1
|
Fonseka O, Gare SR, Chen X, Zhang J, Alatawi NH, Ross C, Liu W. Molecular Mechanisms Underlying Heart Failure and Their Therapeutic Potential. Cells 2025; 14:324. [PMID: 40072053 PMCID: PMC11899429 DOI: 10.3390/cells14050324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Heart failure (HF) is a prominent fatal cardiovascular disorder afflicting 3.4% of the adult population despite the advancement of treatment options. Therefore, a better understanding of the pathogenesis of HF is essential for exploring novel therapeutic strategies. Hypertrophy and fibrosis are significant characteristics of pathological cardiac remodeling, contributing to HF. The mechanisms involved in the development of cardiac remodeling and consequent HF are multifactorial, and in this review, the key underlying mechanisms are discussed. These have been divided into the following categories thusly: (i) mitochondrial dysfunction, including defective dynamics, energy production, and oxidative stress; (ii) cardiac lipotoxicity; (iii) maladaptive endoplasmic reticulum (ER) stress; (iv) impaired autophagy; (v) cardiac inflammatory responses; (vi) programmed cell death, including apoptosis, pyroptosis, and ferroptosis; (vii) endothelial dysfunction; and (viii) defective cardiac contractility. Preclinical data suggest that there is merit in targeting the identified pathways; however, their clinical implications and outcomes regarding treating HF need further investigation in the future. Herein, we introduce the molecular mechanisms pivotal in the onset and progression of HF, as well as compounds targeting the related mechanisms and their therapeutic potential in preventing or rescuing HF. This, therefore, offers an avenue for the design and discovery of novel therapies for the treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (O.F.); (S.R.G.); (X.C.); (J.Z.); (N.H.A.)
| |
Collapse
|
2
|
Li L, Liu B, Zhang H, Wang C, Sun L, Zhang Y, Song L, Yu Y, Zhou K. 4-Phenylbutyric acid suppresses psoralen-induced hepatotoxicity by inhibiting ERS and reestablishing mitochondrial fusion-fission balance in mice. Toxicology 2024; 509:153954. [PMID: 39299507 DOI: 10.1016/j.tox.2024.153954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
Psoralen is a main active molecule of the traditional Chinese herb medicine Fructus Psoraleae. Our previous studies have shown that psoralen induced liver injury through the endoplasmic reticulum stress (ERS) signaling pathways. In this article, we studied whether the ERS inhibitor, 4-phenylbutyrate acid (4-PBA) could inhibit the liver toxicity caused by psoralen, and explored the underlying mechanisms. Mice were given the solvent, 20 mg/kg, 40 mg/kg, 80 mg/kg of psoralen, or 80 mg/kg of psoralen plus 4-PBA for 14 days. We found that 4-PBA significantly reduced the serum LDH and liver tissue MDA level, increased the activities of SOD and CAT, reduced liver weight and coefficient, repaired histopathological damage, and inhibited hepatocytes apoptosis induced by psoralen. RNA-seq transcriptomics found that except for the endoplasmic reticulum, the mitochondria was severely affected by psoralen. And genes involved in mitochondrial fusion, apoptosis, protein folding, and autophagy were found differently expressed in the psoralen group. Further studies found that 4-PBA inhibited the overexpression of GRP78 and CHOP, increased the Bcl-2/Bax ratio, and reduced the expression of Caspase-3. Moreover, 4-PBA reduced the overexpression of mitochondrial fission protein DRP1, increased the expression of fusion proteins Mfn-2 and OPA1, but has no inhibitory effects on autophagy proteins Atg5 or LC3A/B. In conclusion, 4-PBA inhibited ERS and reestablished mitochondrial fusion-fission balance, thereby blocking cell apoptosis, oxidative stress, and mitochondrial dysfunction, thus prevented against psoralen-induced hepatotoxicity.
Collapse
Affiliation(s)
- Li Li
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Bing Liu
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Haorui Zhang
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chen Wang
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Likang Sun
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yue Zhang
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin 301617, China
| | - Lei Song
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin 301617, China
| | - Yingli Yu
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin 301617, China.
| | - Kun Zhou
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
3
|
Mi K, Wang X, Ma C, Tan Y, Zhao G, Cao X, Yuan H. NLRX1 attenuates endoplasmic reticulum stress via STING in cardiac hypertrophy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119852. [PMID: 39357547 DOI: 10.1016/j.bbamcr.2024.119852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Endoplasmic reticulum stress-induced cell apoptosis is a pivotal mechanism underlying the progression of cardiac hypertrophy. NLRX1, a member of the NOD-like receptor family, modulates various cellular processes, including STING, NF-κB, MAPK pathways, reactive oxygen species production, essential metabolic pathways, autophagy and cell death. Emerging evidence suggests that NLRX1 may offer protection against diverse cardiac diseases. However, the impacts and mechanisms of NLRX1 on endoplasmic reticulum stress in cardiac hypertrophy remains largely unexplored. In our study, we observed that the NLRX1 and phosphorylated STING (p-STING) were highly expressed in both hypertrophic mouse heart and cellular model of cardiac hypertrophy. Whereas over-expression of NLRX1 mitigated the expression levels of p-STING, as well as the endoplasmic reticulum stress markers, including transcription activating factor 4 (ATF4), C/EBP homologous protein (CHOP) and the ratios of phosphorylated PERK to PERK, phosphorylated IRE1 to IRE1 and phosphorylated eIF2α to eIF2α in an Angiotensin II (Ang II)-induced cellular model of cardiac hypertrophy. Importantly, the protective effects of NLRX1 were attenuated upon pretreatment with the STING agonist, DMXAA. Our findings provide the evidence that NLRX1 attenuates the PERK-eIF2α-ATF4-CHOP axis of endoplasmic reticulum stress response via inhibition of p-STING in Ang II-treated cardiomyocytes, thereby ameliorating the development of cardiac hypertrophy.
Collapse
Affiliation(s)
- Keying Mi
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, People's Republic of China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Xiaoyan Wang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, People's Republic of China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Chao Ma
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Yinghua Tan
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Gang Zhao
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, People's Republic of China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Xinran Cao
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, People's Republic of China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China.
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, People's Republic of China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China.
| |
Collapse
|
4
|
You JB, Cao Y, You QY, Liu ZY, Wang XC, Ling H, Sha JM, Tao H. The landscape of histone modification in organ fibrosis. Eur J Pharmacol 2024; 977:176748. [PMID: 38897443 DOI: 10.1016/j.ejphar.2024.176748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/05/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
An increase in fibrous connective tissue and a decrease in parenchymal cells in organ tissues are the primary pathological alterations linked to organ fibrosis. If fibrosis is not treated, organ structure is destroyed, function can decline, or even fail, posing a serious risk to human life and health. Numerous organs develop fibrosis, and organ fibroproliferative illnesses account for almost 45% of patient deaths from various diseases in the industrialized world, as well as a major cause of disability and mortality in many other diseases. Recently, it has become evident that histone modification is an important way to regulate gene expression in organ fibrosis. Histone modifications alter the structure of chromatin, thereby affecting gene accessibility. Histone acetylation modifications relax chromatin, making it easier for gene transcription factors to access DNA, thereby promoting gene transcription. In addition, histone modifications recruit other proteins to interact with chromatin to form complexes that further regulate gene expression. Histone methylation modifications recruit methylation-reading proteins that recognize methylation marks and alter gene expression status. It not only affects the normal physiological function of cells, but also plays an important role in organ fibrosis. This article reviews the important role played by histone modifications in organ fibrosis and potential therapeutic approaches.
Collapse
Affiliation(s)
- Jun-Bo You
- Department of Thoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Yi Cao
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Qing-Ye You
- Anhui Women and Children's Medical Center, Hefei, 230001, PR China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Xian-Chen Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Hui Ling
- Department of Thoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Ji-Ming Sha
- Department of Thoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, PR China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, PR China.
| |
Collapse
|
5
|
Jiménez-González S, Delgado-Valero B, Islas F, Romero-Miranda A, Luaces M, Ramchandani B, Cuesta-Corral M, Montoro-Garrido A, Martínez-Martínez E, Cachofeiro V. The detrimental role of galectin-3 and endoplasmic reticulum stress in the cardiac consequences of myocardial ischemia in the context of obesity. FASEB J 2024; 38:e23818. [PMID: 38989572 DOI: 10.1096/fj.202400747r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024]
Abstract
The association between cardiac fibrosis and galectin-3 was evaluated in patients with acute myocardial infarction (MI). The role of galectin-3 and its association with endoplasmic reticulum (ER) stress activation in the progression of cardiovascular fibrosis was also evaluated in obese-infarcted rats. The inhibitor of galectin-3 activity, modified citrus pectin (MCP; 100 mg/kg/day), and the inhibitor of the ER stress activation, 4-phenylbutyric acid (4-PBA; 500 mg/kg/day), were administered for 4 weeks after MI in obese rats. Overweight-obese patients who suffered a first MI showed higher circulating galectin-3 levels, higher extracellular volume, and LV infarcted size, as well as lower E/e'ratio and LVEF compared with normal-weight patients. A correlation was observed between galectin-3 levels and extracellular volume. Obese-infarcted animals presented cardiac hypertrophy and reduction in LVEF, and E/A ratio as compared with control animals. They also showed an increase in galectin-3 gene expression, as well as cardiac fibrosis and reduced autophagic flux. These alterations were associated with ER stress activation characterized by enhanced cardiac levels of binding immunoglobulin protein, which were correlated with those of galectin-3. Both MCP and 4-PBA not only reduced cardiac fibrosis, oxidative stress, galectin-3 levels, and ER stress activation, but also prevented cardiac functional alterations and ameliorated autophagic flux. These results show the relevant role of galectin-3 in the development of diffuse fibrosis associated with MI in the context of obesity in both the animal model and patients. Galectin-3 in tandem with ER stress activation could modulate different downstream mechanisms, including inflammation, oxidative stress, and autophagy.
Collapse
Affiliation(s)
- Sara Jiménez-González
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Beatriz Delgado-Valero
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Fabian Islas
- Unidad de Imagen Cardíaca, Hospital General Universitario de Talavera de la Reina, Toledo, Spain
| | - Ana Romero-Miranda
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - María Luaces
- Servicio de Cardiología, Instituto Cardiovascular, Hospital Clínico San Carlos, Madrid, Spain
| | - Bunty Ramchandani
- Servicio de Cirugía Cardiaca Infantil, Hospital La Paz, Madrid, Spain
| | - María Cuesta-Corral
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Alejandro Montoro-Garrido
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
| | - Ernesto Martínez-Martínez
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Majadahonda, Spain
| | - Victoria Cachofeiro
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, Madrid, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Majadahonda, Spain
| |
Collapse
|
6
|
Wang Z, Lei Z, Wang Q, Jiang Q, Zhang Z, Liu X, Xing B, Li S, Guo X, Liu Y, Li X, Qi Y, Shu K, Zhang H, Huang Y, Lei T. Connexin 36 Mediated Intercellular Endoplasmic Reticulum Stress Transmission Induces SSTA Resistance in Growth Hormone Pituitary Adenoma. Int J Biol Sci 2024; 20:801-817. [PMID: 38169563 PMCID: PMC10758099 DOI: 10.7150/ijbs.86736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 12/10/2023] [Indexed: 01/05/2024] Open
Abstract
Somatostatin analogues (SSTA) are first-line pharmacological treatment choice for acromegaly, which received satisfying tumor shrinkage and normalization of growth hormone. However, there are still patients unresponsive to SSTA, and the underline mechanism remains unknown. Besides, there is no evidence regarding the role of endoplasmic reticulum stress (ERS) and its transmission in SSTA resistance, which also require investigation. Primary growth hormone adenoma cells and cell lines were treated with SSTA; autophagy double-labeled LC3 (mRFP-GFP) adenovirus transfection, flow cytometry sorting, western blotting, calcium imaging as well as immunofluorescence staining were used to determine ERS and autophagy signal transmission; xenograft and syngeneic tumor in vivo model were exploited to confirm the ERS signal transmission mediated effect. Our results revealed that SSTA induces ERS in pituitary growth hormone (GH) adenoma cells. The ERS signals can be intercellularly transmitted, leading to less responsible to SSTA treatment. Moreover, SSTA stimulates inositol triphosphate (IP3) elevation, mediating ERS intercellular transfer. In addition, connexin 36 tunnels ERS transmission, and its blocker, Quinine, exhibits a synergistic effect with SSTA treating GH adenoma. Our study provided insight into ERS intercellular transmission mediated SSTA resistance, which could be translated into clinical usage to improve SSTA efficiency in GH adenoma treatment.
Collapse
Affiliation(s)
- Zihan Wang
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhuowei Lei
- Department of Orthopedics, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Quanji Wang
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qian Jiang
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhuo Zhang
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaojin Liu
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Biao Xing
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sihan Li
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiang Guo
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanchao Liu
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xingbo Li
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yiwei Qi
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kai Shu
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yimin Huang
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ting Lei
- Department of Neurosurgery, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
7
|
Chen J, Ikeda SI, Kang L, Negishi K, Tsubota K, Kurihara T. Bisphenol A exposure triggers endoplasmic reticulum stress pathway leading to ocular axial elongation in mice. Front Med (Lausanne) 2023; 10:1255121. [PMID: 37746069 PMCID: PMC10517050 DOI: 10.3389/fmed.2023.1255121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Background Ocular axial elongation is one of the features of myopia progression. Endoplasmic reticulum (ER) stress-associated scleral remodeling plays an important role in ocular axial elongation. Bisphenol A (BPA) is one of the most common environmental pollutants and is known to affect various human organs through ER stress. However, whether BPA exerts an effect on scleral remodeling remains unknown. The purpose of this study was to determine the effect of BPA on the development of myopia and scleral ER stress. Methods BPA was administered by intraperitoneal injection. 4-PBA was administered as an endoplasmic reticulum stress inhibitor by eye drops. Refraction and axial length were measured by refractometer and SD-OCT system. Western blot was performed to detect the expression level of ER stress-related proteins. Results BPA-administered mice exhibit axial elongation and myopic refractive shift with endoplasmic reticulum stress in the sclera. BPA administration activated scleral PERK and ATF6 pathways, and 4-PBA eye drops attenuated ER stress response and suppressed myopia progression. Conclusion BPA controlled axial elongation during myopia development in a mouse model by inducing scleral ER stress and activation of the PERK/ATF6 pathway. 4-PBA eye drops as ER stress inhibitor suppressed BPA-induced myopia development.
Collapse
Affiliation(s)
- Junhan Chen
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Shin-ichi Ikeda
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Longdan Kang
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Tsubota Laboratory, Inc., Tokyo, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Xiong Y, Wang Y, Xiong Y, Teng L. 4-PBA inhibits hypoxia-induced lipolysis in rat adipose tissue and lipid accumulation in the liver through regulating ER stress. Food Sci Nutr 2023; 11:1223-1231. [PMID: 36911831 PMCID: PMC10002945 DOI: 10.1002/fsn3.3156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 02/12/2023] Open
Abstract
High-altitude hypoxia may disturb the metabolic modulation and function of both adipose tissue and liver. The endoplasmic reticulum (ER) is a crucial organelle in lipid metabolism and ER stress is closely correlated with lipid metabolism dysfunction. The aim of this study is to elucidate whether the inhibition of ER stress could alleviate hypoxia-induced white adipose tissue (WAT) lipolysis and liver lipid accumulation-mediated hepatic injury. A rat model of high-altitude hypoxia (5500 m) was established using hypobaric chamber. The response of ER stress and lipolysis-related pathways were analyzed in WAT under hypoxia exposure with or without 4-phenylbutyric acid (PBA) treatment. Liver lipid accumulation, liver injury, and apoptosis were evaluated. Hypoxia evoked significant ER stress in WAT, evidenced by increased GRP78, CHOP, and phosphorylation of IRE1α, PERK. Moreover, Lipolysis in perirenal WAT significantly increased under hypoxia, accompanied with increased phosphorylation of hormone-sensitive lipase (HSL) and perilipin. Treatment with 4-PBA, inhibitor of ER stress, effectively attenuated hypoxia-induced lipolysis via cAMP-PKA-HSL/perilipin pathway. In addition, 4-PBA treatment significantly inhibited the increase in fatty acid transporters (CD36, FABP1, FABP4) and ameliorated liver FFA accumulation. 4-PBA treatment significantly attenuated liver injury and apoptosis, which is likely resulting from decreased liver lipid accumulation. Our results highlight the importance of ER stress in hypoxia-induced WAT lipolysis and liver lipid accumulation.
Collapse
Affiliation(s)
- Yanlei Xiong
- Department of PathologyXuanwu Hospital, Capital Medical UniversityBeijingChina
- Department of PathophysiologyInstitute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS); School of Basic Medicine, Peking Union Medical College (PUMC)BeijingChina
| | - Yueming Wang
- Department of AnatomySchool of Basic Medicine, Binzhou Medical UniversityYantaiChina
| | - Yanlian Xiong
- Department of AnatomySchool of Basic Medicine, Binzhou Medical UniversityYantaiChina
| | - Lianghong Teng
- Department of PathologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
9
|
Bao M, Feng Q, Zou L, Huang J, Zhu C, Xia W. Endoplasmic reticulum stress promotes endometrial fibrosis through the TGF-β/SMAD pathway. Reproduction 2023; 165:171-182. [PMID: 36342661 DOI: 10.1530/rep-22-0294] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022]
Abstract
In brief Intrauterine adhesion (IUA) is one of the main causes of female infertility. This study reveals that endoplasmic reticulum stress activation upregulates the TGF-β/SMAD pathway to induce epithelial-mesenchymal transition and promote endometrial fibrosis in an IUA model. Abstract IUA is a common gynecological disease and is a leading cause of female infertility. Mechanical or infectious damage to the endometrial basal layer can lead to endometrial fibrosis, which is the most common cause of IUA. Endoplasmic reticulum stress (ERS), the transforming growth factor beta signaling pathway (TGF-β/SMAD) and epithelial-mesenchymal transition (EMT) are important factors promoting endometrial fibrosis. The purpose of this study was to determine the up- and downstream regulatory relationships of the above three in the process of endometrial fibrosis. The rat IUA model was induced by double injury method and prophylactic injection of the ERS inhibitor 4-phenylbutyric acid (4-PBA) was given in vivo. The ERS activator tunicamycin and the TGF-β/SMAD pathway inhibitor A 83-01 were used in human endometrial epithelial cells (HEECs) in vitro. Masson's trichrome, Sirius red staining, immunohistochemistry, immunofluorescence and Western blot analyses were used to determine ERS, TGF-β/SMAD pathway, EMT and fibrosis markers in the uterine tissue and HEECs of the different treatment groups. In animal experiments, ERS and the TGF-β/SMAD pathway had been activated and EMT occurred in an in vivo model of IUA but was suppressed in animals treated with prophylactic 4-PBA. In in vitro experiments, tunicamycin-treated HEECs had increased the activation of ERS, the abundance of TGF-β/SMAD pathway and fibrosis markers while EMT occurred, but the TGF-β/SMAD pathway and EMT were significantly inhibited in the tunicamycin+A 83-01 group. Our data suggest that increased ERS can induce EMT and promote endometrial fibrosis through the TGF-β/SMAD pathway.
Collapse
Affiliation(s)
- Meng Bao
- Institute of Reproductive Health, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Qiwen Feng
- Institute of Reproductive Health, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Liping Zou
- Institute of Reproductive Health, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Jin Huang
- Institute of Reproductive Health, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Changhong Zhu
- Institute of Reproductive Health, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Wei Xia
- Institute of Reproductive Health, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Luo Y, Lu J, Wang Z, Wang L, Wu G, Guo Y, Dong Z. Small ubiquitin-related modifier (SUMO)ylation of SIRT1 mediates (-)-epicatechin inhibited- differentiation of cardiac fibroblasts into myofibroblasts. PHARMACEUTICAL BIOLOGY 2022; 60:1762-1770. [PMID: 36086802 PMCID: PMC9467557 DOI: 10.1080/13880209.2022.2101672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT (-)-Epicatechin (EPI) is a crucial substance involved in the protective effects of flavanol-rich foods. Previous studies have indicated EPI has a cardioprotective effect, but the molecular mechanisms in inhibition of cardiac fibrosis are unclear. OBJECTIVE We evaluated the effect of EPI in preventing cardiac fibrosis and the underlying molecular mechanism related to the SIRT1-SUMO1/AKT/GSK3β pathway. MATERIALS AND METHODS Cardiac fibrosis mice model was established with transaortic constriction (TAC). Male C57BL/6 mice were randomly separated into 4 groups. Mice received 1 mg/kg/day of EPI or vehicle orally for 4 weeks. The acutely isolated cardiac fibroblasts were induced to myofibroblasts with 1 µM angiotensin II (Ang II). The cardiac function was measured with the ultrasonic instrument. Histological analysis of mice's hearts was determined with H&E or Masson method. The protein level of fibrosis markers, SUMOylation of SIRT1, and AKT/GSK3β pathway were quantified by immunofluorescence and western blot. RESULTS EPI treatment (1 mg/kg/day) could reverse the TAC-induced decline in LVEF (TAC, 61.28% ± 1.33% vs. TAC + EPI, 74.00% ± 1.64%), LVFS (TAC, 28.16% ± 0.89% vs. TAC + EPI, 37.18% ± 1.29%). Meantime, we found that 10 µM EPI blocks Ang II-induced transformation of cardiac fibroblasts into myofibroblasts. The underlying mechanism of EPI-inhibited myofibroblasts transformation involves activation of SUMOylation of SIRT1 through SP1. Furthermore, SUMOylation of SIRT1 inhibited Ang II-induced fibrogenic effect via the AKT/GSK3β pathway. CONCLUSION EPI plays a protective effect on cardiac fibrosis by regulating the SUMO1-dependent modulation of SIRT1, which provides a theoretical basis for use in clinical therapies.
Collapse
Affiliation(s)
- Yingchun Luo
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jing Lu
- Department of Pharmacy, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zeng Wang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Lu Wang
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Guodong Wu
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yuanyuan Guo
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zengxiang Dong
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, the First Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
11
|
Stein D, Slobodnik Z, Tam B, Einav M, Akabayov B, Berstein S, Toiber D. 4-phenylbutyric acid-Identity crisis; can it act as a translation inhibitor? Aging Cell 2022; 21:e13738. [PMID: 36373957 PMCID: PMC9741500 DOI: 10.1111/acel.13738] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/30/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
Loss of proteostasis can occur due to mutations, the formation of aggregates, or general deficiency in the correct translation and folding of proteins. These phenomena are commonly observed in pathologies, but most significantly, loss of proteostasis characterizes aging. This loss leads to the chronic activation of stress responses and has a generally deleterious impact on the organism. While finding molecules that can alleviate these symptoms is an important step toward solutions for these conditions, some molecules might be mischaracterized on the way. 4-phenylbutyric acid (4PBA) is known for its role as a chemical chaperone that helps alleviate endoplasmic reticulum (ER) stress, yet a scan of the literature reveals that no biochemical or molecular experiments have shown any protein refolding capacity. Here, we show that 4PBA is a conserved weak inhibitor of mRNA translation, both in vitro and in cellular systems, and furthermore-it does not promote protein folding nor prevents aggregation. 4PBA possibly alleviates proteostatic or ER stress by inhibiting protein synthesis, allowing the cells to cope with misfolded proteins by reducing the protein load. Better understanding of 4PBA biochemical mechanisms will improve its usage in basic science and as a drug in different pathologies, also opening new venues for the treatment of different diseases.
Collapse
Affiliation(s)
- Daniel Stein
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael,The Zlotowski Center for NeuroscienceBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Zeev Slobodnik
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael,The Zlotowski Center for NeuroscienceBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Benjamin Tam
- Department of ChemistryBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Monica Einav
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael,The Zlotowski Center for NeuroscienceBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Barak Akabayov
- Department of ChemistryBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Shimon Berstein
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Debra Toiber
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael,The Zlotowski Center for NeuroscienceBen‐Gurion University of the NegevBeer ShevaIsrael
| |
Collapse
|
12
|
4-PBA Attenuates Fat Accumulation in Cultured Spotted Seabass Fed High-Fat-Diet via Regulating Endoplasmic Reticulum Stress. Metabolites 2022; 12:metabo12121197. [PMID: 36557235 PMCID: PMC9784988 DOI: 10.3390/metabo12121197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/13/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Excessive fat accumulation is a common phenomenon in cultured fish, which can cause metabolic disease such as fatty liver. However, the relative regulatory approach remains to be explored. Based on this, two feeding trials were conducted. Firstly, fish were fed either a normal-fat diet (NFD) or a high-fat diet (HFD) for eight weeks and sampled at the 2nd, 4th, 6th, and 8th week after feeding (Experiment I). In the first four weeks, fish fed an HFD grew faster than those fed an NFD. Conversely, the body weight and weight gain were higher in the NFD group at the 6th and 8th weeks. Under light and transmission electron microscopes, fat accumulation of the liver was accompanied by an obvious endoplasmic reticulum (ER) swell. Accordingly, the expressions of atf-6, ire-1, perk, eif-2α, atf-4, grp78, and chop showed that ER stress was activated at the 6th and 8th weeks. In Experiment II, 50 mg/kg 4-PBA (an ERs inhibitor) was supplemented to an HFD; this was named the 4-PBA group. Then, fish was fed with an NFD, an HFD, and a 4-PBA diet for eight weeks. As the result, the excessive fat deposition caused by an HFD was reversed by 4-PBA. The expression of ER stress-related proteins CHOP and GRP78 was down-regulated by 4-PBA, and the transmission electron microscope images also showed that 4-PBA alleviated ER stress induced by the feeding of an HFD. Furthermore, 4-PBA administration down-regulated SREBP-1C/ACC/FAS, the critical pathways of fat synthesis. In conclusion, the results confirmed that ER stress plays a contributor role in the fat deposition by activating the SREBP-1C/ACC/FAS pathway. 4-PBA as an ER stress inhibitor could reduce fat deposition caused by an HFD via regulating ER stress.
Collapse
|
13
|
Hyperglycemia promotes myocardial dysfunction via the ERS-MAPK10 signaling pathway in db/db mice. J Transl Med 2022; 102:1192-1202. [PMID: 36775445 PMCID: PMC9588458 DOI: 10.1038/s41374-022-00819-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 01/15/2023] Open
Abstract
Recent studies have demonstrated that hyperglycemia is a major risk factor for the development and exacerbation of cardiovascular disease (CVD). However, the molecular mechanisms involved in diabetic cardiomyopathy (DCM) have not been fully elucidated. In this study, we focused on the underlying mechanism of DCM. Leptin receptor-deficient db/db mice were used to model a type 2 diabetes mellitus (T2DM) model in our study. WT mice and db/db mice received 4-phenylbutyric acid (4-PBA) (25 mg/kg/day) and saline by intraperitoneal injection every other day for 4 weeks. WT and db/db mice were given tail vein injections of 100 μL of rAAV9-Sh-MAPK10 and rAAV9-Sh-GFP at the age of 6-8 weeks. Echocardiography was performed to measure cardiac function, histological examinations were used to evaluate ventricular hypertrophy and fibrosis. Quantitative RT-qPCR was used to assess the mRNA expression of Jun N-terminal kinase 3 (JNK3, MAPK10), atrial natriuretic factor (ANF), brain natriuretic peptide (BNP), and collagen I and III. Immunoblotting was performed to measure the levels of cardiac hypertrophy-related proteins, fibrosis-related proteins, endoplasmic reticulum stress (ERS)-related proteins and apoptosis-related proteins. TUNEL staining was performed to examine cardiomyocyte apoptosis. In contrast to 12-week-old db/db mice, 16-week-old db/db mice showed the most severe myocardial dysfunction. The DCM induced by hyperglycemia was largely alleviated by 4-PBA (25 mg/kg/day, intraperitoneal injection). Similarly, tail vein injection of rAAV9-Sh-MAPK10 reversed the phenotype of the heart in db/db mice including cardiac hypertrophy and apoptosis in db/db mice. The mechanistic findings suggested that hyperglycemia initiated the ERS response through the negative regulation of sirtuin 1 (SIRT1), leading to the occurrence of myocardial dysfunction, and specific knockdown of MAPK10 in the heart directly reversed myocardial dysfunction induced by hyperglycemia. We demonstrated that hyperglycemia promotes DCM in db/db mice through the ERS-MAPK10 signaling pathway in diabetic mice.
Collapse
|
14
|
Zhu F, Li P, Sheng Y. Treatment of myocardial interstitial fibrosis in pathological myocardial hypertrophy. Front Pharmacol 2022; 13:1004181. [PMID: 36249793 PMCID: PMC9561344 DOI: 10.3389/fphar.2022.1004181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/14/2022] [Indexed: 01/09/2023] Open
Abstract
Pathological myocardial hypertrophy can be caused by a variety of diseases, mainly accompanied by myocardial interstitial fibrosis (MIF), which is a diffuse and patchy process, appearing as a combination of interstitial micro-scars and perivascular collagen fiber deposition. Different stimuli may trigger MIF without cell death by activating a variety of fibrotic signaling pathways in mesenchymal cells. This manuscript summarizes the current knowledge about the mechanism and harmful outcomes of MIF in pathological myocardial hypertrophy, discusses the circulating and imaging biomarkers that can be used to identify this lesion, and reviews the currently available and potential future treatments that allow the individualized management of patients with pathological myocardial hypertrophy.
Collapse
Affiliation(s)
- Fuyu Zhu
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Peng Li
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China,*Correspondence: Yanhui Sheng, ; Peng Li,
| | - Yanhui Sheng
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China,Department of Cardiology, Jiangsu Province Hospital, Nanjing, China,*Correspondence: Yanhui Sheng, ; Peng Li,
| |
Collapse
|
15
|
Chen K, Wang Z, Liu C, Yang X, Jiang J. Sphingosine-1-phosphate Attenuates Endoplasmic Reticulum Stress-induced Cardiomyocyte Apoptosis Through Sphingosine-1-phosphate Receptor 1. Arch Med Res 2022; 53:562-573. [PMID: 35999060 DOI: 10.1016/j.arcmed.2022.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 07/03/2022] [Accepted: 08/09/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND Endoplasmic reticulum stress (ER stress) is involved in the development and progression of various forms of heart disease and may lead to myocardial apoptosis. Sphingosine-1-phosphate (S1P) possesses cardioprotective properties, including anti-apoptosis. However, little is known about the link between S1P and ER stress-induced myocardial apoptosis. This study investigated the regulatory role of S1P in ER stress-induced apoptosis in cardiomyocytes. METHODS ER stress and myocardial apoptosis were induced by transverse aortic constriction (TAC) or tunicamycin in mice, which were then treated with 2-acetyl-5-tetrahydroxybutyl imidazole (THI) or S1P. AC16 cells were treated with tunicamycin or thapsigargin, or pretreated with S1P, sphingosine-1-phosphate receptor (S1PR) subtype antagonists, S1PR1 agonist, and PI3K and MEK inhibitors. Cardiac function, the level of S1P in plasma and heart, ER stress markers, cell viability, and apoptosis were detected. RESULTS S1P reduced the expression of ER stress-related molecules and ER stress-induced myocardial apoptosis in mice subjected to TAC or an injection of tunicamycin. Furthermore, in AC16 cells exposed to thapsigargin or tunicamycin, S1P decreased the expression of ER stress-related molecules, promoting cell viability and survival. Nevertheless, the S1PR1 antagonist abrogated the protection of S1P. Subsequently, in TAC S1PR1 heterozygous (S1PR1+/-) mice, S1P had no effect on ER stress and apoptosis in cardiomyocytes. Notably, in vitro, the impact of anti-ER stress-induced myocardial apoptosis by the S1PR1 agonist was reversed by PI3K and MEK inhibitors. CONCLUSION This study is the first to demonstrate that S1P relieves ER stress-induced myocardial apoptosis via S1PR1/AKT and S1PR1/ERK1/2, which are potential therapeutic targets for heart disease.
Collapse
Affiliation(s)
- Kengquan Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhongqin Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Liu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Yang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiangang Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
16
|
Azam T, Zhang H, Zhou F, Wang X. Recent Advances on Drug Development and Emerging Therapeutic Agents Through Targeting Cellular Homeostasis for Ageing and Cardiovascular Disease. FRONTIERS IN AGING 2022; 3:888190. [PMID: 35821839 PMCID: PMC9261412 DOI: 10.3389/fragi.2022.888190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/30/2022] [Indexed: 12/11/2022]
Abstract
Ageing is a progressive physiological process mediated by changes in biological pathways, resulting in a decline in tissue and cellular function. It is a driving factor in numerous age-related diseases including cardiovascular diseases (CVDs). Cardiomyopathies, hypertension, ischaemic heart disease, and heart failure are some of the age-related CVDs that are the leading causes of death worldwide. Although individual CVDs have distinct clinical and pathophysiological manifestations, a disturbance in cellular homeostasis underlies the majority of diseases which is further compounded with aging. Three key evolutionary conserved signalling pathways, namely, autophagy, mitophagy and the unfolded protein response (UPR) are involved in eliminating damaged and dysfunctional organelle, misfolded proteins, lipids and nucleic acids, together these molecular processes protect and preserve cellular homeostasis. However, amongst the numerous molecular changes during ageing, a decline in the signalling of these key molecular processes occurs. This decline also increases the susceptibility of damage following a stressful insult, promoting the development and pathogenesis of CVDs. In this review, we discuss the role of autophagy, mitophagy and UPR signalling with respect to ageing and cardiac disease. We also highlight potential therapeutic strategies aimed at restoring/rebalancing autophagy and UPR signalling to maintain cellular homeostasis, thus mitigating the pathological effects of ageing and CVDs. Finally, we highlight some limitations that are likely hindering scientific drug research in this field.
Collapse
Affiliation(s)
- Tayyiba Azam
- Michael Smith Building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Hongyuan Zhang
- Michael Smith Building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Fangchao Zhou
- Michael Smith Building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Xin Wang
- Michael Smith Building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
17
|
Mitigating sarcoplasmic reticulum stress limits disuse-induced muscle loss in hindlimb unloaded mice. NPJ Microgravity 2022; 8:24. [PMID: 35817772 PMCID: PMC9273600 DOI: 10.1038/s41526-022-00211-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/22/2022] [Indexed: 01/31/2023] Open
Abstract
Muscle disuse in the hindlimb unloaded (HU) mice causes significant atrophy and weakness. However, the cellular and molecular mechanisms driving disuse-muscle atrophy remain elusive. We investigated the potential contribution of proteins dysregulation by sarcoplasmic reticulum (SR), a condition called SR stress, to muscle loss during HU. Male, c57BL/6j mice were assigned to ground-based controls or HU groups treated with vehicle or 4-phenylbutyrate (4-PBA), a potent inhibitor of SR stress, once a day for three weeks. We report that the 4-PBA reduced the SR stress and partly reversed the muscle atrophy and weakness in the HU mice. Transcriptome analysis revealed that several genes were switched on (n = 3688) or differentially expressed (n = 1184) due to HU. GO, and KEGG term analysis revealed alterations in pathways associated with the assembly of cilia and microtubules, extracellular matrix proteins regulation, calcium homeostasis, and immune modulation during HU. The muscle restoration with 4-PBA partly reversed these changes along with differential and unique expression of several genes. The analysis of genes among the two comparisons (HU-v vs. control and HU-t vs. HU-v.) shows 841 genes were overlapped between the two comparisons and they may be regulated by 4-PBA. Altogether, our findings suggest that the pharmacological suppression of SR stress may be an effective strategy to prevent disuse-induced muscle weakness and atrophy.
Collapse
|
18
|
Sharma R, Sharma S, Thakur A, Singh A, Singh J, Nepali K, Liou JP. The Role of Epigenetic Mechanisms in Autoimmune, Neurodegenerative, Cardiovascular, and Imprinting Disorders. Mini Rev Med Chem 2022; 22:1977-2011. [PMID: 35176978 DOI: 10.2174/1389557522666220217103441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/01/2021] [Accepted: 11/11/2021] [Indexed: 11/22/2022]
Abstract
Epigenetic mutations like aberrant DNA methylation, histone modifications, or RNA silencing are found in a number of human diseases. This review article discusses the epigenetic mechanisms involved in neurodegenerative disorders, cardiovascular disorders, auto-immune disorder, and genomic imprinting disorders. In addition, emerging epigenetic therapeutic strategies for the treatment of such disorders are presented. Medicinal chemistry campaigns highlighting the efforts of the chemists invested towards the rational design of small molecule inhibitors have also been included. Pleasingly, several classes of epigenetic inhibitors, DNMT, HDAC, BET, HAT, and HMT inhibitors along with RNA based therapies have exhibited the potential to emerge as therapeutics in the longer run. It is quite hopeful that epigenetic modulator-based therapies will advance to clinical stage investigations by leaps and bounds.
Collapse
Affiliation(s)
- Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Sachin Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Arshdeep Singh
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jagjeet Singh
- School of Pharmacy, University of Queensland, Brisbane, QLD, Australia.,Department of Pharmacy, Rayat-Bahara Group of Institutes, Hoshiarpur, India
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
19
|
Liao J, Hu Z, Li Q, Li H, Chen W, Huo H, Han Q, Zhang H, Guo J, Hu L, Pan J, Li Y, Tang Z. Endoplasmic Reticulum Stress Contributes to Copper-Induced Pyroptosis via Regulating the IRE1α-XBP1 Pathway in Pig Jejunal Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1293-1303. [PMID: 35075900 DOI: 10.1021/acs.jafc.1c07927] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Copper (Cu) is a common additive in food products, which poses a potential concern to animal and human health when it is in excess. Here, we investigated the relationship between endoplasmic reticulum (ER) stress and pyroptosis in Cu-induced toxicity of jejunum in vivo and in vitro. In in vivo experiments, excess intake of dietary Cu caused ER cavity expansion, elevated fluorescence signals of GRP78 and Caspase-1, and increased the mRNA and protein expression levels related to ER stress and pyroptosis in pig jejunal epithelium. Simultaneously, similar effects were observed in IPEC-J2 cells under excess Cu treatment. Importantly, 4-phenylbutyric acid (ER stress inhibitor) and MKC-3946 (IRE1α inhibitor) significantly inhibited the ER stress-triggered IRE1α-XBP1 pathway, which also alleviated the Cu-induced pyroptosis in IPEC-J2 cells. In general, these results suggested that ER stress participated in regulating Cu-induced pyroptosis in jejunal epithelial cells via the IRE1α-XBP1 pathway, which provided a novel view into the toxicology of Cu.
Collapse
Affiliation(s)
- Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Zhuoying Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Quanwei Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Hongji Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Weijin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Haihua Huo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Qingyue Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Jianying Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Jiaqiang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| |
Collapse
|
20
|
Tominaga K, Tominaga N, Williams EO, Rufibach L, Schöwel V, Spuler S, Viswanathan M, Guarente LP. 4-Phenylbutyrate restores localization and membrane repair to human dysferlin mutations. iScience 2022; 25:103667. [PMID: 35028538 PMCID: PMC8741482 DOI: 10.1016/j.isci.2021.103667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 12/15/2021] [Indexed: 10/29/2022] Open
Abstract
Dysferlinopathies are muscular dystrophies caused by recessive loss-of-function mutations in dysferlin (DYSF), a membrane protein involved in skeletal muscle membrane repair. We describe a cell-based assay in which human DYSF proteins bearing missense mutations are quantitatively assayed for membrane localization by flow cytometry and identified 64 localization-defective DYSF mutations. Using this platform, we show that the clinically approved drug 4-phenylbutryric acid (4-PBA) partially restores membrane localization to 25 mutations, as well as membrane repair to cultured myotubes expressing 2 different mutations. Two-day oral administration of 4-PBA to mice homozygous for one of these mutations restored myofiber membrane repair. 4-PBA may hold therapeutic potential for treating a subset of humans with muscular dystrophy due to dysferlin deficiency.
Collapse
Affiliation(s)
- Kana Tominaga
- Paul F. Glenn Center for Biology of Aging, Department of Biology, Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Pathology and Oncology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyoku, Tokyo 113-8421, Japan
| | - Naoomi Tominaga
- Paul F. Glenn Center for Biology of Aging, Department of Biology, Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eric O. Williams
- Paul F. Glenn Center for Biology of Aging, Department of Biology, Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Fitchburg State University, School of Heath and Natural Sciences, Antonucci Science Complex 235, Fitchburg, MA 01420, USA
| | - Laura Rufibach
- Jain Foundation, 9706 4th Avenue NE, Suite 101, Seattle, WA 98115, USA
| | - Verena Schöwel
- Muscle Research Unit, Experimental and Clinical Research Center (ECRC), a joint cooperation of Charité Universitätsmedizin Berlin and Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center (ECRC), a joint cooperation of Charité Universitätsmedizin Berlin and Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Mohan Viswanathan
- Paul F. Glenn Center for Biology of Aging, Department of Biology, Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leonard P. Guarente
- Paul F. Glenn Center for Biology of Aging, Department of Biology, Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
21
|
Wang X, Zhang Y, Wuyun K, Gong H. Therapeutic effect and mechanism of 4‑phenyl butyric acid on renal ischemia‑reperfusion injury in mice. Exp Ther Med 2021; 23:144. [PMID: 35069825 PMCID: PMC8756420 DOI: 10.3892/etm.2021.11067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 08/06/2021] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to explore the effects and possible mechanism of 4-phenylbutyric acid (4-PBA) on renal ischemia-reperfusion injury (RIRI) in mice. A RIRI model of HK-2 cells was constructed using hypoxia/reoxygenation (H/R) treatment. Dexmedetomidine and 4-PBA were used to treat the cells before and after modeling. Apoptosis and expression levels of cyclophilin D (CypD), cytochrome c, eukaryotic translation initiation factor 2α (eIF2α), glucose-regulated protein 78 (GRP78), intercellular adhesion molecule (ICAM)-1 and vascular adhesion molecule (VCAM)-1 were measured using flow cytometry, western blotting and immunohistochemistry. The renal volume, weight and renal arterial resistance index (RRI) were determined using the renal ischemia model. Compared with untreated model cells, 4-PBA treatment significantly decreased apoptosis and the expression levels of CypD, Cytochrome c, eIF2α and GRP78 in HK-2 cells. There was no significant change in renal volume and weight after modeling, but RRI was significantly decreased after 4-PBA treatments in the model. Western blotting and immunohistochemistry analysis demonstrated that 4-PBA treatment also significantly decreased the expression of ICAM-1 and VCAM-1. Overall, 4-PBA had a therapeutic effect on RIRI in mice. This protection may be mediated by decreasing the expression levels of CypD, Cytochrome c, eIF2α and GRP78, and subsequent reduction of cellular oxygen free radicals and apoptosis, leading to an alleviated endoplasmic reticulum stress response and RIRI.
Collapse
Affiliation(s)
- Xinlei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Kun Wuyun
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Haixia Gong
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
22
|
Han X, Zhang Y, Qiao O, Ji H, Zhang X, Wang W, Li X, Wang J, Li D, Ju A, Liu C, Gao W. Proteomic Analysis Reveals the Protective Effects of Yiqi Fumai Lyophilized Injection on Chronic Heart Failure by Improving Myocardial Energy Metabolism. Front Pharmacol 2021; 12:719532. [PMID: 34630097 PMCID: PMC8494180 DOI: 10.3389/fphar.2021.719532] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/03/2021] [Indexed: 12/28/2022] Open
Abstract
Yiqi Fumai lyophilized injection (YQFM) is the recombination of Sheng mai san (SMS).YQFM has been applied clinically to efficaciously and safely treat chronic heart failure (CHF). However, the mechanism of YQFM is still not fully elucidated. The purpose of our study was to investigate the protective mechanism of YQFM against abdominal aortic coarctation (AAC) in rats by proteomic methods. After YQFM treatment, the cardiac function were obviously meliorated. One hundred and fifty-seven important differentially expressed proteins (DEPs) were identified, including 109 in model rat compared with that in control rat (M:C) and 48 in YQFM-treated rat compared with that in model rat (T:M) by iTRAQ technology to analyze the proteomic characteristics of heart tissue. Bioinformatics analysis showed that DEPs was mainly involved in the body’s energy metabolism and was closely related to oxidative phosphorylation. YQFM had also displayed efficient mitochondrial dysfunction alleviation properties in hydrogen peroxide (H2O2)-induced cardiomyocyte damage by Transmission Electron Microscope (TEM), Metabolic assay, and Mitotracker staining. What’s more, the levels of total cardiomyocyte apoptosis were markedly reduced following YQFM treatment. Furthermore, Western blot analysis showed that the expressions of peroxisome proliferator activated receptor co-activator-1α(PGC-1α) (p < 0.01 or p < 0.001), perixisome proliferation-activated receptor alpha (PPAR-α) (p < 0.001)and retinoid X receptor alpha (RXR-α) were upregulated (p < 0.001), PGC-1α as well as its downstream effectors were also found to be upregulated in cardiomyocytes after YQFM treatment(p < 0.001).These results provided evidence that YQFM could enhance mitochondrial function of cardiomyocytes to play a role in the treatment of CHF by regulating mitochondrial biogenesis-related proteins.
Collapse
Affiliation(s)
- Xiaoying Han
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Yi Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Ou Qiao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Haixia Ji
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Xinyu Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Wenzhe Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Xia Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Juan Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Dekun Li
- Tasly Pride Pharmaceutical Company Limited, Tianjin, China
| | - Aichun Ju
- Tasly Pride Pharmaceutical Company Limited, Tianjin, China
| | - Changxiao Liu
- Tianjin Pharmaceutical Research Institute, Tianjin, China
| | - Wenyuan Gao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
23
|
Souza-Neto FV, Jiménez-González S, Delgado-Valero B, Jurado-López R, Genty M, Romero-Miranda A, Rodríguez C, Nieto ML, Martínez-Martínez E, Cachofeiro V. The Interplay of Mitochondrial Oxidative Stress and Endoplasmic Reticulum Stress in Cardiovascular Fibrosis in Obese Rats. Antioxidants (Basel) 2021; 10:antiox10081274. [PMID: 34439522 PMCID: PMC8389298 DOI: 10.3390/antiox10081274] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
We have evaluated the role of mitochondrial oxidative stress and its association with endoplasmic reticulum (ER) stress activation in the progression of obesity-related cardiovascular fibrosis. MitoQ (200 µM) was orally administered for 7 weeks to male Wistar rats that were fed a high-fat diet (HFD, 35% fat) or a control diet (CT, 3.5% fat). Obese animals presented cardiovascular fibrosis accompanied by increased levels of extracellular matrix proteins and profibrotic mediators. These alterations were associated with ER stress activation characterized by enhanced levels (in heart and aorta vs. CT group, respectively) of immunoglobulin binding protein (BiP; 2.1-and 2.6-fold, respectively), protein disulfide-isomerase A6 (PDIA6; 1.9-fold) and CCAAT-enhancer-binding homologous protein (CHOP; 1.5- and 1.8-fold, respectively). MitoQ treatment was able to prevent (p < 0.05) these modifications at cardiac and aortic levels. MitoQ (5 nM) and the ER stress inhibitor, 4-phenyl butyric acid (4 µM), were able to block the prooxidant and profibrotic effects of angiotensin II (Ang II, 10−6 M) in cardiac and vascular cells. Therefore, the data show a crosstalk between mitochondrial oxidative stress and ER stress activation, which mediates the development of cardiovascular fibrosis in the context of obesity and in which Ang II can play a relevant role.
Collapse
Affiliation(s)
- Francisco V. Souza-Neto
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (B.D.-V.); (R.J.-L.); (M.G.); (A.R.-M.)
| | - Sara Jiménez-González
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (B.D.-V.); (R.J.-L.); (M.G.); (A.R.-M.)
| | - Beatriz Delgado-Valero
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (B.D.-V.); (R.J.-L.); (M.G.); (A.R.-M.)
| | - Raquel Jurado-López
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (B.D.-V.); (R.J.-L.); (M.G.); (A.R.-M.)
| | - Marie Genty
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (B.D.-V.); (R.J.-L.); (M.G.); (A.R.-M.)
| | - Ana Romero-Miranda
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (B.D.-V.); (R.J.-L.); (M.G.); (A.R.-M.)
| | - Cristina Rodríguez
- Institut de Recerca del Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
- Instituto de Investigación Biomédica Sant Pau (IB Sant Pau), 08025 Barcelona, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28220 Majadahonda, Spain;
| | - María Luisa Nieto
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28220 Majadahonda, Spain;
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, 47002 Valladolid, Spain
| | - Ernesto Martínez-Martínez
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (B.D.-V.); (R.J.-L.); (M.G.); (A.R.-M.)
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28220 Majadahonda, Spain;
- Correspondence: (E.M.-M.); (V.C.); Tel.: +34-913941483 (E.M.-M.); +34-913941489 (V.C.)
| | - Victoria Cachofeiro
- Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28040 Madrid, Spain; (F.V.S.-N.); (S.J.-G.); (B.D.-V.); (R.J.-L.); (M.G.); (A.R.-M.)
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28220 Majadahonda, Spain;
- Correspondence: (E.M.-M.); (V.C.); Tel.: +34-913941483 (E.M.-M.); +34-913941489 (V.C.)
| |
Collapse
|
24
|
Liu M, Liu H, Parthiban P, Kang GJ, Shi G, Feng F, Zhou A, Gu L, Karnopp C, Tolkacheva EG, Dudley SC. Inhibition of the unfolded protein response reduces arrhythmic risk after myocardial infarction. J Clin Invest 2021; 131:e147836. [PMID: 34324437 DOI: 10.1172/jci147836] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Ischemic cardiomyopathy is associated with an increased risk of sudden death, activation of the unfolded protein response (UPR), and reductions in multiple cardiac ion channels. When activated, the protein kinase-like ER kinase (PERK) branch of the UPR reduces protein translation and abundance. We hypothesized that PERK inhibition could prevent ion channel downregulation and reduce arrhythmic risk after myocardial infarct (MI). MI induced by coronary artery ligation resulted in mice exhibited reduced ion channel levels, ventricular tachycardia (VT), and prolonged corrected intervals between the Q and T waves of the ECGs (QTc). Protein levels of major cardiac ion channels were decreased. MI cardiomyocytes showed significantly prolonged action potential duration and decreased maximum upstroke velocity. Cardiac-specific PERK knockout (PERKKO) reduced electrical remodeling in response to MI with shortened QTc intervals, less VT episodes, and higher survival rates (P<0.05 vs. MI). Pharmacological PERK inhibition had similar effects. In conclusion, activated PERK during MI contributed to arrhythmic risk by downregulation of select cardiac ion channels. PERK inhibition prevented these changes and reduced arrhythmic risk. These results suggest that ion channel downregulation during MI is a fundamental arrhythmic mechanism and maintaining ion channel levels is antiarrhythmic.
Collapse
Affiliation(s)
- Man Liu
- Lillehei Heart Institute, University of Minnesota, Minneapolis, United States of America
| | - Hong Liu
- Lillehei Heart Institute, University of Minnesota, Minneapolis, United States of America
| | - Preethy Parthiban
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, United States of America
| | - Gyeoung-Jin Kang
- Lillehei Heart Institute, University of Minnesota, Minneapolis, United States of America
| | - Guangbin Shi
- Department of Medicine, Brown University, Providence, United States of America
| | - Feng Feng
- Lillehei Heart Institute, University of Minnesota, Minneapolis, United States of America
| | - Anyu Zhou
- Department of Medicine, Brown University, Providence, United States of America
| | - Lianzhi Gu
- Lillehei Heart Institute, University of Minnesota, Minneapolis, United States of America
| | - Courtney Karnopp
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, United States of America
| | - Elena G Tolkacheva
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, United States of America
| | - Samuel C Dudley
- Lillehei Heart Institute, University of Minnesota, Minneapolis, United States of America
| |
Collapse
|
25
|
Zhang Y, Ji H, Qiao O, Li Z, Pecoraro L, Zhang X, Han X, Wang W, Zhang X, Man S, Wang J, Li X, Liu C, Huang L, Gao W. Nanoparticle conjugation of ginsenoside Rb3 inhibits myocardial fibrosis by regulating PPARα pathway. Biomed Pharmacother 2021; 139:111630. [PMID: 33945912 DOI: 10.1016/j.biopha.2021.111630] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cardiac fibrosis occurs in ischemic and non-ischemic heart failure, hereditary cardiomyopathy, diabetes and aging. Energy metabolism, which serves a crucial function in the course and treatment of cardiovascular diseases, might have therapeutic benefits for myocardial fibrosis. Ginsenoside Rb3 (G-Rb3) is one of the main components of Ginseng and exhibits poor oral bioavailability but still exerts regulate energy metabolism effects in some diseases. Therefore, the study investigated the effect of chitosan (CS) @ sodium tripolyphosphate (TPP) nanoparticles conjugation with ginsenoside Rb3 (NpRb3) on myocardial fibrosis and studied its possible mechanisms. The results showed that NpRb3 directly participates in the remodeling of myocardial energy metabolism and the regulation of perixisome proliferation-activated receptor alpha (PPARα), thereby improving the degree of myocardial fibrosis. The study also verifies the protective effect of NpRb3 on energy metabolism and mitochondrial function by targeting the PPARα pathway. Therefore, the prepared nanodrug carrier may be a potential solution for the delivery of G-Rb3, which is a promising platform for oral treatment of myocardial fibrosis.
Collapse
Affiliation(s)
- Yi Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Haixia Ji
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Ou Qiao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Zhi Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Lorenzo Pecoraro
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Xueqian Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Xiaoying Han
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Wenzhe Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Xinyu Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Shuli Man
- Tianjin University of Science and Technology, Tianjin, PR China
| | - Juan Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Xia Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Changxiao Liu
- Tianjin Pharmaceutical Research Institute, Tianjin, PR China.
| | - Luqi Huang
- National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, PR China.
| | - Wenyuan Gao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China.
| |
Collapse
|
26
|
MiR-137 regulates low-intensity shear stress-induced human aortic endothelial cell apoptosis via JNK/AP-1 signaling. J Physiol Biochem 2021; 77:451-460. [PMID: 33893994 DOI: 10.1007/s13105-021-00812-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/30/2021] [Indexed: 10/21/2022]
Abstract
The objective of this study is to evaluate the role of miR-137 in low-intensity shear stress-induced endoplasmic reticulum (ER) stress and cell apoptosis in human aortic endothelial cells (HAECs). HAECs were transfected with miR-137 mimic, miR-137 inhibitor, or the corresponding negative control and then exposed to pulsatile shear stress in a parallel-plate flow chamber at 1, 2, 5, 10, and 15 dyn/cm2 for 3 h. Real-time polymerase chain reaction was used to detect mRNA expression of miR-137 and SDS22. A dual-luciferase reporter assay was employed to verify the direct interaction between miR-137 and SDS22. The internal morphology of cells and cell apoptosis was assessed by TUNEL staining observed under a transmission electron microscope. Meanwhile, the protein expression of oxidative stress-related, apoptosis-related, and activated c-Jun N-terminal kinase (JNK)/activator protein-1 (AP-1) signaling-related genes were analyzed by western blotting. Low strength shear stress (0-5 dyn/cm2) caused a negative change of HAEC surface and internal morphology in an intensity-dependent manner, and these changes were gradually weakened when shear stress was increased more than 5 dyn/cm2. Furthermore, low-intensity shear stress promoted oxidative stress response, accelerated cell apoptosis, and upregulated miR-137 expression and JNK/AP-1 signaling in HAECs. MiR-137 directly targets SDS22. Knockdown of miR-137 noticeably reduced activation of JNK/AP-1 signaling, oxidative stress response, and cell apoptosis induced by shear stress. MiR-137 regulated low-intensity shear stress-induced human aortic endothelial cell ER stress and cell apoptosis via JNK/AP-1 signaling.
Collapse
|
27
|
McCarty MF. Nutraceutical, Dietary, and Lifestyle Options for Prevention and Treatment of Ventricular Hypertrophy and Heart Failure. Int J Mol Sci 2021; 22:ijms22073321. [PMID: 33805039 PMCID: PMC8037104 DOI: 10.3390/ijms22073321] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Although well documented drug therapies are available for the management of ventricular hypertrophy (VH) and heart failure (HF), most patients nonetheless experience a downhill course, and further therapeutic measures are needed. Nutraceutical, dietary, and lifestyle measures may have particular merit in this regard, as they are currently available, relatively safe and inexpensive, and can lend themselves to primary prevention as well. A consideration of the pathogenic mechanisms underlying the VH/HF syndrome suggests that measures which control oxidative and endoplasmic reticulum (ER) stress, that support effective nitric oxide and hydrogen sulfide bioactivity, that prevent a reduction in cardiomyocyte pH, and that boost the production of protective hormones, such as fibroblast growth factor 21 (FGF21), while suppressing fibroblast growth factor 23 (FGF23) and marinobufagenin, may have utility for preventing and controlling this syndrome. Agents considered in this essay include phycocyanobilin, N-acetylcysteine, lipoic acid, ferulic acid, zinc, selenium, ubiquinol, astaxanthin, melatonin, tauroursodeoxycholic acid, berberine, citrulline, high-dose folate, cocoa flavanols, hawthorn extract, dietary nitrate, high-dose biotin, soy isoflavones, taurine, carnitine, magnesium orotate, EPA-rich fish oil, glycine, and copper. The potential advantages of whole-food plant-based diets, moderation in salt intake, avoidance of phosphate additives, and regular exercise training and sauna sessions are also discussed. There should be considerable scope for the development of functional foods and supplements which make it more convenient and affordable for patients to consume complementary combinations of the agents discussed here. Research Strategy: Key word searching of PubMed was employed to locate the research papers whose findings are cited in this essay.
Collapse
Affiliation(s)
- Mark F McCarty
- Catalytic Longevity Foundation, 811 B Nahant Ct., San Diego, CA 92109, USA
| |
Collapse
|
28
|
Endoplasmic reticulum stress and unfolded protein response in cardiovascular diseases. Nat Rev Cardiol 2021; 18:499-521. [PMID: 33619348 DOI: 10.1038/s41569-021-00511-w] [Citation(s) in RCA: 421] [Impact Index Per Article: 105.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases (CVDs), such as ischaemic heart disease, cardiomyopathy, atherosclerosis, hypertension, stroke and heart failure, are among the leading causes of morbidity and mortality worldwide. Although specific CVDs and the associated cardiometabolic abnormalities have distinct pathophysiological and clinical manifestations, they often share common traits, including disruption of proteostasis resulting in accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER). ER proteostasis is governed by the unfolded protein response (UPR), a signalling pathway that adjusts the protein-folding capacity of the cell to sustain the cell's secretory function. When the adaptive UPR fails to preserve ER homeostasis, a maladaptive or terminal UPR is engaged, leading to the disruption of ER integrity and to apoptosis. ER stress functions as a double-edged sword, with long-term ER stress resulting in cellular defects causing disturbed cardiovascular function. In this Review, we discuss the distinct roles of the UPR and ER stress response as both causes and consequences of CVD. We also summarize the latest advances in our understanding of the importance of the UPR and ER stress in the pathogenesis of CVD and discuss potential therapeutic strategies aimed at restoring ER proteostasis in CVDs.
Collapse
|
29
|
Liu S, Sun WC, Zhang YL, Lin QY, Liao JW, Song GR, Ma XL, Li HH, Zhang B. SOCS3 Negatively Regulates Cardiac Hypertrophy via Targeting GRP78-Mediated ER Stress During Pressure Overload. Front Cell Dev Biol 2021; 9:629932. [PMID: 33585485 PMCID: PMC7874011 DOI: 10.3389/fcell.2021.629932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/06/2021] [Indexed: 01/17/2023] Open
Abstract
Pressure overload-induced hypertrophic remodeling is a critical pathological process leading to heart failure (HF). Suppressor of cytokine signaling-3 (SOCS3) has been demonstrated to protect against cardiac hypertrophy and dysfunction, but its mechanisms are largely unknown. Using primary cardiomyocytes and cardiac-specific SOCS3 knockout (SOCS3cko) or overexpression mice, we demonstrated that modulation of SOCS3 level influenced cardiomyocyte hypertrophy, apoptosis and cardiac dysfunction induced by hypertrophic stimuli. We found that glucose regulatory protein 78 (GRP78) was a direct target of SOCS3, and that overexpression of SOCS3 inhibited cardiomyocyte hypertrophy and apoptosis through promoting proteasomal degradation of GRP78, thereby inhibiting activation of endoplasmic reticulum (ER) stress and mitophagy in the heart. Thus, our results uncover SOCS3-GRP78-mediated ER stress as a novel mechanism in the transition from cardiac hypertrophy to HF induced by sustained pressure overload, and suggest that modulating this pathway may provide a new therapeutic approach for hypertrophic heart diseases.
Collapse
Affiliation(s)
- Shuang Liu
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Wen-Chang Sun
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yun-Long Zhang
- Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Qiu-Yue Lin
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jia-Wei Liao
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Gui-Rong Song
- Department of Health Statistics, School of Public Health, Dalian Medical University, Dalian, China
| | - Xiao-Lei Ma
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui-Hua Li
- Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bo Zhang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
30
|
Delgado-Valero B, de la Fuente-Chávez L, Romero-Miranda A, Visitación Bartolomé M, Ramchandani B, Islas F, Luaces M, Cachofeiro V, Martínez-Martínez E. Role of endoplasmic reticulum stress in renal damage after myocardial infarction. Clin Sci (Lond) 2021; 135:143-159. [PMID: 33355632 DOI: 10.1042/cs20201137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 02/06/2023]
Abstract
Myocardial infarction (MI) is associated with renal alterations resulting in poor outcomes in patients with MI. Renal fibrosis is a potent predictor of progression in patients and is often accompanied by inflammation and oxidative stress; however, the mechanisms involved in these alterations are not well established. Endoplasmic reticulum (ER) plays a central role in protein processing and folding. An accumulation of unfolded proteins leads to ER dysfunction, termed ER stress. Since the kidney is the organ with highest protein synthesis fractional rate, we herein investigated the effects of MI on ER stress at renal level, as well as the possible role of ER stress on renal alterations after MI. Patients and MI male Wistar rats showed an increase in the kidney injury marker neutrophil gelatinase-associated lipocalin (NGAL) at circulating level or renal level respectively. Four weeks post-MI rats presented renal fibrosis, oxidative stress and inflammation accompanied by ER stress activation characterized by enhanced immunoglobin binding protein (BiP), protein disulfide-isomerase A6 (PDIA6) and activating transcription factor 6-alpha (ATF6α) protein levels. In renal fibroblasts, palmitic acid (PA; 50-200 µM) and angiotensin II (Ang II; 10-8 to 10-6M) promoted extracellular matrix, superoxide anion production and inflammatory markers up-regulation. The presence of the ER stress inhibitor, 4-phenylbutyric acid (4-PBA; 4 µM), was able to prevent all of these modifications in renal cells. Therefore, the data show that ER stress mediates the deleterious effects of PA and Ang II in renal cells and support the potential role of ER stress on renal alterations associated with MI.
Collapse
Affiliation(s)
- Beatriz Delgado-Valero
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid-Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Lucía de la Fuente-Chávez
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid-Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Ana Romero-Miranda
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid-Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - María Visitación Bartolomé
- Departmento de Inmunología, Oftalmología y Otorrinolaringología, Facultad de Psicología, Universidad Complutense Madrid, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Bunty Ramchandani
- Servicio de Cirugía Cardiaca Infantil, Hospital La Paz, Madrid, Spain
| | - Fabián Islas
- Servicio de Cardiología, Instituto Cardiovascular, Hospital Clínico San Carlos, Madrid, Spain
| | - María Luaces
- Servicio de Cardiología, Instituto Cardiovascular, Hospital Clínico San Carlos, Madrid, Spain
| | - Victoria Cachofeiro
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid-Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Ernesto Martínez-Martínez
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid-Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
31
|
Zhang Y, Yang S, Fu J, Liu A, Liu D, Cao S. Inhibition of endoplasmic reticulum stress prevents high-fat diet mediated atrial fibrosis and fibrillation. J Cell Mol Med 2020; 24:13660-13668. [PMID: 33135380 PMCID: PMC7754029 DOI: 10.1111/jcmm.15816] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 06/17/2020] [Accepted: 08/08/2020] [Indexed: 12/14/2022] Open
Abstract
Obesity is a significant risk factor for atrial fibrillation (AF), which is the most common sustained arrhythmia with increased mortality and morbidity. High-fat diet (HFD)-induced obesity is associated with the activation of endoplasmic reticulum stress (ERS). However, the role of ERS in HFD-induced AF remains elusive. Human atrium samples were examined for the ERS activation test. C57BL/6J mice were divided into four groups, including the control group, the HFD group, the 4-phenylbutyric acid (4-PBA) group, and the HFD + 4-PBA group. At the age of 4 weeks, the HFD group and the HFD + 4-PBA group were given HFD to construct the obesity model, while the other two groups were given a normal diet (ND). Transesophageal programmed electrical stimulation was conducted to evaluate the AF inducibility and duration. Atrial fibrosis and ERS activation were also investigated.We found that CHOP and GRP-78 protein were significantly higher in overweight patients than the controls (both P < 0.05). AF inducibility and duration of the HFD group were significantly higher than the other groups (both P < 0.05), while there was no difference between those groups (P > 0.05). The mice of the HFD group had significantly higher collagen volume fraction (CVF%) than the other groups (P < 0.05). ERS marker protein of GRP78, p-PERK, ATF6 and CHOP protein expression level was increased in the HFD group, which were significantly mitigated in the HFD + 4-PBA group. In summary, HFD-induced ERS activation facilitates atrial fibrosis and AF. The inhibition of ERS might alleviate atrial fibrosis and reduce the incidence of AF-associated obesity.
Collapse
Affiliation(s)
- Yan Zhang
- Department of General Practice/VIP Medical ServiceBeijing HospitalNational Center of GerontologyBeijingChina
- Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Shuwen Yang
- Department of General Practice/VIP Medical ServiceBeijing HospitalNational Center of GerontologyBeijingChina
- Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Jing Fu
- Department of General Practice/VIP Medical ServiceBeijing HospitalNational Center of GerontologyBeijingChina
- Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Annan Liu
- Department of General Practice/VIP Medical ServiceBeijing HospitalNational Center of GerontologyBeijingChina
- Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Deping Liu
- Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
- Department of CardiologyBeijing HospitalNational Center of GerontologyBeijingChina
| | - Suyan Cao
- Department of General Practice/VIP Medical ServiceBeijing HospitalNational Center of GerontologyBeijingChina
- Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
32
|
Kennelly JP, Carlin S, Ju T, van der Veen JN, Nelson RC, Buteau J, Thiesen A, Richard C, Willing BP, Jacobs RL. Intestinal Phospholipid Disequilibrium Initiates an ER Stress Response That Drives Goblet Cell Necroptosis and Spontaneous Colitis in Mice. Cell Mol Gastroenterol Hepatol 2020; 11:999-1021. [PMID: 33238221 PMCID: PMC7898069 DOI: 10.1016/j.jcmgh.2020.11.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Patients with ulcerative colitis have low concentrations of the major membrane lipid phosphatidylcholine (PC) in gastrointestinal mucus, suggesting that defects in colonic PC metabolism might be involved in the development of colitis. To determine the precise role that PC plays in colonic barrier function, we examined mice with intestinal epithelial cell (IEC)-specific deletion of the rate-limiting enzyme in the major pathway for PC synthesis: cytidine triphosphate:phosphocholine cytidylyltransferase-α (CTαIKO mice). METHODS Colonic tissue of CTαIKO mice and control mice was analyzed by histology, immunofluorescence, electron microscopy, quantitative polymerase chain reaction, Western blot, and thin-layer chromatography. Histopathologic colitis scores were assigned by a pathologist blinded to the experimental groupings. Intestinal permeability was assessed by fluorescein isothiocyanate-dextran gavage and fecal microbial composition was analyzed by sequencing 16s ribosomal RNA amplicons. Subsets of CTαIKO mice and control mice were treated with dietary PC supplementation, antibiotics, or 4-phenylbutyrate. RESULTS Inducible loss of CTα in the intestinal epithelium reduced colonic PC concentrations and resulted in rapid and spontaneous colitis with 100% penetrance in adult mice. Colitis development in CTαIKO mice was traced to a severe and unresolving endoplasmic reticulum stress response in IECs with altered membrane phospholipid composition. This endoplasmic reticulum stress response was linked to the necroptotic death of IECs, leading to excessive loss of goblet cells, formation of a thin mucus barrier, increased intestinal permeability, and infiltration of the epithelium by microbes. CONCLUSIONS Maintaining the PC content of IEC membranes protects against colitis development in mice, showing a crucial role for IEC phospholipid equilibrium in colonic homeostasis. SRA accession number: PRJNA562603.
Collapse
Affiliation(s)
- John P. Kennelly
- Group on the Molecular and Cell Biology of Lipids, Edmonton, Alberta, Canada,Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada
| | - Stephanie Carlin
- Group on the Molecular and Cell Biology of Lipids, Edmonton, Alberta, Canada,Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada
| | - Tingting Ju
- Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada
| | - Jelske N. van der Veen
- Group on the Molecular and Cell Biology of Lipids, Edmonton, Alberta, Canada,Department of Biochemistry, Edmonton, Alberta, Canada
| | - Randal C. Nelson
- Group on the Molecular and Cell Biology of Lipids, Edmonton, Alberta, Canada,Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada,Department of Biochemistry, Edmonton, Alberta, Canada
| | - Jean Buteau
- Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada
| | - Aducio Thiesen
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Caroline Richard
- Group on the Molecular and Cell Biology of Lipids, Edmonton, Alberta, Canada,Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada
| | - Ben P. Willing
- Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada
| | - René L. Jacobs
- Group on the Molecular and Cell Biology of Lipids, Edmonton, Alberta, Canada,Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada,Department of Biochemistry, Edmonton, Alberta, Canada,Correspondence Address correspondence to: René L. Jacobs, PhD, Department of Agricultural, Food and Nutritional Science, 4-002E Li Ka Shing Centre for Health Research and Innovation, University of Alberta, Alberta, T6G2E1 Canada. fax: (780) 492-2343.
| |
Collapse
|
33
|
Mattke AC, Shikata F, McGill J, Justo R, Venugopal P. Successful management of a neonate with OTC deficiency presenting with hyperammonemia and severe cardiac dysfunction with extracorporeal membrane oxygenation support and continuous renal replacement therapy. JIMD Rep 2020; 55:12-14. [PMID: 32905004 PMCID: PMC7463050 DOI: 10.1002/jmd2.12135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/22/2020] [Accepted: 02/24/2020] [Indexed: 11/24/2022] Open
Abstract
Ornithine transcarbamylase (OTC) deficiency is an X-linked urea cycle disorder which-in severe form-results in rapid accumulation of ammonia and glutamine with subsequent irreversible brain injury. We present a case of severe left ventricular dysfunction with hyperammonemic crisis caused by OTC deficiency which was managed with veno-arterial extracorporeal membrane oxygenation support combined with continuous renal replacement therapy. Aggressive treatment led to normalization of ammonia and full left ventricular recovery.
Collapse
Affiliation(s)
- Adrian C. Mattke
- Paediatric Intensive Care Unit, Queensland Children's HospitalSouth BrisbaneQueenslandAustralia
- Queensland Pediatric Cardiac Service, Queensland Children's HospitalSouth BrisbaneQueenslandAustralia
- University of Queensland, Brisbane, School of MedicineSt LuciaQueenslandAustralia
- Paediatric Critical Care Research Group, CCHRUniversity of QueenslandSt LuciaQueenslandAustralia
| | - Fumiaki Shikata
- Queensland Pediatric Cardiac Service, Queensland Children's HospitalSouth BrisbaneQueenslandAustralia
| | - James McGill
- Metabolic Medicine, Queensland Children's HospitalSouth BrisbaneQueenslandAustralia
| | - Rob Justo
- Queensland Pediatric Cardiac Service, Queensland Children's HospitalSouth BrisbaneQueenslandAustralia
- University of Queensland, Brisbane, School of MedicineSt LuciaQueenslandAustralia
| | - Prem Venugopal
- Queensland Pediatric Cardiac Service, Queensland Children's HospitalSouth BrisbaneQueenslandAustralia
- University of Queensland, Brisbane, School of MedicineSt LuciaQueenslandAustralia
- Paediatric Critical Care Research Group, CCHRUniversity of QueenslandSt LuciaQueenslandAustralia
| |
Collapse
|
34
|
Wang C, Pan Z. Hydrogen-rich saline mitigates pressure overload-induced cardiac hypertrophy and atrial fibrillation in rats via the JAK-STAT signalling pathway. J Int Med Res 2020; 48:300060520936415. [PMID: 32762484 PMCID: PMC7416141 DOI: 10.1177/0300060520936415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objective To investigate if hydrogen-rich saline (HRS), which has been shown to have
antioxidant and anti-inflammatory properties, could mitigate cardiac
remodelling and reduce the incidence of atrial fibrillation (AF) in the rat
model of cardiac hypertrophy. Methods Pressure overload was induced in rats by abdominal aortic constriction (AAC).
The animals were separated into four groups: sham; AAC group; AAC plus low
dose HRS (LHRS); AAC plus high dose HRS (HHRS). The sham and AAC groups
received normal saline intraperitoneally and the LHRS and HHRS groups
received 3 or 6 ml/kg HRS daily for six weeks, respectively. In
vitro research was also performed using cardiotrophin-1
(CT-1)-induced hypertrophy of cultured neonatal rat cardiomyocytes. Results Cardiac hypertrophy was successfully induced by AAC and low and high dose HRS
mitigated the pressure overload as shown by lower heart and atrial weights
in these treatment groups. AF incidence and duration of the HRS groups were
also significantly lower in the HRS groups compared with the AAC group.
Atrial fibrosis was also reduced in the HRS groups and the JAK-STAT
signalling pathway was down-regulated. In vitro experiments
showed that hydrogen-rich medium mitigated the CT-1-induced cardiomyocyte
hypertrophy with a similar effect as the JAK specific antagonists AG490. Conclusions HRS was found to mitigate cardiac hypertrophy induced by pressure overload in
rats and reduce atrial fibrosis and AF which was possibly achieved via
inhibition of the JAK-STAT signalling pathway.
Collapse
Affiliation(s)
- Chufeng Wang
- Clinical medicine and biomedicine, Nanchang Joint Program, Queen Mary University of London, Nanchang, Jiangxi, China
| | - Zezheng Pan
- Department of Biochemistry and Molecular Biology, Medical Faculty of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
35
|
Histone Deacetylase Inhibition Attenuates Aortic Remodeling in Rats under Pressure Overload. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4705615. [PMID: 32775424 PMCID: PMC7397417 DOI: 10.1155/2020/4705615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 06/22/2020] [Accepted: 06/26/2020] [Indexed: 11/18/2022]
Abstract
The use of histone deacetylase (HDAC) inhibitor is a novel therapeutic strategy for cardiovascular disease. Studies have shown that many HDAC inhibitors have the ability to reduce the aortic remodeling in various animal models. We hypothesized that the HDAC inhibitor, MGCD0103 (MGCD), attenuates aortic remodeling in rats under pressure overload-induced by transverse aortic constriction (TAC). The aortic ring tension analysis was conducted using the thoracic aorta. Sections of the aorta were visualized after hematoxylin and eosin, trichrome, and Verhoeff-van Gieson staining, and immunohistochemistry. The expression of genes related to aortic remodeling (αSMA, Mmp2, and Mmp9) and angiotensin receptors (Agtr1 and Agtr2) was determined by quantitative real-time polymerase chain reaction. There was a significant decrease in relaxation of the aorta when treated with MGCD. Fibrosis of the aortic wall and expression of angiotensin receptors increased in TAC rats, which was attenuated by MGCD. These results indicate that MGCD, an HDAC inhibitor, attenuates aortic remodeling in rats with TAC-induced pressure overload rats and may serve as a potential therapeutic target of antiaortic remodeling in pressure overload-induced hypertension-related diseases.
Collapse
|
36
|
Xue X, Ling X, Xi W, Wang P, Sun J, Yang Q, Xiao J. Exogenous hydrogen sulfide reduces atrial remodeling and atrial fibrillation induced by diabetes mellitus via activation of the PI3K/Akt/eNOS pathway. Mol Med Rep 2020; 22:1759-1766. [PMID: 32705232 PMCID: PMC7411292 DOI: 10.3892/mmr.2020.11291] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/14/2020] [Indexed: 11/06/2022] Open
Abstract
Diabetes mellitus (DM) facilitates atrial fibrosis and increases the risk of atrial fibrillation (AF). The underlying mechanism of DM in causing AF remains mostly unknown and potential therapeutic targets for DM‑induced AF are rarely reported. Hydrogen sulfide (H2S) has drawn considerable attention in recent years for its potential as a cardiovascular protector. Thus, the aim of the present study was to investigate the effect of H2S on DM‑induced AF and the mechanism of action. Sprague‑Dawley rats were divided into four groups, including the control group, the DM group, the H2S group and the DM+H2S group. The DM group and the DM+H2S group were administered streptozotocin to induce DM, whereas the other two groups were given citrate buffer as a control. The H2S group and the DM+H2S group were administered with an intraperitoneal injection of sodium hydrosulfide (precursor of H2S). AF inducibility, AF duration, atrial fibrosis and vital protein expression of oxidative stress were compared among the four groups. The DM group showed significantly higher AF incidence rates and duration (P<0.05). Histology results demonstrated severe atrial fibrosis in the DM group, and the PI3K/Akt/endothelial nitric oxide synthase (eNOS) pathway was significantly downregulated (P<0.05). However, when H2S was administered, the rats showed lower AF incidence and duration compared with the DM group. Additionally, H2S was able to mitigate the atrial fibrosis induced by DM, as well as the proliferation and migration of cardiac fibroblasts, as demonstrated by an MTT assay and real‑time cell analyzer migration experiment. Western blotting showed that the expression levels of the PI3K/Akt/eNOS pathway in the DM+H2S group were significantly upregulated compared with those of the DM group (P<0.05). In summary, DM status can lead to the structural remodeling of atrial fibrosis, facilitating AF incidence and persistence. Administration of H2S does not affect the glucose level, but can significantly mitigate atrial fibrosis and reduce the incidence of AF induced by DM, probably via activation of the PI3K/Akt/eNOS pathway.
Collapse
Affiliation(s)
- Xiaofei Xue
- Center for Comprehensive Treatment of Atrial Fibrillation, Department of Cardiothoracic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Xinyu Ling
- Center for Comprehensive Treatment of Atrial Fibrillation, Department of Cardiothoracic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Wang Xi
- Center for Comprehensive Treatment of Atrial Fibrillation, Department of Cardiothoracic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Pei Wang
- Center for Comprehensive Treatment of Atrial Fibrillation, Department of Cardiothoracic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Jianjun Sun
- Department of Rehabilitation, Elderly Rehabilitation Hospital, Suzhou Red Cross Society, Suzhou, Jiangsu 215009, P.R. China
| | - Qian Yang
- Center for Comprehensive Treatment of Atrial Fibrillation, Department of Cardiothoracic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Jian Xiao
- Center for Comprehensive Treatment of Atrial Fibrillation, Department of Cardiothoracic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
37
|
Long F, Wang Q, Yang D, Zhu M, Wang J, Zhu Y, Liu X. Targeting JMJD3 histone demethylase mediates cardiac fibrosis and cardiac function following myocardial infarction. Biochem Biophys Res Commun 2020; 528:671-677. [PMID: 32513540 DOI: 10.1016/j.bbrc.2020.05.115] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 05/17/2020] [Indexed: 01/19/2023]
Abstract
Myocardial fibrosis is the pathological consequence of injury-induced fibroblastto-myofibroblast transition, resulting in increased stiffness and diminished cardiac function. Histone modification has been shown to play an important role in the pathogenesis of cardiac fibrosis. Here, we identified H3K27me3 demethylase JMJD3/KDM6B promotes cardiac fibrosis via regulation of fibrogenic pathways. Using neonatal rat cardiac fibroblasts (NRCF), we show that the expression of endogenous JMJD3 is induced by angiotensin II (Ang II), while the principle extracellular matrix (ECM) such as fibronectin, CTGF, collagen I and III are increased. We find that JMJD3 inhibition markedly enhances the suppressive mark (H3K27me3) at the beta (β)-catenin promoter in activated cardiac fibroblasts, and then substantially decreases expression of fibrogenic gene. Both inhibition of β-catenin-mediated transcription with ICG-001 and genetic loss of β-catenin can prevent Ang II-induced ECM deposition. Most importantly, in vivo inhibition of JMJD3 rescues myocardial ischemia-induced cardiac fibrosis and cardiac dysfunction. Collectively, our findings are the first to report a novel role of histone demethylase JMJD3 in the pro-fibrotic cardiac fibroblast phenotype, pharmacological targeting of JMJD3 might represent a promising therapeutic approach for the treatment of human cardiac fibrosis and other fibrotic diseases.
Collapse
Affiliation(s)
- Fen Long
- Department of Pharmacology, School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, PR China
| | - Qing Wang
- Department of Pharmacology, School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, PR China
| | - Di Yang
- Department of Pharmacology, School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, PR China
| | - Menglin Zhu
- Department of Pharmacology, School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, PR China
| | - Jinghuan Wang
- Department of Pharmacology, School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, PR China
| | - YiZhun Zhu
- Department of Pharmacology, School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, PR China; State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, PR China
| | - Xinhua Liu
- Department of Pharmacology, School of Pharmacy, Human Phenome Institute, Fudan University, Shanghai, 201203, PR China.
| |
Collapse
|
38
|
Kriegermeier A, Green R. Pediatric Cholestatic Liver Disease: Review of Bile Acid Metabolism and Discussion of Current and Emerging Therapies. Front Med (Lausanne) 2020; 7:149. [PMID: 32432119 PMCID: PMC7214672 DOI: 10.3389/fmed.2020.00149] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 04/06/2020] [Indexed: 02/06/2023] Open
Abstract
Cholestatic liver diseases are a significant cause of morbidity and mortality and the leading indication for pediatric liver transplant. These include diseases such as biliary atresia, Alagille syndrome, progressive intrahepatic cholestasis entities, ductal plate abnormalities including Caroli syndrome and congenital hepatic fibrosis, primary sclerosing cholangitis, bile acid synthesis defects, and certain metabolic disease. Medical management of these patients typically includes supportive care for complications of chronic cholestasis including malnutrition, pruritus, and portal hypertension. However, there are limited effective interventions to prevent progressive liver damage in these diseases, leaving clinicians to ultimately rely on liver transplantation in many cases. Agents such as ursodeoxycholic acid, bile acid sequestrants, and rifampicin have been mainstays of treatment for years with the understanding that they may decrease or alter the composition of the bile acid pool, though clinical response to these medications is frequently insufficient and their effects on disease progression remain limited. Recently, animal and human studies have identified potential new therapeutic targets which may disrupt the enterohepatic circulation of bile acids, alter the expression of bile acid transporters or decrease the production of bile acids. In this article, we will review bile formation, bile acid signaling, and the relevance for current and newer therapies for pediatric cholestasis. We will also highlight further areas of potential targets for medical intervention for pediatric cholestatic liver diseases.
Collapse
Affiliation(s)
- Alyssa Kriegermeier
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago, IL, United States
| | - Richard Green
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
39
|
Zhang Y, Chen W, Wang Y. STING is an essential regulator of heart inflammation and fibrosis in mice with pathological cardiac hypertrophy via endoplasmic reticulum (ER) stress. Biomed Pharmacother 2020; 125:110022. [PMID: 32106379 DOI: 10.1016/j.biopha.2020.110022] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/10/2020] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Pathological cardiac hypertrophy is characterized by myocyte enlargement and cardiac dysfunction. However, the pathogenesis for this disease is still poorly understood. Stimulator of interferon genes (STING) could meditate inflammation and immune response in various kinds of diseases. In this work, we demonstrated that STING was critical for pressure overload-induced cardiac hypertrophy. Results showed that STING expression was up-regulated in human and mouse hypertrophic hearts. STING knockout attenuated cardiac hypertrophy induced by aortic banding (AB). The effects of STING deficiency on the improvement of cardiac hypertrophy and dysfunction were associated with the restrained macrophage infiltration, inflammatory response and fibrosis. Moreover, ER stress was detected in hearts of AB-operated mice, as evidenced by the increased expression of phospho-protein kinase RNA-like endoplasmic reticulum kinase (PERK), phospho-eukaryotic initiation factor 2 alpha (eIF2α) and phospho-inositol-requiring kinase (IRE)-1α. Importantly, these proteins were restrained in mice with STING knockout after AB surgery. What's more, angiotensin II (Ang II)-induced STING could be accelerated by ER stress activator, while being markedly abolished by the ER stress inhibitor. We then found that whether co-treated with or without transforming growth factor-beta 1 (TGF-β1), cardiac fibroblasts cultured in the conditional medium (CM) from Ang II-incubated cardiomyocytes with STING knockdown exhibited significantly reduced fibrosis, as displayed by the clearly down-regulated expression of α-SMA, Collagen type I (Col I) and Collagen type III (Col III). Therefore, we defined STING as an important signal contributing to cardiac hypertrophy closely associated with ER stress.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, 710004, China
| | - Wenzhong Chen
- Department of Cardiovascular Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Yan Wang
- Department of General Medicine, The Second Affiliated Hospital of Shenzhen University(People's Hospital of Shenzhen Baoan District), Shenzhen City, 518101, China.
| |
Collapse
|
40
|
Sabirli R, Koseler A, Mansur N, Zeytunluoglu A, Sabirli GT, Turkcuer I, Kilic ID. Predictive Value of Endoplasmic Reticulum Stress Markers in Low Ejection Fractional Heart Failure. In Vivo 2020; 33:1581-1592. [PMID: 31471408 DOI: 10.21873/invivo.11640] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND/AIM Endoplasmic reticulum (ER) stress plays a critical role in the development of cardiac hypertrophy and heart failure. Heart failure is a crucial health problem that affects 23 million people worldwide, causes approximately 2.4 million people to be hospitalized every year in the USA, and leads to the death of more than 300,000 people. In this study, we aimed to investigate the clinical significance of ER stress markers and the predictive value of acute decompensated heart failure in patients with low ejection fraction heart failure (ADHF). PATIENTS AND METHODS This is a prospective case control study. The data included laboratory parameters pertaining to patients with ADHF in the emergency service and lipid parameters obtained during their admission to the hospital. In addition, the same parameters obtained from the control group patients with chronic heart failure (CHF) during their routine polyclinic control were recorded in the data set. Admission time to the hospital and length of hospital stay were included in the data. The levels of glucose regulated protein (GRP78), protein kinase RNA-like endoplasmic reticulum kinase (PERK), and C/EBP homologous protein (CHOP) in peripheral blood serum obtained from the patients and the control group were measured using the ELISA method. RESULTS Serum GRP78 concentration was lower in the HF group (p=0.003) compared to the control. The median value of serum PERK concentration in the HF group was higher than that of the control group (573 pg/ml, IQR=477.5-650 vs. 495.5 pg/ml, IQR=294-648, respectively) (p=0.001). However, there were no statistically significant differences in GRP78 and PERK serum concentrations between ADHF and CHF subgroups. Receiver operating characteristic (ROC) curve analysis showed greater area under the curve (AUC) for the serum GRP78 levels of the healthy individuals (AUC=0.748, 95% CI=0.681-0.814, p=0.0003). The serum GRP78 level was found to be 80% sensitive and 70% specific at 147.5 pg/ml (p=0.0003) for distinguishing healthy individuals from HF patients. In the ADHF subgroup, there was a moderate correlation between hospitalization time and serum CHOP concentrations (Spearman rho=0.586 and p=0.001). CONCLUSION High GRP78 serum concentration may protect the patient from ER stress. In addition, the serum PERK level is high in patients with HF, whereas it is insufficient in predicting acute decompensation. CHOP may be useful in predicting the length of hospital stay in patients with ADHF.
Collapse
Affiliation(s)
- Ramazan Sabirli
- Servergazi State Hospital, Department of Emergency Medicine, Denizli, Turkey
| | - Aylin Koseler
- Department of Biophysics, Pamukkale University Medical Faculty, Denizli, Turkey
| | - Nesteren Mansur
- Department of Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ali Zeytunluoglu
- Department of Electronic and Automation, Denizli Vocational School of Technical Sciences, Pamukkale University, Denizli, Turkey
| | | | - Ibrahim Turkcuer
- Department of Emergency Medicine, Pamukkale University Medical Faculty, Denizli, Turkey
| | - Ismail Dogu Kilic
- Department of Cardiology, Pamukkale University Medical Faculty, Denizli, Turkey
| |
Collapse
|
41
|
Huoxue Qianyang decoction ameliorates cardiac remodeling in obese spontaneously hypertensive rats in association with ATF6-CHOP endoplasmic reticulum stress signaling pathway regulation. Biomed Pharmacother 2019; 121:109518. [PMID: 31689600 DOI: 10.1016/j.biopha.2019.109518] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/10/2019] [Accepted: 10/01/2019] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Endoplasmic reticulum (ER) stress is involved in hypertension related cardiac remodeling. We aimed to evaluate the effects of Huoxue Qianyang (HXQY) decoction on cardiac remodeling in obese spontaneously hypertensive rats (SHRs), and explore its impacts on the activating transcription factor 6 (ATF6)-C/EBP homologous protein (CHOP) ER stress signaling pathway. METHODS Twenty-seven obese SHRs were randomly divided into Obese SHR, Obese SHR + HXQY and Obese SHR + Valsartan groups, and treated with the indicated drugs for 8 weeks. Nine age-matched male SHRs were used as controls. Systolic blood pressure (SBP), body weight (BW), and the left ventricular mass index (LVMI) were measured weekly or at end point. Then, angiotensin II (Ang II), fasting glucose (FPG) and fasting insulin (FIN), total cholesterol (TC), LDL-cholesterol (LDL-C), HDL-cholesterol (HDL-C) and triglyceride (TG) levels were evaluated with commercial kits. Apoptotic cardiomyocytes were detected by the terminal deoxynucleotidyl transferase mediated dUTP nick end labeling (TUNEL) method. The expression levels of GRP78, ATF6, PERK/pPERK and CHOP were assessed by quantitative PCR and Western blot. RESULTS Treatment with HXQY decoction resulted in significantly reduced SBP, BW, LVMI, Ang II, TC and LDL-C levels, as well as the homeostasis model assessment of insulin resistance (HOMA-IR) score in obese SHRs. Apoptosis in heart tissues of obese SHRs was significantly attenuated after HXQY decoction administration, paralleling reduced expression of GRP78, ATF6, PERK/pPERK and CHOP at both mRNA and protein levels. CONCLUSION Cardiac remodeling in obese SHRs is ameliorated by intervention with HXQY decoction in association with inhibited ATF6-CHOP ER stress signaling pathway.
Collapse
|
42
|
Ge CX, Xu MX, Qin YT, Gu TT, Lou DS, Li Q, Hu LF, Wang BC, Tan J. Endoplasmic reticulum stress-induced iRhom2 up-regulation promotes macrophage-regulated cardiac inflammation and lipid deposition in high fat diet (HFD)-challenged mice: Intervention of fisetin and metformin. Free Radic Biol Med 2019; 141:67-83. [PMID: 31153974 DOI: 10.1016/j.freeradbiomed.2019.05.031] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/04/2019] [Accepted: 05/29/2019] [Indexed: 12/15/2022]
Abstract
Endoplasmic reticulum stress (ERS) has been implicated in obesity-associated cardiac remodeling and dysfunction. Inactive rhomboid protein 2 (iRhom2), also known as Rhbdf2, is an inactive member of the rhomboid intramembrane proteinase family, playing an essential role in regulating inflammation. Nevertheless, the role of ERS-meditated iRhom2 pathway in metabolic stress-induced cardiomyopathy remains unknown. In the study, we showed that 4-PBA, as an essential ERS inhibitor, significantly alleviated high fat diet (HFD)-induced metabolic disorder and cardiac dysfunction in mice. Additionally, lipid deposition in heart tissues was prevented by 4-PBA in HFD-challenged mice. Moreover, 4-PBA blunted the expression of iRhom2, TACE, TNFR2 and phosphorylated NF-κB to prevent HFD-induced expression of inflammatory factors. Further, 4-PBA restrained HFD-triggered oxidative stress by promoting Nrf-2 signaling. Importantly, 4-PBA markedly suppressed cardiac ERS in HFD mice. The anti-inflammation, anti-ERS and anti-oxidant effects of 4-PBA were verified in palmitate (PAL)-incubated macrophages and cardiomyocytes. In addition, promoting ERS could obviously enhance iRhom2 signaling in vitro. Intriguingly, our data demonstrated that PAL-induced iRhom2 up-regulation apparently promoted macrophage to generate inflammatory factors that could promote cardiomyocyte inflammation and lipid accumulation. Finally, interventions by adding fisetin or metformin significantly abrogated metabolic stress-induced cardiomyopathy through the mechanisms mentioned above. In conclusion, this study provided a novel mechanism for metabolic stress-induced cardiomyopathy pathogenesis. Therapeutic strategy to restrain ROS/ERS/iRhom2 signaling pathway could be developed to prevent myocardial inflammation and lipid deposition, consequently alleviating obesity-induced cardiomyopathy.
Collapse
Affiliation(s)
- Chen-Xu Ge
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China
| | - Min-Xuan Xu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China.
| | - Yu-Ting Qin
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, PR China
| | - Ting-Ting Gu
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210023, PR China
| | - De-Shuai Lou
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China
| | - Qiang Li
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China
| | - Lin-Feng Hu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China
| | - Bo-Chu Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, China.
| | - Jun Tan
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China.
| |
Collapse
|
43
|
Jung H, Lee E, Kim I, Song JH, Kim GJ. Histone deacetylase inhibition has cardiac and vascular protective effects in rats with pressure overload cardiac hypertrophy. Physiol Res 2019; 68:727-737. [PMID: 31424255 DOI: 10.33549/physiolres.934110] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors have shown beneficial effects in animal models of cardiovascular diseases. We hypothesized that HDAC inhibitor, sodium valproate (VPA), has cardiac and vascular protective effects in rats with pressure overload cardiac hypertrophy induced by transverse aortic constriction (TAC). Sections of the heart were visualized after hematoxylin and eosin staining, picrosirius red staining and immunohistochemistry. The expression of genes related to cardiac hypertrophy, fibrosis, and oxidative stress was determined by quantitative real-time polymerase chain reaction. The aortic ring tension analysis was conducted using both the ascending aorta and descending thoracic aorta. TAC increased the expression of hypertrophic, fibrotic, and oxidative stress genes, which was attenuated by VPA. In the ascending aorta with intact endothelium, there was a significant decrease in the relaxation response, which was recovered by VPA treatment. These results indicate that VPA has cardiac and vascular protective effects in rats with pressure overload cardiac hypertrophy.
Collapse
Affiliation(s)
- H Jung
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.
| | | | | | | | | |
Collapse
|
44
|
Xiao D, Zhang Y, Wang R, Fu Y, Zhou T, Diao H, Wang Z, Lin Y, Li Z, Wen L, Kang X, Kopylov P, Shchekochikhin D, Zhang Y, Yang B. Emodin alleviates cardiac fibrosis by suppressing activation of cardiac fibroblasts via upregulating metastasis associated protein 3. Acta Pharm Sin B 2019; 9:724-733. [PMID: 31384533 PMCID: PMC6664101 DOI: 10.1016/j.apsb.2019.04.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/21/2019] [Accepted: 04/02/2019] [Indexed: 12/15/2022] Open
Abstract
Excess activation of cardiac fibroblasts inevitably induces cardiac fibrosis. Emodin has been used as a natural medicine against several chronic diseases. The objective of this study is to determine the effects of emodin on cardiac fibrosis and the underlying molecular mechanisms. Intragastric administration of emodin markedly decreased left ventricular wall thickness in a mouse model of pathological cardiac hypertrophy with excess fibrosis induced by transaortic constriction (TAC) and suppressed activation of cardiac fibroblasts induced by angiotensin II (AngII). Emodin upregulated expression of metastasis associated protein 3 (MTA3) and restored the MTA3 expression in the setting of cardiac fibrosis. Moreover, overexpression of MTA3 promoted cardiac fibrosis; in contrast, silence of MTA3 abrogated the inhibitory effect of emodin on fibroblast activation. Our findings unraveled the potential of emodin to alleviate cardiac fibrosis via upregulating MTA3 and highlight the regulatory role of MTA3 in the development of cardiac fibrosis.
Collapse
|
45
|
Mohamed AAA, Yang D, Liu S, Lin P, Mohamad OAA, Jin Y. Endoplasmic reticulum stress is involved in lipopolysaccharide-induced inflammatory response and apoptosis in goat endometrial stromal cells. Mol Reprod Dev 2019; 86:908-921. [PMID: 31041824 DOI: 10.1002/mrd.23152] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/22/2019] [Accepted: 04/04/2019] [Indexed: 02/06/2023]
Abstract
Endoplasmic reticulum (ER) stress is involved in regulating cell metabolism, apoptosis, autophagy, and survival. However, there is not enough information about the role of ER stress in lipopolysaccharide (LPS)-induced apoptosis and inflammatory cytokine secretion in the uterus. In this study, we found that LPS induced apoptosis and inflammation in goat endometrial stromal cells (ESCs). LPS treatment inhibited cell viability and cell proliferation. In addition, the genes associated with proliferation, such as proliferating cell nuclear antigen and MKI67, were affected by LPS treatment. Moreover, LPS increased the secretion of interleukin (IL)-1β and IL-8, promoting the levels of MYD88, caspase1, and TRL4. The 4-phenylbutyric acid pretreatment inhibited the expression of unfolded protein response proteins and the secretion of inflammatory cytokines in LPS-treated cells. However, blockage of inositol-requiring enzyme 1 and activating transcription factor 6 did not significantly reduce apoptosis and inflammatory cytokine secretion. Collectively, ER stress involved in LPS-induced apoptosis and inflammatory cytokine increased in goat ESCs. This study provides new insight into the function of ER stress in the pathological process.
Collapse
Affiliation(s)
- Amira Abdalla Abdelshafy Mohamed
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Animal Production, College of Environmental Agricultural Sciences, Arish University, Al-Arish, North-Sinai, Egypt
| | - Diqi Yang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Shouqin Liu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Pengfei Lin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Osama Abdalla Abdelshafy Mohamad
- Department of Biological, Marine Sciences, and Environmental Agriculture, Institute for Post Graduate Environmental Studies, Arish University, Al-Arish, North-Sinai, Egypt
| | - Yaping Jin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
46
|
Shi L, Zhu D, Wang S, Jiang A, Li F. Dapagliflozin Attenuates Cardiac Remodeling in Mice Model of Cardiac Pressure Overload. Am J Hypertens 2019; 32:452-459. [PMID: 30689697 DOI: 10.1093/ajh/hpz016] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/10/2018] [Accepted: 01/18/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Dapagliflozin (DAPA) is an inhibitor of sodium-glucose cotransporter 2 prescribed for type 2 diabetes mellitus. DAPA plays a protective role against cardiovascular diseases. Nevertheless, the effect and mechanism of DAPA on pressure-overload-induced cardiac remodeling has not been determined. METHODS We used a transverse aortic constriction (TAC) induced cardiac remodeling model to evaluate the effect of DAPA. Twenty-four C57BL/6J mice were divided into 3 groups: Sham, TAC, and TAC + DAPA groups (n = 8, each). DAPA was administered by gavage (1.0 mg/kg/day) for 4 weeks in the TAC + DAPA group, and then the myocardial hypertrophy, cardiac systolic function, myocardial fibrosis, and cardiomyocyte apoptosis were evaluated. RESULTS Mice in TAC group showed increased heart weight/body weight, left ventricular (LV) diameter, LV posterior wall thickness, and decreased LV ejection fraction and LV fractional shortening. The collagen volume fraction and perivascular collagen area/luminal area ratio were significantly greater in the TAC group; the TUNEL-positive cell number and PARP level were also increased. We found that DAPA treatment reduced myocardial hypertrophy, myocardial interstitial and perivascular fibrosis, and cardiomyocyte apoptosis. Furthermore, DAPA administration inhibited phosphorylation of P38 and JNK in TAC group. In addition, the inhibited phosphorylation of FoxO1 in the TAC mice was upregulated by DAPA administration. CONCLUSION DAPA administration had a cardioprotective effect by improving cardiac systolic function, inhibiting myocardial fibrosis and cardiomyocyte apoptosis in a TAC mouse model, indicating that it could serve as a new therapy to prevent pathological cardiac remodeling in nondiabetics.
Collapse
Affiliation(s)
- Lin Shi
- Department of Cardiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Diqi Zhu
- Department of Cardiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shoubao Wang
- Department of Cardiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aixia Jiang
- Department of Cardiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fen Li
- Department of Cardiology, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Pediatric Congenital Heart Disease Institute, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
47
|
Yusoff SI, Roman M, Lai FY, Eagle-Hemming B, Murphy GJ, Kumar T, Wozniak M. Systematic review and meta-analysis of experimental studies evaluating the organ protective effects of histone deacetylase inhibitors. Transl Res 2019; 205:1-16. [PMID: 30528323 PMCID: PMC6386580 DOI: 10.1016/j.trsl.2018.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/09/2018] [Accepted: 11/13/2018] [Indexed: 01/07/2023]
Abstract
The clinical efficacy of organ protection interventions are limited by the redundancy of cellular activation mechanisms. Interventions that target epigenetic mechanisms overcome this by eliciting genome wide changes in transcription and signaling. We aimed to review preclinical studies evaluating the organ protection effects of histone deacetylase inhibitors (HDACi) with a view to informing the design of early phase clinical trials. A systematic literature search was performed. Methodological quality was assessed against prespecified criteria. The primary outcome was mortality, with secondary outcomes assessing mechanisms. Prespecified analyses evaluated the effects of likely moderators on heterogeneity. The analysis included 101 experimental studies in rodents (n = 92) and swine (n = 9), exposed to diverse injuries, including: ischemia (n = 72), infection (n = 7), and trauma (n = 22). There were a total of 448 comparisons due to the evaluation of multiple independent interventions within single studies. Sodium valproate (VPA) was the most commonly evaluated HDACi (50 studies, 203 comparisons). All of the studies were judged to have significant methodological limitations. HDACi reduced mortality in experimental models of organ injury (risk ratio = 0.52, 95% confidence interval 0.40-0.68, p < 0.001) without heterogeneity. HDACi administration resulted in myocardial, brain and kidney protection across diverse species and injuries that was attributable to increases in prosurvival cell signaling, and reductions in inflammation and programmed cell death. Heterogeneity in the analyses of secondary outcomes was explained by differences in species, type of injury, HDACi class (Class I better), drug (trichostatin better), and time of administration (at least 6 hours prior to injury better). These findings highlight a potential novel application for HDACi in clinical settings characterized by acute organ injury.
Collapse
Affiliation(s)
- Syabira I Yusoff
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK.
| | - Marius Roman
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Florence Y Lai
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Bryony Eagle-Hemming
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Gavin J Murphy
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Tracy Kumar
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Marcin Wozniak
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| |
Collapse
|
48
|
Hu WS, Ting WJ, Tamilselvi S, Day CH, Wang T, Chiang WD, Viswanadha VP, Yeh YL, Lin WT, Huang CY. Oral administration of alcalase potato protein hydrolysate-APPH attenuates high fat diet-induced cardiac complications via TGF-β/GSN axis in aging rats. ENVIRONMENTAL TOXICOLOGY 2019; 34:5-12. [PMID: 30240538 DOI: 10.1002/tox.22651] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/23/2018] [Accepted: 08/25/2018] [Indexed: 06/08/2023]
Abstract
Consumption of high fat diet (HFD) is associated with increased cardiovascular risk factors among elderly people. Aging and obesity induced-cardiac remodeling includes hypertrophy and fibrosis. Gelsolin (GSN) induces cardiac hypertrophy and TGF-β, a key cytokine, which induces fibrosis. The relationship between TGF-β and GSN in aging induced cardiac remodeling is still unknown. We evaluated the expressions of TGF-β and GSN in HFD fed 22 months old aging SD rats, followed by the administration of either probucol or alcalase potato protein hydrolysate (APPH). Western blotting and Masson trichrome staining showed that APPH (45 and 75 mg/kg/day) and probucol (500 mg/kg/day) treatments significantly reduced the aging and HFD-induced hypertrophy and fibrosis. Echocardiograph showed that the performance of the hearts was improved in APPH, and probucol treated HFD aging rats. Serum from all rats was collected and H9c2 cells were cultured with collected serums separately. The GSN dependent hypertrophy was inhibited with an exogenous TGF-β in H9c2 cells cultured in HFD+ APPH treated serum. Thus, we propose that along with its role in cardiac fibrosis, TGF-β also acts as an upstream activator of GSN dependent hypertrophy. Hence, TGF-β in serum could be a promising therapeutic target for cardiac remodeling in aging and/or obese subjects.
Collapse
Affiliation(s)
- Wei Syun Hu
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University, Hospital, Taichung, Taiwan
| | - Wei Jen Ting
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Shanmugam Tamilselvi
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | | | - Ting Wang
- Department of hospitality management, College of Agriculture, Tunghai University, Taichung, Taiwan
| | - Wen-Dee Chiang
- Department of Food science, College of Agriculture, Tunghai University, Taichung, Taiwan
| | | | - Yu Lan Yeh
- Department of pathology, Changhua Christian Hospital, Changhua, Taiwan
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Wan Teng Lin
- Department of hospitality management, College of Agriculture, Tunghai University, Taichung, Taiwan
| | - Chih Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
49
|
Bozi LH, Takano AP, Campos JC, Rolim N, Dourado PM, Voltarelli VA, Wisløff U, Ferreira JC, Barreto-Chaves ML, Brum PC. Endoplasmic reticulum stress impairs cardiomyocyte contractility through JNK-dependent upregulation of BNIP3. Int J Cardiol 2018; 272:194-201. [DOI: 10.1016/j.ijcard.2018.08.070] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 08/22/2018] [Indexed: 12/22/2022]
|
50
|
Xu XX, Zhang SS, Lin HL, Lin Q, Shen LE, Ansong E, Wu XQ. Metformin Promotes Regeneration of the Injured Endometrium Via Inhibition of Endoplasmic Reticulum Stress-Induced Apoptosis. Reprod Sci 2018; 26:560-568. [PMID: 30466344 DOI: 10.1177/1933719118804424] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intrauterine adhesion (IUA) is now recognized as one of the most common diseases in reproductive-age women. Metformin, a well-known frontline oral antidiabetic drug, has been found effective in numerous different diseases. The aim of this study was to determine the effect of metformin on reducing adhesions in an animal model of IUA. Sprague-Dawley rats were randomized into 4 groups: sham operation, control, metformin-treated for 7 days, and metformin-treated for 14 days. To establish the IUA model, mechanical injury to the endometria of rats was induced with a mini curette. Metformin was injected intraperitoneally after surgery. A significant amelioration in both the number of glands and the fibrotic area, compared to those of the control group, was detected 14 days after metformin intervention. The expression levels of antigen KI-67 and vascular endothelial growth factor were increased at 7 and 14 days after treatment. However, the transforming growth factor-β expression was decreased at 14 days after treatment. Endoplasmic reticulum stress-related apoptosis proteins (glucose-regulated protein 78, caspase-12, and CCAAT/enhancer binding protein (EBP) homologous protein) were downregulated after metformin treatment. Moreover, we determined that the effect of metformin was related to the inhibition of endoplasmic reticulum stress-induced apoptosis via the Phosphatidylinositol 3 kinase (PI3K)/Protein kinase B (AKT) and Extracellular regulated protein kinases1/2 pathways. In conclusion, metformin can attenuate the adhesion and promote the regeneration of the endometrium of the IUA rat, and metformin may serve as a novel therapeutic strategy for IUA patients.
Collapse
Affiliation(s)
- Xin-Xin Xu
- 1 Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,These authors contributed equally to this work
| | - Si-Si Zhang
- 2 Department of Obstetrics and Gynecology, The University of Hong Kong, Pokfulam, Hong Kong, China.,These authors contributed equally to this work
| | - Hui-Long Lin
- 1 Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qi Lin
- 1 Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lai-En Shen
- 1 Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Emmanuel Ansong
- 1 Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xue-Qing Wu
- 1 Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,3 Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Guangdong, China.,4 Clinical Medical Academy, Shenzhen University, Guangdong, China
| |
Collapse
|