic Target in Cardiovascular Disease. Curr Top Med Chem 2019; 19:1902-1917. [PMID: 31109279 PMCID: PMC7024549 DOI: 10.2174/1568026619666190521093049] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 04/09/2019] [Accepted: 05/02/2019] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Despite overwhelming socioeconomic impact and mounting clinical needs, our understanding of the underlying pathophysiology remains incomplete. Multiple forms of cardiovascular disease involve an acute or chronic disturbance in cardiac myocytes, which may lead to potent activation of the Unfolded Protein Response (UPR), a cellular adaptive reaction to accommodate protein-folding stress. Accumulation of unfolded or misfolded proteins in the Endoplasmic Reticulum (ER) elicits three signaling branches of the UPR, which otherwise remain quiescent. This ER stress response then transiently suppresses global protein translation, augments production of protein-folding chaperones, and enhances ER-associated protein degradation, with an aim to restore cellular homeostasis. Ample evidence has established that the UPR is strongly induced in heart disease. Recently, the mechanisms of action and multiple pharmacological means to favorably modulate the UPR are emerging to curb the initiation and progression of cardiovascular disease. Here, we review the current understanding of the UPR in cardiovascular disease and discuss existing therapeutic explorations and future directions.
Collapse
Affiliation(s)
- Guangyu Zhang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Xiaoding Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Thomas G. Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Yingfeng Deng
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Zhao V. Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
216
|
Wang X, Yuan B, Cheng B, Liu Y, Zhang B, Wang X, Lin X, Yang B, Gong G. Crocin Alleviates Myocardial Ischemia/Reperfusion-Induced Endoplasmic Reticulum Stress via Regulation of miR-34a/Sirt1/Nrf2 Pathway. Shock 2019; 51:123-130. [DOI: 10.1097/shk.0000000000001116] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
217
|
Li J, Zhao Y, Zhou N, Li L, Li K. Dexmedetomidine Attenuates Myocardial Ischemia-Reperfusion Injury in Diabetes Mellitus by Inhibiting Endoplasmic Reticulum Stress. J Diabetes Res 2019; 2019:7869318. [PMID: 31886285 PMCID: PMC6914963 DOI: 10.1155/2019/7869318] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/24/2019] [Accepted: 10/25/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE With the increasing incidence of diabetes mellitus (DM) combined with myocardial ischemia, how to reduce myocardial ischemia-reperfusion injury in DM patients has become a major problem faced by clinicians. We investigated the therapeutic effects of dexmedetomidine (DEX) on myocardial ischemia-reperfusion injury in DM rats and its effect on endoplasmic reticulum stress. METHODS SD rats with SPF grade were randomly divided into 6 groups: non-DM rats were divided into the sham operation group (NDM-S group), ischemia-reperfusion group (NDM-IR group), and dexmedetomidine group (NDM-DEX group); DM rats were divided into the diabetic sham operation group (DM-S group), diabetes-reperfusion group (DM-IR group), and diabetes-dexmedetomidine (DM-DEX) group, with 10 rats in each group. Then the effects of DEX on the changes of CK-MB and cTnT levels were examined. The effects of myocardial pathological damage and myocardial infarct size were detected. The apoptosis of cardiomyocytes was detected. The apoptosis of heart tissue cells was also tested through the expressions of cleaved caspase-3, Bcl-2, and Bax proteins. The expression of endoplasmic reticulum stress-related proteins GRP78, CHOP, ERO1α, ERO1β, and PDI was examined. The hypoxia/reoxygenation (H/R) injury cell model was established, the effects of DEX, DEX+ ERS agonist on cell apoptosis was also detected. RESULTS The myocardial damage of DM-IR was more severe than that of NDM-IR rats. DEX could reduce the expression of CK-MB and cTnT, reduce pathological damage, and reduce scar formation and improve fibrosis. DEX can reduce the expression of GRP78, CHOP, ERO1α, ERO1β, and PDI proteins in vivo and in vitro. And the effect of DEX on cell apoptosis could be blocked by ERS agonist. CONCLUSION DEX attenuates myocardial ischemia-reperfusion injury in DM rats and H/R injury cell, which is associated with the reduction of ERS-induced cardiomyocyte apoptosis.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cell Line
- Creatine Kinase, MB Form/blood
- Dexmedetomidine/pharmacology
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Endoplasmic Reticulum Stress/drug effects
- Fibrosis
- Heat-Shock Proteins/metabolism
- Male
- Membrane Glycoproteins/metabolism
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/prevention & control
- Myocardial Reperfusion Injury/complications
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/prevention & control
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Protein Disulfide-Isomerases/metabolism
- Rats, Sprague-Dawley
- Signal Transduction
- Transcription Factor CHOP/metabolism
- Troponin T/blood
Collapse
Affiliation(s)
- Jinjie Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, Jilin 130033, China
| | - Ying Zhao
- Department of Nephrology, The First Hospital of Jilin University, No. 71, Xinmin Street, Changchun Jilin 130021, China
| | - Nan Zhou
- Department of Anesthesia, The General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Longyun Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, Jilin 130033, China
| | - Kai Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, Jilin 130033, China
| |
Collapse
|
218
|
He F, Shu Y, Wang X, Liu X, Liu G, Chen Z, Wang Z, Li L, Liu R, Zhou H, Xu H, Zhang W, Zhou G. Intensive Glucose Control Reduces the Risk Effect of TRIB3, SMARCD3, and ATF6 Genetic Variation on Diabetic Vascular Complications. Front Pharmacol 2018; 9:1422. [PMID: 30618737 PMCID: PMC6297143 DOI: 10.3389/fphar.2018.01422] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 11/19/2018] [Indexed: 02/05/2023] Open
Abstract
Type 2 diabetes mellitus is a complex disease. Our previous study revealed that TRIB3 genetic variations were strongly associated with diabetic vascular complications, although TRIB3 regulation pathways remain poorly understood. We used two extreme treatment groups from a 2 × 2 factorial randomized controlled trial to identify a positive association, which was further validated in patients receiving cross treatment to test the effect of genetic polymorphisms among the different treatment groups. A gene-centric score (GS)-weighted model including the three associated genetic variations TRIB3 rs2295490, ATF6 rs12086247, and SMARCD3 rs58125572 was used. The results of the GS model indicated a 46% reduction in the risk of primary vascular complications in patients bearing more than two risk alleles [hazard ratio (HR) 0.54, 95% confidence interval (CI) 0.38-0.76, p < 0.001], following intensive glucose control treatment when compared with patients who received standard glucose control treatment. Furthermore, these patients benefited from active blood pressure-lowering treatment (HR 0.39, 95% CI 0.24-0.64, p < 0.001). However, no significant difference was observed between the two interventions in patients with fewer than two risk alleles (HR 1.09, 95% CI 0.86-1.39, p = 0.47). These results indicate that genetic variants in these three genes may be useful biomarkers for individualized drug therapy in diabetic patients.
Collapse
Affiliation(s)
- Fazhong He
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Pharmacogenetics Research Institute – Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, United States
| | - Xingyu Wang
- Beijing Hypertension League Institute, Beijing, China
| | - Xin Liu
- Beijing Hypertension League Institute, Beijing, China
| | - Guojing Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Pharmacogenetics Research Institute – Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Zhangren Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Pharmacogenetics Research Institute – Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenmin Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Pharmacogenetics Research Institute – Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Ling Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Pharmacogenetics Research Institute – Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Rong Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Pharmacogenetics Research Institute – Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Pharmacogenetics Research Institute – Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Heng Xu
- Department of Laboratory Medicine, Precision Medicine Center, and Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- Sichuan and Collaborative Innovation Center, Chengdu, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Pharmacogenetics, Pharmacogenetics Research Institute – Institute of Clinical Pharmacology, Central South University, Changsha, China
- National Clinical Research Center for Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Gan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
219
|
Pandey VK, Mathur A, Kakkar P. Emerging role of Unfolded Protein Response (UPR) mediated proteotoxic apoptosis in diabetes. Life Sci 2018; 216:246-258. [PMID: 30471281 DOI: 10.1016/j.lfs.2018.11.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023]
Abstract
Endoplasmic reticulum (ER) is a crucial single membrane organelle that acts as a quality control system for cellular proteins as it is intricately involved in their synthesis, folding and trafficking to the respective targets. Type 2 diabetes is characterized by enhanced blood glucose level that promotes insulin resistance and hampers cellular glucose metabolism. Hyperglycemia provokes mitochondrial ROS production and glycation of proteins which exert a tremendous load on ER for conventional refolding of misfolded/unfolded and nascent proteins that perturb ER homeostasis resulting in apoptotic cell death. Impairment in ER functions is suspected to be through specific ER membrane-bound proteins known as Unfolded Protein Response (UPR) sensor proteins. Conformational changes in these proteins induce oligomerization and cross-autophosphorylation which facilitate processes required for the restoration of ER homeostatic imbalance. Multiple studies have reported the involvement of UPR mediated autophagy and apoptotic pathways in the progression of metabolic disorders including diabetes, cardiac ischemia/reperfusion injury and hypoxia-mediated cell death. In this review, the involvement of UPR pathways in the progression of diabetes associated complications have been addressed, which underscores molecular crosstalks during neuropathy, nephropathy, hepatic injury and retinopathy. A better understanding of these molecular interventions may reveal advanced therapeutic approaches for preventing diabetic comorbidities. The article also highlights the importance of phytochemicals that are emerging as novel ER stress inhibitors and are being explored for targeted interaction in preventing cell death responses during diabetes.
Collapse
Affiliation(s)
- Vivek Kumar Pandey
- Herbal Research Laboratory, Food, Drug & Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan 31, M.G Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Toxicology Research, Lucknow 226001, Uttar Pradesh, India
| | - Alpana Mathur
- Herbal Research Laboratory, Food, Drug & Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan 31, M.G Marg, Lucknow 226001, Uttar Pradesh, India; Babu Banarasi Das University, Lucknow, Uttar Pradesh, India
| | - Poonam Kakkar
- Herbal Research Laboratory, Food, Drug & Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan 31, M.G Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Toxicology Research, Lucknow 226001, Uttar Pradesh, India.
| |
Collapse
|
220
|
Gáll T, Pethő D, Nagy A, Hendrik Z, Méhes G, Potor L, Gram M, Åkerström B, Smith A, Nagy P, Balla G, Balla J. Heme Induces Endoplasmic Reticulum Stress (HIER Stress) in Human Aortic Smooth Muscle Cells. Front Physiol 2018; 9:1595. [PMID: 30515102 PMCID: PMC6255930 DOI: 10.3389/fphys.2018.01595] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/24/2018] [Indexed: 12/17/2022] Open
Abstract
Accumulation of damaged or misfolded proteins resulted from oxidative protein modification induces endoplasmic reticulum (ER) stress by activating the pathways of unfolded protein response. In pathologic hemolytic conditions, extracellular free hemoglobin is submitted to rapid oxidation causing heme release. Resident cells of atherosclerotic lesions, after intraplaque hemorrhage, are exposed to heme leading to oxidative injury. Therefore, we raised the question whether heme can also provoke ER stress. Smooth muscle cells are one of the key players of atherogenesis; thus, human aortic smooth muscle cells (HAoSMCs) were selected as a model cell to reveal the possible link between heme and ER stress. Using immunoblotting, quantitative polymerase chain reaction and immunocytochemistry, we quantitated the markers of ER stress. These were: phosphorylated eIF2α, Activating transcription factor-4 (ATF4), DNA-damage-inducible transcript 3 (also known as C/EBP homology protein, termed CHOP), X-box binding protein-1 (XBP1), Activating transcription factor-6 (ATF6), GRP78 (glucose-regulated protein, 78kDa) and heme responsive genes heme oxygenase-1 and ferritin. In addition, immunohistochemistry was performed on human carotid artery specimens from patients who had undergone carotid endarterectomy. We demonstrate that heme increases the phosphorylation of eiF2α in HAoSMCs and the expression of ATF4. Heme also enhances the splicing of XBP1 and the proteolytic cleavage of ATF6. Consequently, there is up-regulation of target genes increasing both mRNA and protein levels of CHOP and GRP78. However, TGFβ and collagen type I decreased. When the heme binding proteins, alpha-1-microglobulin (A1M) and hemopexin (Hpx) are present in cell media, the ER stress provoked by heme is inhibited. ER stress pathways are also retarded by the antioxidant N-acetyl cysteine (NAC) indicating that reactive oxygen species are involved in heme-induced ER stress. Consistent with these findings, elevated expression of the ER stress marker GRP78 and CHOP were observed in smooth muscle cells of complicated lesions with hemorrhage compared to either atheromas or healthy arteries. In conclusion, heme triggers ER stress in a time- and dose-dependent manner in HAoSMCs. A1M and Hpx as well as NAC effectively hamper heme-induced ER stress, supporting their use as a potential therapeutic approach to reverse such a deleterious effects of heme toxicity.
Collapse
Affiliation(s)
- Tamás Gáll
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dávid Pethő
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Annamária Nagy
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Hendrik
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Potor
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Magnus Gram
- Department of Clinical Sciences Lund, Infection Medicine, Lund University, Lund, Sweden
| | - Bo Åkerström
- Department of Clinical Sciences Lund, Infection Medicine, Lund University, Lund, Sweden
| | - Ann Smith
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Péter Nagy
- Department of Vascular Surgery, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Balla
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - József Balla
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
221
|
Network pharmacology-based identification of major component of Angelica sinensis and its action mechanism for the treatment of acute myocardial infarction. Biosci Rep 2018; 38:BSR20180519. [PMID: 30232231 PMCID: PMC6239257 DOI: 10.1042/bsr20180519] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 01/06/2023] Open
Abstract
Background: To decipher the mechanisms of Angelica sinensis for the treatment of acute myocardial infarction (AMI) using network pharmacology analysis. Methods: Databases were searched for the information on constituents, targets, and diseases. Cytoscape software was used to construct the constituent–target–disease network and screen the major targets, which were annotated with the DAVID (Database for Annotation, Visualization and Integrated Discovery) tool. The cardioprotective effects of Angelica sinensis polysaccharide (ASP), a major component of A. sinensis, were validated both in H9c2 cells subjected to simulated ischemia by oxygen and glucose deprivation and in rats with AMI by ligation of the left anterior coronary artery. Results: We identified 228 major targets against AMI injury for A. sinensis, which regulated multiple pathways and hit multiple targets involved in several biological processes. ASP significantly decreased endoplasmic reticulum (ER) stress-induced cell death both in vitro and in vivo. In ischemia injury rats, ASP treatment reduced infarct size and preserved heart function. ASP enhanced activating transcription factor 6 (ATF6) activity, which improved ER-protein folding capacity. ASP activated the expression of p-AMP-activated protein kinase (p-AMPK) and peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α). Additionally, ASP attenuated levels of proinflammatory cytokines and maintained a balance in the oxidant/antioxidant levels after AMI. Conclusion:In silico analysis revealed the associations between A. sinensis and AMI through multiple targets and several key signaling pathways. Experimental data indicate that ASP protects the heart against ischemic injury by activating ATF6 to ameliorate the detrimental ER stress. ASP’s effects could be mediated via the activation of AMPK-PGC1α pathway.
Collapse
|
222
|
Wu B, Feng J, Yu L, Wang Y, Chen Y, Wei Y, Han J, Feng X, Zhang Y, Di S, Ma Z, Fan C, Ha X. Icariin protects cardiomyocytes against ischaemia/reperfusion injury by attenuating sirtuin 1-dependent mitochondrial oxidative damage. Br J Pharmacol 2018; 175:4137-4153. [PMID: 30051466 PMCID: PMC6177614 DOI: 10.1111/bph.14457] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 07/02/2018] [Accepted: 07/12/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Icariin, a major active ingredient in traditional Chinese medicines, is attracting increasing attention because of its unique pharmacological effects against ischaemic heart disease. The histone deacetylase, sirtuin-1, plays a protective role in ischaemia/reperfusion (I/R) injury, and this study was designed to investigate the protective role of icariin in models of cardiac I/R injury and to elucidate the potential involvement of sirtuin-1. EXPERIMENTAL APPROACH I/R injury was simulated in vivo (mouse hearts), ex vivo (isolated rat hearts) and in vitro (neonatal rat cardiomyocytes and H9c2 cells). Prior to I/R injury, animals or cells were exposed to icariin, with or without inhibitors of sirtuin-1 (sirtinol and SIRT1 siRNA). KEY RESULTS In vivo and in vitro, icariin given before I/R significantly improved post-I/R heart contraction and limited the infarct size and leakage of creatine kinase-MB and LDH from the damaged myocardium. Icariin also attenuated I/R-induced mitochondrial oxidative damage, decreasing malondialdehyde content and increasing superoxide dismutase activity and expression of Mn-superoxide dismutase. Icariin significantly improved mitochondrial membrane homeostasis by increasing mitochondrial membrane potential and cytochrome C stabilization, which further inhibited cell apoptosis. Sirtuin-1 was significantly up-regulated in hearts treated with icariin, whereas Ac-FOXO1 was simultaneously down-regulated. Importantly, sirtinol and SIRT1 siRNA either blocked icariin-induced cardioprotection or disrupted icariin-mediated mitochondrial homeostasis. CONCLUSIONS AND IMPLICATIONS Pretreatment with icariin protected cardiomyocytes from I/R-induced oxidative stress through activation of sirtuin-1 /FOXO1 signalling.
Collapse
Affiliation(s)
- Bing Wu
- Department of GeriatricsLanzhou General Hospital of the People's Liberation ArmyLanzhouChina
- Department of CardiologyTangdu Hospital, Fourth Military Medical UniversityXi'anChina
| | - Jian‐yu Feng
- Department of Cardiovascular Surgery, Xijing HospitalFourth Military Medical UniversityXi'anChina
| | - Li‐ming Yu
- Department of Cardiovascular SurgeryGeneral Hospital of Shenyang Military Area CommandShenyangChina
| | - Yan‐chun Wang
- Department of GeriatricsLanzhou General Hospital of the People's Liberation ArmyLanzhouChina
| | - Yong‐qing Chen
- Department of CardiologyLanzhou General Hospital of the People's Liberation ArmyLanzhouChina
| | - Yan Wei
- Department of ophthalmologyLanzhou General Hospital of the People's Liberation ArmyLanzhouChina
| | - Jin‐song Han
- Department of Cardiovascular SurgeryGeneral Hospital of Shenyang Military Area CommandShenyangChina
| | - Xiao Feng
- Department of Cardiovascular Surgery, Xijing HospitalFourth Military Medical UniversityXi'anChina
| | - Yu Zhang
- Department of Cardiovascular SurgeryLanzhou General Hospital of the People's Liberation ArmyLanzhouChina
| | - Shou‐yin Di
- Department of Thoracic SurgeryTangdu Hospital, Fourth Military Medical UniversityXi'anChina
| | - Zhi‐qiang Ma
- Department of Thoracic SurgeryTangdu Hospital, Fourth Military Medical UniversityXi'anChina
| | - Chong‐xi Fan
- Department of Thoracic SurgeryTangdu Hospital, Fourth Military Medical UniversityXi'anChina
- Department of Biomedical EngineeringFourth Military Medical UniversityXi'anChina
| | - Xiao‐qin Ha
- Department of Clinical LaboratoryLanzhou General Hospital of the People's Liberation ArmyLanzhouChina
| |
Collapse
|
223
|
Hu C, Tian Y, Xu H, Pan B, Terpstra EM, Wu P, Wang H, Li F, Liu J, Wang X. Inadequate ubiquitination-proteasome coupling contributes to myocardial ischemia-reperfusion injury. J Clin Invest 2018; 128:5294-5306. [PMID: 30204128 DOI: 10.1172/jci98287] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 09/04/2018] [Indexed: 12/14/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) degrades a protein molecule via 2 main steps: ubiquitination and proteasomal degradation. Extraproteasomal ubiquitin receptors are thought to couple the 2 steps, but this proposition has not been tested in vivo with vertebrates. More importantly, impaired UPS performance plays a major role in cardiac pathogenesis, including myocardial ischemia-reperfusion injury (IRI), but the molecular basis of UPS impairment remains poorly understood. Ubiquilin1 is a bona fide extraproteasomal ubiquitin receptor. Here, we report that mice with a cardiomyocyte-restricted knockout of Ubiquilin1 (Ubqln1-CKO mice) accumulated a surrogate UPS substrate (GFPdgn) and increased myocardial ubiquitinated proteins without altering proteasome activities, resulting in late-onset cardiomyopathy and a markedly shortened life span. When subject to regional myocardial ischemia-reperfusion, young Ubqln1-CKO mice showed substantially exacerbated cardiac malfunction and enlarged infarct size, and conversely, mice with transgenic Ubqln1 overexpression displayed attenuated IRI. Furthermore, Ubqln1 overexpression facilitated proteasomal degradation of oxidized proteins and the degradation of a UPS surrogate substrate in cultured cardiomyocytes without increasing autophagic flux. These findings demonstrate that Ubiquilin1 is essential to cardiac ubiquitination-proteasome coupling and that an inadequacy in the coupling represents a major pathogenic factor for myocardial IRI; therefore, strategies to strengthen coupling have the potential to reduce IRI.
Collapse
Affiliation(s)
- Chengjun Hu
- Department of Human Anatomy, Wuhan University College of Basic Medical Sciences, Wuhan, Hubei, China.,Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| | - Yihao Tian
- Department of Human Anatomy, Wuhan University College of Basic Medical Sciences, Wuhan, Hubei, China.,Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| | - Hongxin Xu
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Bo Pan
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| | - Erin M Terpstra
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| | - Penglong Wu
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA.,Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hongmin Wang
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| | - Faqian Li
- Department of Pathology and Laboratory Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jinbao Liu
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, South Dakota, USA
| |
Collapse
|
224
|
Abstract
Human heart failure is characterized by arrhythmogenic electrical remodeling consisting mostly of ion channel downregulations. Reversing these downregulations is a logical approach to antiarrhythmic therapy, but understanding the pathophysiological mechanisms of the reduced currents is crucial for finding the proper treatments. The unfolded protein response (UPR) is activated by endoplasmic reticulum (ER) stress and has been found to play pivotal roles in different diseases including neurodegenerative diseases, diabetes mellitus, and heart disease. Recently, the UPR is reported to regulate multiple cardiac ion channels, contributing to arrhythmias in heart disease. In this review, we will discuss which UPR modulators and effectors could be involved in regulation of cardiac ion channels in heart disease, and how the understanding of these regulating mechanisms may lead to new antiarrhythmic therapeutics that lack the proarrhythmic risk of current ion channel blocking therapies.
Collapse
Affiliation(s)
- Man Liu
- a Division of Cardiology, Department of Medicine, The Lillehei Heart Institute , University of Minnesota at Twin Cities , Minneapolis , USA
| | - Samuel C Dudley
- a Division of Cardiology, Department of Medicine, The Lillehei Heart Institute , University of Minnesota at Twin Cities , Minneapolis , USA
| |
Collapse
|
225
|
Yoon Y, Kim TJ, Lee JM, Kim DY. SOD2 is upregulated in periodontitis to reduce further inflammation progression. Oral Dis 2018; 24:1572-1580. [PMID: 29972711 DOI: 10.1111/odi.12933] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 06/22/2018] [Accepted: 06/28/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Periodontitis is a highly prevalent chronic inflammatory disease that results in destruction of tooth-supporting structures followed by tooth-loss. Until now, periodontitis has been regarded to be initiated by bacterial infection followed by aberrant host response. Although increasing evidence suggests a strong association between oxidative stress and periodontitis, precise molecular mechanism has been left unanswered. In this study, we investigated roles of SOD2, the main antioxidant enzyme maintaining reactive oxygen species (ROS) homeostasis, under inflammatory conditions. METHODS We computationally analyzed SOD2 expression in periodontitis. To confirm this data, immunoblot assay was performed with samples from periodontitis patients. The cellular mechanism of change in SOD2 expression was identified through immunoblot assay and immunofluorescence. To evaluate the molecular function of SOD2, we generated SOD2-deficient cells by utilizing the CRISPR/Cas9 system. RESULTS We first determined that SOD2 expression was significantly increased in periodontitis. We also confirmed that SOD2 expression was upregulated through the NF-κB pathway when the inflammatory signal was stronger and extended. Gene manipulation against SOD2 through the CRISPR/Cas9 system showed that the absence of SOD2 increased production of NLRP3 inflammasome components. CONCLUSIONS Our study demonstrates that intracellular SOD2 has a protective role by suppressing NLRP inflammasome-caspase-1-IL-1β axis under inflammatory conditions.
Collapse
Affiliation(s)
- Yong Yoon
- Department of Periodontology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Tae-Jun Kim
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Jae-Mok Lee
- Department of Periodontology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Do-Yeon Kim
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, Korea
| |
Collapse
|
226
|
Paxman R, Plate L, Blackwood EA, Glembotski C, Powers ET, Wiseman RL, Kelly JW. Pharmacologic ATF6 activating compounds are metabolically activated to selectively modify endoplasmic reticulum proteins. eLife 2018; 7:37168. [PMID: 30084354 PMCID: PMC6080950 DOI: 10.7554/elife.37168] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/24/2018] [Indexed: 12/15/2022] Open
Abstract
Pharmacologic arm-selective unfolded protein response (UPR) signaling pathway activation is emerging as a promising strategy to ameliorate imbalances in endoplasmic reticulum (ER) proteostasis implicated in diverse diseases. The small molecule N-(2-hydroxy-5-methylphenyl)-3-phenylpropanamide (147) was previously identified (Plate et al., 2016) to preferentially activate the ATF6 arm of the UPR, promoting protective remodeling of the ER proteostasis network. Here we show that 147-dependent ATF6 activation requires metabolic oxidation to form an electrophile that preferentially reacts with ER proteins. Proteins covalently modified by 147 include protein disulfide isomerases (PDIs), known to regulate ATF6 activation. Genetic depletion of PDIs perturbs 147-dependent induction of the ATF6-target gene, BiP, implicating covalent modifications of PDIs in the preferential activation of ATF6 afforded by treatment with 147. Thus, 147 is a pro-drug that preferentially activates ATF6 signaling through a mechanism involving localized metabolic activation and selective covalent modification of ER resident proteins that regulate ATF6 activity.
Collapse
Affiliation(s)
- Ryan Paxman
- Department of Chemistry, The Scripps Research Institute, La Jolla, United States
| | - Lars Plate
- Department of Chemistry, The Scripps Research Institute, La Jolla, United States.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, United States
| | - Erik A Blackwood
- Department of Biology, San Diego State University, San Diego, United States.,San Diego State University Heart Institute, San Diego State University, San Diego, United States
| | - Chris Glembotski
- Department of Biology, San Diego State University, San Diego, United States.,San Diego State University Heart Institute, San Diego State University, San Diego, United States
| | - Evan T Powers
- Department of Chemistry, The Scripps Research Institute, La Jolla, United States
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, United States
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, United States.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, United States.,The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, United States
| |
Collapse
|
227
|
|
228
|
Hillary RF, FitzGerald U. A lifetime of stress: ATF6 in development and homeostasis. J Biomed Sci 2018; 25:48. [PMID: 29801500 PMCID: PMC5968583 DOI: 10.1186/s12929-018-0453-1] [Citation(s) in RCA: 181] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/22/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Activating transcription factor 6 (ATF6) is an endoplasmic reticulum (ER)-localised protein and member of the leucine zipper family of transcription factors. Best known for its role in transducing signals linked to stress to the endoplasmic reticulum, the 50 kDa activated form of ATF6 is now emerging as a major regulator of organogenesis and tissue homeostasis. Responsible for the correct folding, secretion and membrane insertion of a third of the proteome in eukaryotic cells, the ER encompasses a dynamic, labyrinthine network of regulators, chaperones, foldases and cofactors. Such structures are crucial to the extensive protein synthesis required to undergo normal development and maintenance of tissue homeostasis. When an additional protein synthesis burden is placed on the ER, ATF6, in tandem with ER stress transducers inositol requiring enzyme 1 (IRE1) and PKR-like endoplasmic reticulum kinase (PERK), slows the pace of protein translation and induces the production of stress-reducing chaperones and foldases. MAIN TEXT In the context of development and tissue homeostasis, however, distinct cellular impacts have been attributed to ATF6. Drawing on data published from human, rodent, fish, goat and bovine research, this review first focuses on ATF6-mediated regulation of osteo- and chondrogenesis, ocular development as well as neuro- and myelinogenesis. The purported role of ATF6 in development of the muscular and reproductive systems as well as adipo- and lipogenesis is then described. With relevance to cardiac disease, cancer and brain disorders, the importance of ATF6 in maintaining tissue homeostasis is the subject of the final section. CONCLUSION In conclusion, the review encourages further elucidation of ATF6 regulatory operations during organogenesis and tissue homeostasis, to spawn the development of ATF6-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Robert F Hillary
- Galway Neuroscience Centre, Cúram Centre for Research in Medical Devices, School of Natural Sciences, National University of Ireland, Galway, Ireland
| | - Una FitzGerald
- Galway Neuroscience Centre, Cúram Centre for Research in Medical Devices, School of Natural Sciences, National University of Ireland, Galway, Ireland.
| |
Collapse
|
229
|
Twayana KS, Ravanan P. Eukaryotic cell survival mechanisms: Disease relevance and therapeutic intervention. Life Sci 2018; 205:73-90. [PMID: 29730169 DOI: 10.1016/j.lfs.2018.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/17/2018] [Accepted: 05/01/2018] [Indexed: 01/10/2023]
Abstract
Cell responds to stress by activating various modes of stress responses which aim for minimal damage to cells and speedy recovery from the insults. However, unresolved stresses exceeding the tolerance limit lead to cell death (apoptosis, autophagy etc.) that helps to get rid of damaged cells and protect cell integrity. Furthermore, aberrant stress responses are the hallmarks of several pathophysiologies (neurodegeneration, metabolic diseases, cancer etc.). The catastrophic remodulation of stress responses is observed in cancer cells in favor of their uncontrolled growth. Whereas pro-survival stress responses redirected to death signaling provokes excessive cell death in neurodegeneration. Clear understanding of such mechanistic link to disease progression is required in order to modulate these processes for new therapeutic targets. The current review explains this with respect to novel drug discoveries and other breakthroughs in therapeutics.
Collapse
Affiliation(s)
- Krishna Sundar Twayana
- Apoptosis and Cell Survival Research Laboratory, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu-632014, India
| | - Palaniyandi Ravanan
- Apoptosis and Cell Survival Research Laboratory, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu-632014, India.
| |
Collapse
|
230
|
Bi X, Zhang G, Wang X, Nguyen C, May HI, Li X, Al-Hashimi AA, Austin RC, Gillette TG, Fu G, Wang ZV, Hill JA. Endoplasmic Reticulum Chaperone GRP78 Protects Heart From Ischemia/Reperfusion Injury Through Akt Activation. Circ Res 2018; 122:1545-1554. [PMID: 29669712 DOI: 10.1161/circresaha.117.312641] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/30/2018] [Accepted: 04/17/2018] [Indexed: 12/14/2022]
Abstract
RATIONALE Restoration of coronary artery blood flow is the most effective means of ameliorating myocardial damage triggered by ischemic heart disease. However, coronary reperfusion elicits an increment of additional injury to the myocardium. Accumulating evidence indicates that the unfolded protein response (UPR) in cardiomyocytes is activated by ischemia/reperfusion (I/R) injury. Xbp1s (spliced X-box binding protein 1), the most highly conserved branch of the unfolded protein response, is protective in response to cardiac I/R injury. GRP78 (78 kDa glucose-regulated protein), a master regulator of the UPR and an Xbp1s target, is upregulated after I/R. However, its role in the protective response of Xbp1s during I/R remains largely undefined. OBJECTIVE To elucidate the role of GRP78 in the cardiomyocyte response to I/R using both in vitro and in vivo approaches. METHODS AND RESULTS Simulated I/R injury to cultured neonatal rat ventricular myocytes induced apoptotic cell death and strong activation of the UPR and GRP78. Overexpression of GRP78 in neonatal rat ventricular myocytes significantly protected myocytes from I/R-induced cell death. Furthermore, cardiomyocyte-specific overexpression of GRP78 ameliorated I/R damage to the heart in vivo. Exploration of underlying mechanisms revealed that GRP78 mitigates cellular damage by suppressing the accumulation of reactive oxygen species. We go on to show that the GRP78-mediated cytoprotective response involves plasma membrane translocation of GRP78 and interaction with PI3 kinase, culminating in stimulation of Akt. This response is required as inhibition of the Akt pathway significantly blunted the antioxidant activity and cardioprotective effects of GRP78. CONCLUSIONS I/R induction of GRP78 in cardiomyocytes stimulates Akt signaling and protects against oxidative stress, which together protect cells from I/R damage.
Collapse
Affiliation(s)
- Xukun Bi
- From the Department of Cardiology, Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (X.B., X.L., G.F.).,Division of Cardiology, Department of Internal Medicine (X.B., G.Z., X.W., C.N., H.I.M., T.G.G., Z.V.W., J.A.H.)
| | - Guangyu Zhang
- Division of Cardiology, Department of Internal Medicine (X.B., G.Z., X.W., C.N., H.I.M., T.G.G., Z.V.W., J.A.H.).,University of Texas Southwestern Medical Center, Dallas; Department of Cardiology, Zhongnan Hospital of Wuhan University, Hubei, China (G.Z.)
| | - Xiaoding Wang
- Division of Cardiology, Department of Internal Medicine (X.B., G.Z., X.W., C.N., H.I.M., T.G.G., Z.V.W., J.A.H.).,Department of Cardiology, Renmin Hospital of Wuhan University, Hubei, China (X.W.)
| | - Chau Nguyen
- Division of Cardiology, Department of Internal Medicine (X.B., G.Z., X.W., C.N., H.I.M., T.G.G., Z.V.W., J.A.H.)
| | - Herman I May
- Division of Cardiology, Department of Internal Medicine (X.B., G.Z., X.W., C.N., H.I.M., T.G.G., Z.V.W., J.A.H.)
| | - Xiaoting Li
- From the Department of Cardiology, Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (X.B., X.L., G.F.)
| | - Ali A Al-Hashimi
- Department of Medicine, Hamilton Center for Kidney Research, McMaster University and the Research Institute of St. Joseph's Healthcare Hamilton, ON, Canada (A.A.A.-H., R.C.A.)
| | - Richard C Austin
- Department of Medicine, Hamilton Center for Kidney Research, McMaster University and the Research Institute of St. Joseph's Healthcare Hamilton, ON, Canada (A.A.A.-H., R.C.A.)
| | - Thomas G Gillette
- Division of Cardiology, Department of Internal Medicine (X.B., G.Z., X.W., C.N., H.I.M., T.G.G., Z.V.W., J.A.H.)
| | - Guosheng Fu
- From the Department of Cardiology, Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China (X.B., X.L., G.F.)
| | - Zhao V Wang
- Division of Cardiology, Department of Internal Medicine (X.B., G.Z., X.W., C.N., H.I.M., T.G.G., Z.V.W., J.A.H.)
| | - Joseph A Hill
- Division of Cardiology, Department of Internal Medicine (X.B., G.Z., X.W., C.N., H.I.M., T.G.G., Z.V.W., J.A.H.).,Department of Molecular Biology (J.A.H.)
| |
Collapse
|
231
|
Wang X, Bi X, Zhang G, Deng Y, Luo X, Xu L, Scherer PE, Ferdous A, Fu G, Gillette TG, Lee AS, Jiang X, Wang ZV. Glucose-regulated protein 78 is essential for cardiac myocyte survival. Cell Death Differ 2018; 25:2181-2194. [PMID: 29666470 PMCID: PMC6261960 DOI: 10.1038/s41418-018-0109-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/09/2018] [Accepted: 03/19/2018] [Indexed: 02/03/2023] Open
Abstract
Secretory and transmembrane proteins rely on proper function of the secretory pathway for folding, posttranslational modification, assembly, and secretion. Accumulation of misfolded proteins in the endoplasmic reticulum (ER) stimulates the unfolded protein response (UPR), which communicates between the ER and other organelles to enhance ER-folding capacity and restore cellular homeostasis. Glucose-regulated protein of 78 kDa (GRP78), an ER-resident protein chaperone, is a master regulator of all UPR signaling branches. Accumulating studies have established a fundamental role of GRP78 in protein folding, ER stress response, and cell survival. However, role of GRP78 in the heart remains incompletely characterized. Here we showed that embryos lacking GRP78 specifically in cardiac myocytes manifest cardiovascular malformations and die in utero at late gestation. We went further to show that inducible knockout of GRP78 in adult cardiac myocytes causes early mortality due to cardiac cell death and severe decline in heart performance. At the cellular level, we found that loss of GRP78 increases apoptotic cell death, which is accompanied by reduction in AKT signaling and augmentation of production for reactive oxygen species. Importantly, enhancing AKT phosphorylation and activity leads to decreases in oxidative stress and increases in cardiac myocyte survival. Collectively, our results demonstrate an essential role of GRP78 in ensuring normal cardiogenesis and maintaining cardiac contractility and function.
Collapse
Affiliation(s)
- Xiaoding Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xukun Bi
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Cardiology, Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Guangyu Zhang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yingfeng Deng
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiang Luo
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Anwarul Ferdous
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guosheng Fu
- Department of Cardiology, Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Thomas G Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Zhao V Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
232
|
Valenzuela V, Jackson KL, Sardi SP, Hetz C. Gene Therapy Strategies to Restore ER Proteostasis in Disease. Mol Ther 2018; 26:1404-1413. [PMID: 29728295 DOI: 10.1016/j.ymthe.2018.04.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/01/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022] Open
Abstract
Proteostasis alterations are proposed as a transversal hallmark of several pathological conditions, including metabolic disorders, mechanical injury, cardiac malfunction, neurodegeneration, and cancer. Strategies to improve proteostasis aim to reduce the accumulation of specific disease-related misfolded proteins or bolster the endogenous mechanisms to fold and degrade abnormal proteins. Endoplasmic reticulum (ER) stress is a common pathological signature of a variety of diseases, which engages the unfolded protein response (UPR) as a cellular reaction to mitigate ER stress. Pharmacological modulation of the UPR is challenging considering the physiological importance of the pathway in various organs. However, local targeting of ER stress responses in the affected tissue using gene therapy is emerging as a possible solution to overcome side effects. The delivery of ER chaperones or active UPR components using adeno-associated virus (AAV) has demonstrated outstanding beneficial effects in several disease models (e.g., neurodegenerative conditions, eye disorders, and metabolic diseases). Here, we discuss current efforts to design and optimize gene therapy strategies to improve ER proteostasis in different disease contexts.
Collapse
Affiliation(s)
- Vicente Valenzuela
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Kasey L Jackson
- Neuroscience Therapeutic Area, Sanofi, Framingham, MA 01701, USA
| | - Sergio P Sardi
- Neuroscience Therapeutic Area, Sanofi, Framingham, MA 01701, USA
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA 94945, USA; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
233
|
Liu Y, Li P, Fan L, Wu M. The nuclear transportation routes of membrane-bound transcription factors. Cell Commun Signal 2018; 16:12. [PMID: 29615051 PMCID: PMC5883603 DOI: 10.1186/s12964-018-0224-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/19/2018] [Indexed: 12/12/2022] Open
Abstract
Membrane-bound transcription factors (MTFs) are transcription factors (TFs) that are anchored in membranes in a dormant state. Activated by external or internal stimuli, MTFs are released from parent membranes and are transported to the nucleus. Existing research indicates that some plasma membrane (PM)-bound proteins and some endoplasmic reticulum (ER) membrane-bound proteins have the ability to enter the nucleus. Upon specific signal recognition cues, some PM-bound TFs undergo proteolytic cleavage to liberate the intracellular fragments that enter the nucleus to control gene transcription. However, lipid-anchored PM-bound proteins enter the nucleus in their full length for depalmitoylation. In addition, some PM-bound TFs exist as full-length proteins in cell nucleus via trafficking to the Golgi and the ER, where membrane-releasing mechanisms rely on endocytosis. In contrast, the ER membrane-bound TFs relocate to the nucleus directly or by trafficking to the Golgi. In both of these pathways, only the fragments of the ER membrane-bound TFs transit to the nucleus. Several different nuclear trafficking modes of MTFs are summarized in this review, providing an effective supplement to the mechanisms of signal transduction and gene regulation. Moreover, targeting intracellular movement pathways of disease-associated MTFs may significantly improve the survival of patients.
Collapse
Affiliation(s)
- Yang Liu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410008, Hunan, China
| | - Peiyao Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410008, Hunan, China
| | - Li Fan
- Department of Biochemistry, University of California, Riverside, CA, 92521, USA
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China. .,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
234
|
Zhang L, Yan X, Zhang YL, Bai J, Hidru TH, Wang QS, Li HH. Vitamin D attenuates pressure overload-induced cardiac remodeling and dysfunction in mice. J Steroid Biochem Mol Biol 2018; 178:293-302. [PMID: 29337094 DOI: 10.1016/j.jsbmb.2018.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 12/28/2022]
Abstract
Vitamin D (VD) and its analogues play critical roles in metabolic and cardiovascular diseases. Recent studies have demonstrated that VD exerts a protective role in cardiovascular diseases. However, the beneficial effect of VD on pressure overload-induced cardiac remodeling and dysfunction and its underlying mechanisms are not fully elucidated. In this study, cardiac dysfunction and hypertrophic remodeling in mice were induced by pressure overload. Cardiac function was evaluated by echocardiography, and myocardial histology was detected by H&E and Masson's trichrome staining. Cardiomyocyte size was detected by wheat germ agglutinin staining. The protein levels of signaling mediators were examined by western blotting while mRNA expression of hypertrophic and fibrotic markers was examined by qPCR analysis. Oxidative stress was detected by dihydroethidine staining. Our results showed that administration of VD3 significantly ameliorates pressure overload-induced contractile dysfunction, cardiac hypertrophy, fibrosis and inflammation in mice. In addition, VD3 treatment also markedly inhibited cardiac oxidative stress and apoptosis. Moreover, protein levels of calcineurin A, ERK1/2, AKT, TGF-β, GRP78, cATF6, and CHOP were significantly reduced whereas SERCA2 level was upregulated in the VD3-treated hearts compared with control. These results suggest that VD3 attenuates cardiac remodeling and dysfunction induced by pressure overload, and this protective effect is associated with inhibition of multiple signaling pathways.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China; Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiao Yan
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China; Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yun-Long Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China; Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jie Bai
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China; Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | | | - Qing-Shan Wang
- School of Public Health, Dalian Medical University, Dalian 116044, China.
| | - Hui-Hua Li
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China; Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
235
|
Wang S, Binder P, Fang Q, Wang Z, Xiao W, Liu W, Wang X. Endoplasmic reticulum stress in the heart: insights into mechanisms and drug targets. Br J Pharmacol 2018; 175:1293-1304. [PMID: 28548229 PMCID: PMC5867005 DOI: 10.1111/bph.13888] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/04/2017] [Accepted: 05/16/2017] [Indexed: 01/03/2023] Open
Abstract
The endoplasmic reticulum (ER) serves several essential cellular functions including protein synthesis, protein folding, protein translocation, calcium homoeostasis and lipid biosynthesis. Physiological or pathological stimuli, which disrupt ER homoeostasis and disturb its functions, lead to an accumulation of misfolded and unfolded proteins, a condition referred to as ER stress. ER stress triggers the unfolded protein response to restore the homoeostasis of ER, through activating transcriptional and translational pathways. However, prolonged ER stress will lead to cell dysfunction and apoptosis. Recent evidence revealed that ER stress is involved in the development and progression of various heart diseases, such as cardiac hypertrophy, ischaemic heart diseases and heart failure. Therefore, improved understanding of the molecular mechanisms of ER stress in heart disease will help to investigate more potential targets for new therapeutic interventions and drug discovery. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Shunyao Wang
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Pablo Binder
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Qiru Fang
- State Key Laboratory of New‐tech for Chinese Medicine Pharmaceutical ProcessLianyungangChina
| | - Zhenzhong Wang
- State Key Laboratory of New‐tech for Chinese Medicine Pharmaceutical ProcessLianyungangChina
| | - Wei Xiao
- State Key Laboratory of New‐tech for Chinese Medicine Pharmaceutical ProcessLianyungangChina
| | - Wei Liu
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Xin Wang
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| |
Collapse
|
236
|
Niu X, Zhang J, Ling C, Bai M, Peng Y, Sun S, Li Y, Zhang Z. Polysaccharide from Angelica sinensis protects H9c2 cells against oxidative injury and endoplasmic reticulum stress by activating the ATF6 pathway. J Int Med Res 2018. [PMID: 29517941 PMCID: PMC5991254 DOI: 10.1177/0300060518758863] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objectives Angelica sinensis exerts various pharmacological effects, such as antioxidant and anti-apoptotic activity. This study aimed to investigate the active ingredients in A. sinensis with antioxidant properties and whether A. sinensis polysaccharide (ASP) protects H9c2 cells against oxidative and endoplasmic reticulum (ER) stress. Methods The ingredients of A. sinensis and their targets and related pathways were determined using web-based databases. Markers of oxidative stress, cell viability, apoptosis, and ER stress-related signalling pathways were measured in H9c2 cells treated with hydrogen peroxide (H2O2) and ASP. Results The ingredient–pathway–disease network showed that A. sinensis exerted protective effects against oxidative injury through its various active ingredients on regulation of multiple pathways. Subsequent experiments showed that ASP pretreatment significantly decreased H2O2-induced cytotoxicity and apoptosis in H9c2 cells. ASP pretreatment inhibited H2O2-induced reactive oxygen species generation, lactic dehydrogenase release, and malondialdehyde production. ASP exerted beneficial effects by inducing activating transcription factor 6 (ATF6) and increasing ATF6 target protein levels, which in turn attenuated ER stress and increased antioxidant activity. Conclusions Our findings indicate that ASP, a major water-soluble component of A. sinensis, exerts protective effects against H2O2-induced injury in H9c2 cells by activating the ATF6 pathway, thus ameliorating ER and oxidative stress.
Collapse
Affiliation(s)
- Xiaowei Niu
- 1 The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | | | - Chun Ling
- 3 The First People's Hospital of Chuzhou, Chuzhou, Anhui, China
| | - Ming Bai
- 4 Department of Cardiology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China.,5 Gansu Key Laboratory of Cardiovascular Disease, Lanzhou, Gansu, China
| | - Yu Peng
- 4 Department of Cardiology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China.,5 Gansu Key Laboratory of Cardiovascular Disease, Lanzhou, Gansu, China
| | - Shaobo Sun
- 6 Key Lab of Prevention and Treatment for Chronic Disease, Traditional Chinese Medicine of Gansu Province, Lanzhou, Gansu, China
| | - Yingdong Li
- 6 Key Lab of Prevention and Treatment for Chronic Disease, Traditional Chinese Medicine of Gansu Province, Lanzhou, Gansu, China
| | - Zheng Zhang
- 4 Department of Cardiology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China.,5 Gansu Key Laboratory of Cardiovascular Disease, Lanzhou, Gansu, China
| |
Collapse
|
237
|
Islam O, Patil P, Goswami SK, Razdan R, Inamdar MN, Rizwan M, Mathew J, Inceoglu B, Stephen Lee KS, Hwang SH, Hammock BD. Inhibitors of soluble epoxide hydrolase minimize ischemia-reperfusion-induced cardiac damage in normal, hypertensive, and diabetic rats. Cardiovasc Ther 2018; 35. [PMID: 28296232 DOI: 10.1111/1755-5922.12259] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 02/12/2017] [Accepted: 03/05/2017] [Indexed: 01/29/2023] Open
Abstract
AIM We designed a study to evaluate the cardioprotective effect of two soluble epoxide hydrolase (sEH) inhibitors, 1-(1-propanoylpiperidin-4-yl)-3-(4-trifluoromethoxy)phenyl)urea (TPPU) and trans-4-{4-[3-(4-trifluoromethoxyphenyl)-ureido]cyclohexyloxy}benzoic acid (t-TUCB), in ischemia-reperfusion (IR) model. METHODS Cardioprotective effects of the sEH inhibitors were evaluated against IR-induced myocardial damage in hearts from normal, hypertensive, and diabetic rats using Langendorff's apparatus. In addition, the effect of sEH inhibitors on endothelial function was evaluated in vitro and ex vivo using isolated rat thoracic aorta. RESULTS Ischemia-reperfusion (IR) increased the myocardial damage in hearts from normal rats. IR-induced myocardial damage was augmented in hearts isolated from hypertensive and diabetic rats. Myocardial damage as evident from increase in the activities of lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) in heart perfusate was associated with significant decrease in the heart rate and developed tension, and increase in the resting tension in isolated heart. Both sEH inhibitors protected the heart in normal, hypertensive, and diabetic rats subjected to IR injury. The sEH inhibitor t-TUCB relaxed phenylephrine precontracted aorta from normal rats. Relaxant effect of acetylcholine (ACh) was reduced in aortas from diabetic and hypertensive rats compared to normal rats. Pretreatment of sEH inhibitors to diabetic and hypertensive rats increased relaxant effect of ACh on aortas isolated from these rats. CONCLUSIONS Prophylactic treatment with sEH inhibitors decreased myocardial damage due to IR, hypertension and diabetes, and decreased endothelial dysfunction created by diabetes and hypertension. Therefore, inhibitors of sEH are useful probes to study cardiovascular pathology, and inhibition of the sEH is a potential approach in the management of IR-induced cardiac damage and endothelial dysfunction-related cardiovascular disorders.
Collapse
Affiliation(s)
- Oliul Islam
- Department of Pharmacology, Al-Ameen College of Pharmacy, Bangalore, Karnataka, India
| | - Prashanth Patil
- Department of Pharmacology, Al-Ameen College of Pharmacy, Bangalore, Karnataka, India
| | - Sumanta K Goswami
- Department of Pharmacology, Al-Ameen College of Pharmacy, Bangalore, Karnataka, India.,Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Rema Razdan
- Department of Pharmacology, Al-Ameen College of Pharmacy, Bangalore, Karnataka, India
| | - Mohammed N Inamdar
- Department of Pharmacology, Al-Ameen College of Pharmacy, Bangalore, Karnataka, India.,Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, KSA
| | - Mohammed Rizwan
- Department of Pharmacology, Al-Ameen College of Pharmacy, Bangalore, Karnataka, India
| | - Jubin Mathew
- Department of Pharmacology, Al-Ameen College of Pharmacy, Bangalore, Karnataka, India
| | - Bora Inceoglu
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Kin S Stephen Lee
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Sung H Hwang
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA, USA
| |
Collapse
|
238
|
Wang X, Xu L, Gillette TG, Jiang X, Wang ZV. The unfolded protein response in ischemic heart disease. J Mol Cell Cardiol 2018; 117:19-25. [PMID: 29470977 DOI: 10.1016/j.yjmcc.2018.02.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/10/2018] [Accepted: 02/17/2018] [Indexed: 12/28/2022]
Abstract
Ischemic heart disease is a severe stress condition that causes extensive pathological alterations and triggers cardiac cell death. Accumulating evidence suggests that the unfolded protein response (UPR) is strongly induced by myocardial ischemia. The UPR is an evolutionarily conserved cellular response to cope with protein-folding stress, from yeast to mammals. Endoplasmic reticulum (ER) transmembrane sensors detect the accumulation of unfolded proteins and stimulate a signaling network to accommodate unfolded and misfolded proteins. Distinct mechanisms participate in the activation of three major signal pathways, viz. protein kinase RNA-like ER kinase, inositol-requiring protein 1, and activating transcription factor 6, to transiently suppress protein translation, enhance protein folding capacity of the ER, and augment ER-associated degradation to refold denatured proteins and restore cellular homeostasis. However, if the stress is severe and persistent, the UPR elicits inflammatory and apoptotic pathways to eliminate terminally affected cells. The ER is therefore recognized as a vitally important organelle that determines cell survival or death. Recent studies indicate the UPR plays critical roles in the pathophysiology of ischemic heart disease. The three signaling branches may elicit distinct but overlapping effects in cardiac response to ischemia. Here, we outline the findings and discuss the mechanisms of action and therapeutic potentials of the UPR in the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Xiaoding Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Thomas G Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Zhao V Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
239
|
Kroeger H, Grimsey N, Paxman R, Chiang WC, Plate L, Jones Y, Shaw PX, Trejo J, Tsang SH, Powers E, Kelly JW, Wiseman RL, Lin JH. The unfolded protein response regulator ATF6 promotes mesodermal differentiation. Sci Signal 2018; 11:eaan5785. [PMID: 29440509 PMCID: PMC5957084 DOI: 10.1126/scisignal.aan5785] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
ATF6 encodes a transcription factor that is anchored in the endoplasmic reticulum (ER) and activated during the unfolded protein response (UPR) to protect cells from ER stress. Deletion of the isoform activating transcription factor 6α (ATF6α) and its paralog ATF6β results in embryonic lethality and notochord dysgenesis in nonhuman vertebrates, and loss-of-function mutations in ATF6α are associated with malformed neuroretina and congenital vision loss in humans. These phenotypes implicate an essential role for ATF6 during vertebrate development. We investigated this hypothesis using human stem cells undergoing differentiation into multipotent germ layers, nascent tissues, and organs. We artificially activated ATF6 in stem cells with a small-molecule ATF6 agonist and, conversely, inhibited ATF6 using induced pluripotent stem cells from patients with ATF6 mutations. We found that ATF6 suppressed pluripotency, enhanced differentiation, and unexpectedly directed mesodermal cell fate. Our findings reveal a role for ATF6 during differentiation and identify a new strategy to generate mesodermal tissues through the modulation of the ATF6 arm of the UPR.
Collapse
Affiliation(s)
- Heike Kroeger
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Neil Grimsey
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ryan Paxman
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Wei-Chieh Chiang
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lars Plate
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
- Departments of Chemistry and Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
| | - Ying Jones
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Peter X Shaw
- Department of Ophthalmology, University of California, San Diego, La Jolla, CA 92093, USA
| | - JoAnn Trejo
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephen H Tsang
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology, Pathology and Cell Biology, Edward S. Harkness Eye Institute, New York Presbyterian Hospital, Columbia University, New York, NY 10032, USA
| | - Evan Powers
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jeffery W Kelly
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | - R Luke Wiseman
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jonathan H Lin
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA.
- VA San Diego Healthcare System, San Diego, CA 92161, USA
| |
Collapse
|
240
|
Gao X, Chen W, Li J, Shen C, Zhou P, Che X, Li X, Xie R. The protective effect of alpha-lipoic acid against brain ischemia and reperfusion injury via mTOR signaling pathway in rats. Neurosci Lett 2018; 671:108-113. [PMID: 29432779 DOI: 10.1016/j.neulet.2018.02.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 02/05/2018] [Accepted: 02/06/2018] [Indexed: 10/18/2022]
Abstract
Alpha-lipoic Acid(ALA), an endogenous short-chain fatty acid, has been found inducing a protective effect against ischemia and reperfusion(I/R) injury. Recently, mTOR signaling pathway has been proved to involve in the mechanism of I/R injury. In our previous study, we determined that ALA could protect cerebral endothelial cells against I/R injury via mTOR signaling pathway. However, whether ALA can protect against brain I/R injury in vivo and its mechanisms is uncertain. In this study, we try to explore if the ALA treatment can protect against brain I/R injury and confirm the relationship between ALA and mTOR signaling pathway. ALA was administrated to the animals after dMCAo and reperfusion model established with or without rapamycin pre-treatment. The results showed the infarct size was obviously reduced after ALA treatment in acute stage, neurological functions were also improved distinctly. The mTOR signaling pathway was remarkably blocked after brain I/R injury while it could be activated through ALA treatment. However, rapamycin, can abolish the protective effects induced by ALA treatment in both acute and long-term phase. In conclusion, we demonstrate the protective effects induced by ALA treatment against the brain I/R injury in rats and mTOR signaling pathway is required for the protective effects of ALA against brain I/R injury. The results might contribute to the potential clinical application of ALA and provide a potential therapeutic target on ischemic stroke.
Collapse
Affiliation(s)
- Xinjie Gao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wei Chen
- Department of Neurosurgery, Jingan District Center Hospital, Shanghai 200040, China
| | - Jinquan Li
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chao Shen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ping Zhou
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaoming Che
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaomu Li
- Department of Endocrinology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Rong Xie
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
241
|
Doran AC, Ozcan L, Cai B, Zheng Z, Fredman G, Rymond CC, Dorweiler B, Sluimer JC, Hsieh J, Kuriakose G, Tall AR, Tabas I. CAMKIIγ suppresses an efferocytosis pathway in macrophages and promotes atherosclerotic plaque necrosis. J Clin Invest 2017; 127:4075-4089. [PMID: 28972541 DOI: 10.1172/jci94735] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/10/2017] [Indexed: 01/29/2023] Open
Abstract
Atherosclerosis is the underlying etiology of cardiovascular disease, the leading cause of death worldwide. Atherosclerosis is a heterogeneous disease in which only a small fraction of lesions lead to heart attack, stroke, or sudden cardiac death. A distinct type of plaque containing large necrotic cores with thin fibrous caps often precipitates these acute events. Here, we show that Ca2+/calmodulin-dependent protein kinase γ (CaMKIIγ) in macrophages plays a major role in the development of necrotic, thin-capped plaques. Macrophages in necrotic and symptomatic atherosclerotic plaques in humans as well as advanced atherosclerotic lesions in mice demonstrated activation of CaMKII. Western diet-fed LDL receptor-deficient (Ldlr-/-) mice with myeloid-specific deletion of CaMKII had smaller necrotic cores with concomitantly thicker collagen caps. These lesions demonstrated evidence of enhanced efferocytosis, which was associated with increased expression of the macrophage efferocytosis receptor MerTK. Mechanistic studies revealed that CaMKIIγ-deficient macrophages and atherosclerotic lesions lacking myeloid CaMKIIγ had increased expression of the transcription factor ATF6. We determined that ATF6 induces liver X receptor-α (LXRα), an Mertk-inducing transcription factor, and that increased MerTK expression and efferocytosis in CaMKIIγ-deficient macrophages is dependent on LXRα. These findings identify a macrophage CaMKIIγ/ATF6/LXRα/MerTK pathway as a key factor in the development of necrotic atherosclerotic plaques.
Collapse
Affiliation(s)
- Amanda C Doran
- Department of Medicine, Columbia University, New York, New York, USA
| | - Lale Ozcan
- Department of Medicine, Columbia University, New York, New York, USA
| | - Bishuang Cai
- Department of Medicine, Columbia University, New York, New York, USA
| | - Ze Zheng
- Department of Medicine, Columbia University, New York, New York, USA
| | - Gabrielle Fredman
- Department of Molecular and Cellular Physiology, Center for Cardiovascular Sciences, Albany Medical Center, Albany, New York, USA
| | | | - Bernhard Dorweiler
- Department of Cardiothoracic and Vascular Surgery, Universitätsmedizin Mainz, Johannes-Gutenberg University, Mainz, Germany
| | - Judith C Sluimer
- Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Joanne Hsieh
- Department of Medicine, Columbia University, New York, New York, USA
| | | | - Alan R Tall
- Department of Medicine, Columbia University, New York, New York, USA
| | - Ira Tabas
- Department of Medicine, Columbia University, New York, New York, USA.,Department of Physiology and Cellular Biophysics and.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| |
Collapse
|
242
|
Thrombospondins: A Role in Cardiovascular Disease. Int J Mol Sci 2017; 18:ijms18071540. [PMID: 28714932 PMCID: PMC5536028 DOI: 10.3390/ijms18071540] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/05/2017] [Accepted: 07/13/2017] [Indexed: 12/16/2022] Open
Abstract
Thrombospondins (TSPs) represent extracellular matrix (ECM) proteins belonging to the TSP family that comprises five members. All TSPs have a complex multidomain structure that permits the interaction with various partners including other ECM proteins, cytokines, receptors, growth factors, etc. Among TSPs, TSP1, TSP2, and TSP4 are the most studied and functionally tested. TSP1 possesses anti-angiogenic activity and is able to activate transforming growth factor (TGF)-β, a potent profibrotic and anti-inflammatory factor. Both TSP2 and TSP4 are implicated in the control of ECM composition in hypertrophic hearts. TSP1, TSP2, and TSP4 also influence cardiac remodeling by affecting collagen production, activity of matrix metalloproteinases and TGF-β signaling, myofibroblast differentiation, cardiomyocyte apoptosis, and stretch-mediated enhancement of myocardial contraction. The development and evaluation of TSP-deficient animal models provided an option to assess the contribution of TSPs to cardiovascular pathology such as (myocardial infarction) MI, cardiac hypertrophy, heart failure, atherosclerosis, and aortic valve stenosis. Targeting of TSPs has a significant therapeutic value for treatment of cardiovascular disease. The activation of cardiac TSP signaling in stress and pressure overload may be therefore beneficial.
Collapse
|
243
|
Abstract
The incidence and prevalence of cardiac diseases, which are the main cause of death worldwide, are likely to increase because of population ageing. Prevailing theories about the mechanisms of ageing feature the gradual derailment of cellular protein homeostasis (proteostasis) and loss of protein quality control as central factors. In the heart, loss of protein patency, owing to flaws in genetically-determined design or because of environmentally-induced 'wear and tear', can overwhelm protein quality control, thereby triggering derailment of proteostasis and contributing to cardiac ageing. Failure of protein quality control involves impairment of chaperones, ubiquitin-proteosomal systems, autophagy, and loss of sarcomeric and cytoskeletal proteins, all of which relate to induction of cardiomyocyte senescence. Targeting protein quality control to maintain cardiac proteostasis offers a novel therapeutic strategy to promote cardiac health and combat cardiac disease. Currently marketed drugs are available to explore this concept in the clinical setting.
Collapse
Affiliation(s)
- Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, De Boelelaan 1117, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
244
|
Plate L, Wiseman RL. Regulating Secretory Proteostasis through the Unfolded Protein Response: From Function to Therapy. Trends Cell Biol 2017. [PMID: 28647092 DOI: 10.1016/j.tcb.2017.05.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Imbalances in secretory proteostasis induced by genetic, environmental, or aging-related insults are pathologically associated with etiologically diverse protein misfolding diseases. To protect the secretory proteome from these insults, organisms evolved stress-responsive signaling pathways that regulate the composition and activity of biologic pathways involved in secretory proteostasis maintenance. The most prominent of these is the endoplasmic reticulum (ER) unfolded protein response (UPR), which functions to regulate ER proteostasis in response to ER stress. While the signaling mechanisms involved in UPR activation are well defined, the impact of UPR activation on secretory proteostasis is only now becoming clear. Here, we highlight recent reports defining how activation of select UPR signaling pathways influences proteostasis within the ER and downstream secretory environments. Furthermore, we describe recent evidence that highlights the therapeutic potential for targeting UPR signaling pathways to correct pathologic disruption in secretory proteostasis associated with diverse types of protein misfolding diseases.
Collapse
Affiliation(s)
- Lars Plate
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
245
|
Lindholm D, Korhonen L, Eriksson O, Kõks S. Recent Insights into the Role of Unfolded Protein Response in ER Stress in Health and Disease. Front Cell Dev Biol 2017; 5:48. [PMID: 28540288 PMCID: PMC5423914 DOI: 10.3389/fcell.2017.00048] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/13/2017] [Indexed: 12/20/2022] Open
Abstract
Unfolded stress response (UPR) is a conserved cellular pathway involved in protein quality control to maintain homeostasis under different conditions and disease states characterized by cell stress. Although three general schemes of and genes induced by UPR are rather well-established, open questions remain including the precise role of UPR in human diseases and the interactions between different sensor systems during cell stress signaling. Particularly, the issue how the normally adaptive and pro-survival UPR pathway turns into a deleterious process causing sustained endoplasmic reticulum (ER) stress and cell death requires more studies. UPR is also named a friend with multiple personalities that we need to understand better to fully recognize its role in normal physiology and in disease pathology. UPR interacts with other organelles including mitochondria, and with cell stress signals and degradation pathways such as autophagy and the ubiquitin proteasome system. Here we review current concepts and mechanisms of UPR as studied in different cells and model systems and highlight the relevance of UPR and related stress signals in various human diseases.
Collapse
Affiliation(s)
- Dan Lindholm
- Medicum, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of HelsinkiHelsinki, Finland.,Minerva Foundation Institute for Medical ResearchHelsinki, Finland
| | - Laura Korhonen
- Minerva Foundation Institute for Medical ResearchHelsinki, Finland.,Division of Child Psychiatry, Helsinki University Central HospitalHelsinki, Finland
| | - Ove Eriksson
- Medicum, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of HelsinkiHelsinki, Finland
| | - Sulev Kõks
- Department of Pathophysiology, University of TartuTartu, Estonia.,Department of Reproductive Biology, Estonian University of Life SciencesTartu, Estonia
| |
Collapse
|
246
|
Arrieta A, Blackwood EA, Glembotski CC. ER Protein Quality Control and the Unfolded Protein Response in the Heart. Curr Top Microbiol Immunol 2017; 414:193-213. [PMID: 29026925 DOI: 10.1007/82_2017_54] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiac myocytes are the cells responsible for the robust ability of the heart to pump blood throughout the circulatory system. Cardiac myocytes grow in response to a variety of physiological and pathological conditions; this growth challenges endoplasmic reticulum-protein quality control (ER-PQC), a major feature of which includes the unfolded protein response (UPR). ER-PQC and the UPR in cardiac myocytes growing under physiological conditions, including normal development, exercise, and pregnancy, are sufficient to support hypertrophic growth of each cardiac myocyte. However, the ER-PQC and UPR are insufficient to respond to the challenge of cardiac myocyte growth under pathological conditions, including myocardial infarction and heart failure. In part, this insufficiency is due to a continual decline in the expression levels of important adaptive UPR components as a function of age and during myocardial pathology. This chapter will discuss the physiological and pathological conditions unique to the heart that involves ER-PQC, and whether the UPR is adaptive or maladaptive under these circumstances.
Collapse
Affiliation(s)
- A Arrieta
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, CA, 92182, USA
| | - E A Blackwood
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, CA, 92182, USA
| | - C C Glembotski
- San Diego State University Heart Institute and the Department of Biology, San Diego State University, San Diego, CA, 92182, USA.
| |
Collapse
|