1
|
Li X, Zhang Z, Li C, Liu J, Fang Q, Zhang M, Huang J. Novel applications of metformin in the treatment of septic myocardial injury based on metabolomics and network pharmacology. Eur J Pharmacol 2025; 986:177141. [PMID: 39566813 DOI: 10.1016/j.ejphar.2024.177141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/08/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND While metformin has shown promise in treating septic myocardial injury (SMI), its underlying mechanisms and impact on metabolic disturbances remain poorly understood. METHODS This study employed an integrated approach of metabolomics and network pharmacology to identify key targets and pathways through which metformin may act against SMI. Findings were validated using a lipopolysaccharide (LPS)-induced mouse model. RESULTS Metformin was found to counter myocardial metabolic disruptions, indicated by the reversal of 49 metabolites primarily involved in purine metabolism, pantothenate and CoA biosynthesis, and histidine metabolism. In vivo, metformin significantly improved survival rates and cardiac function, reduced cardiomyocyte apoptosis, and inhibited inflammation and oxidative stress in LPS-induced mice. Integrated analyses identified 27 potential targets for metformin in SMI treatment. KEGG pathway analysis revealed significant enrichment in TNF, HIF-1, IL-17, and PI3K/AKT signaling pathways, while protein-protein interaction analysis pinpointed ten core targets, including IL6, IL1B, CCL2, CASP3, MMP9, HIF1A, IGF1, NOS3, MMP2, and LEP. Molecular docking and dynamics simulations demonstrated metformin's high affinity for these core targets. Further, RT-qPCR and Western blot analyses confirmed that metformin modulates core target expression to mitigate SMI. Notably, our data underscore the importance of PI3K/AKT and MMP2/MMP9 signaling pathways in SMI therapy. CONCLUSION This study elucidates the metabolic and molecular mechanisms of metformin in SMI treatment, supporting its potential repurposing for SMI.
Collapse
Affiliation(s)
- Xingyu Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zihan Zhang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chaohong Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Henan Key Laboratory of Neurorestoratology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Jun Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinghua Fang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Muzi Zhang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Huang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Park JS, Jeon H, Lee Y, Cheon SY, Lee D, Lim SG, Koo H. Rapid DNA Repair in Mesenchymal Stem Cells and Bone Regeneration by Nanoparticle-Based Codelivery of Nrf2-mRNA and Dexamethasone. ACS NANO 2024; 18:31877-31890. [PMID: 39520360 DOI: 10.1021/acsnano.4c08939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Directional differentiation is a key factor determining the result of stem cell therapy. Herein, we developed a polyethylenimine (PEI)-coated poly(lactic-co-glycolic) acid (PLGA) nanoparticle (mPDN) carrying both nuclear factor erythroid 2-related factor 2 (Nrf2) mRNA and dexamethasone (Dex) to human mesenchymal stem cells (hMSCs). The combination of Dex and Nrf2-mRNA delivered by mPDN promoted the osteogenic differentiation of hMSCs. In particular, Nrf2-mRNA rapidly reduced the DNA damage caused by ROS due to early and efficient gene expression at 3 h after treatment, which was not achieved in traditional pDNA systems. High and rapid transfection, effective ROS-scavenging effect, and protection of mitochondrial dynamics were observed in hMSCs after treatment with the resulting Nrf2-mPDN. Osteogenic differentiation was also observed in 3D pellets for up to 5 weeks. Finally, the effects of rapid DNA repair in hMSCs by Nrf2-mPDN and on in vivo bone regeneration were evaluated in a rat femoral bone defect model using CT. This study demonstrated the potential of an NP-based codelivery system and efficient transfection of mRNA at early stages in hMSCs for bone regeneration and stem cell therapy.
Collapse
Affiliation(s)
- Ji Sun Park
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Hayoung Jeon
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yeeun Lee
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seo Young Cheon
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Donghyun Lee
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seong Gi Lim
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Heebeom Koo
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| |
Collapse
|
3
|
Jha G, Sharma RB, Sridhar S, Hayagreev D, Sinha T, Kaur H, Das A, Bollineni RL. Nanoparticle-Based Therapies for Cardiovascular Diseases: A Literature Review of Recent Advances and Clinical Potential. Cureus 2024; 16:e72808. [PMID: 39552990 PMCID: PMC11569831 DOI: 10.7759/cureus.72808] [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] [Accepted: 10/31/2024] [Indexed: 11/19/2024] Open
Abstract
Cardiovascular diseases (CVDs) present a significant global health burden and remain the leading cause of morbidity and mortality worldwide. Conventional pharmacological therapies have yielded limited success in addressing the underlying pathophysiology of these diseases, leading to the exploration of novel therapeutic approaches. Nanotechnology is transforming cardiovascular disease management by enabling the engineering of materials at the atomic and molecular levels. This has led to the development of advanced diagnostic tools with unparalleled accuracy and sensitivity in detecting these diseases. By enabling targeted drug delivery, enhancing imaging techniques, and facilitating personalized therapies, nanotechnology promises significant advancements in the diagnosis, treatment, and prevention of cardiovascular diseases. This narrative review provides a comprehensive outlook on the recent advancements in nanoparticle-based therapies for cardiovascular diseases. We delve into the diverse applications of various nanoparticle types, exploring their potential to surpass the limitations of conventional treatments and improve clinical outcomes. Additionally, we critically examine the challenges and future directions of this rapidly evolving field, emphasizing the need for rigorous clinical evaluation.
Collapse
Affiliation(s)
- Gaurav Jha
- Trauma and Orthopaedics, Leicester Royal Infirmary, Leicester, GBR
| | - Ritika B Sharma
- Geriatrics, Pinderfields General Hospital, MidYorkshire, GBR
| | - Sruthi Sridhar
- Emergency Department, Croydon Health Services NHS Trust, London, GBR
| | - Disha Hayagreev
- Emergency Department, Basingstoke and North Hampshire Hospital, Basingstoke, GBR
| | - Tanya Sinha
- Emergency Medicine, South Tyneside and Sunderland NHS Foundation Trust, South Sheilds, GBR
| | | | - Adrija Das
- Medicine, Newcastle University, Newcastle, GBR
| | | |
Collapse
|
4
|
Cui Y, Wu J, Wang Y, Li D, Zhang F, Jin X, Li M, Zhang J, Liu Z. Protective effects of ginsenoside F 2 on isoproterenol-induced myocardial infarction by activating the Nrf2/HO-1 and PI3K/Akt signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155637. [PMID: 38669969 DOI: 10.1016/j.phymed.2024.155637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/23/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Ginsenoside F2 (GF2) serves as the principal intestinal metabolite resulting from the oral intake of Panax ginseng and Panax quinquefolius, exhibiting antioxidative, hypolipidemic, antitumor, and anti-inflammatory properties. Nevertheless, its effect on myocardial infarction (MI) is still unknown. PURPOSE The purpose of this study is to investigate the protective effect and the underlying mechanisms of GF2 against isoproterenol (ISO)-induced MI. METHODS ISO-induced H9c2 cardiomyocytes and MI rat models were utilized as in vitro and in vivo models to evaluate the impact of anti-MI of GF2. The underlying mechanisms were investigated using a variety of methodologies, including electrocardiography, Western blot analysis, histopathological examination, immunofluorescence, immunohistochemistry, and ELISA techniques. RESULTS In vivo experiments, our results indicated that GF2 significantly ameliorated ISO-induced electrocardiographic (ECG) abnormalities, myocardial fiber necrosis, rupture, fibrosis of myocardial tissues, and suppressed cardiac enzyme activities. Meanwhile, GF2 notably raised the activity of antioxidant enzymes like CAT, GSH, and SOD. Furthermore, it downregulated Keap1 expression level while upregulating NQO1, Nrf2, and HO-1 expression levels. Additionally, GF2 suppressed the expression of the cleaved caspase-3 and pro-apoptotic protein Bax while promoting the expression of anti-apoptotic proteins Bcl-2, p-PI3K, and p-Akt. TUNEL fluorescence results also demonstrated that GF2 effectively inhibited cardiomyocyte apoptosis. Furthermore, consistent with the results of animal experiments, GF2 considerably attenuated ROS generation, changed apoptosis and mitochondrial function, and reduced oxidative stress in ISO-induced H9c2 cardiomyocytes through activating Nrf2/HO-1 and PI3K/Akt signaling pathways. CONCLUSION Taken together, GF2 ameliorated MI by preventing cardiocyte apoptosis, oxidative stress, and mitochondrial dysfunction via modulating the Nrf2/HO-1 and PI3K/Akt signaling pathways, showing potential as a treatment strategy for treating MI.
Collapse
Affiliation(s)
- Ying Cui
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Jianfa Wu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Yanfang Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Dan Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Furui Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Xiaoman Jin
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Meihui Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Jing Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Changchun 130118, China
| | - Zhi Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Changchun 130118, China.
| |
Collapse
|
5
|
Nemati S, Zavvari-Oskuye Z, Bafadam S, Mokhtari B, Badalzadeh R, Vakili A. Impact of combined alpha-lipoic acid and mitoquinone supplementation on myocardial infarction in aged rats: Heart performance and molecular mechanisms. Exp Gerontol 2024; 189:112402. [PMID: 38484905 DOI: 10.1016/j.exger.2024.112402] [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: 11/25/2023] [Revised: 01/16/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND This study aimed to investigate the effects of combined alpha-lipoic acid (ALA) and mitoquinone (Mito Q) supplementation on cardiac function and the underlying mechanisms in aged rats with myocardial infarction (MI). METHODS The aged rats underwent left anterior descending artery (LADA) occlusion for 30 min, followed by reperfusion for 24 h. ALA (100 mg/kg, gavage) and Mito Q (10 mg/kg, IP) were administered daily for two weeks before ischemia. Cardiac function, inflammatory, and apoptotic markers were evaluated 24 h after ischemia. RESULTS The results of this study indicated that the administration of the combination of ALA and Mito Q significantly improved cardiac function. This improvement was linked to a reduction in the expression of pro-inflammatory cytokines TNF-α, IL-6, and IL-1β (P < 0.001) and apoptotic markers (Bax, caspase-3, and Cyt-c), as well as a decrease in the percentage of TUNEL-positive cells (P < 0.001). CONCLUSION The study revealed that combined intervention synergistically mitigated cardiac dysfunction by suppressing inflammatory and apoptotic pathways in aged rats with MI. Further research is needed to validate the potential of ALA and Mito Q as therapeutic options for elderly people at risk of heart attacks.
Collapse
Affiliation(s)
- Samira Nemati
- Research Center of Physiology, Semnan University of Medical Sciences, Iran
| | - Zohreh Zavvari-Oskuye
- Drug Applied Research Center, Tabriz University of Medical Sciences, Iran; Student Research Committee, Tabriz University of Medical Sciences, Iran
| | - Soleyman Bafadam
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Iran; Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Iran; Student Research Committee, Tabriz University of Medical Sciences, Iran
| | - Behnaz Mokhtari
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Iran; Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Iran
| | - Reza Badalzadeh
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Iran; Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Iran
| | - Abedin Vakili
- Research Center of Physiology, Semnan University of Medical Sciences, Iran.
| |
Collapse
|
6
|
Wang H, Han J, Dmitrii G, Zhang XA. Potential Targets of Natural Products for Improving Cardiac Ischemic Injury: The Role of Nrf2 Signaling Transduction. Molecules 2024; 29:2005. [PMID: 38731496 PMCID: PMC11085255 DOI: 10.3390/molecules29092005] [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: 03/22/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Myocardial ischemia is the leading cause of health loss from cardiovascular disease worldwide. Myocardial ischemia and hypoxia during exercise trigger the risk of sudden exercise death which, in severe cases, will further lead to myocardial infarction. The Nrf2 transcription factor is an important antioxidant regulator that is extensively engaged in biological processes such as oxidative stress, inflammatory response, apoptosis, and mitochondrial malfunction. It has a significant role in the prevention and treatment of several cardiovascular illnesses, since it can control not only the expression of several antioxidant genes, but also the target genes of associated pathological processes. Therefore, targeting Nrf2 will have great potential in the treatment of myocardial ischemic injury. Natural products are widely used to treat myocardial ischemic diseases because of their few side effects. A large number of studies have shown that the Nrf2 transcription factor can be used as an important way for natural products to alleviate myocardial ischemia. However, the specific role and related mechanism of Nrf2 in mediating natural products in the treatment of myocardial ischemia is still unclear. Therefore, this review combs the key role and possible mechanism of Nrf2 in myocardial ischemic injury, and emphatically summarizes the significant role of natural products in treating myocardial ischemic symptoms, thus providing a broad foundation for clinical transformation.
Collapse
Affiliation(s)
- Haixia Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, China; (H.W.); (J.H.)
| | - Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, China; (H.W.); (J.H.)
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Gorbachev Dmitrii
- General Hygiene Department, Samara State Medical University, Samara 443000, Russia;
| | - Xin-an Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, China; (H.W.); (J.H.)
| |
Collapse
|
7
|
Oskuye ZZ, Mehri K, Mokhtari B, Bafadam S, Nemati S, Badalzadeh R. Cardioprotective effect of antioxidant combination therapy: A highlight on MitoQ plus alpha-lipoic acid beneficial impact on myocardial ischemia-reperfusion injury in aged rats. Heliyon 2024; 10:e28158. [PMID: 38524576 PMCID: PMC10957437 DOI: 10.1016/j.heliyon.2024.e28158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 03/26/2024] Open
Abstract
Objective (s): Considering the poor prognosis of ischemic heart disease and the diminished effectiveness of cardioprotective interventions in the elderly, it becomes necessary to investigate the interaction of aging with protection during myocardial ischemia/reperfusion injury (IRI). This study was conducted to assess the impact of mitoquinone (MitoQ) and alpha-lipoic acid (ALA) preconditioning on cardioprotection following IRI in aged rats. Methods Fifty aged male Wistar rats (22-24 months old) were divided into five groups including Sham, IR, and treatment groups receiving ALA and/or MitoQ. Treatment groups were received 100 mg/kg/day ALA by oral gavage and/or 10 mg/kg/day MitoQ by intraperitoneal injection for 14 consecutive days. An in vivo model of myocardial IRI was established through ligation of coronary artery for 30 min and it's reopening for 24 h. The left ventricles were removed at the end of reperfusion to assess oxidative stress indicators, mitochondrial function, and expression of mitochondrial dynamic genes. Myocardial infarct size (IS), hemodynamic parameters, and serum lactate dehydrogenase (LDH) level were also measured. Results Combination of MitoQ and ALA reduced oxidative stress, LDH level, and IS in aged hearts subjected to IRI. It also enhanced mitochondrial function and upregulated Mfn1, Mfn2, and Foxo1 and downregulated Drp1 and Fis1 gene expression. Co-administration of MitoQ and ALA partially restored IRI-induced hemodynamic changes to normal state. In all measured parameters, the effect of combined treatment was greater than monotherapies. Conclusion The combination therapy of MitoQ and ALA demonstrated considerable therapeutic potential in protecting the aging heart against IRI by improving oxidative stress, mitochondrial function, and dynamics in aged rats.
Collapse
Affiliation(s)
- Zohreh Zavvari Oskuye
- Drug Applied Research Center, Tabriz University of Medical Sciences, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Iran
| | - Keyvan Mehri
- Student Research Committee, Tabriz University of Medical Sciences, Iran
| | - Behnaz Mokhtari
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Iran
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Iran
| | - Soleyman Bafadam
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Iran
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Iran
| | - Samira Nemati
- Physiology Research Center, Semnan University of Medical Sciences, Iran
| | - Reza Badalzadeh
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Iran
| |
Collapse
|
8
|
Khan SU, Khan SU, Suleman M, Khan MU, Khan MS, Arbi FM, Hussain T, Mohammed Alsuhaibani A, S Refat M. Natural Allies for Heart Health: Nrf2 Activation and Cardiovascular Disease Management. Curr Probl Cardiol 2024; 49:102084. [PMID: 37714318 DOI: 10.1016/j.cpcardiol.2023.102084] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/17/2023]
Abstract
The term "cardiovascular diseases" (CVD) refers to various ailments that affect the heart and blood vessels, including myocardial ischemia, congenital heart defects, heart failure, rheumatic heart disease, hypertension, peripheral artery disease, atherosclerosis, and cardiomyopathies. Despite significant breakthroughs in preventative measures and treatment choices, CVDs significantly contribute to morbidity and mortality, imposing a considerable financial burden. Oxidative stress (OS) is a fundamental contributor to the development and progression of CVDs, resulting from an inherent disparity in generating reactive oxygen species. The disparity above significantly contributes to the aberrant operation of the cardiovascular system. To tackle this issue, therapeutic intervention primarily emphasizes the nuclear erythroid 2-related factor 2 (Nrf2), a transcription factor crucial in regulating endogenous antioxidant defense systems against OS. The Nrf2 exhibits potential as a promising target for effectively managing CVDs. Significantly, an emerging field of study is around the utilization of natural substances to stimulate the activation of Nrf2, hence facilitating the promotion of cardioprotection. This technique introduces a new pathway for treating CVD. The substances above elicit their advantageous effects by mitigating the impact of OS via initiating Nrf2 signaling. The primary objective of our study is to provide significant insights that can contribute to advancing treatment methods, including natural products. These strategies aim to tackle the obstacles associated with CVDs.
Collapse
Affiliation(s)
- Safir Ullah Khan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Shahid Ullah Khan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and South west University, College of Agronomy and Biotechnology, Southwest University, Chongqing, China; Department of Biochemistry, Women Medical and Dental College, Khyber Medical University, Abbottabad, Khyber Pakhtunkhwa, Pakistan.
| | - Muhammad Suleman
- Center for Biotechnology and Microbiology, University of Swat, Swat, Pakistan; Laboratory of Animal Research Center (LARC), Qatar University, Doha, Qatar
| | - Munir Ullah Khan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | | | | | - Talib Hussain
- Women Dental College Abbottabad, Khyber Pakhtunkhwa, Pakistan
| | - Amnah Mohammed Alsuhaibani
- Department of Physical Sport Science, College of Education, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Moamen S Refat
- Department of Chemistry, College of Science, Taif University, Taif, Saudi Arabia
| |
Collapse
|
9
|
Han X, Wang H, Du F, Zeng X, Guo C. Nrf2 for a key member of redox regulation: A novel insight against myocardial ischemia and reperfusion injuries. Biomed Pharmacother 2023; 168:115855. [PMID: 37939614 DOI: 10.1016/j.biopha.2023.115855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/21/2023] [Accepted: 11/05/2023] [Indexed: 11/10/2023] Open
Abstract
Nuclear factor erythroid-2 related factor 2 (Nrf2), a nuclear transcription factor, modulates genes responsible for antioxidant responses against toxic and oxidative stress to maintain redox homeostasis and participates in varieties of cellular processes such as metabolism and inflammation during myocardial ischemia and reperfusion injuries (MIRI). The accumulation of reactive oxygen species (ROS) from damaged mitochondria, xanthine oxidase, NADPH oxidases, and inflammation contributes to depraved myocardial ischemia and reperfusion injuries. Considering that Nrf2 played crucial roles in antagonizing oxidative stress, it is reasonable to delve into the up or down-regulated molecular mechanisms of Nrf2 in the progression of MIRI to provide the possibility of new therapeutic medicine targeting Nrf2 in cardiovascular diseases. This review systematically describes the generation of ROS, the regulatory metabolisms of Nrf2 as well as several natural or synthetic compounds activating Nrf2 during MIRI, which might provide novel insights for the anti-oxidative stress and original ideas targeting Nrf2 for the prevention and treatment in cardiovascular diseases.
Collapse
Affiliation(s)
- Xuejie Han
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 1 Dongjiaomin Lane, Dongcheng District, Beijing 100730, PR China
| | - Hongxia Wang
- Department of Physiology and Pathophysiology, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Fengtai District, Beijing 100069, PR China
| | - Fenghe Du
- Department of Geriatrics, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing 100070, PR China
| | - Xiangjun Zeng
- Department of Physiology and Pathophysiology, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Fengtai District, Beijing 100069, PR China.
| | - Caixia Guo
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 1 Dongjiaomin Lane, Dongcheng District, Beijing 100730, PR China.
| |
Collapse
|
10
|
Wang Y, Zheng Y, Qi B, Liu Y, Cheng X, Feng J, Gao W, Li T. α-Lipoic acid alleviates myocardial injury and induces M2b macrophage polarization after myocardial infarction via HMGB1/NF-kB signaling pathway. Int Immunopharmacol 2023; 121:110435. [PMID: 37320869 DOI: 10.1016/j.intimp.2023.110435] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Myocardial infarction (MI) is a serious cardiovascular disease with a poor prognosis. Macrophages are the predominant immune cells in patients with MI and macrophage regulation during the different phases of MI has important consequences for cardiac recovery. Alpha-lipoic acid (ALA) plays a critical role in MI by modulating the number of cardiomyocytes and macrophages. METHODS MI mice were generated by ligating the left anterior descending coronary artery. Macrophages were exposed to hypoxia to establish a hypoxia model and M1 polarization was induced by LPS and IFN-γ. Different groups of macrophages and MI mice were treated with ALA. The cardiomyocytes were treated with various macrophage supernatants and the cardiac function, cytokine levels, and pathology were also analyzed. Factors related to apoptosis, autophagy, reactive oxygen species (ROS), and the mitochondrial membrane potential (MMP) were assessed. Finally, the HMGB1/NF-κB pathway was identified. RESULTS ALA promoted M2b polarization in normal cells and suppressed inflammatory cytokines during hypoxia. ALA inhibited ROS and MMP production in vitro. Supernatants containing ALA inhibited apoptosis and autophagy in hypoxic cardiomyocytes. Moreover, ALA suppressed the HMGB1/NF-κB pathway in macrophages, which may be a potential mechanism for attenuating MI. CONCLUSION ALA alleviates MI and induces M2b polarization via the HMGB1/NF-κB pathway, impeding inflammation, oxidation, apoptosis, and autophagy, and might be a potential strategy for MI treatment.
Collapse
Affiliation(s)
- Yuchao Wang
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China.
| | - Yue Zheng
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Bingcai Qi
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yanwu Liu
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Xuan Cheng
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Jianyu Feng
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Wenqing Gao
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China.
| | - Tong Li
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China.
| |
Collapse
|
11
|
Sabatino L. Nrf2-Mediated Antioxidant Defense and Thyroid Hormone Signaling: A Focus on Cardioprotective Effects. Antioxidants (Basel) 2023; 12:1177. [PMID: 37371907 DOI: 10.3390/antiox12061177] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/24/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
Thyroid hormones (TH) perform a plethora of actions in numerous tissues and induce an overall increase in metabolism, with an augmentation in energy demand and oxygen expenditure. Oxidants are required for normal thyroid-cell proliferation, as well as for the synthesis of the main hormones secreted by the thyroid gland, triiodothyronine (T3) and thyroxine (T4). However, an uncontrolled excess of oxidants can cause oxidative stress, a major trigger in the pathogenesis of a broad spectrum of diseases, including inflammation and cancer. In particular, oxidative stress is implicated in both hypo- and hyper-thyroid diseases. Furthermore, it is important for the TH system to rely on efficient antioxidant defense, to maintain balance, despite sustained tissue exposure to oxidants. One of the main endogenous antioxidant responses is the pathway centered on the nuclear factor erythroid 2-related factor (Nrf2). The aim of the present review is to explore the multiple links between Nrf2-related pathways and various TH-associated conditions. The main aspect of TH signaling is described and the role of Nrf2 in oxidant-antioxidant homeostasis in the TH system is evaluated. Next, the antioxidant function of Nrf2 associated with oxidative stress induced by TH pathological excess is discussed and, subsequently, particular attention is given to the cardioprotective role of TH, which also acts through the mediation of Nrf2. In conclusion, the interaction between Nrf2 and most common natural antioxidant agents in altered states of TH is briefly evaluated.
Collapse
Affiliation(s)
- Laura Sabatino
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
12
|
Zheng Z, Xian Y, Jin Z, Yao F, Liu Y, Deng Y, Wang B, Chen D, Yang J, Ren L, Lin R. Rhaponticum carthamoides improved energy metabolism and oxidative stress through the SIRT6/Nrf2 pathway to ameliorate myocardial injury. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154197. [PMID: 35917770 DOI: 10.1016/j.phymed.2022.154197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Rhaponticum carthamoides (Willd.) Iljin (Rha) is a member of the family Compositae that is widely used in folk medicine as a dietary supplement to treat cardiovascular diseases (CVDs), such as senile cardiac insufficiency, and to restore myocardial function after surgery. Sirtuin 6 (SIRT6), an NAD+-dependent class III histone deacetylase, plays a considerable role in the administration of CVDs. However, the specific effects and mechanism of Rha on myocardial injury remain unknown. PURPOSE This study aimed to explore the therapeutic potential of Rha against myocardial injury as well as its underlying mechanisms in vivo and in vitro. METHODS A myocardial ischaemia model was established in male SD rats by subcutaneously injecting ISO. The rats were gavaged with Rha (40, 80, 160 mg/kg) or Rho (6 ml/kg) for 14 successive days and then injected subcutaneously with ISO or saline solution on the 13th and 14th days. The positive effects of Rha against myocardial injury in rats were evaluated by ECG assessment, BP measurements, H&E staining, and myocardial enzyme detection. Biochemical indicators of energy metabolism and oxidative stress, such as NAD+/NADH, ATP, and MDA, were analysed by assay kits to assess the effects of Rha. The protein and mRNA expression levels of SIRT6 and Nrf2 in the myocardium were determined by western blotting and real-time PCR. RESULTS Our results showed that Rha ameliorated myocardial ischaemia and inhibited energy metabolism disorders (NAD+/NADH ratio, ATP, and LD) and oxidative stress (SOD, ROS, etc.) in rat myocardial tissue and H9c2 cells. In addition, Rha upregulated SIRT6 and Nrf2 expression in myocardial injury. Mechanistic studies then found that SIRT6 knockdown reduced the expression of Nrf2 as well as the effects of Rha on the levels of ATP, LD, and ROS, whereas activation of Nrf2 improved the effects of Rha in cells. In summary, Rha might exert its cardioprotective effects via the SIRT6-mediated Nrf2 signaling pathway. CONCLUSION The results suggest that Rha regulates energy metabolism and oxidative stress through the SIRT6/Nrf2 signaling pathway to play a protective role in myocardial injury.
Collapse
Affiliation(s)
- Zihan Zheng
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China
| | - Yushan Xian
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China; Department of Pharmacy, Xi'an NO.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, PR China
| | - Zhen Jin
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China
| | - Feng Yao
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China
| | - YiZhen Liu
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China
| | - Ying Deng
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China
| | - Bo Wang
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China
| | - Danli Chen
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China
| | - Jianjun Yang
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China
| | - Lingxuan Ren
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China
| | - Rong Lin
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| |
Collapse
|
13
|
Zhu C, Liu G, Gu X, Yin J, Xia A, Han M, Zhang T, Jiang Q. Effect of quercetin on muscle growth and antioxidant status of the dark sleeper Odontobutis potamophila. Front Genet 2022; 13:938526. [PMID: 35957695 PMCID: PMC9358148 DOI: 10.3389/fgene.2022.938526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Quercetin is a flavanol beneficial in reducing fat, promoting muscle growth, and Anti-oxidation. To study its effects in freshwater fish, the full-length cDNA of the follistatin (FST) and myostatin (MSTN) genes of the dark sleeper Odontobutis potamophila were cloned for the first time. Juvenile individual O. potamophila was exposed to quercetin at one of four concentrations (0, 2.5, 5, and 10 mg/L) for 21 days. The expression level of MSTN which inhibits muscle growth in the quercetin solution was lower than in the unexposed control group. The genes that promote muscle growth are in TGF-β superfamily like FST, TGF-β1 (transforming growth factor-beta 1), and Myogenic regulatory factors (MRFs) like Myf5 (myogenic factor 5), MyoD (myogenic differentiation), MyoG (myogenin), were higher than in the control group. Apolipoprotein and growth hormone receptor transcription levels in the quercetin-treated fish were significantly lower than in the control group. The concentrations of triglyceride, low-density lipoprotein cholesterol, and high-density lipoprotein cholesterol in the muscle tissue decreased, and the lipid-lowering function of quercetin was also demonstrated at the biochemical level. In this study, we analyzed the mRNA levels of AKT, Keap1 (kelch-like ECH-associated protein 1), Nrf2 (NF-E2-related factor 2) oxidation-related genes in the Nrf2/ARE antioxidant pathway, and Malondialdehyde (MDA), catalase (CAT) activity and glutathione (GSH) content in the hepatopancreas of O. potamophila after quercetin treatment, the mRNA expression of AKT, Nrf2 and CAT activity and GSH content are higher than in the control group. Quercetin enhances antioxidant properties and positively affects muscle growth. The results showed that quercetin has no significant effects on the growth performance of O. potamophila, but is effective in increasing muscle growth rate and lowering muscle fat content.
Collapse
Affiliation(s)
- Chenxi Zhu
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, China
| | - Guoxing Liu
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Xiankun Gu
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, China
- The Lowtemperature Germplasm Bank of Important Economic Fish of Jiangsu Provincial Science and TechnologyResources (Agricultural Germplasm Resources) Coordination Service Platform, Freshwater Fisheries Research Institute of JiangsuProvince, NanjingChina
| | - Jiawen Yin
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, China
- The Lowtemperature Germplasm Bank of Important Economic Fish of Jiangsu Provincial Science and TechnologyResources (Agricultural Germplasm Resources) Coordination Service Platform, Freshwater Fisheries Research Institute of JiangsuProvince, NanjingChina
| | - Aijun Xia
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, China
- The Lowtemperature Germplasm Bank of Important Economic Fish of Jiangsu Provincial Science and TechnologyResources (Agricultural Germplasm Resources) Coordination Service Platform, Freshwater Fisheries Research Institute of JiangsuProvince, NanjingChina
| | - Mingming Han
- Biology Program, School of Distance Education, Universiti Sains Malaysia, Minden, Malaysia
| | - Tongqing Zhang
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, China
- The Lowtemperature Germplasm Bank of Important Economic Fish of Jiangsu Provincial Science and TechnologyResources (Agricultural Germplasm Resources) Coordination Service Platform, Freshwater Fisheries Research Institute of JiangsuProvince, NanjingChina
| | - Qichen Jiang
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, China
- The Lowtemperature Germplasm Bank of Important Economic Fish of Jiangsu Provincial Science and TechnologyResources (Agricultural Germplasm Resources) Coordination Service Platform, Freshwater Fisheries Research Institute of JiangsuProvince, NanjingChina
- *Correspondence: Qichen Jiang,
| |
Collapse
|
14
|
ALIOMRANI M, MESRIPOUR A, MEHRJARDI AS. Creatine and Alpha-Lipoic Acid Antidepressant-Like Effect Following Cyclosporine A Administration. Turk J Pharm Sci 2022; 19:196-201. [DOI: 10.4274/tjps.galenos.2021.27217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
15
|
Zhong M, Sun A, Xiao T, Yao G, Sang L, Zheng X, Zhang J, Jin X, Xu L, Yang W, Wang P, Hu K, Zhang D, Ge J. A Randomized, Single-Blind, Group Sequential, Active-Controlled Study to Evaluate the Clinical Efficacy and Safety of α-Lipoic Acid for Critically Ill Patients With Coronavirus Disease 2019 (COVID-19). Front Med (Lausanne) 2022; 8:566609. [PMID: 35186959 PMCID: PMC8854372 DOI: 10.3389/fmed.2021.566609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/15/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECT To evaluate the clinical efficacy and safety of α-Lipoic acid (ALA) for critically ill patients with coronavirus disease 2019 (COVID-19). METHODS A randomized, single-blind, group sequential, active-controlled trial was performed at JinYinTan Hospital, Wuhan, China. Between February 2020 and March 2020, 17 patients with critically ill COVID-19 were enrolled in our study. Eligible patients were randomly assigned in a 1:1 ratio to receive either ALA (1200 mg/d, intravenous infusion) once daily plus standard care or standard care plus equal volume saline infusion (placebo) for 7 days. All patients were monitored within the 7 days therapy and followed up to day 30 after therapy. The primary outcome of this study was the Sequential Organ Failure Estimate (SOFA) score, and the secondary outcome was the all-cause mortality within 30 days. RESULT Nine patients were randomized to placebo group and 8 patients were randomized to ALA group. SOFA score was similar at baseline, increased from 4.3 to 6.0 in the placebo group and increased from 3.8 to 4.0 in the ALA group (P = 0.36) after 7 days. The 30-day all-cause mortality tended to be lower in the ALA group (3/8, 37.5%) compared to that in the placebo group (7/9, 77.8%, P = 0.09). CONCLUSION In our study, ALA use is associated with lower SOFA score increase and lower 30-day all-cause mortality as compared with the placebo group. Although the mortality rate was two-folds higher in placebo group than in ALA group, only borderline statistical difference was evidenced due to the limited patient number. Future studies with larger patient cohort are warranted to validate the role of ALA in critically ill patients with COVID-19. CLINICAL TRIAL REGISTRATION http://www.chictr.org.cn/showproj.aspx?proj=49534.
Collapse
Affiliation(s)
- Ming Zhong
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Wuhan JinYinTan Hospital, Wuhan, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ting Xiao
- Intensive Care Unit of the Six Floor of South Building, Wuhan JinYinTan Hospital, Wuhan, China
| | - Ge Yao
- Intensive Care Unit of the Six Floor of South Building, Wuhan JinYinTan Hospital, Wuhan, China
| | - Ling Sang
- Department of Critical Care Medicine, GuangZhou Institute of Respiratory Health, The First Affiliated Hospital of GuangZhou Medical University, Guangzhou, China
| | - Xia Zheng
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Jinyan Zhang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Xuejuan Jin
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Lei Xu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Wenlong Yang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Peng Wang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | | | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
16
|
MicroRNA-489 Promotes the Apoptosis of Cardiac Muscle Cells in Myocardial Ischemia-Reperfusion Based on Smart Healthcare. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:2538769. [PMID: 35035817 PMCID: PMC8759872 DOI: 10.1155/2022/2538769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/01/2021] [Accepted: 12/09/2021] [Indexed: 01/10/2023]
Abstract
With the development of information technology, the concept of smart healthcare has gradually come to the fore. Smart healthcare uses a new generation of information technologies, such as the Internet of Things (loT), big data, cloud computing, and artificial intelligence, to transform the traditional medical system in an all-around way, making healthcare more efficient, more convenient, and more personalized. miRNAs can regulate the proliferation, differentiation, and apoptosis of human cells. Relevant studies have also shown that miRNAs may play a key role in the occurrence and development of myocardial ischemia-reperfusion injury (MIRI). This study aims to explore the effects of miR-489 in MIRI. In this study, miR-489 expression in a myocardial ischemia-reperfusion animal model and H9C2 cells induced by H/R was detected by qRT-PCR. The release of lactate dehydrogenase (LDH) and the activity of creatine kinase (CK) was detected after miR-489 knockdown in H9C2 cells induced by H/R. The apoptosis of H9C2 cells and animal models were determined by ELISA. The relationship between miR-489 and SPIN1 was verified by a double fluorescence reporter enzyme assay. The expression of the PI3K/AKT pathway-related proteins was detected by Western blot. Experimental results showed that miR-489 was highly expressed in cardiac muscle cells of the animal model and in H9C2 cells induced by H/R of the myocardial infarction group, which was positively associated with the apoptosis of cardiac muscle cells with ischemia-reperfusion. miR-489 knockdown can reduce the apoptosis of cardiac muscle cells caused by ischemia-reperfusion. In downstream targeting studies, it was found that miR-489 promotes the apoptosis of cardiac muscle cells after ischemia-reperfusion by targeting the inhibition of the SPIN1-mediated PI3K/AKT pathway. In conclusion, high expression of miR-489 is associated with increased apoptosis of cardiac muscle cells after ischemia-reperfusion, which can promote the apoptosis after ischemia-reperfusion by targeting the inhibition of the SPIN1-mediated PI3K/AKT pathway. Therefore, miR-489 can be one of the potential therapeutic targets for reducing the apoptosis of cardiac muscle cells after ischemia-reperfusion.
Collapse
|
17
|
Zhang Y, Liu S, Ma JL, Chen C, Huang P, Ji JH, Wu D, Ren LQ. Apocynum venetum leaf extract alleviated doxorubicin-induced cardiotoxicity through the AKT/Bcl-2 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 94:153815. [PMID: 34781232 DOI: 10.1016/j.phymed.2021.153815] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 10/01/2021] [Accepted: 10/17/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Doxorubicin (DOX) is a broad-spectrum anti-tumor drug that has been associated with cardiotoxicity. Plant extracts have been shown to confer protection against DOX-induced cardiotoxicity. Apocynum venetum L. belongs to the Apocynaceae family. Flavonoid extracted from Apocynum venetum L. possess various biological effects, such as lowering blood pressure levels, sedation, diuresis, anti-aging, and improving immunity. PURPOSE This study investigated the mechanism by which dry leaf extract of Apocynum venetum L. (AVLE) alleviates DOX-induced cardiomyocyte apoptosis. METHODS HPLC-MS/MS and HPLC methods were used to analyze the components of AVLE. The effects of DOX and AVLE on apoptosis of H9c2 and HMC cells were assessed using the MTT assay. Calcein AM/PI, TUNEL, and flow cytometry were carried out to determine the effects of AVLE on DOX-induced apoptosis. The effect of AVLE on DOX-induced oxidative stress in cardiomyocytes was investigated using ELISA test. Mito-Tracker Red CMXRos, JC-1, and RT-qPCR assays were performed to evaluate the impact of AVLE on DOX-induced cardiomyocyte mitochondrial activity and membrane permeability. Western blot assay was carried out to determine the activation of multiple signaling molecules, including phosphorylated-protein kinase B (p-AKT), Cytochrome c, Bcl-2 family, and caspase family in the apoptosis pathway. The AKT inhibitor was used to block AKT/Bcl-2 signaling pathway to investigate the role of AKT in the protection conferred by AVLE against DOX-induced cardiotoxicity. RESULTS A total of 8 compounds, including rutin, hyperoside, isoquercetin, unidentified compounds, myricetin, quercetin, quercetin-3-O-glucuronide and kaempferol, were detected in AVLE. Of note, DOX suppressed lactate dehydrogenase (LDH) levels, aggravated oxidative stress, and promoted cardiomyocyte apoptosis. It also upregulated the mRNA expression levels of voltage-dependent anion channel 1 (VDAC1), adenosine nucleotide transporter 1 (ANT1), and cyclophilin D (CYPD), while suppressing mitochondrial activity and mitochondrial membrane permeability. Treatment with DOX altered the expression levels of apoptosis-associated proteins, Bcl-2 and Bax. However, AVLE treatment alleviated DOX-induced effects on cardiomyocytes. In addition, application of AKT inhibitors promoted DOX-induced apoptosis and reversed the inhibitory effects of AVLE on DOX-induced apoptosis. CONCLUSIONS AVLE confer cardio protection by suppressing oxidative stress and apoptosis of cardiomyocytes via AKT/Bcl-2 signaling pathway.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Shan Liu
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Jiu-Long Ma
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Chen Chen
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Peng Huang
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Jia-Hua Ji
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Di Wu
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Li-Qun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China.
| |
Collapse
|
18
|
Effects of Lipoic Acid on Ischemia-Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5093216. [PMID: 34650663 PMCID: PMC8510805 DOI: 10.1155/2021/5093216] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/07/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022]
Abstract
Ischemia-reperfusion (I/R) injury often occurred in some pathologies and surgeries. I/R injury not only harmed to physiological functions of corresponding organ and tissue but also induced multiple tissue or organ dysfunctions (even these in distant locations). Although the reperfusion of blood attenuated I/R injury to a certain degree, the risk of secondary damages was difficult to be controlled and it even caused failures of these tissues and organs. Lipoic acid (LA), as an endogenous active substance and a functional agent in food, owns better safety and effects in our body (e.g., enhancing antioxidant activity, improving cognition and dementia, controlling weight, and preventing multiple sclerosis, diabetes complication, and cancer). The literature searching was conducted in PubMed, Embase, Cochrane Library, Web of Science, and SCOPUS from inception to 20 May 2021. It had showed that endogenous LA was exhausted in the process of I/R, which further aggravated I/R injury. Thus, supplements with LA timely (especially pretreatments) may be the prospective way to prevent I/R injury. Recently, studies had demonstrated that LA supplements significantly attenuated I/R injuries of many organs, though clinic investigations were short at present. Hence, it was urgent to summarize these progresses about the effects of LA on different I/R organs as well as the potential mechanisms, which would enlighten further investigations and prepare for clinic applications in the future.
Collapse
|
19
|
Mauerhofer C, Grumet L, Schemmer P, Leber B, Stiegler P. Combating Ischemia-Reperfusion Injury with Micronutrients and Natural Compounds during Solid Organ Transplantation: Data of Clinical Trials and Lessons of Preclinical Findings. Int J Mol Sci 2021; 22:10675. [PMID: 34639016 PMCID: PMC8508760 DOI: 10.3390/ijms221910675] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Although extended donor criteria grafts bear a higher risk of complications such as graft dysfunction, the exceeding demand requires to extent the pool of potential donors. The risk of complications is highly associated with ischemia-reperfusion injury, a condition characterized by high loads of oxidative stress exceeding antioxidative defense mechanisms. The antioxidative properties, along with other beneficial effects like anti-inflammatory, antiapoptotic or antiarrhythmic effects of several micronutrients and natural compounds, have recently emerged increasing research interest resulting in various preclinical and clinical studies. Preclinical studies reported about ameliorated oxidative stress and inflammatory status, resulting in improved graft survival. Although the majority of clinical studies confirmed these results, reporting about improved recovery and superior organ function, others failed to do so. Yet, only a limited number of micronutrients and natural compounds have been investigated in a (large) clinical trial. Despite some ambiguous clinical results and modest clinical data availability, the vast majority of convincing animal and in vitro data, along with low cost and easy availability, encourage the conductance of future clinical trials. These should implement insights gained from animal data.
Collapse
Affiliation(s)
- Christina Mauerhofer
- Department of Science and Product Development, pro medico HandelsGmbH, Liebenauer Tangente 6, 8041 Graz, Austria; (C.M.); (L.G.)
| | - Lukas Grumet
- Department of Science and Product Development, pro medico HandelsGmbH, Liebenauer Tangente 6, 8041 Graz, Austria; (C.M.); (L.G.)
| | - Peter Schemmer
- Division of Transplant Surgery, Department of Surgery, Medical University, 8036 Graz, Austria; (P.S.); (B.L.)
| | - Bettina Leber
- Division of Transplant Surgery, Department of Surgery, Medical University, 8036 Graz, Austria; (P.S.); (B.L.)
| | - Philipp Stiegler
- Division of Transplant Surgery, Department of Surgery, Medical University, 8036 Graz, Austria; (P.S.); (B.L.)
| |
Collapse
|
20
|
Theodosis-Nobelos P, Papagiouvannis G, Tziona P, Rekka EA. Lipoic acid. Kinetics and pluripotent biological properties and derivatives. Mol Biol Rep 2021; 48:6539-6550. [PMID: 34420148 DOI: 10.1007/s11033-021-06643-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/10/2021] [Indexed: 12/14/2022]
Abstract
Lipoic acid (LA) is globally known and its supplements are widely used. Despite its importance for the organism it is not considered a vitamin any more. The multiple metabolic forms and the differences in kinetics (absorption, distribution and excretion), as well as the actions of its enantiomers are analysed in the present article together with its biosynthetic path. The proteins involved in the transfer, biotransformation and activity of LA are mentioned. Furthermore, the safety and the toxicological profile of the compound are commented, together with its stability issues. Mechanisms of lipoic acid intervention in the human body are analysed considering the antioxidant and non-antioxidant characteristics of the compound. The chelating properties, the regenerative ability of other antioxidants, the co-enzyme activity and the signal transduction by the implication in various pathways will be discussed in order to be elucidated the pleiotropic effects of LA. Finally, lipoic acid integrating analogues are mentioned under the scope of the multiple pharmacological actions they acquire towards degenerative conditions.
Collapse
Affiliation(s)
| | - Georgios Papagiouvannis
- Department of Pharmacy, School of Health Sciences, Frederick University, 1036, Nicosia, Cyprus
| | - Paraskevi Tziona
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotelian University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Eleni A Rekka
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotelian University of Thessaloniki, 54124, Thessaloniki, Greece
| |
Collapse
|
21
|
Li K, Zhou P, Li S, Zheng S, Wang D. MicroRNA-29b reduces myocardial ischemia-reperfusion injury in rats via down-regulating PTEN and activating the Akt/eNOS signaling pathway. J Thromb Thrombolysis 2021; 53:123-135. [PMID: 34370169 DOI: 10.1007/s11239-021-02535-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/16/2021] [Indexed: 01/20/2023]
Abstract
Reperfusion may cause injuries to the myocardium in ischemia situation, which is called ischemia/reperfusion (I/R) injury. The study aimed to explore the roles of microRNA-29b (miR-29b) in myocardial I/R injury. Myocardial I/R injury rat model was established. Differentially expressed miRNAs between the model rats and the sham-operated rats were analyzed. miR-29b expression in myocardial tissues was measured. Gain-of-function of miR-29b was performed, and then the morphological changes, infarct size, myocardial function, oxidative stress, and the cell apoptosis in myocardial tissues were detected. The target relation between miR-29b and PTEN was detected through bio-information prediction and dual luciferase reporter gene assay. Activation of Akt/eNOS signaling was detected. H9C2 cells were subjected to hypoxia/reoxygenation treatment to perform in vitro experiments. I/R rats presented severe inflammatory infiltration, increased infarct size and cell apoptosis, increased oxidative stress and decreased myocardial function. miR-29b was downregulated in I/R rats, and up-regulation of miR-29b reversed the above changes. miR-29b directly bound to PTEN, and overexpression of miR-29b reduced PTEN expression level and increased the protein levels of p-Akt/Akt and p-eNOS/eNOS. In vivo results were confirmed in in vitro experiments. This study provided evidence that miR-29b could alleviate the myocardial I/R injury in vivo and in vitro by inhibiting PTEN expression and activating the Akt/eNOS signaling pathway.
Collapse
Affiliation(s)
- Kunsheng Li
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, People's Republic of China
| | - Pengyu Zhou
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 515000, Guangdong Province, People's Republic of China
| | - Shiliang Li
- Department of Cardiac Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 515000, Guangdong Province, People's Republic of China.
| | - Dongjin Wang
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, People's Republic of China.
| |
Collapse
|
22
|
Jin AP, Zhang QR, Yang CL, Ye S, Cheng HJ, Zheng YY. Up-regulation of CTRP12 ameliorates hypoxia/re-oxygenation-induced cardiomyocyte injury by inhibiting apoptosis, oxidative stress, and inflammation via the enhancement of Nrf2 signaling. Hum Exp Toxicol 2021; 40:2087-2098. [PMID: 34085554 DOI: 10.1177/09603271211021880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
C1q/TNF-related protein 12 (CTRP12) has been reported to play a key role in coronary artery disease. However, whether CTRP12 plays a role in the regulation of myocardial ischemia-reperfusion injury is not fully understood. The goals of this work were to assess the possible relationship between CTRP12 and myocardial ischemia-reperfusion injury. Here, we exposed cardiomyocytes to hypoxia/re-oxygenation (H/R) to establish an in vitro cardiomyocyte injury model of myocardial ischemia-reperfusion injury. Our results showed that H/R treatment resulted in a decrease in CTRP12 expression in cardiomyocytes. The up-regulation of CTRP12 ameliorated H/R-induced cardiomyocyte injury via the down-regulation of apoptosis, oxidative stress, and inflammation. In contrast, the knockdown of CTRP12 enhanced cardiomyocyte sensitivity to H/R-induced cardiomyocyte injury. Further investigation showed that CTRP12 enhanced the levels of nuclear Nrf2 and increased the expression of Nrf2 target genes in cardiomyocytes exposed to H/R. However, the inhibition of Nrf2 markedly diminished CTRP12-overexpression-mediated cardioprotective effects against H/R injury. Overall, these data indicate that CTRP12 protects against H/R-induced cardiomyocyte injury by inhibiting apoptosis, oxidative stress, and inflammation via the enhancement of Nrf2 signaling. This work suggests a potential role of CTRP12 in myocardial ischemia-reperfusion injury and proposes it as an attractive target for cardioprotection.
Collapse
Affiliation(s)
- Ai-Ping Jin
- Geriatric Cardiovascular Department, 117799The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, Shaanxi Province, China
| | - Qian-Rong Zhang
- Geriatric Cardiovascular Department, 117799The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, Shaanxi Province, China
| | - Cui-Ling Yang
- Geriatric Cardiovascular Department, 117799The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, Shaanxi Province, China
| | - Sha Ye
- Geriatric Cardiovascular Department, 117799The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, Shaanxi Province, China
| | - Hai-Juan Cheng
- Geriatric Cardiovascular Department, 117799The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, Shaanxi Province, China
| | - Yuan-Yuan Zheng
- Geriatric Cardiovascular Department, 117799The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, Shaanxi Province, China
| |
Collapse
|
23
|
Salama A, Mansour D, Hegazy R. The cardio and renoprotective role of ginseng against epinephrine-induced myocardial infarction in rats: Involvement of angiotensin II type 1 receptor/protein kinase C. Toxicol Rep 2021; 8:908-919. [PMID: 33996504 PMCID: PMC8099916 DOI: 10.1016/j.toxrep.2021.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/26/2021] [Accepted: 04/16/2021] [Indexed: 11/30/2022] Open
Abstract
Epinephrine induced MI with renal complication through Nrf2/NF-κB imbalance and PKC/AT1R. Ginseng abolished ECG changes induced by epinephrine and stimulated Nrf2. Ginseng reduced upregulation of PKC, NF-κB, and AT1R induced by epinephrine. Ginseng inhibited iNOS and corrected renal disorder in epinephrine model of MI.
The expression of angiotensin II type 1 receptor (AT1 receptor)/protein kinase C (PKC) in heart tissues has a vital role in myocardial infarction (MI). The current work aimed to clarify the renal complication enhanced by MI following epinephrine injection via AT1 receptor/ PKC expression; in addition, the impact of ginseng extract on epinephrine-induced MI and its renal complication was assessed. Adult male albino Wistar rats were pretreated orally with ginseng extract (200 & 400 mg/kg/day) for 14 days, then two successive doses of epinephrine injection (100 mg/kg, i.p.). Epinephrine evoked electrocardiographic (ECG) and renal changes accompanied with a significant increase in heart and kidney contents of malodialdehyde (MDA), nitric oxide (NO), protein kinase C (PKC), heart contents of nuclear factor-kabba B (NF-κB) and angiotensin 1receptor (AT1R), as well as a decrease in heart and kidney reduced glutathione (GSH) and nuclear factor-erythroid-related factor 2 (Nrf2) contents. In histopathological investigations epinephrine exhibited deleterious heart changes in the form of acute MI with the presence of necrosis of cardiomyocytes with iNOS expression and produced glomerulus and renal tubules degeneration. Pretreatment of rats with ginseng extract in both doses significantly corrected epinephrine-induced heart and renal changes. The current work revealed that epinephrine-induced MI associated with aggravated renal complication and ginseng extract has cardio and reno protective role against this as it reduces infarct size, preserves cardiac and renal tissues and functions through activating Nrf2 and down-regulating NF-κB, PKC, AT1R and iNOS.
Collapse
Affiliation(s)
- Abeer Salama
- Pharmacology Department, Medical Division, National Research Centre, 33 El Bohouth St. (Former El-Tahrir St.), 12622 Dokki, Cairo, Egypt
| | - Dina Mansour
- Pharmacology Department, Medical Division, National Research Centre, 33 El Bohouth St. (Former El-Tahrir St.), 12622 Dokki, Cairo, Egypt
| | - Rehab Hegazy
- Pharmacology Department, Medical Division, National Research Centre, 33 El Bohouth St. (Former El-Tahrir St.), 12622 Dokki, Cairo, Egypt
| |
Collapse
|
24
|
Syed AM, Ram C, Murty US, Sahu BD. A review on herbal Nrf2 activators with preclinical evidence in cardiovascular diseases. Phytother Res 2021; 35:5068-5102. [PMID: 33894007 DOI: 10.1002/ptr.7137] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/24/2021] [Accepted: 04/10/2021] [Indexed: 12/31/2022]
Abstract
Cardiovascular diseases (CVDs) are an ever-growing problem and are the most common cause of death worldwide. The uncontrolled production of reactive oxygen species (ROS) and the activation of ROS associated with various cell signaling pathways with oxidative cellular damage are the most common pathological conditions connected with CVDs including endothelial dysfunction, hypercontractility of vascular smooth muscle, cardiac hypertrophy and heart failure. The nuclear factor E2-related factor 2 (Nrf2) is a basic leucine zipper redox transcription factor, together with its negative regulator, kelch-like ECH-associated protein 1 (Keap1), which serves as a key regulator of cellular defense mechanisms to combat oxidative stress and associated diseases. Multiple lines of evidence described here support the cardiac protective property of Nrf2 in various experimental models of cardiac related disease conditions. In this review, we emphasized the molecular mechanisms of Nrf2 and described the detailed outline of current findings on the therapeutic possibilities of the Nrf2 activators specifically from herbal origin in various CVDs. Based on evidence from various preclinical experimental models, we have highlighted the activation of Nrf2 pathway as a budding therapeutic option for the prevention and treatment of CVDs, which needs further investigation and validation in the clinical settings.
Collapse
Affiliation(s)
- Abu Mohammad Syed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Guwahati, Assam, India
| | - Chetan Ram
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Guwahati, Assam, India
| | - Upadhyayula Suryanarayana Murty
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Guwahati, Assam, India
| | - Bidya Dhar Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Guwahati, Assam, India
| |
Collapse
|
25
|
Simultaneous Quantitative Analysis of Ginsenosides Isolated from the Fruit of Panax ginseng C.A. Meyer and Regulation of HO-1 Expression through EGFR Signaling Has Anti-Inflammatory and Osteogenic Induction Effects in HPDL Cells. Molecules 2021; 26:molecules26072092. [PMID: 33917440 PMCID: PMC8038702 DOI: 10.3390/molecules26072092] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/03/2021] [Accepted: 04/03/2021] [Indexed: 01/09/2023] Open
Abstract
Periodontitis is a set of chronic inflammatory diseases caused by the accumulation of Gram-negative bacteria on teeth, resulting in gingivitis, pocket formation, alveolar bone loss, tissue destruction, and tooth loss. In this study, the contents of ginsenosides isolated from Panax ginseng fruit extract were quantitatively analyzed, and the anti-inflammatory effects were evaluated in human periodontal ligament cells. The major ginsenosides, Re, Ra8, and Rf, present in ginseng fruit were simultaneously analyzed by a validated method using high-performance liquid chromatography with a diode-array detector; Re, Ra8, and Rf content per 1 g of P. ginseng fruit extract was 1.01 ± 0.03, 0.33 ± 0.01, and 0.55 ± 0.04 mg, respectively. Ginsenosides-Re, -Ra8, and -Rf inhibited the production of pro-inflammatory factors and the expression of important cytokines in periodontitis by inducing the expression of heme oxygenase 1 (HO-1), promoting osteoblast differentiation of periodontal ligament cells, suppressing alveolar bone loss, and promoting the expression of osteoblast-specific genes, such as alp, opn, and runx2. An inhibitory effect of these ginsenosides on periodontitis and alveolar bone loss was observed via the regulation of HO-1 and subsequent epidermal growth factor receptor (EGFR) signaling. Silencing EGFR with EGFR siRNA confirmed that the effect of ginsenosides on HO-1 is mediated by EGFR. In conclusion, this study evaluated the contents of ginsenosides-Re, -Ra8, and -Rf isolated from P. ginseng fruit extract. Therefore, these results provide important basic data for future P. ginseng fruit component studies and suggest that ginsenosides Re, Ra8, and Rf have potential as future treatment options for periodontitis.
Collapse
|
26
|
SUPPORT-1 (Subjects Undergoing PCI and Perioperative Reperfusion Treatment): A Prospective, Randomized Trial of CMX-2043 in Patients Undergoing Elective Percutaneous Coronary Intervention. J Cardiovasc Pharmacol 2021; 76:189-196. [PMID: 32769849 DOI: 10.1097/fjc.0000000000000830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The natural molecule α-lipoic acid has been shown to be partially cytoprotective through antioxidant and antiapoptotic mechanisms. To obtain an initial assessment of the safety and potential efficacy of a synthetic derivative, CMX-2043, in preventing ischemic complications of percutaneous coronary intervention (PCI) we conducted the Subjects Undergoing PCI and Perioperative Reperfusion Treatment (SUPPORT-1) trial, the first patient experience with this agent. METHODS AND RESULTS SUPPORT-1 was a phase 2a, 6-center, international, placebo-controlled, randomized, double-blind trial. A total of 142 patients were randomized to receive a single intravenous bolus dose of drug or placebo administered 15-60 minutes before PCI. Cardiac biomarker assessments included serial measurements of creatine kinase myocardial band (CK-MB) at 6, 12, 18, and 24 hours after PCI and a single measurement of troponin T (TnT) at 24 hours. Peak concentrations of CK-MB and TnT were significantly reduced in the 2.4 mg/kg group compared with placebo (P = 0.05 and 0.03, respectively). No subject administered 2.4 mg/kg of CMX-2043 had an increase of CK-MB to ≥3X upper limit of normal versus 16% for placebo (P = 0.02); 16% of the 2.4-mg/kg dose group developed an elevation of TnT to ≥3X upper limit of normal versus 39% in the placebo group (P = 0.05). No drug-related serious adverse events were observed in any group. CONCLUSION These data suggest that CMX-2043 may reduce PCI periprocedural myonecrosis and support further clinical evaluation of this novel agent for its potential cytoprotective effects.
Collapse
|
27
|
Hedrich WD, Wang H. Friend or Foe: Xenobiotic Activation of Nrf2 in Disease Control and Cardioprotection. Pharm Res 2021; 38:213-241. [PMID: 33619640 DOI: 10.1007/s11095-021-02997-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/08/2020] [Indexed: 12/30/2022]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that governs a highly conserved pathway central to the protection of cells against various oxidative stresses. However, the biological impact of xenobiotic intervention of Nrf2 in physiological and pathophysiological conditions remains debatable. Activation of Nrf2 in cancer cells has been shown to elevate drug resistance and increase cell survival and proliferation, while inhibition of Nrf2 sensitizes cancer cells to drug treatment. On the other hand, activation of Nrf2 in normal healthy cells has been explored as a rather successful strategy for cancer chemoprevention. Selective activation of Nrf2 in off-target cells has recently been investigated as an approach for protecting off-target tissues from untoward drug toxicity. Specifically, induction of antioxidant response element genes via Nrf2 activation in cardiac cells is being explored as a means to limit the well-documented cardiotoxicity accompanied by cancer treatment with commonly prescribed anthracycline drugs. In addition to cancers, Nrf2 has been implicated in many other diseases including Alzheimer's and Parkinson's Diseases, diabetes, and cardiovascular disease. In this review, we discuss the roles of Nrf2 and its downstream target genes in the treatment of various diseases, and its recently explored potential for increasing the benefit: risk ratio of commonly utilized cancer treatments.
Collapse
Affiliation(s)
- William D Hedrich
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland, 21201, USA.,Bristol-Myers Squibb Company, Pharmaceutical Candidate Optimization, Metabolism and Pharmacokinetics, Rt. 206 and Province Line Road, Princeton, New Jersey, 08543, USA
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland, 21201, USA.
| |
Collapse
|
28
|
Behl T, Kaur I, Sehgal A, Sharma E, Kumar A, Grover M, Bungau S. Unfolding Nrf2 in diabetes mellitus. Mol Biol Rep 2021; 48:927-939. [PMID: 33389540 DOI: 10.1007/s11033-020-06081-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/10/2020] [Indexed: 11/24/2022]
Abstract
In spite of much awareness, diabetes mellitus continues to remain one of major reasons for mortality and morbidity rate all over the globe. Free radicals cause oxidative stress which is responsible for causing diabetes. The recent advancements in elucidation of ARE/keap1/Nrf2 pathway can help in better understanding of diabetes mellitus. Various clinical trials and animal studies have shown the promising effect of Nrf2 pathway in reversing diabetes by counteracting with the oxidative stress produced. The gene is known to dissociate from Keap1 on coming in contact with such stresses to show preventive and prognosis effect. The Nrf2 gene has been marked as a molecular player in dealing with wide intracellular as well as extracellular cellular interactions in different diseases. The regulation of this gene gives some transcription factor that contain antioxidant response elements (ARE) in their promoter region and thus are responsible for encoding certain proteins involved in regulation of metabolic and detoxifying enzymes.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Eshita Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhuri Grover
- B.S. Anangpuria Institute of Pharmacy, Alampur, Haryana, India
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
29
|
Tan X, Jiao PL, Sun JC, Wang W, Ye P, Wang YK, Leng YQ, Wang WZ. β-Arrestin1 Reduces Oxidative Stress via Nrf2 Activation in the Rostral Ventrolateral Medulla in Hypertension. Front Neurosci 2021; 15:657825. [PMID: 33897365 PMCID: PMC8059792 DOI: 10.3389/fnins.2021.657825] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/04/2021] [Indexed: 12/31/2022] Open
Abstract
Oxidative stress in the rostral ventrolateral medulla (RVLM), a key region for blood pressure (BP) regulation, has been demonstrated to be responsible for the overactivity of the sympathetic nervous system in hypertension and heart failure. Nuclear factor-erythroid-2-related factor 2 (Nrf2) is a key transcription factor that maintains redox homeostasis by governing a broad array of antioxidant genes in response to oxidative stress. β-Arrestin1 is a multifunctional scaffold protein with the ability to interact with diverse signaling molecules independent of G protein-coupled receptors (GPCRs), and its overexpression in the RVLM could reduce BP and renal sympathetic nerve activity (RSNA) in spontaneously hypertensive rats (SHR). The goal of this study was to investigate whether Nrf2-mediated antioxidative stress is involved in the antihypertensive effect of β-arrestin1 in the RVLM. It was found that the activation level of Nrf2 in the RVLM of SHR was significantly reduced, compared with normotensive Wistar-Kyoko (WKY) rats. Overexpression of β-arrestin1 in the RVLM significantly decreased ROS production and facilitated the Nrf2 activation in the RVLM of SHR, accompanied by upregulating the expression of HO-1 and NQO-1. However, Nrf2 knockdown attenuated the antioxidant effect of β-arrestin1 overexpression in the RVLM by downregulating HO-1 and NQO-1 expression levels. In conclusion, the current results suggested that the antihypertensive effect of β-arrestin1 overexpression in the RVLM is mediated by decreased ROS production, which is associated with Nrf2 activation.
Collapse
Affiliation(s)
- Xing Tan
- Department of Marine Biomedicine and Polar Medicine, Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Pei-Lei Jiao
- Department of Marine Biomedicine and Polar Medicine, Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai, China
- Department of Orthopedics, The 962th Hospital of People’s Liberation Army, Harbin, China
| | - Jia-Cen Sun
- Department of Marine Biomedicine and Polar Medicine, Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wen Wang
- Department of Marine Biomedicine and Polar Medicine, Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Peng Ye
- Department of Marine Biomedicine and Polar Medicine, Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yang-Kai Wang
- Department of Marine Biomedicine and Polar Medicine, Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yue-Qi Leng
- Department of Marine Biomedicine and Polar Medicine, Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wei-Zhong Wang
- Department of Marine Biomedicine and Polar Medicine, Naval Medical Center, Naval Medical University (Second Military Medical University), Shanghai, China
- *Correspondence: Wei-Zhong Wang,
| |
Collapse
|
30
|
An S, Wang X, Shi H, Zhang X, Meng H, Li W, Chen D, Ge J. Apelin protects against ischemia-reperfusion injury in diabetic myocardium via inhibiting apoptosis and oxidative stress through PI3K and p38-MAPK signaling pathways. Aging (Albany NY) 2020; 12:25120-25137. [PMID: 33342766 PMCID: PMC7803490 DOI: 10.18632/aging.104106] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
Among all diabetes mellitus-associated cardiovascular diseases, morbidity of diabetic myocardium with ischemia reperfusion injury (D-IRI) is increasing year by year. We aimed to discover a therapeutic biomarker and investigate its mechanism in D-IRI. High-fat diet and streptozotocin-induced diabetes rats were operated with IRI or sham. Recombined lentiviral vector encoding Apelin was injected into D-IRI rat via tail vein. Cardiac function, infarct size, cellular death and oxidative stress were major outcome measures. Cardiomyocyte ischemia reperfusion injury was more serious in D-IRI rats than in non-diabetes ischemia reperfusion injury (ND-IRI) rats. The secretion of NTproBNP was increased in D-IRI compared with ND-IRI. Bcl-2 expression was decreased, and Bax and cleaved caspase-3 expression was increased in D-IRI rats compared with ND-IRI rats, which were reversed after treatment with Apelin. Apelin-upregulation improved cardiomyocyte ischemia reperfusion injury and decreased NT-proBNP levels in D-IRI rats. Apelin overexpression enhanced PI3K and eNOS levels while reduced those of p38-MAPK and iNOS in D-IRI rats. Apelin overexpression protected against D-IRI through inhibiting apoptosis and oxidative stress via PI3K and p38MAPK signaling pathways in D-IRI rats. These findings provide critical new insight into understanding of Apelin's cardio-protective effects, which may become a novel therapeutic target for the diabetic IRI patients.
Collapse
Affiliation(s)
- Songtao An
- Department of Cardiology, Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Xi Wang
- Department of Cardiology, Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Huairui Shi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xueqiang Zhang
- Department of Cardiology, Hongxing Hospital, Hami 839000, Xinjiang, China
| | - Hua Meng
- Department of Cardiology, Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Wenbo Li
- Department of Cardiology, Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Dongchang Chen
- Department of Cardiology, Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
31
|
Liu L, Yang S, Wang H. α-Lipoic acid alleviates ferroptosis in the MPP + -induced PC12 cells via activating the PI3K/Akt/Nrf2 pathway. Cell Biol Int 2020; 45:422-431. [PMID: 33241887 DOI: 10.1002/cbin.11505] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/27/2020] [Accepted: 10/31/2020] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is a typical neurodegenerative disease. α-Lipoic acid (α-LA) can reduce the incidence of neuropathy. The present study explored the role and mechanism of α-LA in 1-methyl-4-phenylpyridinium (MPP+ )-induced cell model of PD. The PD model was induced via treating PC12 cells with MPP+ at different concentrations. MPP+ and α-LA effects on PC12 cells were assessed from cell viability and ferroptosis. Cell viability was detected using the cell counting kit-8 assay. Malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), iron, reactive xygen species (ROS), and glutathione (GSH) concentrations, and ferroptosis-related protein SLC7A11 and GPx4 expressions were used for ferroptosis evaluation. p-PI3K, p-Akt, and nuclear factor erythroid 2-related factor 2 (Nrf2) protein levels were detected. The PI3K/Akt/Nrf2 pathway inhibitors were applied to verify the role of the PI3K/Akt/Nrf2 pathway in α-LA protection against MPP+ -induced decreased cell viability and ferroptosis. MPP+ -reduced cell viability and induced ferroptosis as presented by increased MDA, 4-HNE, iron, and ROS concentrations, and reduced levels of GSH and ferroptosis marker proteins (SLC7A11 and GPx4). α-LA attenuated MPP+ -induced cell viability decline and ferroptosis. The PI3K/Akt/Nrf2 pathway was activated after α-LA treatment. Inhibiting the PI3K/Akt/Nrf2 pathway weakened the protection of α-LA against MPP+ treatment. We highlighted that α-LA alleviated MPP+ -induced cell viability decrease and ferroptosis in PC12 cells via activating the PI3K/Akt/Nrf2 pathway.
Collapse
Affiliation(s)
- Lin Liu
- Department of Neurology, Nankai University Affiliated Nankai Hospital, Changjiang Dao, Nankai, Tianjin, China
| | - Songqi Yang
- Department of Neurology, Nankai University Affiliated Nankai Hospital, Changjiang Dao, Nankai, Tianjin, China
| | - Heng Wang
- Department of Neurology, Nankai University Affiliated Nankai Hospital, Changjiang Dao, Nankai, Tianjin, China
| |
Collapse
|
32
|
Bencsik P, Gömöri K, Szabados T, Sántha P, Helyes Z, Jancsó G, Ferdinandy P, Görbe A. Myocardial ischaemia reperfusion injury and cardioprotection in the presence of sensory neuropathy: Therapeutic options. Br J Pharmacol 2020; 177:5336-5356. [PMID: 32059259 PMCID: PMC7680004 DOI: 10.1111/bph.15021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
During the last decades, mortality from acute myocardial infarction has been dramatically reduced. However, the incidence of post-infarction heart failure is still increasing. Cardioprotection by ischaemic conditioning had been discovered more than three decades ago. Its clinical translation, however, is still an unmet need. This is mainly due to the disrupted cardioprotective signalling pathways in the presence of different cardiovascular risk factors, co-morbidities and the medication being taken. Sensory neuropathy is one of the co-morbidities that has been shown to interfere with cardioprotection. In the present review, we summarize the diverse aetiology of sensory neuropathies and the mechanisms by which these neuropathies may interfere with ischaemic heart disease and cardioprotective signalling. Finally, we suggest future therapeutic options targeting both ischaemic heart and sensory neuropathy simultaneously. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Péter Bencsik
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of MedicineUniversity of SzegedSzegedHungary
- Pharmahungary GroupSzegedHungary
| | - Kamilla Gömöri
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of MedicineUniversity of SzegedSzegedHungary
- Pharmahungary GroupSzegedHungary
| | - Tamara Szabados
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of MedicineUniversity of SzegedSzegedHungary
- Pharmahungary GroupSzegedHungary
| | - Péter Sántha
- Department of Physiology, Faculty of MedicineUniversity of SzegedSzegedHungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical SchoolUniversity of PécsPécsHungary
- Molecular Pharmacology Research Group, Centre for Neuroscience, János Szentágothai Research CentreUniversity of PécsPécsHungary
| | - Gábor Jancsó
- Department of Physiology, Faculty of MedicineUniversity of SzegedSzegedHungary
| | - Péter Ferdinandy
- Pharmahungary GroupSzegedHungary
- Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of MedicineUniversity of SzegedSzegedHungary
- Pharmahungary GroupSzegedHungary
- Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
| |
Collapse
|
33
|
Gunata M, Parlakpinar H. A review of myocardial ischaemia/reperfusion injury: Pathophysiology, experimental models, biomarkers, genetics and pharmacological treatment. Cell Biochem Funct 2020; 39:190-217. [PMID: 32892450 DOI: 10.1002/cbf.3587] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/03/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases are known to be the most fatal diseases worldwide. Ischaemia/reperfusion (I/R) injury is at the centre of the pathology of the most common cardiovascular diseases. According to the World Health Organization estimates, ischaemic heart disease is the leading global cause of death, causing more than 9 million deaths in 2016. After cardiovascular events, thrombolysis, percutaneous transluminal coronary angioplasty or coronary bypass surgery are applied as treatment. However, after restoring coronary blood flow, myocardial I/R injury may occur. It is known that this damage occurs due to many pathophysiological mechanisms, especially increasing reactive oxygen types. Besides causing cardiomyocyte death through multiple mechanisms, it may be an important reason for affecting other cell types such as platelets, fibroblasts, endothelial and smooth muscle cells and immune cells. Also, polymorphonuclear leukocytes are associated with myocardial I/R damage during reperfusion. This damage may be insufficient in patients with co-morbidity, as it is demonstrated that it can be prevented by various endogenous antioxidant systems. In this context, the resulting data suggest that optimal cardioprotection may require a combination of additional or synergistic multi-target treatments. In this review, we discussed the pathophysiology, experimental models, biomarkers, treatment and its relationship with genetics in myocardial I/R injury. SIGNIFICANCE OF THE STUDY: This review summarized current information on myocardial ischaemia/reperfusion injury (pathophysiology, experimental models, biomarkers, genetics and pharmacological therapy) for researchers and reveals guiding data for researchers, especially in the field of cardiovascular system and pharmacology.
Collapse
Affiliation(s)
- Mehmet Gunata
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Hakan Parlakpinar
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
34
|
Role of Nrf2 and Its Activators in Cardiocerebral Vascular Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4683943. [PMID: 32831999 PMCID: PMC7428967 DOI: 10.1155/2020/4683943] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/16/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
Abstract
Cardiocerebral vascular disease (CCVD) is a common disease with high morbidity, disability, and mortality. Oxidative stress (OS) is closely related to the progression of CCVD. Abnormal redox regulation leads to OS and overproduction of reactive oxygen species (ROS), which can cause biomolecular and cellular damage. The Nrf2/antioxidant response element (ARE) signaling pathway is one of the most important defense systems against exogenous and endogenous OS injury, and Nrf2 is regarded as a vital pharmacological target. The complexity of the CCVD pathological process and the current difficulties in conducting clinical trials have hindered the development of therapeutic drugs. Furthermore, little is known about the role of the Nrf2/ARE signaling pathway in CCVD. Clarifying the role of the Nrf2/ARE signaling pathway in CCVD can provide new ideas for drug design. This review details the recent advancements in the regulation of the Nrf2/ARE system and its role and activators in common CCVD development.
Collapse
|
35
|
Effects of alpha-lipoic acid on skeletal muscle ischemia-reperfusion injury in mice. JOURNAL OF SURGERY AND MEDICINE 2020. [DOI: 10.28982/josam.764953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
36
|
Wang S, Yang X. Eleutheroside E decreases oxidative stress and NF-κB activation and reprograms the metabolic response against hypoxia-reoxygenation injury in H9c2 cells. Int Immunopharmacol 2020; 84:106513. [PMID: 32330867 DOI: 10.1016/j.intimp.2020.106513] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/21/2020] [Accepted: 04/13/2020] [Indexed: 01/23/2023]
Abstract
Ischemia-reperfusion (I/R) injury causes cardiac dysfunction through several mechanisms including oxidative stress and pro-inflammation. Eleutheroside E (EE) has protective effects in ischemia tissue and anti-inflammatory action. However, the effect of EE on I/R-injured cardiomyocytes is unknown. In this study, we used in vitro H9c2 cell model to investigate the favorable role of EE on myocardial I/R injury. We found that EE administration attenuated the cardiomyocyte apoptosis induced by hypoxia-reoxygenation (H/R) injury. Further, pre-treatment with EE dramatically inhibited mitochondrial oxidative stress, IκBα phosphorylation and nuclear factor kappa B (NF-κB) subunit p65 translocation into nuclei. EE might suppress the MAPK signaling pathway to inhibit the H/R-induced NF-κB activation. Moreover, we had analyzed the metabolomic profile of H/R-injured and H/R + 100 EE-treated H9c2 cells and found that the abundance of most metabolites changed by H/R could be re-modulated by EE treatment. Pathway analysis highlighted the inhibition of fatty acid biosynthesis and alternation of arginine and proline metabolism as two potential links to the favorable effect of EE on H/R-injured cardiomyocytes. The further demonstration showed that nitric oxide (NO), a product that is solely catabolized by l-arginine and has profound anti-oxidative stress activity during H/R in cardiomyocytes, was augmented by EE. Altogether, our results provide evidence that EE may be a potential drug for myocardial I/R injury by reducing oxidative stress, NF-κB activation, and metabolic reprogramming.
Collapse
Affiliation(s)
- Shanyue Wang
- Department Cardiovascular Medicine, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Xuming Yang
- Department Cardiovascular Medicine, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| |
Collapse
|
37
|
Wu PF, Chen GL. Interleukin-6 Response of C 2C 12 Myotubes Stimulated with Lipopolysaccharide and Lipoic Acid. J Interferon Cytokine Res 2020; 40:254-261. [PMID: 32176561 DOI: 10.1089/jir.2019.0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Our previous study explored the dual effect of lipoic acid on the regulation of IL-6 expression in C2C12 myotubes. However, the specific mechanism remains unclear. To evaluate IL-6 signaling in skeletal muscle, pCMV6-IL-6 was overexpressed in C2C12 myotubes. The real-time quantitative polymerase chain reaction was used to detect mRNA expression. Immunohistochemistry and a DeadEnd colorimetric TUNEL system were used to detect IL-6 localization and analyze the apoptosis in IL-6-overexpressing cells, respectively. A caspase-3/CPP32 colorimetric assay and Western blotting were used to analyze caspase-3 activity and protein expression, respectively. Our results showed the overexpressed IL-6 was not only located in the cytosol but also on the intracellular side of the cell membrane. Moreover, the nucleus did not demonstrate IL-6 overexpression. The DeadEnd colorimetric apoptosis detection assay results demonstrated that apoptotic nuclei were present in IL-6-overexpressing cells. However, the overexpressed IL-6 failed to promote caspase-3 activity. Notably, the exogenous pyrogen lipopolysaccharide (LPS) significantly promoted IL-6 mRNA expression and caspase-3 activity but did not induce apoptotic cell formation. Moreover, lipoic acid significantly upregulated IL-6, IL-6Ra, and gp130 mRNA expression and significantly increased caspase-3 activity but did not induce apoptotic cell formation. Lipoic acid significantly increased the p-Akt level in untreated cells but not in LY294002-treated cells. Taken together, our results suggesting that the overexpressed IL-6-induced apoptosis may not be mediated by caspase-3. LPS-induced IL-6 mRNA expression may not be involved in IL-6 classical signaling or trans-signaling in C2C12 myotubes. Lipoic acid-induced IL-6 mRNA expression may be mediated by IL-6 classical signaling in C2C12 myotubes. [Figure: see text].
Collapse
Affiliation(s)
- Pei-Fung Wu
- Department of Kinesiology, Health and Leisure Studies, National University of Kaohsiung, Kaohsiung, Taiwan
| | - Guan-Lin Chen
- Department of Kinesiology, Health and Leisure Studies, National University of Kaohsiung, Kaohsiung, Taiwan
| |
Collapse
|
38
|
Billah M, Ridiandries A, Allahwala UK, Mudaliar H, Dona A, Hunyor S, Khachigian LM, Bhindi R. Remote Ischemic Preconditioning induces Cardioprotective Autophagy and Signals through the IL-6-Dependent JAK-STAT Pathway. Int J Mol Sci 2020; 21:ijms21051692. [PMID: 32121587 PMCID: PMC7084188 DOI: 10.3390/ijms21051692] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/19/2020] [Accepted: 02/27/2020] [Indexed: 02/08/2023] Open
Abstract
Autophagy is a cellular process by which mammalian cells degrade and assist in recycling damaged organelles and proteins. This study aimed to ascertain the role of autophagy in remote ischemic preconditioning (RIPC)-induced cardioprotection. Sprague Dawley rats were subjected to RIPC at the hindlimb followed by a 30-min transient blockade of the left coronary artery to simulate ischemia reperfusion (I/R) injury. Hindlimb muscle and the heart were excised 24 h post reperfusion. RIPC prior to I/R upregulated autophagy in the rat heart at 24 h post reperfusion. In vitro, autophagy inhibition or stimulation prior to RIPC, respectively, either ameliorated or stimulated the cardioprotective effect, measured as improved cell viability to mimic the preconditioning effect. Recombinant interleukin-6 (IL-6) treatment prior to I/R increased in vitro autophagy in a dose-dependent manner, activating the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathway without affecting the other kinase pathways, such as p38 mitogen-activated protein kinases (MAPK), and glycogen synthase kinase 3 Beta (GSK-3β) pathways. Prior to I/R, in vitro inhibition of the JAK-STAT pathway reduced autophagy upregulation despite recombinant IL-6 pre-treatment. Autophagy is an essential component of RIPC-induced cardioprotection that may upregulate autophagy through an IL-6/JAK-STAT-dependent mechanism, thus identifying a potentially new therapeutic option for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Muntasir Billah
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
- Sydney Medical School Northern, University of Sydney, Sydney, NSW 2006, Australia
- School of Life Sciences, Independent University Bangladesh, Dhaka 1229, Bangladesh
- Correspondence:
| | - Anisyah Ridiandries
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
- Sydney Medical School Northern, University of Sydney, Sydney, NSW 2006, Australia
| | - Usaid K Allahwala
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
- Sydney Medical School Northern, University of Sydney, Sydney, NSW 2006, Australia
| | - Harshini Mudaliar
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
| | - Anthony Dona
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
| | - Stephen Hunyor
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
| | - Levon M. Khachigian
- Vascular Biology and Translational Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia;
| | - Ravinay Bhindi
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia; (A.R.); (U.K.A.); (H.M.); (A.D.); (S.H.); (R.B.)
- Sydney Medical School Northern, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
39
|
Nyunt T, Britton M, Wanichthanarak K, Budamagunta M, Voss JC, Wilson DW, Rutledge JC, Aung HH. Mitochondrial oxidative stress-induced transcript variants of ATF3 mediate lipotoxic brain microvascular injury. Free Radic Biol Med 2019; 143:25-46. [PMID: 31356870 PMCID: PMC6848793 DOI: 10.1016/j.freeradbiomed.2019.07.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/30/2019] [Accepted: 07/23/2019] [Indexed: 12/06/2022]
Abstract
Elevation of blood triglycerides, primarily triglyceride-rich lipoproteins (TGRL), is an independent risk factor for cardiovascular disease and vascular dementia (VaD). Accumulating evidence indicates that both atherosclerosis and VaD are linked to vascular inflammation. However, the role of TGRL in vascular inflammation, which increases risk for VaD, remains largely unknown and its underlying mechanisms are still unclear. We strived to determine the effects of postprandial TGRL exposure on brain microvascular endothelial cells, the potential risk factor of vascular inflammation, resulting in VaD. We showed in Aung et al., J Lipid Res., 2016 that postprandial TGRL lipolysis products (TL) activate mitochondrial reactive oxygen species (ROS) and increase the expression of the stress-responsive protein, activating transcription factor 3 (ATF3), which injures human brain microvascular endothelial cells (HBMECs) in vitro. In this study, we deployed high-throughput sequencing (HTS)-based RNA sequencing methods and mito stress and glycolytic rate assays with an Agilent Seahorse XF analyzer and profiled the differential expression of transcripts, constructed signaling pathways, and measured mitochondrial respiration, ATP production, proton leak, and glycolysis of HBMECs treated with TL. Conclusions: TL potentiate ROS by mitochondria which activate mitochondrial oxidative stress, decrease ATP production, increase mitochondrial proton leak and glycolysis rate, and mitochondria DNA damage. Additionally, CPT1A1 siRNA knockdown suppresses oxidative stress and prevents mitochondrial dysfunction and vascular inflammation in TL treated HBMECs. TL activates ATF3-MAPKinase, TNF, and NRF2 signaling pathways. Furthermore, the NRF2 signaling pathway which is upstream of the ATF3-MAPKinase signaling pathway, is also regulated by the mitochondrial oxidative stress. We are the first to report differential inflammatory characteristics of transcript variants 4 (ATF3-T4) and 5 (ATF3-T5) of the stress responsive gene ATF3 in HBMECs induced by postprandial TL. Specifically, our data indicates that ATF3-T4 predominantly regulates the TL-induced brain microvascular inflammation and TNF signaling. Both siRNAs of ATF3-T4 and ATF3-T5 suppress cells apoptosis and lipotoxic brain microvascular endothelial cells. These novel signaling pathways triggered by oxidative stress-responsive transcript variants, ATF3-T4 and ATF3-T5, in the brain microvascular inflammation induced by TGRL lipolysis products may contribute to pathophysiological processes of vascular dementia.
Collapse
Affiliation(s)
- Tun Nyunt
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Monica Britton
- Genome Center and Bioinformatics Core Facility, University of California, Davis, CA, 95616, USA
| | - Kwanjeera Wanichthanarak
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA, USA; Department of Biochemistry and Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Madhu Budamagunta
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - John C Voss
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Dennis W Wilson
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - John C Rutledge
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Hnin H Aung
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
40
|
Zhang S, Zhao Y. Lentinan protects cardiomyocytes against hypoxia-induced injury by regulation of microRNA-22/Sirt1. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3938-3946. [PMID: 31581847 DOI: 10.1080/21691401.2019.1666863] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Shaohui Zhang
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Yongliang Zhao
- Department of Cardiac Surgery, Affiliated Hospital of Jining Medical University, Jining, China
| |
Collapse
|
41
|
Chai L, Huang T, Wang Z, Xu R, Deng C. AAVC-I promotes apoptosis of human oral squamous cell carcinoma through the mitochondrial pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:3968-3974. [PMID: 31933792 PMCID: PMC6949759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 09/24/2019] [Indexed: 06/10/2023]
Abstract
This study aimed to explore the role and possible mechanism of component I from agkistrodon acutus venom (AAVC-I) in promoting the apoptosis of oral squamous cell carcinoma (OSCC). HN4 cells (a human OSCC cell line) were randomly divided into four experimental groups: low AAVC-I (2.5 μg/mL) group, medium AAVC-I (5 μg/mL) group and high AAVC-I (10 μg/mL) group, as well as control group (AAVC-I, 0 μg/mL). AVVC-I was dissolved in RPMI-1640 medium and added to the culture wells at different concentrations when tumor cells had reached the logarithmic growth phase. After 24 hours, cells were harvested and the inhibitory rate of cell proliferation and the mitochondrial membrane depolarization were measured. Western blotting was used to detect the expression levels of cytochrome c, Bcl-2 associated X protein (Bax), and caspase-3 in the cellular cytoplasm either containing mitochondria or following the removal of mitochondria. Cellular apoptosis was detected by flow cytometry. Compared to the control group, AAVC-I treatment not only inhibited NH4 growth, but also upgraded the expression of caspase-3 in NH4 cells. Meanwhile, it was observed that Bax translocation to mitochondria and cytochrome c release into the cytosol increased in AAVC-I treatment. This indicated that AAVC-I could disrupt mitochondrial membrane depolarization and result in cellular apoptosis, and the apoptosis rate of NH4 increased with the concentration of AAVC-I. The data suggested that AAVC-I promotes the apoptosis of HN4 cells through the mitochondrial pathway in a dose-dependent manner, which provides experimental data and new ideas for future research and clinical treatment options for OSCC.
Collapse
Affiliation(s)
- Lin Chai
- School of Stomatology, Wannan Medical College22 West Wenchang Road, Wuhu 241002, Anhui Province, China
| | - Tingting Huang
- Department of Pathophysiology, Wannan Medical College22 West Wenchang Road, Wuhu 241002, Anhui Province, China
| | - Zhenjie Wang
- Department of Pathophysiology, Wannan Medical College22 West Wenchang Road, Wuhu 241002, Anhui Province, China
| | - Ran Xu
- School of Stomatology, Wannan Medical College22 West Wenchang Road, Wuhu 241002, Anhui Province, China
| | - Chao Deng
- School of Stomatology, Wannan Medical College22 West Wenchang Road, Wuhu 241002, Anhui Province, China
| |
Collapse
|
42
|
Kurumazuka D, Kitada K, Tanaka R, Mori T, Ohkita M, Takaoka M, Matsumura Y. α-Lipoic acid exerts a primary prevention for the cardiac dysfunction in aortocaval fistula-created rat hearts. Heliyon 2019; 5:e02371. [PMID: 31517099 PMCID: PMC6728770 DOI: 10.1016/j.heliyon.2019.e02371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 07/04/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023] Open
Abstract
Aim α-Lipoic acid exerts a powerful antioxidant effect by acting as a free radical scavenger and inducing endogenous antioxidants such as vitamin E and glutathione. In the present study, we examined the effects of α-lipoic acid on cardiac dysfunction in rat hearts with aortocaval fistulae. Main methods Aortocaval fistulae were created between the abdominal aorta and inferior vena cava in male rats. Hemodynamic parameters were measured 14 days after surgery using an intravascular pressure transducer, and then these hearts were harvested for tissue weight measurement, pathological evaluation, and mRNA isolation. Results In vehicle-treated rats, left ventricular end-diastolic pressure and left ventricular weight significantly increased at 14 days after fistula creation. Fistula-creation resulted in expression of 4-hydroxy-2-nonenal, NADPH oxidase subunit p67phox and BNP mRNA in a time-dependent manner in the left ventricle. Long-term treatment (initiated 2 days before surgery, and continued for 14 days after fistula creation; days -2 to 14) with α-lipoic acid (30 mg/kg/day) markedly suppressed the increases in left and right ventricular weight, and left ventricular end-diastolic pressure. α-Lipoic acid treatment from days -2 to 14 prominently prevented the expression of 4-hydroxy-2-nonenal and NADPH oxidase subunit p67phox, and significantly raised BNP mRNA levels. Short-term treatment with α-lipoic acid from day - 2 to 7 was effective in preventing cardiac enlargement and dysfunction, similar to long-term treatment, but treatment from days 7–14 was not effective. Conclusions Treatment with α-lipoic acid can prevent cardiac hyperplasia and dysfunction, probably by inhibiting superoxide production and enhancing BNP mRNA expression in an early phase after fistula creation.
Collapse
Affiliation(s)
- Daisuke Kurumazuka
- Laboratory of Pathological and Molecular Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Kento Kitada
- Laboratory of Pathological and Molecular Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Ryosuke Tanaka
- Laboratory of Pathological and Molecular Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Tatsuhiko Mori
- Division of Nephrology, Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan
| | - Mamoru Ohkita
- Laboratory of Pathological and Molecular Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Masanori Takaoka
- Laboratory of Cell Biology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Yasuo Matsumura
- Laboratory of Pathological and Molecular Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| |
Collapse
|
43
|
Li Y, Zhu X, Liu X, Du A, Yu B. miR-200a mediates protection of thymosin β-4 in cardiac microvascular endothelial cells as a novel mechanism under hypoxia-reoxygenation injury. J Cell Biochem 2019; 120:19098-19106. [PMID: 31265170 DOI: 10.1002/jcb.29237] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 12/29/2022]
Abstract
Thymosin β-4 (Tβ4) is a ubiquitous protein, which has been suggested to regulate multiple cell signal pathways and a variety of cellular functions. However, the role Tβ4 plays in the cardiac microvascular endothelial cells (CMECs) under myocardial ischemia/reperfusion injury is currently unknown. Here we investigated the effects of Tβ4 on hypoxia/reoxygenation (H/R) induced CMECs injury and its potential molecular mechanism. Cultured CMECs were positively identified by flow cytometry using antibody against CD31 and VWF/Factor VIII, which are constitutively expressed on the surface of CMECs. Then the reduced level of Tβ4 was detected in H/R-CMECs by a real-time quantitative polymerase chain reaction. To determine the effects of Tβ4 on H/R-CMECs, we transfected the overexpression or silence vector of Tβ4 into CMECs under H/R condition. Our results indicated that H/R treatment could reduce proliferation, increased apoptosis, adhesion, and reactive oxygen species (ROS) production in CMECs, which were attenuated by Tβ4 overexpression or aggravated by Tβ4 silencing, implying Tβ4 is able to promote CMECs against H/R-induced cell injury. Furthermore, the microRNA-200a (miR-200a) level was also increased by Tβ4 in H/R-CMECs or reduced by Tβ4 small interfering RNA. To investigated the mechanism of protective effects of Tβ4 on CMECs injury, the miR-200a inhibitor was transfected into H/R-CMECs. The results indicated that inhibition of miR-200a inversed the protection of Tβ4 on H/R-CMECs, specifically including cell proliferation, cell adhesion, cell apoptosis, and ROS production, as well as nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation. In conclusion, our results determined that Tβ4 attenuated H/R-induced CMECs injury by miR-200a-Nrf2 signaling.
Collapse
Affiliation(s)
- Yang Li
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Xiaolong Zhu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Xiping Liu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Aolin Du
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Bo Yu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| |
Collapse
|
44
|
Abstract
OBJECTIVES To assess the effect of vitamin D2 and to elucidate the underlying mechanisms on acute myocardial injury induced by isoproterenol (ISO) in diabetic rats. METHODS Rats were divided into control rats, diabetic rats (DM), diabetic rats received ISO (DM-ISO), and diabetic rats pretreated with vitamin D2 and received ISO (DM-D2-ISO). RESULTS Vitamin D2 pretreatment significantly decreased fasting glucose and myocardial malondialdehyde, associated with increased insulin, myocardial glutathione and superoxide dismutase in DM-D2-ISO versus DM-ISO. The serum triglycerides, total cholesterol, and LDL were significantly decreased, along with increased HDL and adiponectin. Poly-ADP ribose polymerase, cyclooxygenase-2, tumour necrosis factor alpha, interleukin-6, caspase-3, BAX, and p53 were significantly downregulated in myocardium of DM-D2-ISO versus DM-ISO. Histological studies showed diminished inflammatory cells infiltration in myocardium of DM-D2-ISO versus DM-ISO. CONCLUSION Vitamin D2 ameliorates hyperglycaemia, dyslipidaemia, redox imbalance, inflammatory and apoptotic processes, protecting the myocardium of diabetic rats against acute myocardial infarction.
Collapse
Affiliation(s)
- Sahar M El Agaty
- a Physiology Department, Faculty of Medicine , Ain Shams University , Cairo , Egypt
| |
Collapse
|
45
|
Aksoy U, Savtekin G, Şehirli AÖ, Kermeoğlu F, Kalender A, Özkayalar H, Sayıner S, Orhan K. Effects of alpha-lipoic acid therapy on experimentally induced apical periodontitis: a biochemical, histopathological and micro-CT analysis. Int Endod J 2019; 52:1317-1326. [PMID: 30900747 DOI: 10.1111/iej.13121] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 03/18/2019] [Indexed: 12/23/2022]
Abstract
AIM To investigate the possible therapeutic effects of alpha-lipoic acid (ALA) in a model of chronic apical periodontitis in rats by analysing biochemical, histopathological and micro-CT parameters. METHODOLOGY The study was approved by the Animal Ethics Committee of the Near East University. Thirty-two Wistar rats were divided into four groups of eight rats each: Control Group; ALA Group; AP Group; AP + ALA Group. In the AP and AP + ALA groups, the pulp chambers of the mandibular first molars were surgically exposed and were left open to the oral environment for 4-weeks to allow the establishment of periapical lesions. The rats in the Control and AP groups were treated intraperitoneally with saline solution (with a daily dose of 100 mg kg-1 , for 28 days after periapical lesion induction). The rats in the ALA and AP + ALA groups were treated intraperitoneally with ALA (with a daily dose of 100 mg kg-1 , for 28 days after periapical lesion induction). After decapitation, the trunk blood was collected for the assessment of biochemical parameters. The mandibles were surgically removed and dissected for histopathologic analysis and further scanned with micro-CT. Groups of data were compared with a two-way analysis of variance (two-way anova) followed by Sidak's multiple comparison tests. Values of P < 0.05 were regarded as significant. RESULTS TNF-α, IL-1β, MMP-1, MMP-2 levels were significantly lower in AP + ALA group compared with AP group (P < 0.05). There was a significant difference between the AP and AP + ALA groups according to assessment of the inflammatory scores (P < 0.05). The periapical inflammatory infiltrates were significantly more severe (P < 0.05) in the AP group. The AP + ALA group exhibited lower values both in terms of surface area and volume of resorption cavities than the AP group and this difference was significant (P < 0.05). CONCLUSION alpha-lipoic acid treatment provided therapeutic effects on the inhibition of periapical bone loss.
Collapse
Affiliation(s)
- U Aksoy
- Department of Endodontics, Faculty of Dentistry, Near East University, Mersin 10, Turkey
| | - G Savtekin
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Near East University, Mersin 10, Turkey
| | - A Ö Şehirli
- Departments of Pharmacology, Faculty of Dentistry, Near East University, Mersin 10, Turkey
| | - F Kermeoğlu
- Department of Endodontics, Faculty of Dentistry, Near East University, Mersin 10, Turkey
| | - A Kalender
- Department of Endodontics, Faculty of Dentistry, Near East University, Mersin 10, Turkey
| | - H Özkayalar
- Department of Pathology, Faculty of Medicine, Near East University, Mersin 10, Turkey
| | - S Sayıner
- Department of Biochemistry, Faculty of Veterinary Medicine, Near East University, Mersin 10, Turkey
| | - K Orhan
- OMFS IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, University of Leuven, Leuven, Belgium.,Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium.,Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Ankara University, Ankara, Turkey
| |
Collapse
|
46
|
The Immunomodulatory Effect of Alpha-Lipoic Acid in Autoimmune Diseases. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8086257. [PMID: 31016198 PMCID: PMC6446120 DOI: 10.1155/2019/8086257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 02/11/2019] [Accepted: 02/21/2019] [Indexed: 01/08/2023]
Abstract
Αlpha-lipoic acid is a naturally occurring antioxidant in human body and has been widely used as an antioxidant clinically. Accumulating evidences suggested that α-lipoic acid might have immunomodulatory effects on both adaptive and innate immune systems. This review focuses on the evidences and potential targets involved in the immunomodulatory effects of α-lipoic acid. It highlights the fact that α-lipoic acid may have beneficial effects in autoimmune diseases once the immunomodulatory effects can be confirmed by further investigation.
Collapse
|
47
|
Zhang Y, Lv YL, Si YN, Zhou J, Qian Y, Bao HG. α-lipoic acid attenuates spatial learning and memory impairment induced by hepatectomy. Exp Ther Med 2019; 17:2329-2333. [PMID: 30867718 DOI: 10.3892/etm.2019.7202] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 11/19/2018] [Indexed: 01/07/2023] Open
Abstract
The aim of the present study was to compare the effects of α-lipoic acid (ALA) on postoperative cognitive dysfunction (POCD) between wild type (WT) and leptin receptor-deficient (db/db) mice and to elucidate the underlying mechanism of treatment with ALA. The present study compared the effects of ALA on spatial learning and memory of WT and db/db mice using a Morris water maze following hepatectomy. The expression levels of proteins, including cyclin-dependent kinase 5 (Cdk5), tau, phosphorylated tau and amyloid β (Aβ) were measured in the hippocampus. Surgery impaired postoperative cognitive function in both WT and db/db mice. Furthermore, the expression levels of Cdk5 and Aβ, and the phosphorylation of tau in the hippocampus increased after the surgery in both WT and db/db mice. The ultrastructure of hippocampal neurons and synapses was analyzed by transmission electron microscopy and the results revealed that surgery damaged the structure of neurons and synapses in both WT and db/db mice. Treatment with ALA protected the postoperative cognitive function and the structure of hippocampal neurons and synapses, and prevented the increase in protein expression levels of Cdk5 and Aβ, and the phosphorylation of tau in the hippocampus of WT but not db/db mice. The results of the present study suggest that ALA may be used for the treatment of POCD. The molecular mechanisms underlying the activity of ALA require further investigation.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Yun-Luo Lv
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Yan-Na Si
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Jianwei Zhou
- Deparment of Molecular Cell Biology and Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Yanning Qian
- Department of Anesthesiology, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hong-Guang Bao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| |
Collapse
|
48
|
Cong P, Tong C, Liu Y, Shi L, Shi X, Zhao Y, Xiao K, Jin H, Liu Y, Hou M. CD28 Deficiency Ameliorates Thoracic Blast Exposure-Induced Oxidative Stress and Apoptosis in the Brain through the PI3K/Nrf2/Keap1 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8460290. [PMID: 31885821 PMCID: PMC6915017 DOI: 10.1155/2019/8460290] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/25/2019] [Accepted: 10/24/2019] [Indexed: 12/22/2022]
Abstract
Blast exposure is a worldwide public health concern, but most related research has been focused on direct injury. Thoracic blast exposure-induced neurotrauma is a type of indirect injuries where research is lacking. As CD28 stimulates T cell activation and survival and contributes to inflammation initiation, it may play a role in thoracic blast exposure-induced neurotrauma. However, it has not been investigated. To explore the effects of CD28 on thoracic blast exposure-induced brain injury and its potential molecular mechanisms, a mouse model of thoracic blast exposure-induced brain injury was established. Fifty C57BL/6 wild-type (WT) and fifty CD28 knockout (CD28-/-) mice were randomly divided into five groups (one control group and four model groups), with ten mice (from each of the two models) for each group. Lung and brain tissue and serum samples were collected at 12 h, 24 h, 48 h, and 1 week after thoracic blast exposure. Histopathological changes were detected by hematoxylin-eosin staining. The expressions of inflammatory-related factors were detected by ELISA. Oxidative stress in the brain tissue was evaluated by determining the generation of reactive oxygen species (ROS) and the expressions of thioredoxin (TRX), malondialdehyde (MDA), SOD-1, and SOD-2. Apoptosis in the brain tissue was evaluated by TUNEL staining and the levels of Bax, Bcl-xL, Bad, Cytochrome C, and caspase-3. In addition, proteins of related pathways were also studied by western blotting and immunofluorescence. We found that CD28 deficiency significantly reduced thoracic blast exposure-induced histopathological changes and decreased the levels of inflammatory-related factors, including IL-1β, TNF-α, and S100β. In the brain tissue, CD28 deficiency also significantly attenuated thoracic blast exposure-induced generation of ROS and expressions of MDA, TRX, SOD-1, and SOD-2; lowered the number of apoptotic cells and the expression of Bax, cleaved caspase-3, Cytochrome C, and Bad; and maintained Bcl-xL expression. Additionally, CD28 deficiency significantly ameliorated thoracic blast exposure-induced increases of p-PI3K and Keap1 and the decrease of Nrf2 expression in the brain. Our results indicate that CD28 deficiency has a protective effect on thoracic blast exposure-induced brain injury that might be associated with the PI3K/Nrf2/Keap1 signaling pathway.
Collapse
Affiliation(s)
- Peifang Cong
- 1College of Medicine and Biological Information Engineering, Northeastern University, No. 195, Chuangxin Road, Hunnan District, Shenyang l10016, China
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Changci Tong
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Ying Liu
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Lin Shi
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Xiuyun Shi
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Yan Zhao
- 3Institute of Metal Research, Chinese Academy of Sciences, No. 72, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Keshen Xiao
- 3Institute of Metal Research, Chinese Academy of Sciences, No. 72, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Hongxu Jin
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Yunen Liu
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Mingxiao Hou
- 1College of Medicine and Biological Information Engineering, Northeastern University, No. 195, Chuangxin Road, Hunnan District, Shenyang l10016, China
| |
Collapse
|
49
|
Zheng J, Li H, He L, Huang Y, Cai J, Chen L, Zhou C, Fu H, Lu T, Zhang Y, Yao J, Yang Y. Preconditioning of umbilical cord-derived mesenchymal stem cells by rapamycin increases cell migration and ameliorates liver ischaemia/reperfusion injury in mice via the CXCR4/CXCL12 axis. Cell Prolif 2018; 52:e12546. [PMID: 30537044 PMCID: PMC6496237 DOI: 10.1111/cpr.12546] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 08/11/2018] [Accepted: 08/13/2018] [Indexed: 12/18/2022] Open
Abstract
Objectives Transfusion of umbilical cord‐derived mesenchymal stem cells (UC‐MSCs) is a novel strategy for treatment of various liver diseases. However, the therapeutic effect of UC‐MSCs is limited because only a few UC‐MSCs migrate towards the damaged regions. In this study, we observed the effects of autophagy on the migration of UC‐MSCs in vitro and in a model of liver ischaemia/reperfusion (I/R) injury. Materials and Methods We investigated the effects of autophagy on the status of the cell, release of anti‐inflammatory factors and migration of UC‐MSCs in vitro. The therapeutic effects and in vivo migration of rapamycin‐preconditioned UC‐MSCs were observed in a C57/B6 mouse model of liver I/R injury. Results Induction of autophagy by rapamycin enhanced the ability of UC‐MSCs to migrate and release anti‐inflammatory cytokines as well as increased expression of CXCR4 without affecting cell viability. Inhibition of CXCR4 activation markedly decreased migration of these cells. In a mouse model of liver I/R injury, we found significantly upregulated expression of CXCR12 in the damaged liver. More rapamycin‐preconditioned UC‐MSCs migrated towards the ischaemic regions than 3‐methyladenine‐preconditioned or non‐preconditioned UC‐MSCs, leading to improvement in hepatic performance, pathological changes and levels of inflammatory cytokines. These effects were abolished by AMD3100. Conclusions Preconditioning of UC‐MSCs by rapamycin afforded increased protection against liver I/R injury by enhancing immunosuppression and strengthening the homing and migratory capacity of these cells via the CXCR4/CXCL12 axis.
Collapse
Affiliation(s)
- Jun Zheng
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Organ Transplantation Research Center of Guangdong Province, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hui Li
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Organ Transplantation Research Center of Guangdong Province, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liying He
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Yiming Huang
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Organ Transplantation Research Center of Guangdong Province, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jianye Cai
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Organ Transplantation Research Center of Guangdong Province, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liang Chen
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Organ Transplantation Research Center of Guangdong Province, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chaorong Zhou
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Organ Transplantation Research Center of Guangdong Province, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hongyuan Fu
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Organ Transplantation Research Center of Guangdong Province, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tongyu Lu
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Organ Transplantation Research Center of Guangdong Province, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yingcai Zhang
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Organ Transplantation Research Center of Guangdong Province, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jia Yao
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Organ Transplantation Research Center of Guangdong Province, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yang Yang
- Department of Hepatic Surgery and Liver Transplantation Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Organ Transplantation Research Center of Guangdong Province, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
50
|
Zhan C, Dai X, Shen G, Lu X, Wang X, Lu L, Qian X, Rao J. Preoperative short-term fasting protects liver injury in patients undergoing hepatectomy. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:449. [PMID: 30603637 DOI: 10.21037/atm.2018.10.64] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Our previous study demonstrated that preoperative short-term fasting attenuates mice hepatic ischemia/reperfusion injury (IRI), which greatly piqued our interest in verifying if fasting produces similar protective effects in patients undergoing hepatectomy. Methods Eighty patients with liver tumors were randomized into control (Ctrl, n=40, preoperative fasting for 6 h) or fasting group (Fasting, n=40, preoperative fasting for 24 h). Serum was collected at pre-operation (Pre-Op), post-operation 1 day (POD-1), post-operation 3 days (POD-3), and post-operation 7 days (POD-7). Liver tissue was removed from the resected specimen. Results Sixty-three patients were eventually enrolled, with 33 in Ctrl and 30 in Fasting group. Our data showed that 24 h fasting effectively attenuated elevated sALT and sAST levels after operation (P<0.05), but serum total bilirubin was significantly lower at only POD-3 (P<0.05); and serum albumin was not markedly different in either of the groups. Interestingly, 24 h fasting partially attenuates expression of pro-inflammatory cytokine (TNF-α) and improves oxidative stress (MDA and SOD). Our data further showed short-term fasting triggered Nrf2 signaling pathway. Conclusions This study demonstrates preoperative short-term fasting effectively improves clinical outcomes and markedly attenuates inflammatory responses and oxidative stress in patients undergoing hepatectomy, and Nrf2 signaling pathway may play a key role in fasting against inflammatory responses and oxidant stress.
Collapse
Affiliation(s)
- Chuanfei Zhan
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Xinzheng Dai
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Gefengqiang Shen
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Xu Lu
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Xuehao Wang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Ling Lu
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Xiaofeng Qian
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Jianhua Rao
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|