1
|
Du G, Zheng K, Sun C, Sun M, Pan J, Meng D, Guan W, Zhao H. The relationship mammalian p38 with human health and its homolog Hog1 in response to environmental stresses in Saccharomyces cerevisiae. Front Cell Dev Biol 2025; 13:1522294. [PMID: 40129568 PMCID: PMC11931143 DOI: 10.3389/fcell.2025.1522294] [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: 11/09/2024] [Accepted: 02/13/2025] [Indexed: 03/26/2025] Open
Abstract
The mammalian p38 MAPK pathway plays a vital role in transducing extracellular environmental stresses into numerous intracellular biological processes. The p38 MAPK have been linked to a variety of cellular processes including inflammation, cell cycle, apoptosis, development and tumorigenesis in specific cell types. The p38 MAPK pathway has been implicated in the development of many human diseases and become a target for treatment of cancer. Although MAPK p38 pathway has been extensively studied, many questions still await clarification. More comprehensive understanding of the MAPK p38 pathway will provide new possibilities for the treatment of human diseases. Hog1 in S. cerevisiae is the conserved homolog of p38 in mammalian cells and the HOG MAPK signaling pathway in S. cerevisiae has been extensively studied. The deep understanding of HOG MAPK signaling pathway will help provide clues for clarifying the p38 signaling pathway, thereby furthering our understanding of the relationship between p38 and disease. In this review, we elaborate the functions of p38 and the relationship between p38 and human disease. while also analyzing how Hog1 regulates cellular processes in response to environmental stresses. 1, p38 in response to various stresses in mammalian cells.2, The functions of mammalian p38 in human health.3, Hog1 as conserved homolog of p38 in response to environmental stresses in Saccharomyces cerevisiae. 1, p38 in response to various stresses in mammalian cells. 2, The functions of mammalian p38 in human health. 3, Hog1 as conserved homolog of p38 in response to environmental stresses in S. cerevisiae.
Collapse
Affiliation(s)
- Gang Du
- *Correspondence: Gang Du, ; Wenqiang Guan, ; Hui Zhao,
| | | | | | | | | | | | - Wenqiang Guan
- Tianjin Key Laboratory of Food Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, China
| | - Hui Zhao
- Tianjin Key Laboratory of Food Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, China
| |
Collapse
|
2
|
Zhou Y, Ling T, Shi W. Current state of signaling pathways associated with the pathogenesis of idiopathic pulmonary fibrosis. Respir Res 2024; 25:245. [PMID: 38886743 PMCID: PMC11184855 DOI: 10.1186/s12931-024-02878-z] [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: 04/13/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) represents a chronic and progressive pulmonary disorder distinguished by a notable mortality rate. Despite the elusive nature of the pathogenic mechanisms, several signaling pathways have been elucidated for their pivotal roles in the progression of this ailment. This manuscript aims to comprehensively review the existing literature on the signaling pathways linked to the pathogenesis of IPF, both within national and international contexts. The objective is to enhance the comprehension of the pathogenic mechanisms underlying IPF and offer a scholarly foundation for the advancement of more efficacious therapeutic strategies, thereby fostering research and clinical practices within this domain.
Collapse
Affiliation(s)
- Yang Zhou
- School of Medicine, Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, 224005, China
| | - Tingting Ling
- School of Medicine, Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, 224005, China
| | - Weihong Shi
- School of Medicine, Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, 224005, China.
| |
Collapse
|
3
|
Gao T, Wang J, Xiao M, Wang J, Wang S, Tang Y, Zhang J, Lu G, Guo H, Guo Y, Liu Q, Li J, Gu J. SESN2-Mediated AKT/GSK-3β/NRF2 Activation to Ameliorate Adriamycin Cardiotoxicity in High-Fat Diet-Induced Obese Mice. Antioxid Redox Signal 2024; 40:598-615. [PMID: 37265150 DOI: 10.1089/ars.2022.0156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Aims: Obese patients are highly sensitive to adriamycin (ADR)-induced cardiotoxicity. However, the potential mechanism of superimposed toxicity remains to be elucidated. Sestrin 2 (SESN2), a potential antioxidant, could attenuate stress-induced cardiomyopathy; therefore, this study aims to explore whether SESN2 enhances cardiac resistance to ADR-induced oxidative damage in high-fat diet (HFD)-induced obese mice. Results: The results revealed that obesity decreased SESN2 expression in ADR-exposed heart. And, HFD mice may predispose to ADR-induced cardiotoxicity, which was probably associated with inhibiting protein kinase B (AKT), glycogen synthase kinase-3 beta (GSK-3β) phosphorylation and subsequently blocking nuclear localization of nuclear factor erythroid-2 related factor 2 (NRF2), ultimately resulting in cardiac oxidative damage. However, these destructive cascades and cardiac oxidative damage effects induced by HFD/sodium palmitate combined with ADR were blocked by overexpression of SESN2. Moreover, the antioxidant effect of SESN2 could be largely abolished by sh-Nrf2 or wortmannin. And sulforaphane, an NRF2 agonist, could remarkably reverse cardiac pathological and functional abnormalities caused by ADR in obese mice. Innovation and Conclusion: This study demonstrated that SESN2 might be a promising therapeutic target for improving anthracycline-related cardiotoxicity in obesity by upregulating activity of NRF2 via AKT/GSK-3β/Src family tyrosine kinase signaling pathway. Antioxid. Redox Signal. 40, 598-615.
Collapse
Affiliation(s)
- Ting Gao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mengjie Xiao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shudong Wang
- Department of Cardiology at the First Hospital of Jilin University, Changchun, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Jingjing Zhang
- Department of Cardiology at the First Hospital of China Medical University, Shenyang, China
- Department of Cardiology at the People's Hospital of Liaoning Province, Shenyang, China
| | - Guangping Lu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hua Guo
- Department of Nursing, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qingbo Liu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiahao Li
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
4
|
Luan R, Luo M, Ding D, Su X, Yang J. Zinc deficiency increases lung inflammation and fibrosis in obese mice by promoting oxidative stress. Biochim Biophys Acta Gen Subj 2024; 1868:130518. [PMID: 37951369 DOI: 10.1016/j.bbagen.2023.130518] [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: 06/30/2023] [Revised: 10/22/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Zinc deficiency can lead to multiple organ damage. In this study, we investigated the effects of zinc deficiency on obesity-related lung damage. METHODS C57BL/6 J mice were fed a diet with differing amounts of zinc and fat over a 6-month period. Palmitic acid was used to stimulate A549 cells to construct a high-fat alveolar epithelial cell model. Western blotting and histopathological staining were performed on animal tissues. Nuclear expression of nuclear factor erythroid 2-related factor 2 (Nrf2) was detected in cultured cells. A reactive oxygen species (ROS) assay kit was used to detect intracellular ROS. Furthermore, Nrf2 siRNA was used to examine zinc deficiency effects on A549 cells. RESULTS Pathological results showed significant damage to the lung structure of mice in the high-fat and low-zinc diet group, with a significant increase in the expression of inflammatory (IL-6, TNF-α) and fibrosis (TGFβ1, PAI-1) factors, combined with a decrease in the expression of Nrf2, HO-1 and NQO1 in the antioxidant pathway. In A549 cells, high fat and low zinc levels aggravated ROS production. Western blot and immunofluorescence results showed that high fat and zinc deficiency inhibited Nrf2 expression. After Nrf2-specific knockout in A549 cells, the protective effect of zinc on oxidant conditions induced by high fat was reduced. Phosphorylated Akt and PI3K levels were downregulated on the high-fat and low-zinc group compared with the high-fat group. CONCLUSIONS Zinc attenuated lung oxidative damage in obesity-related lung injury and Nrf2 activation is one of the important mechanisms of this effect. GENERAL SIGNIFICANCE Regulating zinc homeostasis through dietary modifications or supplemental nutritional therapy can contribute to the prevention and treatment of obesity-related lung injury.
Collapse
Affiliation(s)
- Rumei Luan
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun, Jilin, China
| | - Manyu Luo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Dongyan Ding
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xin Su
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun, Jilin, China
| | - Junling Yang
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
5
|
Cai L, Tan Y, Watson S, Wintergerst K. Diabetic cardiomyopathy - Zinc preventive and therapeutic potentials by its anti-oxidative stress and sensitizing insulin signaling pathways. Toxicol Appl Pharmacol 2023; 477:116694. [PMID: 37739320 PMCID: PMC10616760 DOI: 10.1016/j.taap.2023.116694] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Oxidative stress and insulin resistance are two key mechanisms for the development of diabetic cardiomyopathy (DCM, cardiac remodeling and dysfunction). In this review, we discussed how zinc and metallothionein (MT) protect the heart from type 1 or type 2 diabetes (T1D or T2D) through its anti-oxidative function and insulin-mediated PI3K/Akt signaling activation. Both T1D and T2D-induced DCM, shown by cardiac structural remodeling and dysfunction, in wild-type mice, but not in cardiomyocyte-specific overexpressing MT mice. In contrast, mice with global MT gene deletion were more susceptible to the development of DCM. When we used zinc to treat mice with either T1D or T2D, cardiac remodeling and dysfunction were significantly prevented along with increased cardiac MT expression. To support the role of zinc homeostasis in insulin signaling pathways, treatment of diabetic mice with zinc showed the preservation of phosphorylation levels of insulin-mediated glucose metabolism-related Akt2 and GSK-3β and even rescued cardiac pathogenesis induced by global deletion of Akt2 gene in a MT-dependent manner. These results suggest the protection by zinc from DCM is through both the induction of MT and sensitization of insulin signaling. Combined our own and other works, this review comprehensively summarized the roles of zinc homeostasis in the development and progression of DCM and its therapeutic implications. At the end, we provided pre-clinical and clinical evidence for the preventive and therapeutic potential of zinc supplementation through its anti-oxidative stress and sensitizing insulin signaling actions. Understanding the intricate connections between zinc and DCM provides insights for the future interventional approaches.
Collapse
Affiliation(s)
- Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America; Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, United States of America.
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America.
| | - Sara Watson
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, United States of America
| | - Kupper Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, United States of America
| |
Collapse
|
6
|
Zhou W, Young JL, Men H, Zhang H, Yu H, Lin Q, Xu H, Xu J, Tan Y, Zheng Y, Cai L. Sex differences in the effects of whole-life, low-dose cadmium exposure on postweaning high-fat diet-induced cardiac pathogeneses. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 809:152176. [PMID: 34875320 PMCID: PMC11871371 DOI: 10.1016/j.scitotenv.2021.152176] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/17/2021] [Accepted: 11/30/2021] [Indexed: 06/13/2023]
Abstract
We previously showed the development of cardiac remodeling (hypertrophy or fibrosis) in mice with either post-weaning high-fat diet (HFD, 60% kcal fat) feeding or exposure to chronic low-dose cadmium. Here, we determined whether whole-life exposure to environmentally relevant, low-dose cadmium affects the susceptibility of offspring to post-weaning HFD-induced cardiac pathologies and function. Besides, we also determined whether these effects are sex-dependent. Male and female mice were exposed to cadmium-containing (0, 0.5, or 5 parts per million [ppm]) drinking water before breeding; the pregnant mice and dams with offspring continually drank the same cadmium-containing water. After weaning, the offspring were continued on the same regime as their parents and fed either a HFD or normal fat diet for 24 weeks. Cardiac function was examined with echocardiography. Cardiac tissues were used for the histopathological and biochemical (gene and protein expression by real-time PCR and Western blotting) assays. Results showed a dose-dependent cadmium accumulation in the hearts of male and female mice along with decreased cardiac zinc and copper levels only in female offspring. Exposure to 5 ppm, but not 0.5 ppm, cadmium significantly enhanced HFD cardiac effects only in female mice, shown by worsened cardiac systolic and diastolic dysfunction (ejection fraction, mitral E-to-annular e' ratio), increased fibrosis (collagen, fibronectin, collagen1A1), hypertrophy (cardiomyocyte size, atrial natriuretic peptide, β-myosin heavy chain), and inflammation (intercellular adhesion molecule-1, tumor necrosis factor-α, plasminogen activator inhibitor type 1), compared to the HFD group. These synergistic effects were associated with activation of the p38 mitogen-activated protein kinases (MAPK) signaling pathway and increased oxidative stress, shown by 3-nitrotyrosine and malondialdehyde, along with decreased metallothionein expression. These results suggest that whole-life 5 ppm cadmium exposure significantly increases the susceptibility of female offspring to HFD-induced cardiac remodeling and dysfunction. The underlying mechanism and potential intervention will be further explored in the future.
Collapse
Affiliation(s)
- Wenqian Zhou
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA; The Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun 130021, China.
| | - Jamie L Young
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA; Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA..
| | - Hongbo Men
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA; The Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun 130021, China.
| | - Haina Zhang
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA; The Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun 130021, China.
| | - Haitao Yu
- The Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun 130021, China.
| | - Qian Lin
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA.
| | - He Xu
- Department of Respiratory Medicine, the First Hospital of Jilin University (Eastern Division), Changchun 130031, China.
| | - Jianxiang Xu
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA.
| | - Yi Tan
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA; Wendy L. Novak Diabetes Care Center, Louisville, KY, USA.
| | - Yang Zheng
- The Center of Cardiovascular Diseases, the First Hospital of Jilin University, Changchun 130021, China.
| | - Lu Cai
- Pediatric Research Institute, the Department of Pediatrics of University of Louisville, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA; Wendy L. Novak Diabetes Care Center, Louisville, KY, USA.
| |
Collapse
|
7
|
Fang Y, Wang S, Lv J, Zhao Z, Guo N, Wu G, Tong J, Wang Z. Slc39a2-Mediated Zinc Homeostasis Modulates Innate Immune Signaling in Phenylephrine-Induced Cardiomyocyte Hypertrophy. Front Cardiovasc Med 2021; 8:736911. [PMID: 34790705 PMCID: PMC8592093 DOI: 10.3389/fcvm.2021.736911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/12/2021] [Indexed: 01/05/2023] Open
Abstract
Zinc dyshomeostasis has been involved in the pathogenesis of cardiac hypertrophy; however, the dynamic regulation of intracellular zinc and its downstream signaling in cardiac hypertrophy remain largely unknown. Using Zincpyr1 staining, we found a significant decrease of intracellular Zinc concentration in phenylephrine (PE)-induced hypertrophy of neonatal rat ventricular myocytes (NRVMs). We then screened SLC39 family members responsible for zinc uptake and identified Slc39a2 as the only one altered by PE treatment. Slc39a2 knockdown in NRVMs reduced the intracellular Zinc level, and exacerbated the hypertrophic responses to PE treatment. In contrast, adenovirus-mediated Slc39a2 overexpression enhanced zinc uptake and suppressed PE-induced Nppb expression. RNA sequencing analysis showed a pro-hypertrophic transcriptome reprogramming after Slc39a2 knockdown. Interestingly, the innate immune signaling pathways, including NOD signaling, TOLL-like receptor, NFκB, and IRFs, were remarkably enriched in the Slc39a2-regulated genes. Slc39a2 deficiency enhanced the phosphorylation of P65 NFκB and STAT3, and reduced the expression of IκBα. Finally, the expression of IRF7 was significantly increased by Slc39a2 knockdown, which was in turn suppressed by IRF7 knockdown. Our data demonstrate that zinc homeostasis mediated by a Slc39a2/IRF7 regulatory circuit contributes to the alteration of innate immune signaling in cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Yu Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jian Lv
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhenyi Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,School of Pharmacy, Health Science Center, Shenzhen University, Shenzhen, China
| | - Ningning Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingjing Tong
- School of Life Sciences, Central China Normal University, Wuhan, China
| | - Zhihua Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Henze LA, Estepa M, Pieske B, Lang F, Eckardt KU, Alesutan I, Voelkl J. Zinc Ameliorates the Osteogenic Effects of High Glucose in Vascular Smooth Muscle Cells. Cells 2021; 10:cells10113083. [PMID: 34831306 PMCID: PMC8623153 DOI: 10.3390/cells10113083] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/23/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
In diabetic patients, medial vascular calcification is common and associated with increased cardiovascular mortality. Excessive glucose concentrations can activate the nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-kB) and trigger pro-calcific effects in vascular smooth muscle cells (VSMCs), which may actively augment vascular calcification. Zinc is able to mitigate phosphate-induced VSMC calcification. Reduced serum zinc levels have been reported in diabetes mellitus. Therefore, in this study the effects of zinc supplementation were investigated in primary human aortic VSMCs exposed to excessive glucose concentrations. Zinc treatment was found to abrogate the stimulating effects of high glucose on VSMC calcification. Furthermore, zinc was found to blunt the increased expression of osteogenic and chondrogenic markers in high glucose-treated VSMCs. High glucose exposure was shown to activate NF-kB in VSMCs, an effect that was blunted by additional zinc treatment. Zinc was further found to increase the expression of TNFα-induced protein 3 (TNFAIP3) in high glucose-treated VSMCs. The silencing of TNFAIP3 was shown to abolish the protective effects of zinc on high glucose-induced NF-kB-dependent transcriptional activation, osteogenic marker expression, and the calcification of VSMCs. Silencing of the zinc-sensing receptor G protein-coupled receptor 39 (GPR39) was shown to abolish zinc-induced TNFAIP3 expression and the effects of zinc on high glucose-induced osteogenic marker expression. These observations indicate that zinc may be a protective factor during vascular calcification in hyperglycemic conditions.
Collapse
Affiliation(s)
- Laura A. Henze
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum, 13353 Berlin, Germany; (L.A.H.); (M.E.); (B.P.)
| | - Misael Estepa
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum, 13353 Berlin, Germany; (L.A.H.); (M.E.); (B.P.)
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum, 13353 Berlin, Germany; (L.A.H.); (M.E.); (B.P.)
| | - Florian Lang
- Department of Vegetative and Clinical Physiology, Eberhard Karls University Tübingen, 72076 Tübingen, Germany;
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.-U.E.); (J.V.)
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, 4040 Linz, Austria
- Correspondence: ; Tel.: +43-732-2468-8990
| | - Jakob Voelkl
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.-U.E.); (J.V.)
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, 4040 Linz, Austria
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347 Berlin, Germany
| |
Collapse
|
9
|
Abstract
Since the discovery of manifest Zn deficiency in 1961, the increasing number of studies demonstrated the association between altered Zn status and multiple diseases. In this chapter, we provide a review of the most recent advances on the role of Zn in health and disease (2010-20), with a special focus on the role of Zn in neurodegenerative and neurodevelopmental disorders, diabetes and obesity, male and female reproduction, as well as COVID-19. In parallel with the revealed tight association between ASD risk and severity and Zn status, the particular mechanisms linking Zn2+ and ASD pathogenesis like modulation of synaptic plasticity through ProSAP/Shank scaffold, neurotransmitter metabolism, and gut microbiota, have been elucidated. The increasing body of data indicate the potential involvement of Zn2+ metabolism in neurodegeneration. Systemic Zn levels in Alzheimer's and Parkinson's disease were found to be reduced, whereas its sequestration in brain may result in modulation of amyloid β and α-synuclein processing with subsequent toxic effects. Zn2+ was shown to possess adipotropic effects through the role of zinc transporters, zinc finger proteins, and Zn-α2-glycoprotein in adipose tissue physiology, underlying its particular role in pathogenesis of obesity and diabetes mellitus type 2. Recent findings also contribute to further understanding of the role of Zn2+ in spermatogenesis and sperm functioning, as well as oocyte development and fertilization. Finally, Zn2+ was shown to be the potential adjuvant therapy in management of novel coronavirus infection (COVID-19), underlining the perspectives of zinc in management of old and new threats.
Collapse
Affiliation(s)
- Anatoly V Skalny
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia
| | - Michael Aschner
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia.
| |
Collapse
|
10
|
Hu Y, Wang Y, Wang X, Wu X, Fu L, Liu X, Wen Y, Sheng J, Zhang J. The Role of Cation Diffusion Facilitator CDF-1 in Lipid Metabolism in Caenorhabditis elegans. G3-GENES GENOMES GENETICS 2021; 11:6237889. [PMID: 33871589 PMCID: PMC8495940 DOI: 10.1093/g3journal/jkab120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/08/2021] [Indexed: 11/20/2022]
Abstract
Zinc is one of the most important trace elements as it plays a vital role in many biological processes. As well, aberrant zinc metabolism has been implicated in lipid-related metabolic diseases. Previously, we showed that zinc antagonizes iron to regulate sterol regulatory element-binding proteins and the stearoyl-CoA desaturase (SREBP-SCD) pathway in lipid metabolism in the model organism Caenorhabditis elegans. In this study, we present the identification of another cation diffusion facilitator, CDF-1, which regulates lipid metabolism along with SUR-7 in response to zinc. Inactivation of SBP-1, the only homolog of SREBPs, leads to an increased zinc level but decreased lipid accumulation. However, either the cdf-1(n2527) or sur-7(tm6523) mutation could successfully restore the altered fatty acid profile, fat content, and zinc level of the sbp-1(ep79) mutant. Furthermore, we found that CDF-1/SUR-7 may functionally bypass SBP-1 to directly affect the conversion activity of SCD in the biosynthesis of unsaturated fatty acids and lipid accumulation. Collectively, these results consistently support the link between zinc homeostasis and lipid metabolism via the SREBP-SCD axis by the cation diffusion facilitators CDF-1 and SUR-7.
Collapse
Affiliation(s)
- Ying Hu
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Yanli Wang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Xuanjun Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Xiaoyun Wu
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Lin Fu
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Xiayu Liu
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Yu Wen
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Jun Sheng
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Jingjing Zhang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| |
Collapse
|
11
|
Guo Z, Gao WS, Wang YF, Gao F, Wang W, Ding WY. MiR-502 Suppresses TNF- α-Induced Nucleus Pulposus Cell Apoptosis by Targeting TARF2. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5558369. [PMID: 33869626 PMCID: PMC8035021 DOI: 10.1155/2021/5558369] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/02/2021] [Accepted: 03/18/2021] [Indexed: 12/31/2022]
Abstract
Intervertebral disc degeneration (IVDD) is a common cause of low back pain. This study is aimed at investigating the role of microRNAs (miRNAs) in regulating human nucleus pulposus (NP) cell injury induced by tumor necrosis factor- (TNF-) α in IVDD. In this study, we induced NP cells with 20 ng/mL TNF-α in vitro, which promoted the obvious apoptosis of NP cells and the activation of nuclear transcription factor (NF)-κB. In contrast, using the specific NF-κB inhibitor BAY 11-7082 to treat cells greatly impaired the activation of NF-κB and increased the sensitivity of NP cells to TNF-α-induced apoptosis. Moreover, both TNF-α and BAY 11-7082 treatments were associated with marked miRNA dysregulation, with miR-502 being upregulated by TNF-α treatment and downregulated by BAY 11-7082 treatment, respectively. And the overexpression of miR-502 enhanced NF-κB activation and suppressed apoptosis of human NP cells induced by TNF-α, whereas the opposite was observed following miR-502 inhibition. Last, through bioinformatic analyses and luciferase reporter gene experiments, we identified TRAF2, an important activator of NF-κB, as a miR-502 target gene. Similarly, siRNA-mediated knockdown of the TRAF2 expression also suppressed TNF-α-induced apoptosis and enhanced NF-κB activation. Our findings provide evidence indicating that miR-502 is a key regulator of apoptosis of human NP cells induced by TNF-α by targeting TRAF2 and activating NF-κB.
Collapse
Affiliation(s)
- Zhao Guo
- Orthopedics Department, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, China
- Orthopedics Department, Affiliated Hospital of Hebei University, Baoding, Hebei 071002, China
| | - Wen-Shan Gao
- Orthopedics Department, Affiliated Hospital of Hebei University, Baoding, Hebei 071002, China
| | - Yun-Fei Wang
- Orthopedics Department, Affiliated Hospital of Hebei University, Baoding, Hebei 071002, China
| | - Fei Gao
- Orthopedics Department, Affiliated Hospital of Hebei University, Baoding, Hebei 071002, China
| | - Wei Wang
- Orthopedics Department, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, China
| | - Wen-Yuan Ding
- Orthopedics Department, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, China
| |
Collapse
|
12
|
Santos HO, Teixeira FJ. Use of medicinal doses of zinc as a safe and efficient coadjutant in the treatment of male hypogonadism. Aging Male 2020; 23:669-678. [PMID: 30767598 DOI: 10.1080/13685538.2019.1573220] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Hypogonadism affects an extensive part of the male population, especially among the elderly. The quest for treatment regarding low levels of serum testosterone and male infertility has, therefore, worldwide relevance. Zinc has important biological actions insofar as the male reproductive physiology and endocrine system. In general, a common and safe recommendation for zinc in the treatment of male hypogonadism is 220 mg of zinc sulfate (equivalent to 50 mg of elemental zinc) twice a day, over one to four months. Additionally, it may be further required to extend, both the treatment, dose and daily fractionation of this mineral. Albeit medicinal doses of zinc may increase total testosterone and improve sperm count, the current body of evidence does not suggest broad recommendations regarding the use of zinc for all types of hypogonadism. In many cases, the use of zinc supplements is insufficient, with the use of surgery and drugs being required for an effective treatment.
Collapse
Affiliation(s)
- Heitor O Santos
- School of Medicine, Federal University of Uberlandia (UFU), Uberlandia, Brazil
| | - Filipe J Teixeira
- Laboratory of Physiology and Biochemistry of Exercise Faculty of Human Kinetics, Universidade de Lisboa, Cruz Quebrada, Portugal
| |
Collapse
|
13
|
Zhang H, Cai L. Zinc homeostasis plays an important role in the prevention of obesity-induced cardiac inflammation, remodeling and dysfunction. J Trace Elem Med Biol 2020; 62:126615. [PMID: 32683230 DOI: 10.1016/j.jtemb.2020.126615] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 06/09/2020] [Accepted: 07/03/2020] [Indexed: 01/21/2023]
Abstract
Obesity often leads to cardiovascular diseases, such as obesity-related cardiac hypertrophy (ORCH), due to chronic cardiac inflammation. Zinc is structurally and functionally essential for many transcription factors, therefore it not only has anti-inflammatory and anti-oxidative stress functions, but also has insulin-like function, however, its role in the development of obesity-associated cardiac pathogenesis and the potentially underlying mechanism(s) remains unclear. This review aims to summarize the available evidence on the role of zinc homeostasis in the prevention of ORCH. It was recently reported that when four-week old mice were fed either high fat diet (HFD) or normal diet containing deficient, adequate or supplemented zinc, HFD induced obesity and ORCH along with increased phosphorylation of p38 MAPK and increased expression of B-cell lymphoma/ leukemia 10 (BCL10) and caspase recruitment domain family member 9 (CARD9). These effects were further aggravated by zinc deficiency and significantly alleviated by zinc supplementation. Mechanistically administration of a p38 MAPK specific inhibitor in HFD-fed mice for 3 months did not affect HFD-induced obesity and increased expression of BCL10 and CARD9, but completely abolished HFD/obesity-induced cardiac hypertrophy and inflammation. In cultured cardiomyocytes, inhibition of BCL10 expression by siRNA prevented palmitate-induced increased p38 MAPK activation and atrial natriuretic peptide expression. Deletion of metallothionein abolished the protective effect of zinc on palmitate-induced up-regulation of BCL10 and phospho-p38 MAPK. Taken together with other recent studies, we concluded that HFD and zinc deficiency synergistically induce ORCH by increasing oxidative stress-mediated activation of BCL10/CARD9/p38 MAPK signaling. Zinc supplementation ameliorates ORCH through activation of metallothionein to repress oxidative stress-activated BCL10 expression and p38 MAPK activation.
Collapse
Affiliation(s)
- Haina Zhang
- Pediatric Research Institute, Departments of Pediatric, University of Louisville School of Medicine, Louisville, KY, USA; Center of Cardiovascular Disorders, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Lu Cai
- Pediatric Research Institute, Departments of Pediatric, University of Louisville School of Medicine, Louisville, KY, USA; Departments of Radiation Oncology, Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
14
|
Zhang SY, Yang KL, Long ZY, Li WQ, Huang HY. Use of a Systematic Pharmacological Methodology to Explore the Mechanism of Shengmai Powder in Treating Diabetic Cardiomyopathy. Med Sci Monit 2020; 26:e919029. [PMID: 32023237 PMCID: PMC7020766 DOI: 10.12659/msm.919029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/08/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cardiovascular complications, such as diabetic cardiomyopathy (DCM), are the leading cause of death in diabetic patients. Shengmai Powder (SMP) was found to have cardioprotective effects. MATERIAL AND METHODS Based on the systematic pharmacological methodology, this research determined the genes of DCM and the known targets of SMP, predicted potential compounds and targets of SMP, constructed networks for DCM and SMP, and performed network analysis. RESULTS Five network were constructed: (1) the DCM gene PPI network; (2) the Compound-compound target network of SMP; (3) the SMP-DCM PPI network; (4) the Compound-known target network of SMP; (5) and the SMP known target-DCM PPI network. Several DCM and treatment related targets, clusters, signaling pathways, and biological processes were found. CONCLUSIONS SMP is able to regulate glycometabolism-related, lipid metabolism-related, inflammatory response-related, oxidative stress-related signaling pathways, and biological processes and targets, which suggests that SMP may have a therapeutic effect on DCM.
Collapse
Affiliation(s)
- Shi-ying Zhang
- Department of Traditional Chinese Medicine, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, P.R. China
- Hunan University of Chinese Medicine, Changsha, Hunan, P.R. China
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
- Department of Traditional Chinese Medicine, Shenzhen Luohu Hospital Group Luohu People’s Hospital, Shenzhen, Guangdong, P.R. China
| | - Kai-lin Yang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, P.R. China
- Capital Medical University, Beijing, P.R. China
| | - Zhi-yong Long
- Shantou University Medical College, Shantou University, Shantou, Guangdong, P.R. China
- Department of Rehabilitation Medicine, Guangdong Geriatric Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, P.R. China
| | - Wei-qing Li
- Department of Traditional Chinese Medicine, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, P.R. China
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
- Department of Traditional Chinese Medicine, Shenzhen Luohu Hospital Group Luohu People’s Hospital, Shenzhen, Guangdong, P.R. China
| | - Hui-yong Huang
- Hunan University of Chinese Medicine, Changsha, Hunan, P.R. China
| |
Collapse
|
15
|
Liang Y, Young JL, Kong M, Tong Y, Qian Y, Freedman JH, Cai L. Gender Differences in Cardiac Remodeling Induced by a High-Fat Diet and Lifelong, Low-Dose Cadmium Exposure. Chem Res Toxicol 2019; 32:1070-1081. [PMID: 30912652 PMCID: PMC7060501 DOI: 10.1021/acs.chemrestox.8b00386] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Childhood obesity, which is prevalent in developed countries, is a metabolic risk factor for cardiovascular disease. Cadmium (Cd), a ubiquitous environmental toxic metal, also has deleterious effects on the cardiovascular system. However, the combined effects of a high-fat diet (HFD) and lifelong, low-dose Cd exposure on cardiac remodeling remain unclear. This study aims to determine the effects of combined HFD and Cd exposure on cardiac remodeling, as well as gender-specific differences in the response. C57BL/6J mice were exposed to Cd at a low dose (L-Cd, 0.5 ppm) or high dose (H-Cd, 5 ppm) via drinking water from conception to sacrifice. After being weaned, the offspring mice were fed with a HFD (42% kcal from fat) for an additional 10 weeks. H-Cd exposure significantly increased Cd accumulation in the hearts of both parents and their offspring; a HFD showed no added effects on cardiac Cd content. H-Cd exposure increased cardiac metallothionein protein levels only in female mice, regardless of dietary intake. Histological analysis revealed that H-Cd exposure combined with a HFD induced cardiac hypertrophy and fibrosis only in female mice. This was further supported by elevated expression of ANP and COL1A1 protein levels along with COL1A1, COL1A2, and COL3A1 mRNA levels. Profibrotic markers PAI-1, CTGF, and FN were also elevated in HFD/H-Cd-exposed female mice. Levels of the oxidative stress marker 3-NT significantly increased in the hearts of HFD-fed female mice, whereas Cd exposure showed no additional effects. Of all the antioxidant markers examined, levels of CAT significantly increased in mice fed a HFD, regardless of gender and Cd exposure. In summary, a HFD combined with lifelong, low-dose Cd exposure induces cardiac hypertrophy and fibrosis in female but not male mice, a response that is independent of oxidative stress.
Collapse
Affiliation(s)
- Yaqin Liang
- Department of Pediatrics, First Affiliated Hospital of Wenzhou Medical University, Zhejiang 325000, China
- Pediatric Research Institute, Department of Pediatrics, School of Public Health and Information Sciences, University of Louisville School of Medicine, Louisville, Kentucky 40202, United States
| | - Jamie L. Young
- Department of Pharmacology and Toxicology, School of Public Health and Information Sciences, University of Louisville School of Medicine, Louisville, Kentucky 40202, United States
| | - Maiying Kong
- Department of Bioinformatics and Biostatistics, School of Public Health and Information Sciences, University of Louisville School of Medicine, Louisville, Kentucky 40202, United States
| | - Yongguang Tong
- Department of Pharmacology and Toxicology, School of Public Health and Information Sciences, University of Louisville School of Medicine, Louisville, Kentucky 40202, United States
| | - Yan Qian
- Department of Pediatrics, First Affiliated Hospital of Wenzhou Medical University, Zhejiang 325000, China
| | - Jonathan H. Freedman
- Department of Pharmacology and Toxicology, School of Public Health and Information Sciences, University of Louisville School of Medicine, Louisville, Kentucky 40202, United States
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, School of Public Health and Information Sciences, University of Louisville School of Medicine, Louisville, Kentucky 40202, United States
- Department of Pharmacology and Toxicology, School of Public Health and Information Sciences, University of Louisville School of Medicine, Louisville, Kentucky 40202, United States
| |
Collapse
|
16
|
Oh JE, Jun JH, Hwang HJ, Shin EJ, Oh YJ, Choi YS. Dexmedetomidine restores autophagy and cardiac dysfunction in rats with streptozotocin-induced diabetes mellitus. Acta Diabetol 2019; 56:105-114. [PMID: 30206697 DOI: 10.1007/s00592-018-1225-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/03/2018] [Indexed: 12/17/2022]
Abstract
AIMS Dexmedetomidine (DEX), a highly selective and potent α2-adrenergic receptor agonist, has anti-apoptotic, anti-inflammatory, and anti-oxidative stress effects in diabetes mellitus (DM) rats. The underlying molecular mechanisms and signaling pathways of diabetic cardiomyopathy remain poorly understood. This study aimed to elucidate the effect of DEX on cardiac function in DM rats. METHODS Eight-week-old male Sprague Dawley rats were divided into three groups: control (n = 5), diabetes (DM, n = 7), and diabetes + DEX (DM + DEX, n = 10). DM was induced via intraperitoneal injection of streptozotocin (70 mg/kg); at 3 days later, DEX (1 µg/kg/h) was administered for 4 weeks. Cardiac function was evaluated using pressure-volume loop analysis and echocardiography. Left ventricular (LV) histological sections were used to analyze the interstitial collagen fraction. Using the LV samples, we performed a western blot analysis to evaluate signaling pathways and autophagic markers. RESULTS The DM group had lower body weight and higher blood glucose level and heart weight/body weight ratio than the control group. However, metabolic changes did not differ between the DM and DM + DEX groups. Pressure-volume loop analysis and echocardiography showed impaired cardiac function, evidenced by a decrease in systolic and diastolic function, in both DM groups. DEX treatment in DM rats was associated with increased LV end-systolic pressure, LV contractility, cardiac output, and relaxed LV function compared with that in non-treated DM rats. LC3B and autophagy-related gene (ATG) proteins increased in the hearts of DM rats compared with the hearts of control rats. However, DEX reduced the expression of LC3B and ATG proteins in the hearts of DM rats. Increased p-ERK and decreased p-AKT were reduced in the hearts of DEX-treated DM rats. CONCLUSIONS DEX reduces cardiac dysfunction and impaired autophagy in DM rats. This study reinforces our understanding of the potential anti-autophagic effect of DEX in patients with diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Ju Eun Oh
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Ji Hae Jun
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hye Jeong Hwang
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Eun Jung Shin
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Young Jun Oh
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yong Seon Choi
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
17
|
Wang W, Guo Z, Yang S, Wang H, Ding W. Upregulation of miR-199 attenuates TNF-α-induced Human nucleus pulposus cell apoptosis by downregulating MAP3K5. Biochem Biophys Res Commun 2018; 505:917-924. [PMID: 30309653 DOI: 10.1016/j.bbrc.2018.09.194] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 09/30/2018] [Indexed: 01/09/2023]
Abstract
MicroRNA-199 has been reported to play a potential role in the apoptosis of Human nucleus pulposus cells. However, the effect of miR-199 in regulating Human nucleus pulposus cell injury induced by TNF-α has not been previously illustrated. This study searched to probe the effect and the molecular mechanism of miR-199 on Human nucleus pulposus cell injury induced by TNF-α. Using the TNF-α model of Human nucleus pulposus cell in vitro, we found that miR-199 was extremely decreased in Human nucleus pulposus cells after TNF-α treatment. Knockdown the expression of miR-199 by recombinant adeno-associated viral vector infection markedly promoted the apoptosis of Human nucleus pulposus cells induced by TNF-α treatment, whereas miR-199 overexpression significantly decreased Human nucleus pulposus cell apoptosis. Both Dual-luciferase reporter and western blot assay proved that MAP3K5 was a direct target gene of miR-199, and miR-199 inhibited the expression of MAP3K5 via binding to its 3'-UTR. Furthermore, we proved that overexpression of miR-199 could inhibit the expression of MAP3K5 at the transcription and translation levels, whereas the inhibition of miR-199 could upregulate the expression of MAP3K5. Moreover, MAP3K5 was highly expressed in TNF-α treated Human nucleus pulposus cells and the apoptosis rate induced by TNF-α was associated with the increase in MAP3K5 expression. Importantly, knockdown the expression of MAP3K5 apparently abrogated the inhibitory effect of miR-199 mimics on TNF-α induced Human nucleus pulposus cell apoptosis. In conclusion, these results indicate that upregulation of miR-199 could inhibit Human nucleus pulposus cells injury through downregulation of MAP3K5 expression, providing an important molecular target mechanism for Human nucleus pulposus cells injury.
Collapse
Affiliation(s)
- Wei Wang
- Orthopedics Department, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhao Guo
- Orthopedics Department, Affiliated Hospital of Hebei University, Baoding, Hebei, China
| | - Sidong Yang
- Orthopedics Department, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hui Wang
- Orthopedics Department, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wenyuan Ding
- Orthopedics Department, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
18
|
Pedersen TM, Boardman NT, Hafstad AD, Aasum E. Isolated perfused working hearts provide valuable additional information during phenotypic assessment of the diabetic mouse heart. PLoS One 2018; 13:e0204843. [PMID: 30273374 PMCID: PMC6166959 DOI: 10.1371/journal.pone.0204843] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 09/14/2018] [Indexed: 12/18/2022] Open
Abstract
Although murine models for studying the development of cardiac dysfunction in diabetes mellitus are well established, their reported cardiac phenotypes vary. These reported divergences may, in addition to the severity of different models, also be linked to the methods used for cardiac functional assessment. In the present study, we examined the functional changes using conventional transthoracic echocardiography (in vivo) and isolated heart perfusion techniques (ex vivo), in hearts from two mouse models; one with an overt type 2 diabetes (the db/db mouse) and one with a prediabetic state, where obesity was induced by a high-fat diet (HFD). Analysis of left ventricular function in the isolated working hearts from HFD-fed mice, suggested that these hearts develop diastolic dysfunction with preserved systolic function. Accordingly, in vivo examination demonstrated maintained systolic function, but we did not find parameters of diastolic function to be altered. In db/db mice, ex vivo working hearts showed both diastolic and systolic dysfunction. Although in vivo functional assessment revealed signs of diastolic dysfunction, the hearts did not display reduced systolic function. The contrasting results between ex vivo and in vivo function could be due to systemic changes that may sustain in vivo function, or a lack of sensitivity using conventional transthoracic echocardiography. Thus, this study demonstrates that the isolated perfused working heart preparation provides unique additional information related to the development of cardiomyopathy, which might otherwise go unnoticed when only using conventional echocardiographic assessment.
Collapse
Affiliation(s)
- Tina M. Pedersen
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Neoma T. Boardman
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Anne D. Hafstad
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Ellen Aasum
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
- * E-mail:
| |
Collapse
|
19
|
Al-Damry NT, Attia HA, Al-Rasheed NM, Al-Rasheed NM, Mohamad RA, Al-Amin MA, Dizmiri N, Atteya M. Sitagliptin attenuates myocardial apoptosis via activating LKB-1/AMPK/Akt pathway and suppressing the activity of GSK-3β and p38α/MAPK in a rat model of diabetic cardiomyopathy. Biomed Pharmacother 2018; 107:347-358. [PMID: 30099338 DOI: 10.1016/j.biopha.2018.07.126] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 07/07/2018] [Accepted: 07/24/2018] [Indexed: 12/27/2022] Open
Abstract
The present study aimed to investigate the protective effect of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on diabetic cardiomyopathy (DCM)-associated apoptosis and if this effect is mediated via modulating the activity of the survival kinases; AMP-activated protein kinase (AMPK) and Akt & the apoptotic kinases; glycogen synthase kinase-3 β (GSK-3β) and p38 mitogen-activated protein kinase (p38MAPK). Diabetes was induced by a single intraperitoneal injection of streptozotocin (55 mg/kg). Diabetic rats were treated with sitagliptin (10 mg/kg/day, p.o.) and metformin (200 mg/kg/day, p.o. as positive control) for six weeks. Chronic hyperglycemia resulted in elevation of serum cardiac biomarkers reflecting cardiac damage which was supported by H&E stain. The mRNA levels of collagen types I and III were augmented reflecting cardiac fibrosis and hypertrophy which was supported by Masson trichome stain and enhanced phosphorylation of p38MAPK. Cardiac protein levels of cleaved casapse-3, BAX were elevated, whereas, the levels of Bcl-2 and p-BAD were reduced indicating cardiac apoptosis which could be attributed to the diabetes-induced reduced phosphorylation of Akt and AMPK with concomitant augmented activation of GSK-3β and p38MAPK. Protein levels of liver kinase B-1, the upstream kinase of AMPK were also supressed. Sitagliptin administration alleviated the decreased phosphorylation of AMPK and Akt, inactivated the GSK-3β and p38 AMPK, therefore, attenuating the apoptosis and hypertrophy induced by hyperglycemia in the diabetic heart. In conclusion, sitagliptin exhibits valuable therapeutic potential in the management of DCM by attenuating apoptosis. The underlying mechanism may involve the modulating activity of AMPK, Akt, GSK-3β and p38MAPK.
Collapse
Affiliation(s)
- Nouf T Al-Damry
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hala A Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Nawal M Al-Rasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nouf M Al-Rasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Raeesa A Mohamad
- Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maha A Al-Amin
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nduna Dizmiri
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Muhammad Atteya
- Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
20
|
Liang Y, Lang AL, Zhang J, Chen J, Wang K, Chen L, Beier JI, Qian Y, Cai L. Exposure to Vinyl Chloride and Its Influence on Western Diet-Induced Cardiac Remodeling. Chem Res Toxicol 2018; 31:482-493. [PMID: 29727174 PMCID: PMC6167925 DOI: 10.1021/acs.chemrestox.8b00043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Obesity, usually caused by high fat diets (HFD), is a major public health issue worldwide, causing obesity associated cardiomyopathy. Moreover, the environmental toxicant vinyl chloride (VC) can exacerbate HFD-induced fatty liver disease. However, whether VC serves to enhance obesity-associated cardiomyopathy remains unclear. This study aims to investigate the interaction of western diet (WD) containing relatively low fat (42%) with VC on cardiac remodeling and its underling mechanisms. Adult male C57BL/6J mice were exposed to WD coinhalation of low-dose VC (<1 ppm/d) for 12 weeks. Results showed that WD feeding for 12 weeks caused slight cardiac systolic dysfunction without significant hypertrophy or fibrosis, even with VC. Nevertheless, WD upregulated NF-κB function and expression of IL-1β and PAI-1, while VC showed no significant impact on these effects. In contrast, WD together with VC significantly increased the expression of CHOP and TGF-β1, key markers for endoplasmic reticulum stress and profibrotic cytokine, respectively. In summary, exposure to low-dose of environmental toxicant VC while a WD is consumed for a relatively short time does not have significant impact on cardiac remodeling except for a mild systolic dysfunction of the heart.
Collapse
Affiliation(s)
- Yaqin Liang
- Department of Pediatrics, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
| | - Anna L. Lang
- Department of Pharmacology and Toxicology, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
- Department of Hepatobiology and Toxicology Program, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
| | - Jian Zhang
- Department of Pediatrics, Pediatric Research Institute, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
- The Center of Cardiovascular Disorders, The First Hospital of the Jilin University, Changchun 130021, China
| | - Jing Chen
- Department of Pediatrics, Pediatric Research Institute, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
| | - Kai Wang
- Department of Pediatrics, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
| | - Liya Chen
- Department of Pharmacology and Toxicology, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
- Department of Hepatobiology and Toxicology Program, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
| | - Juliane I. Beier
- Department of Pharmacology and Toxicology, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
- Department of Hepatobiology and Toxicology Program, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
| | - Yan Qian
- Department of Pediatrics, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Lu Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
- Department of Pharmacology and Toxicology, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
- The Center of Cardiovascular Disorders, The First Hospital of the Jilin University, Changchun 130021, China
| |
Collapse
|
21
|
Park Y, Zhang J, Cai L. Reappraisal of metallothionein: Clinical implications for patients with diabetes mellitus. J Diabetes 2018; 10:213-231. [PMID: 29072367 DOI: 10.1111/1753-0407.12620] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 08/29/2017] [Accepted: 10/20/2017] [Indexed: 12/22/2022] Open
Abstract
Reactive oxygen and nitrogen species (ROS and RNS, respectively) are byproducts of cellular physiological processes of the metabolism of intermediary nutrients. Although physiological defense mechanisms readily convert these species into water or urea, an improper balance between their production and removal leads to oxidative stress (OS), which is harmful to cellular components. This OS may result in uncontrolled growth or, ultimately, cell death. In addition, ROS and RNS are closely related to the development of diabetes and its complications. Therefore, numerous researchers have proposed the development of strategies for the removal of ROS/RNS to prevent or treat diabetes and its complications. Some molecules that are synthesized in the body or obtained from food participate in the removal and neutralization of ROS and RNS. Metallothionein, a cysteine-rich protein, is a metal-binding protein that has a wide range of functions in cellular homeostasis and immunity. Metallothionein can be induced by a variety of conditions, including zinc supplementation, and plays a crucial role in mediating anti-OS, anti-apoptotic, detoxification, and anti-inflammatory effects. Metallothionein can modulate various stress-induced signaling pathways (mitogen-activated protein kinase, Wnt, nuclear factor-κB, phosphatidylinositol 3-kinase, sirtuin 1/AMP-activated protein kinase and fibroblast growth factor 21) to alleviate diabetes and diabetic complications. However, a deeper understanding of the functional, biochemical, and molecular characteristics of metallothionein is needed to bring about new opportunities for OS therapy. This review focuses on newly proposed functions of a metallothionein and their implications relevant to diabetes and its complications.
Collapse
Affiliation(s)
- Yongsoo Park
- Department of Pediatrics, Pediatrics Research Institute, University of Louisville, Louisville, Kentucky, USA
- Hanyang University, College of Medicine and Engineering, Seoul, South Korea
| | - Jian Zhang
- Department of Pediatrics, Pediatrics Research Institute, University of Louisville, Louisville, Kentucky, USA
- The Center of Cardiovascular Disorders, The First Hospital of Jilin University, Changchun, China
| | - Lu Cai
- Department of Pediatrics, Pediatrics Research Institute, University of Louisville, Louisville, Kentucky, USA
- Department of Radiation Oncology, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
22
|
Wu X, Li M, Chen SQ, Li S, Guo F. Pin1 facilitates isoproterenol‑induced cardiac fibrosis and collagen deposition by promoting oxidative stress and activating the MEK1/2‑ERK1/2 signal transduction pathway in rats. Int J Mol Med 2017; 41:1573-1583. [PMID: 29286102 PMCID: PMC5819929 DOI: 10.3892/ijmm.2017.3354] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 12/15/2017] [Indexed: 01/08/2023] Open
Abstract
Peptidyl-prolyl cis/trans isomerase, NIMA-interacting 1 (Pin1) is a member of a large superfamily of phosphorylation-dependent peptidyl-prolyl cis/trans isomerases, which not only regulates multiple targets at various stages of cellular processes, but is also involved in the pathogenesis of several diseases, including microbial infection, cancer, asthma and Alzheimer's disease. However, the role of Pin1 in cardiac fibrosis remains to be fully elucidated. The present study investigated the potential mechanism of Pin1 in isoprenaline (ISO)-induced myocardial fibrosis in rats. The rats were randomly divided into three groups. Echocardiography was used to evaluate changes in the size, shape and function of the heart, and histological staining was performed to visualize inflammatory cell infiltration and fibrosis. Reverse transcription-quantitative polymerase chain reaction analysis, immunohistochemistry and Picrosirius red staining were used to differentiate collagen subtypes. Additionally, cardiac-specific phosphorylation of mitogen-activated protein kinase kinase 1/2 (MEK1/2) and extracellular-signal regulated protein kinase 1/2 (ERK1/2), and the activities of Pin1 and α-smooth muscle actin (α-SMA) and other oxidative stress parameters were estimated in the heart. The administration of ISO resulted in an increase in cardiac parameters and elevated the heart-to-body weight ratio. Histopathological examination of heart tissues revealed interstitial inflammatory cellular infiltrate and disorganized collagen fiber deposition. In addition, lipid peroxidation products and oxidative stress marker activity in plasma and tissues were significantly increased in the ISO-treated rats. Western blot analysis showed significantly elevated protein levels of phosphorylated Pin1, MEK1/2, ERK1/2 and α-SMA in remodeling hearts. Treatment with juglone following intraperitoneal injection of ISO significantly prevented inflammatory cell infiltration, improved cardiac function, and suppressed oxidative stresses and fibrotic alterations. In conclusion, the results of the present study suggested that the activation of Pin1 promoted cardiac extracellular matrix deposition and oxidative stress damage by regulating the phosphorylation of the MEK1/2-ERK1/2 signaling pathway and the expression of α-SMA. By contrast, the inhibition of Pin1 alleviated cardiac damage and fibrosis in the experimental models, suggesting that Pin1 contributed to the development of cardiac remodeling in ISO-administered rats, and that the inactivation of Pin1 may be a novel therapeutic candidate for the treatment of cardiovascular disease and heart failure.
Collapse
Affiliation(s)
- Xian Wu
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Mingjiang Li
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Su-Qin Chen
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Sha Li
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Furong Guo
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
23
|
Turan B, Tuncay E. Impact of Labile Zinc on Heart Function: From Physiology to Pathophysiology. Int J Mol Sci 2017; 18:ijms18112395. [PMID: 29137144 PMCID: PMC5713363 DOI: 10.3390/ijms18112395] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/02/2017] [Accepted: 11/08/2017] [Indexed: 12/15/2022] Open
Abstract
Zinc plays an important role in biological systems as bound and histochemically reactive labile Zn2+. Although Zn2+ concentration is in the nM range in cardiomyocytes at rest and increases dramatically under stimulation, very little is known about precise mechanisms controlling the intracellular distribution of Zn2+ and its variations during cardiac function. Recent studies are focused on molecular and cellular aspects of labile Zn2+ and its homeostasis in mammalian cells and growing evidence clarified the molecular mechanisms underlying Zn2+-diverse functions in the heart, leading to the discovery of novel physiological functions of labile Zn2+ in parallel to the discovery of subcellular localization of Zn2+-transporters in cardiomyocytes. Additionally, important experimental data suggest a central role of intracellular labile Zn2+ in excitation-contraction coupling in cardiomyocytes by shaping Ca2+ dynamics. Cellular labile Zn2+ is tightly regulated against its adverse effects through either Zn2+-transporters, Zn2+-binding molecules or Zn2+-sensors, and, therefore plays a critical role in cellular signaling pathways. The present review summarizes the current understanding of the physiological role of cellular labile Zn2+ distribution in cardiomyocytes and how a remodeling of cellular Zn2+-homeostasis can be important in proper cell function with Zn2+-transporters under hyperglycemia. We also emphasize the recent investigations on Zn2+-transporter functions from the standpoint of human heart health to diseases together with their clinical interest as target proteins in the heart under pathological condition, such as diabetes.
Collapse
Affiliation(s)
- Belma Turan
- Department of Biophysics, Ankara University, Faculty of Medicine, 06100 Ankara, Turkey.
| | - Erkan Tuncay
- Department of Biophysics, Ankara University, Faculty of Medicine, 06100 Ankara, Turkey.
| |
Collapse
|
24
|
Zinc enhances the cellular energy supply to improve cell motility and restore impaired energetic metabolism in a toxic environment induced by OTA. Sci Rep 2017; 7:14669. [PMID: 29116164 PMCID: PMC5676743 DOI: 10.1038/s41598-017-14868-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/12/2017] [Indexed: 02/08/2023] Open
Abstract
Exogenous nutrient elements modulate the energetic metabolism responses that are prerequisites for cellular homeostasis and metabolic physiology. Although zinc is important in oxidative stress and cytoprotection processes, its role in the regulation of energetic metabolism remains largely unknown. In this study, we found that zinc stimulated aspect in cell motility and was essential in restoring the Ochratoxin A (OTA)-induced energetic metabolism damage in HEK293 cells. Moreover, using zinc supplementation and zinc deficiency models, we observed that zinc is conducive to mitochondrial pyruvate transport, oxidative phosphorylation, carbohydrate metabolism, lipid metabolism and ultimate energy metabolism in both normal and toxic-induced oxidative stress conditions in vitro, and it plays an important role in restoring impaired energetic metabolism. This zinc-mediated energetic metabolism regulation could also be helpful for DNA maintenance, cytoprotection and hereditary cancer traceability. Therefore, zinc can widely adjust energetic metabolism and is essential in restoring the impaired energetic metabolism of cellular physiology.
Collapse
|
25
|
Cardioprotective effects of fibroblast growth factor 21 against doxorubicin-induced toxicity via the SIRT1/LKB1/AMPK pathway. Cell Death Dis 2017; 8:e3018. [PMID: 28837153 PMCID: PMC5596591 DOI: 10.1038/cddis.2017.410] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/11/2017] [Accepted: 07/12/2017] [Indexed: 12/12/2022]
Abstract
Doxorubicin (DOX) is a highly effective antineoplastic anthracycline drug; however, the adverse effect of the cardiotoxicity has limited its widespread application. Fibroblast growth factor 21 (FGF21), as a well-known regulator of glucose and lipid metabolism, was recently shown to exert cardioprotective effects. The aim of this study was to investigate the possible protective effects of FGF21 against DOX-induced cardiomyopathy. We preliminarily established DOX-induced cardiotoxicity models in H9c2 cells, adult mouse cardiomyocytes, and 129S1/SyImJ mice, which clearly showed cardiac dysfunction and myocardial collagen accumulation accompanying by inflammatory, oxidative stress, and apoptotic damage. Treatment with FGF21 obviously attenuated the DOX-induced cardiac dysfunction and pathological changes. Its effective anti-inflammatory activity was revealed by downregulation of inflammatory factors (tumor necrosis factor-α and interleukin-6) via the IKK/IκBα/nuclear factor-κB pathway. The anti-oxidative stress activity of FGF21 was achieved via reduced generation of reactive oxygen species through regulation of nuclear transcription factor erythroid 2-related factor 2 transcription. Its anti-apoptotic activity was shown by reductions in the number of TUNEL-positive cells and DNA fragments along with a decreased ratio of Bax/Bcl-2 expression. In a further mechanistic study, FGF21 enhanced sirtuin 1 (SIRT1) binding to liver kinase B1 (LKB1) and then decreased LKB1 acetylation, subsequently inducing AMP-activated protein kinase (AMPK) activation, which improved the cardiac inflammation, oxidative stress, and apoptosis. These alterations were significantly prohibited by SIRT1 RNAi. The present work demonstrates for the first time that FGF21 obviously prevented DOX-induced cardiotoxicity via the suppression of oxidative stress, inflammation, and apoptosis through the SIRT1/LKB1/AMPK signaling pathway.
Collapse
|
26
|
Wilde E, Aubdool AA, Thakore P, Baldissera L, Alawi KM, Keeble J, Nandi M, Brain SD. Tail-Cuff Technique and Its Influence on Central Blood Pressure in the Mouse. J Am Heart Assoc 2017; 6:e005204. [PMID: 28655735 PMCID: PMC5669161 DOI: 10.1161/jaha.116.005204] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/24/2017] [Indexed: 12/05/2022]
Abstract
BACKGROUND Reliable measurement of blood pressure in conscious mice is essential in cardiovascular research. Telemetry, the "gold-standard" technique, is invasive and expensive and therefore tail-cuff, a noninvasive alternative, is widely used. However, tail-cuff requires handling and restraint during measurement, which may cause stress affecting blood pressure and undermining reliability of the results. METHODS AND RESULTS C57Bl/6J mice were implanted with radio-telemetry probes to investigate the effects of the steps of the tail-cuff technique on central blood pressure, heart rate, and temperature. This included comparison of handling techniques, operator's sex, habituation, and influence of hypertension induced by angiotensin II. Direct comparison of measurements obtained by telemetry and tail-cuff were made in the same mouse. The results revealed significant increases in central blood pressure, heart rate, and core body temperature from baseline following handling interventions without significant difference among the different handling technique, habituation, or sex of the investigator. Restraint induced the largest and sustained increase in cardiovascular parameters and temperature. The tail-cuff readings significantly underestimated those from simultaneous telemetry recordings; however, "nonsimultaneous" telemetry, obtained in undisturbed mice, were similar to tail-cuff readings obtained in undisturbed mice on the same day. CONCLUSIONS This study reveals that the tail-cuff technique underestimates the core blood pressure changes that occur simultaneously during the restraint and measurement phases. However, the measurements between the 2 techniques are similar when tail-cuff readings are compared with telemetry readings in the nondisturbed mice. The differences between the simultaneous recordings by the 2 techniques should be recognized by researchers.
Collapse
Affiliation(s)
- Elena Wilde
- Vascular Biology and Inflammation Section, BHF Cardiovascular Centre of Research Excellence, Cardiovascular Division, King's College London, London, United Kingdom
| | - Aisah A Aubdool
- Vascular Biology and Inflammation Section, BHF Cardiovascular Centre of Research Excellence, Cardiovascular Division, King's College London, London, United Kingdom
| | - Pratish Thakore
- Pharmaceutical Sciences Division, King's College London, London, United Kingdom
| | - Lineu Baldissera
- Vascular Biology and Inflammation Section, BHF Cardiovascular Centre of Research Excellence, Cardiovascular Division, King's College London, London, United Kingdom
| | - Khadija M Alawi
- Vascular Biology and Inflammation Section, BHF Cardiovascular Centre of Research Excellence, Cardiovascular Division, King's College London, London, United Kingdom
| | - Julie Keeble
- Pharmaceutical Sciences Division, King's College London, London, United Kingdom
| | - Manasi Nandi
- Pharmaceutical Sciences Division, King's College London, London, United Kingdom
| | - Susan D Brain
- Vascular Biology and Inflammation Section, BHF Cardiovascular Centre of Research Excellence, Cardiovascular Division, King's College London, London, United Kingdom
| |
Collapse
|
27
|
Wang S, Gu J, Xu Z, Zhang Z, Bai T, Xu J, Cai J, Barnes G, Liu Q, Freedman JH, Wang Y, Liu Q, Zheng Y, Cai L. Zinc rescues obesity-induced cardiac hypertrophy via stimulating metallothionein to suppress oxidative stress-activated BCL10/CARD9/p38 MAPK pathway. J Cell Mol Med 2017; 21:1182-1192. [PMID: 28158919 PMCID: PMC5431126 DOI: 10.1111/jcmm.13050] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 10/30/2016] [Indexed: 12/13/2022] Open
Abstract
Obesity often leads to obesity-related cardiac hypertrophy (ORCH), which is suppressed by zinc-induced inactivation of p38 mitogen-activated protein kinase (p38 MAPK). In this study, we investigated the mechanisms by which zinc inactivates p38 MAPK to prevent ORCH. Mice (4-week old) were fed either high fat diet (HFD, 60% kcal fat) or normal diet (ND, 10% kcal fat) containing variable amounts of zinc (deficiency, normal and supplement) for 3 and 6 months. P38 MAPK siRNA and the p38 MAPK inhibitor SB203580 were used to suppress p38 MAPK activity in vitro and in vivo, respectively. HFD activated p38 MAPK and increased expression of B-cell lymphoma/CLL 10 (BCL10) and caspase recruitment domain family member 9 (CARD9). These responses were enhanced by zinc deficiency and attenuated by zinc supplement. Administration of SB203580 to HFD mice or specific siRNA in palmitate-treated cardiomyocytes eliminated the HFD and zinc deficiency activation of p38 MAPK, but did not significantly impact the expression of BCL10 and CARD9. In cultured cardiomyocytes, inhibition of BCL10 expression by siRNA prevented palmitate-induced increased p38 MAPK activation and atrial natriuretic peptide (ANP) expression. In contrast, inhibition of p38 MAPK prevented ANP expression, but did not affect BCL10 expression. Deletion of metallothionein abolished the protective effect of zinc on palmitate-induced up-regulation of BCL10 and phospho-p38 MAPK. HFD and zinc deficiency synergistically induce ORCH by increasing oxidative stress-mediated activation of BCL10/CARD9/p38 MAPK signalling. Zinc supplement ameliorates ORCH through activation of metallothionein to repress oxidative stress-activated BCL10 expression and p38 MAPK activation.
Collapse
Affiliation(s)
- Shudong Wang
- Cardiovascular CenterThe First Hospital of Jilin UniversityChangchunJilinChina
- Department of PediatricsKosair Children's Hospital Research InstituteUniversity of LouisvilleLouisvilleKYUSA
| | - Junlian Gu
- Department of PediatricsKosair Children's Hospital Research InstituteUniversity of LouisvilleLouisvilleKYUSA
| | - Zheng Xu
- Cardiovascular CenterThe First Hospital of Jilin UniversityChangchunJilinChina
- Department of PediatricsKosair Children's Hospital Research InstituteUniversity of LouisvilleLouisvilleKYUSA
| | - Zhiguo Zhang
- Cardiovascular CenterThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Tao Bai
- Cardiovascular CenterThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Jianxiang Xu
- Department of PediatricsKosair Children's Hospital Research InstituteUniversity of LouisvilleLouisvilleKYUSA
| | - Jun Cai
- Department of PediatricsKosair Children's Hospital Research InstituteUniversity of LouisvilleLouisvilleKYUSA
| | - Gregory Barnes
- Department of PediatricsKosair Children's Hospital Research InstituteUniversity of LouisvilleLouisvilleKYUSA
- Autism CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Qiu‐Ju Liu
- Department of Hematology DisordersThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Jonathan H. Freedman
- Department of Pharmacology and ToxicologyUniversity of LouisvilleLouisvilleKYUSA
| | - Yonggang Wang
- Cardiovascular CenterThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Quan Liu
- Cardiovascular CenterThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Yang Zheng
- Cardiovascular CenterThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Lu Cai
- Department of PediatricsKosair Children's Hospital Research InstituteUniversity of LouisvilleLouisvilleKYUSA
- Department of Pharmacology and ToxicologyUniversity of LouisvilleLouisvilleKYUSA
- Wendy Novak Diabetes Care CenterUniversity of LouisvilleLouisvilleKYUSA
| |
Collapse
|
28
|
Wang S, Wang B, Wang Y, Tong Q, Liu Q, Sun J, Zheng Y, Cai L. Zinc Prevents the Development of Diabetic Cardiomyopathy in db/db Mice. Int J Mol Sci 2017; 18:580. [PMID: 28272348 PMCID: PMC5372596 DOI: 10.3390/ijms18030580] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 02/23/2017] [Accepted: 02/26/2017] [Indexed: 01/04/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is highly prevalent in type 2 diabetes (T2DM) patients. Zinc is an important essential trace metal, whose deficiency is associated with various chronic ailments, including vascular diseases. We assessed T2DM B6.BKS(D)-Leprdb/J (db/db) mice fed for six months on a normal diet containing three zinc levels (deficient, adequate, and supplemented), to explore the role of zinc in DCM development and progression. Cardiac function, reflected by ejection fraction, was significantly decreased, along with increased left ventricle mass and heart weight to tibial length ratio, in db/db mice. As a molecular cardiac hypertrophy marker, atrial natriuretic peptide levels were also significantly increased. Cardiac dysfunction and hypertrophy were accompanied by significantly increased fibrotic (elevated collagen accumulation as well as transforming growth factor β and connective tissue growth factor levels) and inflammatory (enhanced expression of tumor necrosis factor alpha, interleukin-1β, caspase recruitment domain family member 9, and B-cell lymphoma/leukemia 10, and activated p38 mitogen-activated protein kinase) responses in the heart. All these diabetic effects were exacerbated by zinc deficiency, and not affected by zinc supplementation, respectively. Mechanistically, oxidative stress and damage, mirrored by the accumulation of 3-nitrotyrosine and 4-hydroxy-2-nonenal, was significantly increased along with significantly decreased expression of Nrf2 and its downstream antioxidants (NQO-1 and catalase). This was also exacerbated by zinc deficiency in the db/db mouse heart. These results suggested that zinc deficiency promotes the development and progression of DCM in T2DM db/db mice. The exacerbated effects by zinc deficiency on the heart of db/db mice may be related to further suppression of Nrf2 expression and function.
Collapse
Affiliation(s)
- Shudong Wang
- Cardiovascular Center & Geriatric Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin, China.
- Pediatric Research Institute, The Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA.
| | - Bowei Wang
- Pediatric Research Institute, The Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA.
- Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, Jilin, China.
| | - Yuehui Wang
- Cardiovascular Center & Geriatric Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Qian Tong
- Cardiovascular Center & Geriatric Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Quan Liu
- Cardiovascular Center & Geriatric Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Jian Sun
- Cardiovascular Center & Geriatric Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Yang Zheng
- Cardiovascular Center & Geriatric Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Lu Cai
- Pediatric Research Institute, The Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA.
- Wendy Novak Diabetes Care Center, Departments of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
29
|
Xu Z, Sun J, Tong Q, Lin Q, Qian L, Park Y, Zheng Y. The Role of ERK1/2 in the Development of Diabetic Cardiomyopathy. Int J Mol Sci 2016; 17:2001. [PMID: 27941647 PMCID: PMC5187801 DOI: 10.3390/ijms17122001] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/14/2016] [Accepted: 11/22/2016] [Indexed: 12/23/2022] Open
Abstract
Diabetes mellitus is a chronic metabolic condition that affects carbohydrate, lipid and protein metabolism and may impair numerous organs and functions of the organism. Cardiac dysfunction afflicts many patients who experience the oxidative stress of the heart. Diabetic cardiomyopathy (DCM) is one of the major complications that accounts for more than half of diabetes-related morbidity and mortality cases. Chronic hyperglycemia and hyperlipidemia from diabetes mellitus cause cardiac oxidative stress, endothelial dysfunction, impaired cellular calcium handling, mitochondrial dysfunction, metabolic disturbances, and remodeling of the extracellular matrix, which ultimately lead to DCM. Although many studies have explored the mechanisms leading to DCM, the pathophysiology of DCM has not yet been fully clarified. In fact, as a potential mechanism, the associations between DCM development and mitogen-activated protein kinase (MAPK) activation have been the subjects of tremendous interest. Nonetheless, much remains to be investigated, such as tissue- and cell-specific processes of selection of MAPK activation between pro-apoptotic vs. pro-survival fate, as well as their relation with the pathogenesis of diabetes and associated complications. In general, it turns out that MAPK signaling pathways, such as extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal protein kinase (JNK) and p38 MAP kinase, are demonstrated to be actively involved in myocardial dysfunction, hypertrophy, fibrosis and heart failure. As one of MAPK family members, the activation of ERK1/2 has also been known to be involved in cardiac hypertrophy and dysfunction. However, many recent studies have demonstrated that ERK1/2 signaling activation also plays a crucial role in FGF21 signaling and exerts a protective environment of glucose and lipid metabolism, therefore preventing abnormal healing and cardiac dysfunction. The duration, extent, and subcellular compartment of ERK1/2 activation are vital to differential biological effects of ERK1/2. Moreover, many intracellular events, including mitochondrial signaling and protein kinases, manipulate signaling upstream and downstream of MAPK, to influence myocardial survival or death. In this review, we will summarize the roles of ERK1/2 pathways in DCM development by the evidence from current studies and will present novel opinions on "differential influence of ERK1/2 action in cardiac dysfunction, and protection against myocardial ischemia-reperfusion injury".
Collapse
Affiliation(s)
- Zheng Xu
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
- Department of Pediatrics, Kosair Children's Hospital Research Institute, University of Louisville, Louisville, KY 40202, USA.
| | - Jian Sun
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Qian Tong
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| | - Qian Lin
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40202, USA.
| | - Lingbo Qian
- Department of Pediatrics, Kosair Children's Hospital Research Institute, University of Louisville, Louisville, KY 40202, USA.
- Department of Basic Medical Sciences, Hangzhou Medical College, Hangzhou 310053, China.
| | - Yongsoo Park
- Department of Pediatrics, Kosair Children's Hospital Research Institute, University of Louisville, Louisville, KY 40202, USA.
- College of Medicine & Engineering, Hanyang University, Seoul 04963, Korea.
| | - Yang Zheng
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
30
|
Jagadapillai R, Rane MJ, Lin X, Roberts AM, Hoyle GW, Cai L, Gozal E. Diabetic Microvascular Disease and Pulmonary Fibrosis: The Contribution of Platelets and Systemic Inflammation. Int J Mol Sci 2016; 17:1853. [PMID: 27834824 PMCID: PMC5133853 DOI: 10.3390/ijms17111853] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/24/2016] [Accepted: 11/01/2016] [Indexed: 12/29/2022] Open
Abstract
Diabetes is strongly associated with systemic inflammation and oxidative stress, but its effect on pulmonary vascular disease and lung function has often been disregarded. Several studies identified restrictive lung disease and fibrotic changes in diabetic patients and in animal models of diabetes. While microvascular dysfunction is a well-known complication of diabetes, the mechanisms leading to diabetes-induced lung injury have largely been disregarded. We described the potential involvement of diabetes-induced platelet-endothelial interactions in perpetuating vascular inflammation and oxidative injury leading to fibrotic changes in the lung. Changes in nitric oxide synthase (NOS) activation and decreased NO bioavailability in the diabetic lung increase platelet activation and vascular injury and may account for platelet hyperreactivity reported in diabetic patients. Additionally, the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway has been reported to mediate pancreatic islet damage, and is implicated in the onset of diabetes, inflammation and vascular injury. Many growth factors and diabetes-induced agonists act via the JAK/STAT pathway. Other studies reported the contribution of the JAK/STAT pathway to the regulation of the pulmonary fibrotic process but the role of this pathway in the development of diabetic lung fibrosis has not been considered. These observations may open new therapeutic perspectives for modulating multiple pathways to mitigate diabetes onset or its pulmonary consequences.
Collapse
Affiliation(s)
- Rekha Jagadapillai
- Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
| | - Madhavi J Rane
- Medicine/Nephrology, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
- Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
| | - Xingyu Lin
- Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
- Department of Thoracic Surgery, the First Hospital of Jilin University, Changchun 130021, China.
| | - Andrew M Roberts
- Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
- Physiology, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
| | - Gary W Hoyle
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY 40292, USA.
| | - Lu Cai
- Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
- Pharmacology & Toxicology, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
- Radiation Oncology, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
| | - Evelyne Gozal
- Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
- Physiology, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
- Pharmacology & Toxicology, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|