1
|
Tian H, Wang T, Zhang Y, Pan T, Yao S, Yu H, Ma K, Wang S. Astragaloside IV protects against C/EBP homologous protein-mediated apoptosis in oxidized low-density lipoprotein-treated macrophages by promoting autophagy. Eur J Pharmacol 2022; 923:174912. [PMID: 35339476 DOI: 10.1016/j.ejphar.2022.174912] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/19/2022] [Accepted: 03/19/2022] [Indexed: 11/03/2022]
Abstract
Astragaloside Ⅳ (AS-Ⅳ) is one of the main active components extracted from Astragalus membranaceus that exerts an antiatherosclerotic effect. Our study explored the underlying anti-apoptotic effects and the mechanisms of action of AS-Ⅳ in oxidized low-density lipoprotein (oxLDL)-stimulated macrophages and in vulnerable plaques. The results showed that AS-Ⅳ lowered the oxLDL-induced lipid content and reversed the oxLDL-induced reduction in cell viability and elevation in lactate dehydrogenase (LDH) leakage and apoptosis in RAW264.7 macrophages, similar to the effects of 4-phenylbutyric acid (PBA, an ER stress inhibitor). In addition, consistent with the effect exerted by PBA, AS-Ⅳ inhibited oxLDL-triggered ER stress activation by decreasing the level of inositol-requiring enzyme1 phosphorylation and transcription factor 6 nuclear translocation and upregulating the protein and mRNA expression of glucose-regulated protein 78 (GPR78) and C/EBP homologous protein (CHOP). As expected, autophagy activation was induced by AS-IV, evidenced by increased expression of microtubule-associated protein 1 light chain 3-Ⅱ (LC3-Ⅱ), autophagy-related gene 5, and beclin-1 in macrophages. Furthermore, after pretreatment with 3-methyladenine and beclin-1 small interfering RNA, the inhibitory role played by AS-Ⅳ in oxLDL-induced ER stress-CHOP-mediated macrophage apoptosis was weakened, while its inhibitory effect was further enhanced by rapamycin pretreatment. Moreover, administration of AS-Ⅳ or rapamycin to Apoe-/- mice upregulated LC3-Ⅱ expression and collagen content but decreased CHOP expression, macrophage apoptosis, and lipid areas. Overall, by promoting autophagy, AS-Ⅳ effectively protects macrophages from oxLDL-induced apoptosis mediated by ER stress-CHOP, which may reinforce the stability of atherosclerotic plaques.
Collapse
Affiliation(s)
- Hua Tian
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China; Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China.
| | - Tong Wang
- College of Nursing, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Yumei Zhang
- Department of Follow-up Visit, Binzhou People's Hospital Affiliated to Shandong First Medical University & Shandong Academy of Medical Sciences, Binzhou, 256610, Shandong, China
| | - Tianqi Pan
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Shutong Yao
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Huayun Yu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Ke Ma
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Shijun Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
| |
Collapse
|
2
|
Tian H, Zhang Z, Han X, Pan T, Tao G, Jiao P, Zhai L, Yang L, Wang X, Yao Y, Qin S, Yao S. D4F alleviates the C/EBP homologous protein-mediated apoptosis in glycated high-density lipoprotein-treated macrophages by facilitating autophagy. Exp Biol Med (Maywood) 2021; 246:2595-2609. [PMID: 34525858 DOI: 10.1177/15353702211045323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The present study aimed to investigate the role of D4F, an apolipoprotein A-I mimetic peptide, in macrophage apoptosis induced by the glycated high-density lipoprotein (gly-HDL)-induced endoplasmic reticulum (ER) stress C/EBP homologous protein (CHOP) pathway, and unravel the regulatory role of autophagy in this process. Our results revealed that except for suppressing the accumulation of lipids within RAW264.7 macrophages caused by gly-HDL, D4F inhibited gly-HDL-induced decrease in the cell viability and increase in lactate dehydrogenase leakage and cell apoptosis, which were similar to 4-phenylbutyric acid (PBA, an ER stress inhibitor). Besides, similar to PBA, D4F inhibited gly-HDL-induced ER stress response activation evaluated through the decreased PERK and eIF2α phosphorylation, together with reduced ATF6 nuclear translocation as well as the downregulation of GRP78 and CHOP. Interestingly, D4F facilitated gly-HDL-triggered activation of autophagy, measured as elevated levels of beclin-1, LC3-II, and ATG5 expressions in macrophages. Furthermore, the inhibition effect of D4F on gly-HDL-induced ER stress-CHOP-induced apoptosis of macrophages was restrained after beclin-1 siRNA and 3-methyladenine (3-MA, an inhibitor of autophagy) treatments, while this effect was further reinforced after rapamycin (Rapa, an inducer of autophagy) treatment. Furthermore, administering D4F or Rapa to T2DM mice upregulated LC3-II and attenuated CHOP expression, cell apoptosis, and atherosclerotic lesions. However, the opposite results were obtained when 3-MA was administered to these mice. These results support that D4F effectively protects macrophages against gly-HDL-induced ER stress-CHOP-mediated apoptosis by promoting autophagy.
Collapse
Affiliation(s)
- Hua Tian
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China.,College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zhaoqiang Zhang
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Xiaoyan Han
- College of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Tianqi Pan
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Geru Tao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Peng Jiao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Lei Zhai
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Libo Yang
- Department of Endocrinology, Central Hospital of Taian, Taian 271000, China
| | - Xiaoxu Wang
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Yilin Yao
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Shutong Yao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China.,College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| |
Collapse
|
3
|
Che X, Xiao Q, Song W, Zhang H, Sun B, Geng N, Tao Z, Shao Q, Pu J. Protective Functions of Liver X Receptor α in Established Vulnerable Plaques: Involvement of Regulating Endoplasmic Reticulum-Mediated Macrophage Apoptosis and Efferocytosis. J Am Heart Assoc 2021; 10:e018455. [PMID: 33969692 PMCID: PMC8200716 DOI: 10.1161/jaha.120.018455] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Liver X receptor (LXR) belongs to the metabolic nuclear receptor superfamily, which plays a critical regulatory role in vascular physiology/pathology. However, effects of systemic LXR activation on established vulnerable plaques and the potential isotype‐specific role involved remain unclear. Methods and Results The 8‐week‐old male apolipoprotein E−/− mice went through carotid branch ligation and renal artery constriction, combined with a high‐fat diet. Plaques in the left carotid artery acquired vulnerable features 4 weeks later, confirmed by magnetic resonance imaging scans and histological analysis. From that time on, mice were injected intraperitoneally daily with PBS or GW3965 (10 mg/kg per day) for an additional 4 weeks. Treatment with LXR agonists reduced the lesion volume by 52.61%, compared with the vehicle group. More important, a profile of less intraplaque hemorrhage detection and necrotic core formation was found. These actions collectively attenuated the incidence of plaque rupture. Mechanistically, reduced lesional apoptosis, enhanced efferocytosis, and alleviated endoplasmic reticulum stress are involved in the process. Furthermore, genetic ablation of LXRα, but not LXRβ, blunted the protective effects of LXR on the endoplasmic reticulum stress–elicited C/EBP‐homologous protein pathway in peritoneal macrophages. In concert with the LXRα‐predominant role in vitro, activated LXR failed to stabilize vulnerable plaques and correct the acquired cellular anomalies in LXRα−/− apolipoprotein E−/− mice. Conclusions Our results revealed that LXRα mediates the capacity of LXR activation to stabilize vulnerable plaques and prevent plaque rupture via amelioration of macrophage endoplasmic reticulum stress, lesional apoptosis, and defective efferocytosis. These findings might expand the application scenarios of LXR therapeutics for atherosclerosis.
Collapse
Affiliation(s)
- Xinyu Che
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Qingqing Xiao
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Wei Song
- Cardiovascular Department of Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Shanghai China
| | - Hengyuan Zhang
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Beibei Sun
- Department of Radiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Na Geng
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Zhenyu Tao
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Qin Shao
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Jun Pu
- Department of Cardiology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai China
| |
Collapse
|
4
|
Intermedin 1-53 attenuates atherosclerotic plaque vulnerability by inhibiting CHOP-mediated apoptosis and inflammasome in macrophages. Cell Death Dis 2021; 12:436. [PMID: 33934111 PMCID: PMC8088440 DOI: 10.1038/s41419-021-03712-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/19/2022]
Abstract
Atherosclerotic plaque vulnerability and rupture increase the risk of acute coronary syndromes. Advanced lesion macrophage apoptosis plays important role in the rupture of atherosclerotic plaque, and endoplasmic reticulum stress (ERS) has been proved to be a key mechanism of macrophage apoptosis. Intermedin (IMD) is a regulator of ERS. Here, we investigated whether IMD enhances atherosclerotic plaque stability by inhibiting ERS-CHOP-mediated apoptosis and subsequent inflammasome in macrophages. We studied the effects of IMD on features of plaque vulnerability in hyperlipemia apolipoprotein E-deficient (ApoE−/−) mice. Six-week IMD1-53 infusion significantly reduced atherosclerotic lesion size. Of note, IMD1-53 lowered lesion macrophage content and necrotic core size and increased fibrous cap thickness and vascular smooth muscle cells (VSMCs) content thus reducing overall plaque vulnerability. Immunohistochemical analysis indicated that IMD1-53 administration prevented ERS activation in aortic lesions of ApoE−/− mice, which was further confirmed in oxidized low-density lipoproteins (ox-LDL) induced macrophages. Similar to IMD, taurine (Tau), a non-selective ERS inhibitor significantly reduced atherosclerotic lesion size and plaque vulnerability. Moreover, C/EBP-homologous protein (CHOP), a pro-apoptosis transcription factor involved in ERS, was significantly increased in advanced lesion macrophages, and deficiency of CHOP stabilized atherosclerotic plaques in AopE−/− mice. IMD1-53 decreased CHOP level and apoptosis in vivo and in macrophages treated with ox-LDL. In addition, IMD1-53 infusion ameliorated NLRP3 inflammasome and subsequent proinflammatory cytokines in vivo and in vitro. IMD may attenuate the progression of atherosclerotic lesions and plaque vulnerability by inhibiting ERS-CHOP-mediated macrophage apoptosis, and subsequent NLRP3 triggered inflammation. The inhibitory effect of IMD on ERS-induced macrophages apoptosis was probably mediated by blocking CHOP activation.
Collapse
|
5
|
Yang D, Wang Q, Wei G, Wu J, Zhu YC, Zhu Q, Ni T, Liu X, Zhu YZ. Smyd3-PARP16 axis accelerates unfolded protein response and vascular aging. Aging (Albany NY) 2020; 12:21423-21445. [PMID: 33144524 PMCID: PMC7695420 DOI: 10.18632/aging.103895] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022]
Abstract
Vascular endothelial cell senescence and endoplasmic reticulum (ER) stress induced unfolded protein response (UPR) are two critical contributors to individual aging. However, whether these two biological events have crosstalk and are controlled by shared upstream regulators are largely unknown. Here, we found PARP16, a member of the Poly (ADP-ribose) polymerases family that tail-anchored ER transmembrane, was upregulated in angiotensin II (Ang II)-induced vascular aging and promoted UPR. Further, PARP16 was epigenetically upregulated by Smyd3, a histone H3 lysine 4 methyltransferase that bound to the promotor region of Parp16 gene and increased H3K4me3 level to activate its host gene’s transcription. Intervention of either Smyd3 or PARP16 ameliorated vascular aging associated phenotypes in both cell and mice models. This study identified Smyd3-PARP16 as a novel signal axis in regulating UPR and endothelial senescence, and targeting this axis has implications in preventing vascular aging and related diseases.
Collapse
Affiliation(s)
- Di Yang
- Department of Pharmacology, Human Phenome Institute, School of Pharmacy, Fudan University, Shanghai, P.R. China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, P.R. China.,Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, P.R. China
| | - Qing Wang
- Department of Pharmacology, Human Phenome Institute, School of Pharmacy, Fudan University, Shanghai, P.R. China
| | - Gang Wei
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Jiaxue Wu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Yi Chun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, P.R. China
| | - Qing Zhu
- School of Pharmacy, Nantong University, Nantong, P.R. China
| | - Ting Ni
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Xinhua Liu
- Department of Pharmacology, Human Phenome Institute, School of Pharmacy, Fudan University, Shanghai, P.R. China
| | - Yi Zhun Zhu
- Department of Pharmacology, Human Phenome Institute, School of Pharmacy, Fudan University, Shanghai, P.R. China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, P.R. China.,Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, P.R. China
| |
Collapse
|
6
|
Role of Endoplasmic Reticulum Stress in Atherosclerosis and Its Potential as a Therapeutic Target. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9270107. [PMID: 32963706 PMCID: PMC7499294 DOI: 10.1155/2020/9270107] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/29/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022]
Abstract
Endoplasmic reticulum (ER) stress is closely associated with atherosclerosis and related cardiovascular diseases (CVDs). It occurs due to various pathological factors that interfere with ER homeostasis, resulting in the accumulation of unfolded or misfolded proteins in the ER lumen, thereby causing ER dysfunction. Here, we discuss the role of ER stress in different types of cells in atherosclerotic lesions. This discussion includes the activation of apoptotic and inflammatory pathways induced by prolonged ER stress, especially in advanced lesional macrophages and endothelial cells (ECs), as well as common atherosclerosis-related ER stressors in different lesional cells, which all contribute to the clinical progression of atherosclerosis. In view of the important role of ER stress and the unfolded protein response (UPR) signaling pathways in atherosclerosis and CVDs, targeting these processes to reduce ER stress may be a novel therapeutic strategy.
Collapse
|
7
|
Vaisar T, Hu JH, Airhart N, Fox K, Heinecke J, Nicosia RF, Kohler T, Potter ZE, Simon GM, Dix MM, Cravatt BF, Gharib SA, Dichek DA. Parallel Murine and Human Plaque Proteomics Reveals Pathways of Plaque Rupture. Circ Res 2020; 127:997-1022. [PMID: 32762496 PMCID: PMC7508285 DOI: 10.1161/circresaha.120.317295] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
RATIONALE Plaque rupture is the proximate cause of most myocardial infarctions and many strokes. However, the molecular mechanisms that precipitate plaque rupture are unknown. OBJECTIVE By applying proteomic and bioinformatic approaches in mouse models of protease-induced plaque rupture and in ruptured human plaques, we aimed to illuminate biochemical pathways through which proteolysis causes plaque rupture and identify substrates that are cleaved in ruptured plaques. METHODS AND RESULTS We performed shotgun proteomics analyses of aortas of transgenic mice with macrophage-specific overexpression of urokinase (SR-uPA+/0 mice) and of SR-uPA+/0 bone marrow transplant recipients, and we used bioinformatic tools to evaluate protein abundance and functional category enrichment in these aortas. In parallel, we performed shotgun proteomics and bioinformatics studies on extracts of ruptured and stable areas of freshly harvested human carotid plaques. We also applied a separate protein-analysis method (protein topography and migration analysis platform) to attempt to identify substrates and proteolytic fragments in mouse and human plaque extracts. Approximately 10% of extracted aortic proteins were reproducibly altered in SR-uPA+/0 aortas. Proteases, inflammatory signaling molecules, as well as proteins involved with cell adhesion, the cytoskeleton, and apoptosis, were increased. ECM (Extracellular matrix) proteins, including basement-membrane proteins, were decreased. Approximately 40% of proteins were altered in ruptured versus stable areas of human carotid plaques, including many of the same functional categories that were altered in SR-uPA+/0 aortas. Collagens were minimally altered in SR-uPA+/0 aortas and ruptured human plaques; however, several basement-membrane proteins were reduced in both SR-uPA+/0 aortas and ruptured human plaques. Protein topography and migration analysis platform did not detect robust increases in proteolytic fragments of ECM proteins in either setting. CONCLUSIONS Parallel studies of SR-uPA+/0 mouse aortas and human plaques identify mechanisms that connect proteolysis with plaque rupture, including inflammation, basement-membrane protein loss, and apoptosis. Basement-membrane protein loss is a prominent feature of ruptured human plaques, suggesting a major role for basement-membrane proteins in maintaining plaque stability.
Collapse
Affiliation(s)
- Tomáš Vaisar
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle
| | - Jie H Hu
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle
| | - Nathan Airhart
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle
| | - Kate Fox
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle
| | - Jay Heinecke
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle
| | - Roberto F Nicosia
- Departments of Pathology and Laboratory Medicine (D.A.D., R.F.N.), University of Washington, Seattle.,Departments of Pathology and Laboratory Medicine (R.F.N.), VA Puget Sound Health Care System, Seattle, WA
| | - Ted Kohler
- Departments of Surgery (T.K.), University of Washington, Seattle.,Departments of Surgery (T.K.), VA Puget Sound Health Care System, Seattle, WA
| | - Zachary E Potter
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA (Z.E.P., M.M.D., B.F.C.)
| | | | - Melissa M Dix
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA (Z.E.P., M.M.D., B.F.C.)
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA (Z.E.P., M.M.D., B.F.C.)
| | - Sina A Gharib
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle
| | - David A Dichek
- Departments of Medicine (T.V., J.H.H., N.A., K.F., J.H., S.A.G., D.A.D.), University of Washington, Seattle.,Departments of Pathology and Laboratory Medicine (D.A.D., R.F.N.), University of Washington, Seattle
| |
Collapse
|
8
|
Lynn EG, Lhoták Š, Lebeau P, Byun JH, Chen J, Platko K, Shi C, O'Brien RE, Austin RC. 4‐Phenylbutyrate protects against atherosclerotic lesion growth by increasing the expression of HSP25 in macrophages and in the circulation of
Apoe
−/−
mice. FASEB J 2019; 33:8406-8422. [DOI: 10.1096/fj.201802293rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Edward G. Lynn
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| | - Šárka Lhoták
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| | - Paul Lebeau
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| | - Jae Hyun Byun
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| | - Jack Chen
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| | - Khrystyna Platko
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| | - Chunhua Shi
- Department of Cardiac SciencesLibin Cardiovascular Institute of AlbertaCumming School of MedicineUniversity of Calgary Calgary Alberta Canada
| | - R. Edward O'Brien
- Department of Cardiac SciencesLibin Cardiovascular Institute of AlbertaCumming School of MedicineUniversity of Calgary Calgary Alberta Canada
| | - Richard C. Austin
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| |
Collapse
|
9
|
Tian H, Li Y, Kang P, Wang Z, Yue F, Jiao P, Yang N, Qin S, Yao S. Endoplasmic reticulum stress-dependent autophagy inhibits glycated high-density lipoprotein-induced macrophage apoptosis by inhibiting CHOP pathway. J Cell Mol Med 2019; 23:2954-2969. [PMID: 30746902 PMCID: PMC6433656 DOI: 10.1111/jcmm.14203] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/07/2018] [Accepted: 01/13/2019] [Indexed: 12/15/2022] Open
Abstract
This study was designed to explore the inductive effect of glycated high‐density lipoprotein (gly‐HDL) on endoplasmic reticulum (ER) stress‐C/EBP homologous protein (CHOP)‐mediated macrophage apoptosis and its relationship with autophagy. Our results showed that gly‐HDL caused macrophage apoptosis with concomitant activation of ER stress pathway, including nuclear translocation of activating transcription factor 6, phosphorylation of protein kinase‐like ER kinase (PERK) and eukaryotic translation initiation factor 2α, and CHOP up‐regulation, which were inhibited by 4‐phenylbutyric acid (PBA, an ER stress inhibitor) and the gene silencing of PERK and CHOP. Similar data were obtained from macrophages treated by HDL isolated from diabetic patients. Gly‐HDL induced macrophage autophagy as assessed by up‐regulation of beclin‐1, autophagy‐related gene 5 and microtubule‐associated protein one light chain 3‐II, which were depressed by PBA and PERK siRNA. Gly‐HDL‐induced apoptosis, PERK phosphorylation and CHOP up‐regulation were suppressed by rapamycin (an autophagy inducer), whereas aggravated by 3‐methyladenine (an autophagy inhibitor) and beclin‐1 siRNA. Administration of diabetic apoE−/− mice with rapamycin attenuated MOMA‐2 and CHOP up‐regulation and apoptosis in atherosclerotic lesions. These data indicate that gly‐HDL may induce macrophage apoptosis through activating ER stress‐CHOP pathway and ER stress mediates gly‐HDL‐induced autophagy, which in turn protects macrophages against apoptosis by alleviating CHOP pathway.
Collapse
Affiliation(s)
- Hua Tian
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Yanyan Li
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Panpan Kang
- Affiliated hospital of Chengde Medical University, Chengde Medical University, Chengde, China
| | - Zhichao Wang
- College of Nursing, Taishan Medical University, Taian, China
| | - Feng Yue
- Department of Endocrinology, Central Hospital of Taian, Taian, China
| | - Peng Jiao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Nana Yang
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Shutong Yao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Taian, China.,College of Basic Medical Sciences, Taishan Medical University, Taian, China
| |
Collapse
|
10
|
Jiang Q, Liu H, Wang S, Wang J, Tang Y, He Z, Wu F, Huang Z, Cong X, Ding R, Liang C. Circadian locomotor output cycles kaput accelerates atherosclerotic plaque formation by upregulating plasminogen activator inhibitor-1 expression. Acta Biochim Biophys Sin (Shanghai) 2018; 50:869-879. [PMID: 30124738 DOI: 10.1093/abbs/gmy087] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Indexed: 11/13/2022] Open
Abstract
To explore the association between clock circadian regulator circadian locomotor output cycles kaput gene (CLOCK) and the forming of atherosclerotic plaques and its underlying mechanisms, mouse aortic endothelial cells (MAECs) and atherosclerosis (AS) mouse model were recruited for our study. The apoE gene knockout mouse was used as the model of AS and we accelerated the formation of unstable plaques through the combination of carotid artery ligation and high-fat (HF) diet administration (0.2% cholesterol, 20% fat). The mRNA and protein expressions of CLOCK in peripheral blood monouclear cells of acute coronary syndrome (ACS) patients or mouse AS model were detected by qPCR, western blot analysis and immunohistochemical staining. The number of adherent cells and atherosclerotic plaques was counted to assess the effects of CLOCK on the progression of ACS, and adherence-associated genes, such as vascular cell adhesion molecule (VCAM)-1, C-C motif chemokine ligand 2 (CCL-2), and CCL-5. The results showed that CLOCK expression was significantly increased in both ACS patients and AS mouse model. The levels of CLOCK, leukemia inhibitory factor (LIF), intercellular adhesion molecule 1 (ICAM-1), perilipin 2 (ADFP), nuclear factor kappa B (NF-κB), and plasminogen activator inhibitor-1 (PAI-1), as well as the number of atherosclerotic plaques were elevated in the AS mouse model, as compared with the control group. Chromatin immunoprecipitation assay showed that CLOCK bound directly to the promoter of PAI-1 gene and CLOCK could positively regulate the expressions of LIF, ICAM-1, ADFP, NF-κB, and PAI-1. Reduction of CLOCK expression would decrease the expressions of VCAM-1, CCL-2, and CCL-5, and the number of adherent cells and atherosclerotic plaques, but these effects were neutralized when PAI-1 was simultaneously overexpressed in either mouse model or MAECs. Our results demonstrate that CLOCK overexpression triggers the formation of atherosclerotic plaques by directly upregulating PAI-1 expression.
Collapse
Affiliation(s)
- Qixia Jiang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hua Liu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Shengyun Wang
- Department of Emergency and Critical Care Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jiamei Wang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yehua Tang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhiqing He
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Feng Wu
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhigang Huang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiaoliang Cong
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ru Ding
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Chun Liang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
11
|
Woo M, Kim M, Noh JS, Park CH, Song YO. Kimchi attenuates fatty streak formation in the aorta of low-density lipoprotein receptor knockout mice via inhibition of endoplasmic reticulum stress and apoptosis. Nutr Res Pract 2017; 11:445-451. [PMID: 29209454 PMCID: PMC5712494 DOI: 10.4162/nrp.2017.11.6.445] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/22/2017] [Accepted: 08/31/2017] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND/OBJECTIVES Endoplasmic reticulum (ER) stress is positively associated with atherosclerosis via elevating macrophage cell death and plaque formation, in which oxidative stress plays a pivotal role. Antioxidative, lipid-lowering, and anti-atherogenic effects of kimchi, a Korean fermented vegetable, have been established, wherein capsaicin, ascorbic acid, quercetin, 3-(4'-hydroxyl-3',5'-dimethoxyphenyl)propionic acid, and lactic acids were identified. In this study, mechanisms of action of kimchi methanol extracts (KME) on fatty streak formation via suppression of ER stress and apoptosis in aorta were examined in low-density lipoprotein receptor knockout mice. MATERIALS AND METHODS Mice fed a high cholesterol diet with an oral administration of KME (KME group, 200 mg·kg-bw−1·day−1) or distilled water (control group) for 8 weeks (n = 20 for group). Plasma lipid and oxidative stress levels were evaluated. Protein expression was measured by western blot assay. Fatty streak lesion size and the degree of apoptosis were examined in the aorta. RESULTS Compared to the control group, in the KME group, plasma lipids levels were decreased and oxidative stress was alleviated (P < 0.05). Protein expression levels of nuclear factor (erythroid-derived 2)-like 2-mediated antioxidants in aorta were increased whereas those for ER stress markers, glucose regulated protein 78, phospho-protein kinase RNA-like ER kinase, phospho-eukaryotic initiation factor 2 subunit α, X-box binding protein 1, and C/EBP homologous protein were decreased in the KME group (P < 0.05). Moreover, apoptosis was suppressed via downregulation of phospho-c-Jun N-terminal kinase, bcl-2-associated X protein, caspases-9, and -3 with a concomitant upregulation of anti-apoptotic protein, B-cell lymphoma 2 (P < 0.05). Fatty streak lesion size was reduced and the degree of apoptosis was less severe in the KME group (P < 0.05). CONCLUSIONS In conclusion, antioxidant activity of KME might prevent fatty streak formation through, in part, inhibition of ER stress and apoptosis in aortic sinus where macrophages are harbored.
Collapse
Affiliation(s)
- Minji Woo
- Department of Food Science and Nutrition, Kimchi Research Institute, Pusan National University, 2, Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 46241, Korea
| | - Mijeong Kim
- Department of Food Science and Nutrition, Kimchi Research Institute, Pusan National University, 2, Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 46241, Korea
| | - Jeong Sook Noh
- Department of Food Science and Nutrition, Tongmyong University, Busan 48520, Korea
| | - Chan Hum Park
- Department of Medicinal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Eumseong 55365, Korea
| | - Yeong Ok Song
- Department of Food Science and Nutrition, Kimchi Research Institute, Pusan National University, 2, Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 46241, Korea
| |
Collapse
|
12
|
Grechowa I, Horke S, Wallrath A, Vahl C, Dorweiler B. Human neutrophil elastase induces endothelial cell apoptosis by activating the PERK‐CHOP branch of the unfolded protein response. FASEB J 2017; 31:3868-3881. [DOI: 10.1096/fj.201700012r] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 04/24/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Irina Grechowa
- Division of Vascular SurgeryDepartment of Cardiothoracic and Vascular SurgeryUniversity Medical CenterJohannes‐Gutenberg University Mainz Germany
- Institute of PharmacologyUniversity Medical CenterJohannes‐Gutenberg University Mainz Germany
| | - Sven Horke
- Institute of PharmacologyUniversity Medical CenterJohannes‐Gutenberg University Mainz Germany
- Center for Thrombosis and HemostasisUniversity Medical CenterJohannes‐Gutenberg University Mainz Germany
| | - Anja Wallrath
- Division of Vascular SurgeryDepartment of Cardiothoracic and Vascular SurgeryUniversity Medical CenterJohannes‐Gutenberg University Mainz Germany
| | - Christian‐Friedrich Vahl
- Division of Vascular SurgeryDepartment of Cardiothoracic and Vascular SurgeryUniversity Medical CenterJohannes‐Gutenberg University Mainz Germany
| | - Bernhard Dorweiler
- Division of Vascular SurgeryDepartment of Cardiothoracic and Vascular SurgeryUniversity Medical CenterJohannes‐Gutenberg University Mainz Germany
| |
Collapse
|
13
|
Hoseini Z, Sepahvand F, Rashidi B, Sahebkar A, Masoudifar A, Mirzaei H. NLRP3 inflammasome: Its regulation and involvement in atherosclerosis. J Cell Physiol 2017; 233:2116-2132. [DOI: 10.1002/jcp.25930] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 03/22/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Zahra Hoseini
- Faculty of Medicine, Students Research Center; Isfahan University of Medical Sciences; Isfahan Iran
| | - Fatemeh Sepahvand
- Faculty of Medicine, Students Research Center; Isfahan University of Medical Sciences; Isfahan Iran
| | - Bahman Rashidi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine; Isfahan University of Medical Sciences; Isfahan Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| | - Aria Masoudifar
- Department of Molecular Biotechnology, Cell Science Research Center; Royan Institute for Biotechnology; ACECR; Isfahan Iran
| | - Hamed Mirzaei
- Department of Medical Biotechnology, School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
14
|
Endoplasmic reticulum stress inhibition reduces hypertension through the preservation of resistance blood vessel structure and function. J Hypertens 2016; 34:1556-69. [DOI: 10.1097/hjh.0000000000000943] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
15
|
de Pedro-Cuesta J, Martínez-Martín P, Rábano A, Ruiz-Tovar M, Alcalde-Cabero E, Calero M. Etiologic Framework for the Study of Neurodegenerative Disorders as Well as Vascular and Metabolic Comorbidities on the Grounds of Shared Epidemiologic and Biologic Features. Front Aging Neurosci 2016; 8:138. [PMID: 27378910 PMCID: PMC4904010 DOI: 10.3389/fnagi.2016.00138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/27/2016] [Indexed: 12/12/2022] Open
Abstract
Background: During the last two decades, protein aggregation at all organismal levels, from viruses to humans, has emerged from a neglected area of protein science to become a central issue in biology and biomedicine. This article constitutes a risk-based review aimed at supporting an etiologic scenario of selected, sporadic, protein-associated, i.e., conformational, neurodegenerative disorders (NDDs), and their vascular- and metabolic-associated ailments. Methods: A rationale is adopted, to incorporate selected clinical data and results from animal-model research, complementing epidemiologic evidences reported in two prior articles. Findings: Theory is formulated assuming an underlying conformational transmission mechanism, mediated either by horizontal transfer of mammalian genes coding for specific aggregation-prone proteins, or by xeno-templating between bacterial and host proteins. We build a few population-based and experimentally-testable hypotheses focusing on: (1) non-disposable surgical instruments for sporadic Creutzfeldt-Jakob disease (sCJD) and other rapid progressive neurodegenerative dementia (sRPNDd), multiple system atrophy (MSA), and motor neuron disease (MND); and (2) specific bacterial infections such as B. pertussis and E. coli for all forms, but particularly for late-life sporadic conformational, NDDs, type 2 diabetes mellitus (T2DM), and atherosclerosis where natural protein fibrils present in such organisms as a result of adaptation to the human host induce prion-like mechanisms. Conclusion: Implications for cohort alignment and experimental animal research are discussed and research lines proposed.
Collapse
Affiliation(s)
- Jesús de Pedro-Cuesta
- Department of Applied Epidemiology, National Center for Epidemiology, Carlos III Institute of HealthMadrid, Spain; Consortium for Biomedical Research in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos IIIMadrid, Spain
| | - Pablo Martínez-Martín
- Department of Applied Epidemiology, National Center for Epidemiology, Carlos III Institute of HealthMadrid, Spain; Consortium for Biomedical Research in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos IIIMadrid, Spain
| | - Alberto Rábano
- Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center Madrid, Spain
| | - María Ruiz-Tovar
- Department of Applied Epidemiology, National Center for Epidemiology, Carlos III Institute of HealthMadrid, Spain; Consortium for Biomedical Research in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos IIIMadrid, Spain
| | - Enrique Alcalde-Cabero
- Department of Applied Epidemiology, National Center for Epidemiology, Carlos III Institute of HealthMadrid, Spain; Consortium for Biomedical Research in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos IIIMadrid, Spain
| | - Miguel Calero
- Consortium for Biomedical Research in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos IIIMadrid, Spain; Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer CenterMadrid, Spain; Chronic Disease Programme, Carlos III Institute of Health, MajadahondaMadrid, Spain
| |
Collapse
|
16
|
Liu MQ, Chen Z, Chen LX. Endoplasmic reticulum stress: a novel mechanism and therapeutic target for cardiovascular diseases. Acta Pharmacol Sin 2016; 37:425-43. [PMID: 26838072 DOI: 10.1038/aps.2015.145] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/23/2015] [Indexed: 12/13/2022]
Abstract
Endoplasmic reticulum is a principal organelle responsible for folding, post-translational modifications and transport of secretory, luminal and membrane proteins, thus palys an important rale in maintaining cellular homeostasis. Endoplasmic reticulum stress (ERS) is a condition that is accelerated by accumulation of unfolded/misfolded proteins after endoplasmic reticulum environment disturbance, triggered by a variety of physiological and pathological factors, such as nutrient deprivation, altered glycosylation, calcium depletion, oxidative stress, DNA damage and energy disturbance, etc. ERS may initiate the unfolded protein response (UPR) to restore cellular homeostasis or lead to apoptosis. Numerous studies have clarified the link between ERS and cardiovascular diseases. This review focuses on ERS-associated molecular mechanisms that participate in physiological and pathophysiological processes of heart and blood vessels. In addition, a number of drugs that regulate ERS was introduced, which may be used to treat cardiovascular diseases. This review may open new avenues for studying the pathogenesis of cardiovascular diseases and discovering novel drugs targeting ERS.
Collapse
|
17
|
Low dose tunicamycin enhances atherosclerotic plaque stability by inducing autophagy. Biochem Pharmacol 2016; 100:51-60. [DOI: 10.1016/j.bcp.2015.11.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 11/18/2015] [Indexed: 11/19/2022]
|
18
|
Acyl-CoA:cholesterol acyltransferase 1 mediates liver fibrosis by regulating free cholesterol accumulation in hepatic stellate cells. J Hepatol 2014; 61:98-106. [PMID: 24657401 DOI: 10.1016/j.jhep.2014.03.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/22/2014] [Accepted: 03/10/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Acyl-coenzyme A: cholesterol acyltransferase (ACAT) catalyzes the conversion of free cholesterol (FC) to cholesterol ester, which prevents excess accumulation of FC. We recently found that FC accumulation in hepatic stellate cells (HSCs) plays a role in progression of liver fibrosis, but the effect of ACAT1 on liver fibrosis has not been clarified. In this study, we aimed to define the role of ACAT1 in the pathogenesis of liver fibrosis. METHODS ACAT1-deficient and wild-type mice, or Toll-like receptor 4 (TLR4)(-/-)ACAT1(+/+) and TLR4(-/-)ACAT1(-/-) mice were subjected to bile duct ligation (BDL) for 3 weeks or were given carbon tetrachloride (CCl4) for 4 weeks to induce liver fibrosis. RESULTS ACAT1 was the major isozyme in mice and human primary HSCs, and ACAT2 was the major isozyme in mouse primary hepatocytes and Kupffer cells. ACAT1 deficiency significantly exaggerated liver fibrosis in the mouse models of liver fibrosis, without affecting the degree of hepatocellular injury or liver inflammation, including hepatocyte apoptosis or Kupffer cell activation. ACAT1 deficiency significantly increased FC levels in HSCs, augmenting TLR4 protein and downregulating expression of transforming growth factor-β (TGFβ) pseudoreceptor Bambi (bone morphogenetic protein and activin membrane-bound inhibitor), leading to sensitization of HSCs to TGFβ activation. Exacerbation of liver fibrosis by ACAT1 deficiency was dependent on FC accumulation-induced enhancement of TLR4 signaling. CONCLUSIONS ACAT1 deficiency exaggerates liver fibrosis mainly through enhanced FC accumulation in HSCs. Regulation of ACAT1 activities in HSCs could be a target for treatment of liver fibrosis.
Collapse
|
19
|
Panax quinquefolium saponin attenuates ventricular remodeling after acute myocardial infarction by inhibiting chop-mediated apoptosis. Shock 2014; 40:339-44. [PMID: 23856922 DOI: 10.1097/shk.0b013e3182a3f9e5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Panax quinquefolium saponin (PQS) alleviates hypoxia-reoxygenation injury of cardiomyocytes in vitro by inhibiting excessive endoplasmic reticulum stress (ERS)-related apoptosis. We hypothesized that inhibition of excessive ERS-related apoptosis contributes to cardioprotection in ventricular remodeling after acute myocardial infarction (AMI). Sprague-Dawley rats subjected to AMI were randomly treated with water, PQS (50 mg/kg per day, 100 mg/kg per day, or 200 mg/kg per day), or taurine (300 mg/kg per day), an ERS inhibitor, for 4 weeks. Left ventricular (LV) fractional shortening, ejection fraction, and structure were then evaluated using echocardiography. Myocardial infarct size was measured by Evans blue and 2,3,5-triphenyhetrazolium chloride staining. The hydroxyproline level was assayed using the colorimetric method. Cardiomyocyte apoptosis was detected using terminal deoxynucleotidyl transferase-mediated dUTP biotin nick end labeling. In addition, expression of ERS molecules in the noninfarcted myocardium was detected using Western blotting. We found that PQS treatment significantly reduced infarct size and LV dilation and improved LV ejection fraction and fractional shortening in rat hearts. Panax quinquefolium saponin treatment also decreased hydroxyproline level in noninfarcted myocardium. Panax quinquefolium saponin treatment significantly decreased expression of glucose regulating protein 78, calreticulin, C/EBP homologous protein (CHOP), and Bax protein, as well as increased Bcl-2 protein expression in noninfarcted myocardium. Panax quinquefolium saponin treatment (200 mg/kg per day) mimicked the results achieved from the taurine-treated rats. Expression of CHOP positively correlated with the apoptosis index of cardiomyocytes in the noninfarcted myocardium (r = 0.797, P < 0.01). Taken together, PQS treatment significantly improves AMI-induced LV remodeling, and this may be attributed to inhibiting CHOP-mediated ERS-related apoptosis.
Collapse
|
20
|
Rong JX, Blachford C, Feig JE, Bander I, Mayne J, Kusunoki J, Miller C, Davis M, Wilson M, Dehn S, Thorp E, Tabas I, Taubman MB, Rudel LL, Fisher EA. ACAT inhibition reduces the progression of preexisting, advanced atherosclerotic mouse lesions without plaque or systemic toxicity. Arterioscler Thromb Vasc Biol 2012; 33:4-12. [PMID: 23139293 DOI: 10.1161/atvbaha.112.252056] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Acyl-CoA:cholesterol acyltransferase (ACAT) converts cholesterol to cholesteryl esters in plaque foam cells. Complete deficiency of macrophage ACAT has been shown to increase atherosclerosis in hypercholesterolemic mice because of cytotoxicity from free cholesterol accumulation, whereas we previously showed that partial ACAT inhibition by Fujirebio compound F1394 decreased early atherosclerosis development. In this report, we tested F1394 effects on preestablished, advanced lesions of apolipoprotein-E-deficient mice. METHODS AND RESULTS Apolipoprotein-E-deficient mice on Western diet for 14 weeks developed advanced plaques, and were either euthanized (Baseline), or continued on Western diet with or without F1394 and euthanized after 14 more weeks. F1394 was not associated with systemic toxicity. Compared with the baseline group, lesion size progressed in both groups; however, F1394 significantly retarded plaque progression and reduced plaque macrophage, free and esterified cholesterol, and tissue factor contents compared with the untreated group. Apoptosis of plaque cells was not increased, consistent with the decrease in lesional free cholesterol. There was no increase in plaque necrosis and unimpaired efferocytosis (phagocytic clearance of apoptotic cells). The effects of F1394 were independent of changes in plasma cholesterol levels. CONCLUSIONS Partial ACAT inhibition by F1394 lowered plaque cholesterol content and had other antiatherogenic effects in advanced lesions in apolipoprotein-E-deficient mice without overt systemic or plaque toxicity, suggesting the continued potential of ACAT inhibition for the clinical treatment of atherosclerosis, in spite of recent trial data.
Collapse
Affiliation(s)
- James X Rong
- Marc and Ruti Bell Vascular Biology and Disease Research Program of the Leon H. Charney Division of Cardiology and the Department of Medicine (Cardiology), New York University School of Medicine, Smilow 7, 522 First Ave, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Carlisle RE, Heffernan A, Brimble E, Liu L, Jerome D, Collins CA, Mohammed-Ali Z, Margetts PJ, Austin RC, Dickhout JG. TDAG51 mediates epithelial-to-mesenchymal transition in human proximal tubular epithelium. Am J Physiol Renal Physiol 2012; 303:F467-81. [DOI: 10.1152/ajprenal.00481.2011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) contributes to renal fibrosis in chronic kidney disease. Endoplasmic reticulum (ER) stress, a feature of many forms of kidney disease, results from the accumulation of misfolded proteins in the ER and leads to the unfolded protein response (UPR). We hypothesized that ER stress mediates EMT in human renal proximal tubules. ER stress is induced by a variety of stressors differing in their mechanism of action, including tunicamycin, thapsigargin, and the calcineurin inhibitor cyclosporine A. These ER stressors increased the UPR markers GRP78, GRP94, and phospho-eIF2α in human proximal tubular cells. Thapsigargin and cyclosporine A also increased cytosolic Ca2+ concentration and T cell death-associated gene 51 (TDAG51) expression, whereas tunicamycin did not. Thapsigargin was also shown to increase levels of active transforming growth factor (TGF)-β1 in the media of cultured human proximal tubular cells. Thapsigargin induced cytoskeletal rearrangement, β-catenin nuclear translocation, and α-smooth muscle actin and vinculin expression in proximal tubular cells, indicating an EMT response. Subconfluent primary human proximal tubular cells were induced to undergo EMT by TGF-β1 treatment. In contrast, tunicamycin treatment did not produce an EMT response. Plasmid-mediated overexpression of TDAG51 resulted in cell shape change and β-catenin nuclear translocation. These results allowed us to develop a two-hit model of ER stress-induced EMT, where Ca2+ dysregulation-mediated TDAG51 upregulation primes the cell for mesenchymal transformation via Wnt signaling and then TGF-β1 activation leads to a complete EMT response. Thus the release of Ca2+ from ER stores mediates EMT in human proximal tubular epithelium via the induction of TDAG51.
Collapse
Affiliation(s)
- Rachel E. Carlisle
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Alana Heffernan
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Elise Brimble
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Limin Liu
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Danielle Jerome
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Celeste A. Collins
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Zahraa Mohammed-Ali
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Peter J. Margetts
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Richard C. Austin
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Jeffrey G. Dickhout
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| |
Collapse
|
22
|
Dickhout JG, Carlisle RE, Austin RC. Interrelationship between cardiac hypertrophy, heart failure, and chronic kidney disease: endoplasmic reticulum stress as a mediator of pathogenesis. Circ Res 2011; 108:629-42. [PMID: 21372294 DOI: 10.1161/circresaha.110.226803] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Synthesis of transmembrane and secretory proteins occurs within the endoplasmic reticulum (ER) and is extremely important in the normal functioning of both the heart and kidney. The dysregulation of protein synthesis/processing within the ER causes the accumulation of unfolded proteins, thereby leading to ER stress and the activation of the unfolded protein response. Sarcoplasmic reticulum/ER Ca2+ disequilibrium can lead to cardiac hypertrophy via cytosolic Ca2+ elevation and stimulation of the Ca2+/calmodulin, calcineurin, NF-AT3 pathway. Although cardiac hypertrophy may be initially adaptive, prolonged or severe ER stress resulting from the increased protein synthesis associated with cardiac hypertrophy can lead to apoptosis of cardiac myocytes and result in reduced cardiac output and chronic heart failure. The failing heart has a dramatic effect on renal function because of inadequate perfusion and stimulates the release of many neurohumoral factors that may lead to further ER stress within the heart, including angiotensin II and arginine-vasopressin. Renal failure attributable to proteinuria and uremia also induces ER stress within the kidney, which contributes to the transformation of tubular epithelial cells to a fibroblast-like phenotype, fibrosis, and tubular cell apoptosis, further diminishing renal function. As a consequence, cardiorenal syndrome may develop into a vicious circle with poor prognosis. New therapeutic modalities to alleviate ER stress through stimulation of the cytoprotective components of the unfolded protein response, including GRP78 upregulation and eukaryotic initiation factor 2α phosphorylation, may hold promise to reduce the high morbidity and mortality associated with cardiorenal syndrome.
Collapse
Affiliation(s)
- Jeffrey G Dickhout
- Department of Medicine, Division of Nephrology McMaster University and St Joseph's Healthcare Hamilton, 50 Charlton Ave, East Hamilton, Ontario, Canada, L8N 4A6
| | | | | |
Collapse
|
23
|
Zhou G, Isoe J, Day WA, Miesfeld RL. Alpha-COPI coatomer protein is required for rough endoplasmic reticulum whorl formation in mosquito midgut epithelial cells. PLoS One 2011; 6:e18150. [PMID: 21483820 PMCID: PMC3069061 DOI: 10.1371/journal.pone.0018150] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Accepted: 02/22/2011] [Indexed: 11/18/2022] Open
Abstract
Background One of the early events in midgut epithelial cells of Aedes aegypti mosquitoes is the dynamic reorganization of rough endoplasmic reticulum (RER) whorl structures coincident with the onset of blood meal digestion. Based on our previous studies showing that feeding on an amino acid meal induces TOR signaling in Ae. aegypti, we used proteomics and RNAi to functionally identify midgut epithelial cell proteins that contribute to RER whorl formation. Methodology/Principal Findings Adult female Ae. aegypti mosquitoes were maintained on sugar alone (unfed), or fed an amino acid meal, and then midgut epithelial cells were analyzed by electron microscopy and protein biochemistry. The size and number of RER whorls in midgut epithelial cells were found to decrease significantly after feeding, and several KDEL-containing proteins were shown to have altered expression levels. LC-MS/MS mass spectrometry was used to analyze midgut microsomal proteins isolated from unfed and amino acid fed mosquitoes, and of the 127 proteins identified, 8 were chosen as candidate whorl forming proteins. Three candidate proteins were COPI coatomer subunits (alpha, beta, beta'), all of which appeared to be present at higher levels in microsomal fractions from unfed mosquitoes. Using RNAi to knockdown alpha-COPI expression, electron microscopy revealed that both the size and number of RER whorls were dramatically reduced in unfed mosquitoes, and moreover, that extended regions of swollen RER were prevalent in fed mosquitoes. Lastly, while a deficiency in alpha-COPI had no effect on early trypsin protein synthesis or secretion 3 hr post blood meal (PBM), expression of late phase proteases at 24 hr PBM was completely blocked. Conclusions alpha-COPI was found to be required for the formation of RER whorls in midgut epithelial cells of unfed Aa. aegypti mosquitoes, as well as for the expression of late phase midgut proteases.
Collapse
Affiliation(s)
- Guoli Zhou
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona, United States of America
| | - Jun Isoe
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona, United States of America
| | - W. Antony Day
- Arizona Research Labs, The University of Arizona, Tucson, Arizona, United States of America
| | - Roger L. Miesfeld
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| |
Collapse
|
24
|
Immunohistochemical Detection of the Unfolded Protein Response in Atherosclerotic Plaques. Methods Enzymol 2011; 489:23-46. [DOI: 10.1016/b978-0-12-385116-1.00002-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
25
|
Dickhout JG, Lhoták Š, Hilditch BA, Basseri S, Colgan SM, Lynn EG, Carlisle RE, Zhou J, Sood SK, Ingram AJ, Austin RC. Induction of the unfolded protein response after monocyte to macrophage differentiation augments cell survival in early atherosclerotic lesions. FASEB J 2010; 25:576-89. [PMID: 20966213 DOI: 10.1096/fj.10-159319] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endoplasmic reticulum (ER) stress causes macrophage cell death within advanced atherosclerotic lesions, thereby contributing to necrotic core formation and increasing the risk of atherothrombotic disease. However, unlike in advanced lesions, the appearance of dead/apoptotic macrophages in early lesions is less prominent. Given that activation of the unfolded protein response (UPR) is detected in early lesion-resident macrophages and can enhance cell survival against ER stress, we investigated whether UPR activation occurs after monocyte to macrophage differentiation and confers a cytoprotective advantage to the macrophage. Human peripheral blood monocytes were treated with monocyte colony-stimulating factor to induce macrophage differentiation, as assessed by changes in ultrastructure and scavenger receptor expression. UPR markers, including GRP78, GRP94, and spliced XBP-1, were induced after macrophage differentiation and occurred after a significant increase in de novo protein synthesis. UPR activation after differentiation reduced macrophage cell death by ER stress-inducing agents. Further, GRP78 overexpression in macrophages was sufficient to reduce ER stress-induced cell death. Consistent with these in vitro findings, UPR activation was observed in viable lesion-resident macrophages from human carotid arteries and from the aortas of apoE(-/-) mice. However, no evidence of apoptosis was observed in early lesion-resident macrophages from the aortas of apoE(-/-) mice. Thus, our findings that UPR activation occurs during macrophage differentiation and is cytoprotective against ER stress-inducing agents suggest an important cellular mechanism for macrophage survival within early atherosclerotic lesions.
Collapse
Affiliation(s)
- Jeffrey G Dickhout
- Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, 50 Charlton Ave. East, Hamilton, ON, Canada L8N 4A6
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Engin F, Hotamisligil GS. Restoring endoplasmic reticulum function by chemical chaperones: an emerging therapeutic approach for metabolic diseases. Diabetes Obes Metab 2010; 12 Suppl 2:108-15. [PMID: 21029307 DOI: 10.1111/j.1463-1326.2010.01282.x] [Citation(s) in RCA: 199] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The endoplasmic reticulum (ER) is a eukaryotic organelle that plays important roles in protein synthesis, folding and trafficking, calcium homoeostasis and lipid and steroid synthesis. It is the major protein synthesis compartment for secreted, plasma membrane and organelle proteins. Perturbations of ER homeostasis such as the accumulation of unfolded or misfolded proteins cause ER stress. To alleviate this stress, ER triggers an evolutionarily conserved signalling cascade called the unfolded protein response (UPR). As an initial response, the UPR aims at adapting and restoring ER function by translational attenuation, upregulation of ER chaperones and degradation of unfolded proteins. However, if the ER function is severely impaired because of excessive or prolonged exposure to stress, then the inflicted cells may undergo programmed cell death. During ER stress, unstable or partially folded mutant proteins are prevented from trafficking to their proper subcellular localizations and usually rapidly degraded. The small molecules named chemical chaperones help to stabilize these mutant proteins and facilitate their folding and proper trafficking from the ER to their final destinations. Because increasing number of studies suggest that ER stress is involved in a number of disease pathogenesis including neurodegenerative diseases, cancer, obesity, diabetes and atherosclerosis, promoting ER folding capacity through chemical chaperones emerges as a novel therapeutic approach. In this review, we provide insight into the many important functions of chemical chaperones during ER stress, their impact on the ER-stress-related pathologies and their potential as a new drug targets, especially in the context of metabolic disorders.
Collapse
Affiliation(s)
- F Engin
- Department of Genetics & Complex Diseases, Harvard School of Public Health, Boston, MA 02115, USA
| | | |
Collapse
|
27
|
Santos CXC, Tanaka LY, Wosniak J, Laurindo FRM. Mechanisms and implications of reactive oxygen species generation during the unfolded protein response: roles of endoplasmic reticulum oxidoreductases, mitochondrial electron transport, and NADPH oxidase. Antioxid Redox Signal 2009; 11:2409-27. [PMID: 19388824 DOI: 10.1089/ars.2009.2625] [Citation(s) in RCA: 426] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cellular mechanisms governing redox homeostasis likely involve their integration with other stresses. Endoplasmic reticulum (ER) stress triggers complex adaptive or proapoptotic signaling defined as the unfolded protein response (UPR), involved in several pathophysiological processes. Since protein folding is highly redox-dependent, convergence between ER stress and oxidative stress has attracted interest. Evidence suggests that ROS production and oxidative stress are not only coincidental to ER stress, but are integral UPR components, being triggered by distinct types of ER stressors and contributing to support proapoptotic, as well as proadaptive UPR signaling. Thus, ROS generation can be upstream or downstream UPR targets and may display a UPR-specific plus a nonspecific component. Enzymatic mechanisms of ROS generation during UPR include: (a) Multiple thiol-disulfide exchanges involving ER oxidoreductases including flavooxidase Ero1 and protein disulfide isomerase (PDI); (b) Mitochondrial electron transport; (c) Nox4 NADPH oxidase complex, particularly Nox4. Understanding the roles of such mechanisms and how they interconnect with the UPR requires more investigation. Integration among such ROS sources may depend on Ca(2+) levels, ROS themselves, and PDI, which associates with NADPH oxidase and regulates its function. Oxidative stress may frequently integrate with a background of ER stress/UPR in several diseases; here we discuss a focus in the vascular system.
Collapse
Affiliation(s)
- Célio X C Santos
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, CEP 05403-000, São Paulo, Brazil
| | | | | | | |
Collapse
|