1
|
Rani N, Arya DS. Modulation of PPAR-γ/Nrf2 and AGE/RAGE signaling contributes to the chrysin cardioprotection against myocardial damage following ischemia/reperfusion in diabetic rats. J Pharm Pharmacol 2025; 77:794-804. [PMID: 39673242 DOI: 10.1093/jpp/rgae140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/15/2024] [Indexed: 12/16/2024]
Abstract
OBJECTIVE Advanced glycation end products/receptor for AGEs (AGE/RAGE) signaling has a well-established role in the etiology of diabetic-related cardiovascular disorders. The purpose of the study was to elucidate the role of chrysin, a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist, against ischemia/reperfusion (IR) injury in diabetic rats and its functional interaction with the AGE/RAGE signaling pathway. METHODS A single intraperitoneal injection of streptozotocin (STZ, 70 mg/kg) was administered to rats for induction of diabetes. Rats having blood glucose levels more than 300 mg/dl following a 72 hr STZ injection were classified as diabetic. PPAR-γ antagonist GW9662 (1 mg/kg, i.p.), chrysin (60 mg/kg, p.o.), or both were administered to diabetic rats for 4 weeks. On the 29th day, rats were given ischemia for 45 min and then reperfusion for 1 hr to induce myocardial infarction (MI). KEY FINDINGS Pretreatment with chrysin significantly improved hemodynamic status, ventricular functions, and cardiac injury markers in diabetic myocardium. Increased PPAR-γ/Nrf2 and decreased RAGE protein expressions were linked to this improvement. Chrysin pretreatment resulted in the upregulation of endogenous antioxidants and reduced TBARS levels. Moreover, chrysin significantly decreased inflammation and apoptosis in diabetic myocardium. CONCLUSION PPAR-γ/Nrf2 co-activation by chrysin ameliorated IR-induced MI in diabetic rats, possibly via modulating AGE/RAGE signaling.
Collapse
Affiliation(s)
- Neha Rani
- Department of Pharmacology, Kalpana Chawla Government Medical College, Karnal, Haryana, 132001, India
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Dharamvir Singh Arya
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| |
Collapse
|
2
|
Jiang J, Hu S, Hu K, Xiao L, Lin J, Chen Y, Zhang D, Ou Y, Zhang J, Yuan L, Wang W, Yu P. Novel impact of metal ion-induced cell death on diabetic cardiomyopathy pathogenesis and therapy. Apoptosis 2025; 30:1152-1181. [PMID: 40042744 DOI: 10.1007/s10495-025-02090-4] [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] [Accepted: 02/07/2025] [Indexed: 06/16/2025]
Abstract
Diabetes mellitus is a common chronic metabolic disease, with its prevalence escalating annually. Diabetic cardiomyopathy is a leading cause of mortality among diabetic patients, characterized by intricate metabolic disturbances and myocardial cell demise. Various forms of cellular death pathways including apoptosis, pyroptosis, autophagic cell death, necroptosis, ferroptosis, and entosis have been identified in diabetic cardiomyopathy. Inhibiting myocardial cell death pathways has shown promise in mitigating diabetic cardiomyopathy progression. However, there are still gaps in understanding the role of metal ions in diabetic cardiomyopathy pathogenesis. Recent research endeavors have found that iron, copper, zinc, calcium, manganese and other metal elements related to cell death play an intricate and critical role in the pathogenesis and progression of diabetic cardiomyopathy. Notably, many animal studies have shown that the development and progression of diabetic cardiomyopathy can be alleviated by inhibiting the cell death process induced by these metal ions. Therefore, we review the molecular mechanisms underlying the death of various metal ions and the potential pathophysiological roles they play in diabetic cardiomyopathy. In addition, the value of these metal ions in the treatment of diabetic cardiomyopathy is also described.
Collapse
Affiliation(s)
- Jingjing Jiang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, China
| | - Shengnan Hu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Kaibo Hu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Leyang Xiao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Jitao Lin
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, 999007, Hong Kong
| | - Yangliu Ou
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Linhui Yuan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Wenting Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Hainan University, Haikou, 570311, China.
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
3
|
Li R, Yan X, Zhao Y, Liu H, Wang J, Yuan Y, Li Q, Su J. Oxidative Stress Induced by Nuclear Factor Erythroid 2-Related Factor 2 (NRF2) Dysfunction Aggravates Chronic Inflammation Through the NAD +/SIRT3 Axis and Promotes Renal Injury in Diabetes. Antioxidants (Basel) 2025; 14:267. [PMID: 40227196 PMCID: PMC11939224 DOI: 10.3390/antiox14030267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 04/15/2025] Open
Abstract
Diabetic nephropathy (DN), one of the most common and severe microvascular complications of diabetes, significantly increases the risk of renal failure and cardiovascular events. A high-glucose environment can lead to mitochondrial dysfunction in macrophages, which, through remodeling of energy metabolism, mediates the polarization of a pro-inflammatory phenotype and contributes to the formation of a chronic inflammatory microenvironment. Recent studies have found that high-glucose stimulation induces dysregulation of the nuclear factor erythroid 2-related factor 2 (NRF2) redox pathway in macrophages, leading to the generation of oxidative stress (OS) that further drives chronic inflammation. Therefore, it is crucial to fully understand how OS affects macrophage phenotypes and functions following NRF2 inhibition. This review analyzes the role of OS induced by NRF2 dysfunction in the chronic inflammation of DN and explores the relationship between OS and macrophage mitochondrial energy metabolism through the NAD⁺/NADH-SIRT3 axis, providing new therapeutic targets for targeting OS to improve the inflammatory microenvironment and vascular damage in DN.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jing Su
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basical Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130012, China
| |
Collapse
|
4
|
Emil AB, Hassan NH, Ibrahim S, Hassanen EI, Eldin ZE, Ali SE. Propolis extract nanoparticles alleviate diabetes-induced reproductive dysfunction in male rats: antidiabetic, antioxidant, and steroidogenesis modulatory role. Sci Rep 2024; 14:30607. [PMID: 39715797 DOI: 10.1038/s41598-024-81949-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/02/2024] [Indexed: 12/25/2024] Open
Abstract
Diabetes can affect male fertility via oxidative stress and endocrine system disruption. Nanomedicine based on natural products is employed to address diabetes complications. The current study aims to investigate the potential beneficial effect of propolis extract nanoparticles against diabetes-induced testicular damage in male rats. Sixty male rats were randomly allocated to six groups (n = 10). The first group served as a control group. The second and third received propolis extract (Pr) and propolis extract nanoparticles (PrNPs). The fourth group is the diabetic group that received streptozotocin (STZ) (55 mg kg/bwt) single-dose i/p. The fifth and sixth groups are diabetic rats treated with Pr and PrNPs. Both Pr and PrNPs were received at a dose (100 mg/kg bwt) orally. After 60 days, animals were euthanized, then pancreatic and testicular tissues were collected for redox status evaluation, gene expression analysis, and histopathological examination. Also, hormonal analysis (Insulin, total testosterone, and luteinizing hormone (LH) ) along with semen quality evaluation were done. Results showed that the induction of diabetes led to testicular and pancreatic redox status deterioration showing a reduction in reduced glutathione (GSH) as well as elevation of malondialdehyde (MDA), and nitric oxide (NO) levels. Also, relative transcript levels of testicular cytochrome P450 family 11 subfamily A member 1 (CYP11A1), 3β-Hydroxysteroid dehydrogenase (HSD-3β), and nuclear factor (erythroid-derived 2)-like 2 (NFE2L2) were significantly down-regulated, While the advanced glycation end-product receptor (AGER) relative gene expression was significantly upregulated. Furthermore, hormonal and semen analysis disturbances were observed. Upon treatment with Pr and PrNPs, a marked upregulation of testicular gene expression of CYP11A1, HSD-3β, and NFE2L2 as well as a downregulation of AGER, was observed. Hormones and semen analysis were improved. In addition, the testicular and pancreatic redox status was enhanced. Results were confirmed via histopathological investigations. PrNPs outperformed Pr in terms of steroidogenesis pathway improvement, testicular antioxidant defense mechanism augmentation, and prospective antidiabetic activity.
Collapse
Affiliation(s)
- Abram B Emil
- Department of Physiology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Neven H Hassan
- Department of Physiology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| | - Sally Ibrahim
- Department of Animal Reproduction and AI, Veterinary Research Institute, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Eman I Hassanen
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Zienab E Eldin
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Science (PSAS), Beni-Suef, 62511, Egypt
| | - Sara E Ali
- Department of Physiology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| |
Collapse
|
5
|
Tabandeh MR, Davoodi E, Bayati V, Dayer D. Betaine regulates steroidogenesis, endoplasmic reticulum stress response and Nrf2/HO-1 antioxidant pathways in mouse Leydig cells under hyperglycaemia condition. Arch Physiol Biochem 2024; 130:768-778. [PMID: 37870938 DOI: 10.1080/13813455.2023.2272588] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 09/15/2023] [Indexed: 10/25/2023]
Abstract
We studied the effects of betaine on steroidogenesis, endoplasmic reticulum stress and Nrf2 antioxidant pathways of mice Leydig cells under hyperglycaemia conditions. Leydig cells were grown in low and high glucose concentrations (5 mM and 30 mM) in the presence of 5 mM of betaine for 24 h. Gene expression was determined using a real-time PCR method. The protein levels were determined by Western blot analysis. The testosterone production was evaluated by the ELISA method. Cellular contents of reduced and oxidised glutathione were measured by colorimetric method. Hyperglycaemia caused impaired steroidogenesis and ERS in Leydig cells associated with the down-regulation of 3β-HSD, StAR, P450scc, LH receptor and increased expression of GRP78, CHOP, ATF6 and IRE1. Betaine could improve cell viability, attenuate the ERS, and restore testosterone production in Leydig cells under hyperglycaemia conditions. Betaine can protect Leydig cells against the adverse effects of hyperglycaemia by regulating steroidogenesis, antioxidants, and ERS.
Collapse
Affiliation(s)
- Mohammad Reza Tabandeh
- Department of Basic Sciences, Division of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Stem Cells and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Elahe Davoodi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Vahid Bayati
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Anatomy, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Dian Dayer
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
6
|
Tian M, Huang X, Li M, Lou P, Ma H, Jiang X, Zhou Y, Liu Y. Ferroptosis in diabetic cardiomyopathy: from its mechanisms to therapeutic strategies. Front Endocrinol (Lausanne) 2024; 15:1421838. [PMID: 39588340 PMCID: PMC11586197 DOI: 10.3389/fendo.2024.1421838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/15/2024] [Indexed: 11/27/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is defined as structural and functional cardiac abnormalities in diabetes, and cardiomyocyte death is the terminal event of DCM. Ferroptosis is iron-dependent oxidative cell death. Evidence has indicated that iron overload and ferroptosis play important roles in the pathogenesis of DCM. Mitochondria, an important organelle in iron homeostasis and ROS production, play a crucial role in cardiomyocyte ferroptosis in diabetes. Studies have shown some anti-diabetic medicines, plant extracts, and ferroptosis inhibitors might improve DCM by alleviating ferroptosis. In this review, we systematically reviewed the evidence of ferroptosis in DCM. Anti-ferroptosis might be a promising therapeutic strategy for the treatment of DCM.
Collapse
Affiliation(s)
- Meimei Tian
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinli Huang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Min Li
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Pingping Lou
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huijie Ma
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, China
| | - Xinli Jiang
- Department of Ophthalmology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yaru Zhou
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yan Liu
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
7
|
Hashiesh HM, Azimullah S, Nagoor Meeran MF, Saraswathiamma D, Arunachalam S, Jha NK, Sadek B, Adeghate E, Sethi G, Albawardi A, Al Marzooqi S, Ojha S. Cannabinoid 2 Receptor Activation Protects against Diabetic Cardiomyopathy through Inhibition of AGE/RAGE-Induced Oxidative Stress, Fibrosis, and Inflammasome Activation. J Pharmacol Exp Ther 2024; 391:241-257. [PMID: 38955492 DOI: 10.1124/jpet.123.002037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
Oxidative stress, fibrosis, and inflammasome activation from advanced glycation end product (AGE)-receptor of advanced glycation end product (RAGE) interaction contribute to diabetic cardiomyopathy (DCM) formation and progression. Our study revealed the impact of β-caryophyllene (BCP) on activating cannabinoid type 2 receptors (CB2Rs) against diabetic complication, mainly cardiomyopathy and investigated the underlying cell signaling pathways in mice. The murine model of DCM was developed by feeding a high-fat diet with streptozotocin injections. After the development of diabetes, the animals received a 12-week oral BCP treatment at a dose of 50 mg/kg/body weight. BCP treatment showed significant improvement in glucose tolerance and insulin resistance and enhanced serum insulin levels in diabetic animals. BCP treatment effectively reversed the heart remodeling and restored the phosphorylated troponin I and sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a expression. Ultrastructural examination showed reduced myocardial cell injury in DCM mice treated with BCP. The preserved myocytes were found to be associated with reduced expression of AGE/RAGE in DCM mice hearts. BCP treatment mitigated oxidative stress by inhibiting expression of NADPH oxidase 4 and activating phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. Also, BCP suppressed cardiac fibrosis and endothelial-to-mesenchymal transition in DCM mice by inhibiting transforming growth factor β (TGF-β)/suppressor of mothers against decapentaplegic (Smad) signaling. Further, BCP treatment suppressed nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome activation in DCM mice and alleviated cellular injury to the pancreatic tissues evidenced by significant elevation of the number of insulin-positive cells. To demonstrate a CB2R-dependent mechanism of BCP, another group of DCM mice were pretreated with AM630, a CB2R antagonist. AM630 was observed to abrogate the beneficial effects of BCP in DCM mice. Taken together, BCP demonstrated the potential to protect the myocardium and pancreas of DCM mice mediating CB2R-dependent mechanisms. SIGNIFICANCE STATEMENT: BCP, a CB2R agonist, shows protection against DCM. BCP attenuates oxidative stress, inflammation, and fibrosis in DCM via activating CB2Rs. BCP mediating CB2R activation favorably modulates AGE/RAGE, PI3K/AKT/Nrf2β and TGF-β/Smad and (NLRP3) inflammasome in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Hebaallah Mamdouh Hashiesh
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Sheikh Azimullah
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Mohamed Fizur Nagoor Meeran
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Dhanya Saraswathiamma
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Seenipandi Arunachalam
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Niraj Kumar Jha
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Bassem Sadek
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Ernest Adeghate
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Gautam Sethi
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Alia Albawardi
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Saeeda Al Marzooqi
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Shreesh Ojha
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| |
Collapse
|
8
|
Li H, Bao L, Pan Y, Zhu X, Cheng J, Zhang J, Chu W. The role of miR-216a-mediated Nrf2 pathway in muscle oxidative stress of Siniperca chuatsi induced by cadmium. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116863. [PMID: 39128454 DOI: 10.1016/j.ecoenv.2024.116863] [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: 04/10/2024] [Revised: 08/01/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Cadmium (Cd) is a toxic heavy metal pollutant in the environment. Excessive Cd in water has toxic effects on fish, endangering their healthy growth and ultimately affecting the quality and safety of aquatic products. To evaluate the toxicity of excessive Cd to fish through potential oxidative damage, Siniperca chuatsi was exposed to Cd in water for 15 days. It was found that Cd exposure significantly decreased the survival rate of S. chuatsi and Cd was detected in their muscle. Meanwhile, Cd disrupts the redox balance by reducing antioxidant enzyme activities, increasing reactive oxygen species (ROS) and malondialdehyde (MDA) levels in muscle, and promoting oxidative damage. Histomorphology showed that enlargement of muscle fiber gaps, cell swelling and vacuolar degeneration after Cd exposure. In addition, Cd toxicity induced up-regulating the expression of miR-216a, while down-regulation of Nrf2 protein and its downstream antioxidant enzyme genes expression. Further analysis revealed that miR-216a was significantly negatively correlated with the expression of Nrf2, and injection of miR-216a antagomir significantly enhanced the expression of Nrf2 and antioxidant enzyme genes, as well as the activity of antioxidant enzymes, thereby reducing the damage of Cd to fish. These results suggested that miR-216a-mediated Nrf2 signaling pathway plays an important role in Cd-induced oxidative stress of S. chuatsi muscle.
Collapse
Affiliation(s)
- Honghui Li
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China.
| | - Lingsheng Bao
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China.
| | - Yaxiong Pan
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China
| | - Xin Zhu
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China
| | - Jia Cheng
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China
| | - Jianshe Zhang
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China
| | - Wuying Chu
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, PR China; Hunan Engineering Technology Research Center for Amphibian and Reptile Resource Protection and Product Processing, Changsha 410022, PR China.
| |
Collapse
|
9
|
Cheng PP, Wang XT, Liu Q, Hu YR, Dai ER, Zhang MH, Yang TS, Qu HY, Zhou H. Nrf2 mediated signaling axis in heart failure: Potential pharmacological receptor. Pharmacol Res 2024; 206:107268. [PMID: 38908614 DOI: 10.1016/j.phrs.2024.107268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
Heart failure (HF) has emerged as the most pressing health concerns globally, and extant clinical therapies are accompanied by side effects and patients have a high burden of financial. The protein products of nuclear factor erythroid 2-related factor 2 (Nrf2) target genes have a variety of cardioprotective effects, including antioxidant, metabolic functions and anti-inflammatory. By evaluating established preclinical and clinical research in HF to date, we explored the potential of Nrf2 to exert unique cardioprotective functions as a novel therapeutic receptor for HF. In this review, we generalize the progression, structure, and function of Nrf2 research in the cardiovascular system. The mechanism of action of Nrf2 involved in HF as well as agonists of Nrf2 in natural compounds are summarized. Additionally, we discuss the challenges and implications for future clinical translation and application of pharmacology targeting Nrf2. It's critical to developing new drugs for HF.
Collapse
Affiliation(s)
- Pei-Pei Cheng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xin-Ting Wang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qian Liu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi-Ran Hu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - En-Rui Dai
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ming-Hao Zhang
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tian-Shu Yang
- Department of Cardiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai 200071, China
| | - Hui-Yan Qu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
10
|
Visanji M, Venegas-Pino DE, Werstuck GH. Understanding One Half of the Sex Difference Equation: The Modulatory Effects of Testosterone on Diabetic Cardiomyopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:551-561. [PMID: 38061627 DOI: 10.1016/j.ajpath.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/31/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023]
Abstract
Diabetes is a prevalent disease, primarily characterized by high blood sugar (hyperglycemia). Significantly higher rates of myocardial dysfunction have been noted in individuals with diabetes, even in those without coronary artery disease or high blood pressure (hypertension). Numerous molecular mechanisms have been identified through which diabetes contributes to the pathology of diabetic cardiomyopathy, which presents as cardiac hypertrophy and fibrosis. At the cellular level, oxidative stress and inflammation in cardiomyocytes are triggered by hyperglycemia. Although males are generally more likely to develop cardiovascular disease than females, diabetic males are less likely to develop diabetic cardiomyopathy than are diabetic females. One reason for these differences may be the higher levels of serum testosterone in males compared with females. Although testosterone appears to protect against cardiomyocyte oxidative stress and exacerbate hypertrophy, its role in inflammation and fibrosis is much less clear. Additional preclinical and clinical studies will be required to delineate testosterone's effect on the diabetic heart.
Collapse
Affiliation(s)
- Mika'il Visanji
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | - Geoff H Werstuck
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
11
|
Bao J, Gao Z, Hu Y, Ye L, Wang L. Transient receptor potential vanilloid type 1: cardioprotective effects in diabetic models. Channels (Austin) 2023; 17:2281743. [PMID: 37983306 PMCID: PMC10761101 DOI: 10.1080/19336950.2023.2281743] [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: 05/05/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023] Open
Abstract
Cardiovascular disease, especially heart failure (HF) is the leading cause of death in patients with diabetes. Individuals with diabetes are prone to a special type of cardiomyopathy called diabetic cardiomyopathy (DCM), which cannot be explained by heart diseases such as hypertension or coronary artery disease, and can contribute to HF. Unfortunately, the current treatment strategy for diabetes-related cardiovascular complications is mainly to control blood glucose levels; nonetheless, the improvement of cardiac structure and function is not ideal. The transient receptor potential cation channel subfamily V member 1 (TRPV1), a nonselective cation channel, has been shown to be universally expressed in the cardiovascular system. Increasing evidence has shown that the activation of TRPV1 channel has a potential protective influence on the cardiovascular system. Numerous studies show that activating TRPV1 channels can improve the occurrence and progression of diabetes-related complications, including cardiomyopathy; however, the specific mechanisms and effects are unclear. In this review, we summarize that TRPV1 channel activation plays a protective role in the heart of diabetic models from oxidation/nitrification stress, mitochondrial function, endothelial function, inflammation, and cardiac energy metabolism to inhibit the occurrence and progression of DCM. Therefore, TRPV1 may become a latent target for the prevention and treatment of diabetes-induced cardiovascular complications.
Collapse
Affiliation(s)
- Jiaqi Bao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhicheng Gao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yilan Hu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lifang Ye
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lihong Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Cai L, Tan Y, Watson S, Wintergerst K. Diabetic cardiomyopathy - Zinc preventive and therapeutic potentials by its anti-oxidative stress and sensitizing insulin signaling pathways. Toxicol Appl Pharmacol 2023; 477:116694. [PMID: 37739320 PMCID: PMC10616760 DOI: 10.1016/j.taap.2023.116694] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Oxidative stress and insulin resistance are two key mechanisms for the development of diabetic cardiomyopathy (DCM, cardiac remodeling and dysfunction). In this review, we discussed how zinc and metallothionein (MT) protect the heart from type 1 or type 2 diabetes (T1D or T2D) through its anti-oxidative function and insulin-mediated PI3K/Akt signaling activation. Both T1D and T2D-induced DCM, shown by cardiac structural remodeling and dysfunction, in wild-type mice, but not in cardiomyocyte-specific overexpressing MT mice. In contrast, mice with global MT gene deletion were more susceptible to the development of DCM. When we used zinc to treat mice with either T1D or T2D, cardiac remodeling and dysfunction were significantly prevented along with increased cardiac MT expression. To support the role of zinc homeostasis in insulin signaling pathways, treatment of diabetic mice with zinc showed the preservation of phosphorylation levels of insulin-mediated glucose metabolism-related Akt2 and GSK-3β and even rescued cardiac pathogenesis induced by global deletion of Akt2 gene in a MT-dependent manner. These results suggest the protection by zinc from DCM is through both the induction of MT and sensitization of insulin signaling. Combined our own and other works, this review comprehensively summarized the roles of zinc homeostasis in the development and progression of DCM and its therapeutic implications. At the end, we provided pre-clinical and clinical evidence for the preventive and therapeutic potential of zinc supplementation through its anti-oxidative stress and sensitizing insulin signaling actions. Understanding the intricate connections between zinc and DCM provides insights for the future interventional approaches.
Collapse
Affiliation(s)
- Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America; Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, United States of America.
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America.
| | - Sara Watson
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, United States of America
| | - Kupper Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States of America; Wendy Novak Diabetes Institute, Norton Healthcare, Louisville, KY, United States of America; Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY, United States of America
| |
Collapse
|
13
|
ALTamimi JZ, AlFaris NA, Alshammari GM, Alagal RI, Aljabryn DH, Yahya MA. Esculeoside A Decreases Diabetic Cardiomyopathy in Streptozotocin-Treated Rats by Attenuating Oxidative Stress, Inflammation, Fibrosis, and Apoptosis: Impressive Role of Nrf2. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1830. [PMID: 37893548 PMCID: PMC10608477 DOI: 10.3390/medicina59101830] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/27/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: This experiment evaluated the preventative influence of the tomato-derived Esculeoside A (ESA) on diabetic cardiomyopathy in type 1 diabetes mellitus (T1DM) in rats induced by streptozotocin (STZ). It also examined whether the activation of Nrf2 signaling affords this protection. Materials and Methods: Adult male Wistar control nondiabetic rats and rats with T1DM (STZ-T1DM) were given either carboxymethylcellulose as a vehicle or ESA (100 mg/kg) (eight rats/group) orally daily for 12 weeks. A group of STZ-T1DM rats was also treated with 100 mg/kg ESA and co-treated i.p. with 2 mg/kg (twice/week), brusatol, and Nrf2 inhibitors for 12 weeks. Results and Conclusions: Treatment with ESA prevented the gain in heart weight and cardiomyocyte hypertrophy and improved the left ventricular (LV) systolic and diastolic function (LV) in the STZ-T1DM rat group. Likewise, it reduced their serum levels of triglycerides, cholesterol, and low-density lipoproteins (LDL-c), as well as their LV mRNA, cytoplasmic total, and nuclear total levels of NF-κB. ESA also reduced the total levels of malondialdehyde, tumor necrosis factor-α, interleukine-6 (IL-6), Bax, cytochrome-c, and caspase-3 in the LV of the STZ-T1DM rats. In parallel, ESA enhanced the nuclear and cytoplasmic levels of Nrf2 and the levels of superoxide dismutase, glutathione, and heme oxygenase-1, but decreased the mRNA and cytoplasmic levels of keap-1 in the LVs of the STZ-T1DM rats. Interestingly, ESA did not affect the fasting insulin and glucose levels of the diabetic rats. All of these beneficially protective effects of ESA were not seen in the ESA-treated rats that received brusatol. In conclusion, ESA represses diabetic cardiomyopathy in STZ-diabetic hearts by activating the Nrf2/antioxidant/NF-κB axis.
Collapse
Affiliation(s)
- Jozaa Z. ALTamimi
- Department of Physical Sports Sciences, College of Education, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia; (J.Z.A.); (D.H.A.)
| | - Nora A. AlFaris
- Department of Physical Sports Sciences, College of Education, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia; (J.Z.A.); (D.H.A.)
| | - Ghedeir M. Alshammari
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (G.M.A.); (M.A.Y.)
| | - Reham I. Alagal
- Department of Health Sciences, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Dalal H. Aljabryn
- Department of Physical Sports Sciences, College of Education, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia; (J.Z.A.); (D.H.A.)
| | - Mohammed Abdo Yahya
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (G.M.A.); (M.A.Y.)
| |
Collapse
|
14
|
Xi C, Pang J, Barrett A, Horuzsko A, Ande S, Mivechi NF, Zhu X. Nrf2 Drives Hepatocellular Carcinoma Progression through Acetyl-CoA-Mediated Metabolic and Epigenetic Regulatory Networks. Mol Cancer Res 2023; 21:1079-1092. [PMID: 37364049 PMCID: PMC10592407 DOI: 10.1158/1541-7786.mcr-22-0935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/17/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023]
Abstract
Correlations between the oxidative stress response and metabolic reprogramming have been observed during malignant tumor formation; however, the detailed mechanism remains elusive. The transcription factor Nrf2, a master regulator of the oxidative stress response, mediates metabolic reprogramming in multiple cancers. In a mouse model of hepatocellular carcinoma (HCC), through metabolic profiling, genome-wide gene expression, and chromatin structure analyses, we present new evidence showing that in addition to altering antioxidative stress response signaling, Nrf2 ablation impairs multiple metabolic pathways to reduce the generation of acetyl-CoA and suppress histone acetylation in tumors, but not in tumor-adjacent normal tissue. Nrf2 ablation and dysregulated histone acetylation impair transcription complex assembly on downstream target antioxidant and metabolic regulatory genes for expression regulation. Mechanistic studies indicate that the regulatory function of Nrf2 is low glucose dependent, the effect of which is demolished under energy refeeding. Together, our results implicate an unexpected effect of Nrf2 on acetyl-CoA generation, in addition to its classic antioxidative stress response regulatory activity, integrates metabolic and epigenetic programs to drive HCC progression. IMPLICATIONS This study highlights that Nrf2 integrates metabolic and epigenetic regulatory networks to dictate tumor progression and that Nrf2 targeting is therapeutically exploitable in HCC treatment.
Collapse
Affiliation(s)
- Caixia Xi
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
- Molecular Chaperone Biology, Medical College of Georgia, Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Junfeng Pang
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
- Molecular Chaperone Biology, Medical College of Georgia, Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Amanda Barrett
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | | | | | - Nahid F. Mivechi
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
- Molecular Chaperone Biology, Medical College of Georgia, Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
- Department of Radiation Oncology, Augusta University, Augusta, GA 30912, USA
| | - Xingguo Zhu
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
- Department of Pediatrics, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
15
|
Wang L, Chen P, Pan Y, Wang Z, Xu J, Wu X, Yang Q, Long M, Liu S, Huang W, Ou C, Wu Y. Injectable photocurable Janus hydrogel delivering hiPSC cardiomyocyte-derived exosome for post-heart surgery adhesion reduction. SCIENCE ADVANCES 2023; 9:eadh1753. [PMID: 37540739 PMCID: PMC10403204 DOI: 10.1126/sciadv.adh1753] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 07/06/2023] [Indexed: 08/06/2023]
Abstract
Postsurgical pericardial adhesions pose increased risks of sequelae, prolonged reoperation time, and reduced visibility in the surgical field. Here, we introduce an injectable Janus hydrogel, which exhibits asymmetric adhesiveness properties after photocrosslinking, sustained delivering induced pluripotent stem cell-derived cardiomyocyte exosomes (iCM-EXOs) for post-heart surgery adhesion reduction. Our findings reveal that iCM-EXOs effectively attenuate oxidative stress in hydrogen peroxide-treated primary cardiomyocytes by inhibiting the activation of the transcription factor nuclear factor erythroid 2-related factor 2. Notably, in rat cardiac postsurgery models, the Janus hydrogels loaded with iCM-EXOs demonstrate dual functionality, acting as antioxidants and antipericardial adhesion agents. These hydrogels effectively protect iCM-EXOs from GATA6+ cavity macrophage clearance by inhibiting the recruitment of macrophages from the thoracic cavity. These results highlight the promising potential of iCM-EXO-laden Janus hydrogels for clinical safety and efficacy validation in trials involving heart surgery patients, with the ultimate goal of routine administration during open-heart surgeries.
Collapse
Affiliation(s)
- Ling Wang
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Peier Chen
- Affiliated Dongguan Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan 523058, China
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuxuan Pan
- Affiliated Dongguan Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan 523058, China
| | - Zihan Wang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jie Xu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Xiaoqi Wu
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Qiao Yang
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Meng Long
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sitian Liu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wenhua Huang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Caiwen Ou
- Affiliated Dongguan Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan 523058, China
| | - Yaobin Wu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
16
|
Thiruvengadam R, Venkidasamy B, Samynathan R, Govindasamy R, Thiruvengadam M, Kim JH. Association of nanoparticles and Nrf2 with various oxidative stress-mediated diseases. Chem Biol Interact 2023; 380:110535. [PMID: 37187268 DOI: 10.1016/j.cbi.2023.110535] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/18/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that regultes the cellular antioxidant defense system at the posttranscriptional level. During oxidative stress, Nrf2 is released from its negative regulator Kelch-like ECH-associated protein 1 (Keap1) and binds to antioxidant response element (ARE) to transcribe antioxidative metabolizing/detoxifying genes. Various transcription factors like aryl hydrocarbon receptor (AhR) and nuclear factor kappa light chain enhancer of activated B cells (NF-kB) and epigenetic modification including DNA methylation and histone methylation might also regulate the expression of Nrf2. Despite its protective role, Keap1/Nrf2/ARE signaling is considered as a pharmacological target due to its involvement in various pathophysiological conditions such as diabetes, cardiovascular disease, cancer, neurodegenerative diseases, hepatotoxicity and kidney disorders. Recently, nanomaterials have received a lot of attention due to their unique physiochemical properties and are also used in various biological applications, for example, biosensors, drug delivery systems, cancer therapy, etc. In this review, we will be discussing the functions of nanoparticles and Nrf2 as a combined therapy or sensitizing agent and their significance in various diseases such as diabetes, cancer and oxidative stress-mediated diseases.
Collapse
Affiliation(s)
- Rekha Thiruvengadam
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul, 05006, Republic of Korea
| | - Baskar Venkidasamy
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 600077, India
| | - Ramkumar Samynathan
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 600077, India
| | - Rajakumar Govindasamy
- Department of Periodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 600077, India
| | - Muthu Thiruvengadam
- Department of Applied Bioscience, College of Life and Environmental Sciences, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul, 05006, Republic of Korea.
| |
Collapse
|
17
|
Jiang B, Zhou X, Yang T, Wang L, Feng L, Wang Z, Xu J, Jing W, Wang T, Su H, Yang G, Zhang Z. The role of autophagy in cardiovascular disease: Cross-interference of signaling pathways and underlying therapeutic targets. Front Cardiovasc Med 2023; 10:1088575. [PMID: 37063954 PMCID: PMC10090687 DOI: 10.3389/fcvm.2023.1088575] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
Autophagy is a conserved lysosomal pathway for the degradation of cytoplasmic proteins and organelles, which realizes the metabolic needs of cells and the renewal of organelles. Autophagy-related genes (ATGs) are the main molecular mechanisms controlling autophagy, and their functions can coordinate the whole autophagic process. Autophagy can also play a role in cardiovascular disease through several key signaling pathways, including PI3K/Akt/mTOR, IGF/EGF, AMPK/mTOR, MAPKs, p53, Nrf2/p62, Wnt/β-catenin and NF-κB pathways. In this paper, we reviewed the signaling pathway of cross-interference between autophagy and cardiovascular diseases, and analyzed the development status of novel cardiovascular disease treatment by targeting the core molecular mechanism of autophagy as well as the critical signaling pathway. Induction or inhibition of autophagy through molecular mechanisms and signaling pathways can provide therapeutic benefits for patients. Meanwhile, we hope to provide a unique insight into cardiovascular treatment strategies by understanding the molecular mechanism and signaling pathway of crosstalk between autophagy and cardiovascular diseases.
Collapse
Affiliation(s)
- Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xuan Zhou
- Department of First Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tao Yang
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linlin Wang
- Department of First Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Longfei Feng
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zheng Wang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Jin Xu
- Department of First Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tao Wang
- Research Center for Translational Medicine, Gansu Province Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Haixiang Su
- Research Center for Translational Medicine, Gansu Province Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - GuoWei Yang
- Center for Heart, First Hospital of Lanzhou University, Lanzhou, China
| | - Zheng Zhang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Center for Heart, First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
18
|
LncRNA MHRT Prevents Angiotensin II-Induced Myocardial Oxidative Stress and NLRP3 Inflammasome via Nrf2 Activation. Antioxidants (Basel) 2023; 12:antiox12030672. [PMID: 36978920 PMCID: PMC10044972 DOI: 10.3390/antiox12030672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/19/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
The development of angiotensin II (Ang II)-induced cardiomyopathies is reportedly mediated via oxidative stress and inflammation. Nuclear factor erythroid 2-related factor (Nrf2) is an important regulator of cellular antioxidant defense, and reactive oxygen species (ROS) can activate the NLRP3 inflammasome. MHRT is a newly discovered lncRNA exhibiting cardioprotective effects, demonstrated by inhibiting myocardial hypertrophy via Brg1 and myocardial apoptosis via Nrf2 upregulation. However, the underlying mechanism of MHRT remains unclear. We explored the potential protective effects of MHRT against Ang II-induced myocardial oxidative stress and NLRP3-mediated inflammation by targeting Nrf2. Chronic Ang II administration induced NLRP3 inflammasome activation (increased NLRP3, caspase-1 and interleukin-1β expression), oxidative stress (increased 3-nitrotyrosine and 4-hydroxy-2-nonenal), cardiac dysfunction and decreased MHRT and Nrf2 expression. Lentivirus-mediated MHRT overexpression inhibited Ang II (100 nM)-induced oxidative stress and NLRP3 inflammasome activation in AC16 human cardiomyocyte cells. Mechanistically, MHRT overexpression upregulated the expression and function of Nrf2, as determined by the increased transcription of downstream genes HO-1 and CAT, subsequently decreasing intracellular ROS accumulation and inhibiting the expression of thioredoxin-interacting protein (NLRP3 activator) and its direct binding to NLRP3. Accordingly, MHRT could protect against Ang II-induced myocardial injury by decreasing oxidative stress and NLRP3 inflammasome activation via Nrf2 activation.
Collapse
|
19
|
Liu M, Sun X, Chen B, Dai R, Xi Z, Xu H. Insights into Manganese Superoxide Dismutase and Human Diseases. Int J Mol Sci 2022; 23:ijms232415893. [PMID: 36555531 PMCID: PMC9786916 DOI: 10.3390/ijms232415893] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Redox equilibria and the modulation of redox signalling play crucial roles in physiological processes. Overproduction of reactive oxygen species (ROS) disrupts the body's antioxidant defence, compromising redox homeostasis and increasing oxidative stress, leading to the development of several diseases. Manganese superoxide dismutase (MnSOD) is a principal antioxidant enzyme that protects cells from oxidative damage by converting superoxide anion radicals to hydrogen peroxide and oxygen in mitochondria. Systematic studies have demonstrated that MnSOD plays an indispensable role in multiple diseases. This review focuses on preclinical evidence that describes the mechanisms of MnSOD in diseases accompanied with an imbalanced redox status, including fibrotic diseases, inflammation, diabetes, vascular diseases, neurodegenerative diseases, and cancer. The potential therapeutic effects of MnSOD activators and MnSOD mimetics are also discussed. Targeting this specific superoxide anion radical scavenger may be a clinically beneficial strategy, and understanding the therapeutic role of MnSOD may provide a positive insight into preventing and treating related diseases.
Collapse
Affiliation(s)
- Mengfan Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Xueyang Sun
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Boya Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Rongchen Dai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
- Correspondence: (Z.X.); (H.X.)
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
- Correspondence: (Z.X.); (H.X.)
| |
Collapse
|
20
|
Kim YK, Ning X, Munir KM, Davis SN. Emerging drugs for the treatment of diabetic nephropathy. Expert Opin Emerg Drugs 2022; 27:417-430. [PMID: 36472144 DOI: 10.1080/14728214.2022.2155632] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Diabetic nephropathy remains a significant economic and social burden on both the individual patient and health-care systems as the prevalence of diabetes increases in the general population. The complex pathophysiology of diabetic kidney disease poses a challenge in the development of effective medical treatments for the disease. However, the multiple facets of diabetic nephropathy also offer a variety of potential strategies to manage this condition. AREAS COVERED We retrieved PubMed, Cochrane Library, Scopus, Google Scholar, and ClinicalTrials.gov records to identify studies and articles focused on new pharmacologic advances to treat diabetic nephropathy. EXPERT OPINION RAAS blockers have remained the mainstay of therapy for DM nephropathy for many years, with only recent advancements with SGLT2 inhibitors and nonsteroidal MRAs. Better understanding of the long-term renal effects of ambient hyperglycemia, ranging from hemodynamic changes to increased production of oxidative and pro-inflammatory substances, has evolved our approach to the treatment of diabetic nephropathy. With continuing research for new therapeutics as well as combination therapy, the medical community may be able to better ease the burden of diabetic kidney disease.
Collapse
Affiliation(s)
- Yoon Kook Kim
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Center for Diabetes and Endocrinology, Baltimore, MD, USA
| | - Xinyuan Ning
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Center for Diabetes and Endocrinology, Baltimore, MD, USA
| | - Kashif M Munir
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Center for Diabetes and Endocrinology, Baltimore, MD, USA
| | - Stephen N Davis
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
21
|
Yosri H, El-Kashef DH, El-Sherbiny M, Said E, Salem HA. Calycosin modulates NLRP3 and TXNIP-mediated pyroptotic signaling and attenuates diabetic nephropathy progression in diabetic rats; An insight. Biomed Pharmacother 2022; 155:113758. [DOI: 10.1016/j.biopha.2022.113758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/27/2022] Open
|
22
|
Wang H, Tian Y, Zhang Q, Liu W, Meng L, Jiang X, Xin Y. Essential role of Nrf2 in sulforaphane-induced protection against angiotensin II-induced aortic injury. Life Sci 2022; 306:120780. [PMID: 35839861 DOI: 10.1016/j.lfs.2022.120780] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022]
Abstract
AIMS Cardiovascular disease (CVD) is the leading cause of death worldwide. Inflammation and oxidative stress are the primary factors underlying angiotensin II (Ang II)-induced aortic damage. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important antioxidative stress factor. Sulforaphane (SFN), which is naturally found in cruciferous vegetables, is an Nrf2 agonist that is safe for oral administration. Here, we aimed to explore the potential of SFN in protecting against Ang II-induced aortic damage by upregulating Nrf2 expression via the extracellular signal-regulated kinase (ERK)/glycogen synthase kinase-3 beta (GSK-3β)/Fyn pathway. MAIN METHODS AND KEY FINDINGS Wild-type (WT) C57BL/6J and Nrf2-knockout (Nrf2-KO) mice were injected with Ang II to induce aortic inflammation, oxidative stress, and cardiac remodeling (increased fibrosis and wall thickness). SFN treatment prevented aortic damage via Nrf2 activation in the WT mice. However, the protective effect of SFN on Ang II-induced aortic damage and upregulation of genes downstream of Nrf2 were not observed in Nrf2-KO mice. SFN induced the upregulation of aortic Nrf2 and inhibited the accumulation of ERK, GSK-3β, and Fyn in the nuclei. SIGNIFICANCE These results revealed that Nrf2 plays a central role in protecting against Ang II-induced aortic injury. Furthermore, SFN prevented Ang II-induced aortic damage by activating Nrf2 through the ERK/GSK-3β/Fyn pathway.
Collapse
Affiliation(s)
- Huanhuan Wang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Yuan Tian
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China; Department of Gynecology, The Second Hospital of Jilin University, Changchun 130041, China.
| | - Qihe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Wenyun Liu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA.
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| |
Collapse
|
23
|
Nasehi L, Morassaei B, Ghaffari M, Sharafi A, Dehpour AR, Hosseini MJ. The impacts of vorinostat on NADPH oxidase and mitochondrial biogenesis gene expression in the heart of mice model of depression. Can J Physiol Pharmacol 2022; 100:1077-1085. [PMID: 36166834 DOI: 10.1139/cjpp-2022-0098] [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: 11/22/2022]
Abstract
The comorbidity of depression and high risk of cardiovascular diseases (CVD) have been reported as major health problems. Our previous study confirmed that fluoxetine (FLX) therapy had a significant influence on brain function but not on the heart in depression. In the present study, suberoyanilide hydroxamic acid (SAHA) was proposed as another therapeutic candidate for treatment of depression comorbid CVD in maternal separation model, following behavioral analyses and gene expression level in the heart. Our data demonstrated that SAHA significantly attenuates the NOX-4 gene expression level in treated mice with SAHA and FLX without significant change in NOX-2 expression level. SAHA decreased the gene expression level of peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α) and nuclear respiratory factors (Nrf2) in heart tissues of maternally separated mice. It supposed that non-effectiveness of FLX on mitochondrial biogenesis and NOX gene expression level in the heart of depressed patient can be related to recurrence of depression. It revealed that SAHA not only reversed the depressive-like behavior similar to our previous data but also recovered the heart mitochondrial function via effect on NOX-2, NOX-4, and mitochondrial biogenesis genes' (PGC-1α, Nrf-2, and peroxisome proliferator-activated receptor-α (PPAR-α)) expression levels. We suggest performing more studies to confirm SAHA as a therapeutic candidate in depression comorbid CVD.
Collapse
Affiliation(s)
- Leila Nasehi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Zanjan University of Medical sciences, Zanjan, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Bahareh Morassaei
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical sciences, Zanjan, Iran
| | - Maryam Ghaffari
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical sciences, Zanjan, Iran
| | - Ali Sharafi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical sciences, Zanjan, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Zanjan University of Medical sciences, Zanjan, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mir-Jamal Hosseini
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical sciences, Zanjan, Iran
| |
Collapse
|
24
|
Grün B, Tirre M, Pyschny S, Singh V, Kehl HG, Jux C, Drenckhahn JD. Inhibition of mitochondrial respiration has fundamentally different effects on proliferation, cell survival and stress response in immature versus differentiated cardiomyocyte cell lines. Front Cell Dev Biol 2022; 10:1011639. [PMID: 36211452 PMCID: PMC9538794 DOI: 10.3389/fcell.2022.1011639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022] Open
Abstract
Myocardial tissue homeostasis is critically important for heart development, growth and function throughout the life course. The loss of cardiomyocytes under pathological conditions ultimately leads to cardiovascular disease due to the limited regenerative capacity of the postnatal mammalian heart. Inhibition of electron transport along the mitochondrial respiratory chain causes cellular stress characterized by ATP depletion as well as excessive generation of reactive oxygen species. Adult cardiomyocytes are highly susceptible to mitochondrial dysfunction whereas embryonic cardiomyocytes in the mouse heart have been shown to be resistant towards mitochondrial complex III inhibition. To functionally characterize the molecular mechanisms mediating this stress tolerance, we used H9c2 cells as an in vitro model for immature cardiomyoblasts and treated them with various inhibitors of mitochondrial respiration. The complex I inhibitor rotenone rapidly induced cell cycle arrest and apoptosis whereas the complex III inhibitor antimycin A (AMA) had no effect on proliferation and only mildly increased cell death. HL-1 cells, a differentiated and contractile cardiomyocyte cell line from mouse atrium, were highly susceptible to AMA treatment evident by cell cycle arrest and death. AMA induced various stress response mechanisms in H9c2 cells, such as the mitochondrial unfolded protein response (UPRmt), integrated stress response (ISR), heat shock response (HSR) and antioxidative defense. Inhibition of the UPR, ISR and HSR by siRNA mediated knock down of key components does not impair growth of H9c2 cells upon AMA treatment. In contrast, knock down of NRF2, an important transcriptional regulator of genes involved in detoxification of reactive oxygen species, reduces growth of H9c2 cells upon AMA treatment. Various approaches to activate cell protective mechanisms and alleviate oxidative stress in HL-1 cells failed to rescue them from AMA induced growth arrest and death. In summary, these data show that the site of electron transport interruption along the mitochondrial respiratory chain determines cell fate in immature cardiomyoblasts. The study furthermore points to fundamental differences in stress tolerance and cell survival between immature and differentiated cardiomyocytes which may underlie the growth plasticity of embryonic cardiomyocytes during heart development but also highlight the obstacles of cardioprotective therapies in the adult heart.
Collapse
Affiliation(s)
- Bent Grün
- Department of Pediatric Cardiology, University Hospital Münster, Münster, Germany
| | - Michaela Tirre
- Department of Pediatric Cardiology, University Hospital Münster, Münster, Germany
| | - Simon Pyschny
- Department of Pediatric Cardiology, University Hospital Münster, Münster, Germany
| | - Vijay Singh
- Department of Pediatric Hematology and Oncology, Justus Liebig University, Gießen, Germany
| | - Hans-Gerd Kehl
- Department of Pediatric Cardiology, University Hospital Münster, Münster, Germany
| | - Christian Jux
- Department of Pediatric Cardiology, University Hospital Münster, Münster, Germany
- Department of Pediatric Cardiology, Justus Liebig University, Gießen, Germany
| | - Jörg-Detlef Drenckhahn
- Department of Pediatric Cardiology, University Hospital Münster, Münster, Germany
- Department of Pediatric Cardiology, Justus Liebig University, Gießen, Germany
- *Correspondence: Jörg-Detlef Drenckhahn,
| |
Collapse
|
25
|
Wu PY, Lai SY, Su YT, Yang KC, Chau YP, Don MJ, Lu KH, Shy HT, Lai SM, Kung HN. β-Lapachone, an NQO1 activator, alleviates diabetic cardiomyopathy by regulating antioxidant ability and mitochondrial function. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154255. [PMID: 35738116 DOI: 10.1016/j.phymed.2022.154255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 05/29/2022] [Accepted: 06/06/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Diabetic cardiomyopathy (DC) is one of the major lethal complications in patients with diabetes mellitus (DM); however, no specific strategy for preventing or treating DC has been identified. PURPOSE This study aimed to investigate the effects of β-lapachone (Lap), a natural compound that increases antioxidant activity in various tissues, on DC and explore the underlying mechanisms. STUDY DESIGN AND METHODS As an in vivo model, C57BL/6 mice were fed with the high-fat diet (HF) for 10 weeks to induce type 2 DM. Mice were fed Lap with the HF or after 5 weeks of HF treatment to investigate the protective effects of Lap against DC. RESULTS In the two in vivo models, Lap decreased heart weight, increased heart function, reduced oxidative stress, and elevated mitochondrial content under the HF. In the in vitro model, palmitic acid (PA) was used to mimic the effects of an HF on the differentiated-cardiomyoblast cell line H9c2. The results demonstrated that Lap reduced PA-induced ROS production by increasing the expression of antioxidant regulators and enzymes, inhibiting inflammation, increasing mitochondrial activity, and thus reducing cell damage. Via the use of specific inhibitors and siRNA, the protective effects of Lap were determined to be mediated mainly by NQO1, Sirt1 and mitochondrial activity. CONCLUSION Heart damage in DM is usually caused by excessive oxidative stress. This study showed that Lap can protect the heart from DC by upregulating antioxidant ability and mitochondrial activity in cardiomyocytes. Lap has the potential to serve as a novel therapeutic agent for both the prevention and treatment of DC.
Collapse
Affiliation(s)
- Pei-Yu Wu
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University
| | - Shin-Yu Lai
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University
| | - Yi-Ting Su
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University
| | - Kai-Chien Yang
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University
| | | | | | - Kai-Hsi Lu
- Department of Medical Research and Education, Cheng-Hsin General Hospital
| | - Horng-Tzer Shy
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University
| | - Shu-Mei Lai
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University
| | - Hsiu-Ni Kung
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University.
| |
Collapse
|
26
|
Sulforaphane inhibits angiotensin II-induced cardiomyocyte apoptosis by acetylation modification of Nrf2. Aging (Albany NY) 2022; 14:6740-6755. [PMID: 36006435 PMCID: PMC9467410 DOI: 10.18632/aging.204247] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/15/2022] [Indexed: 12/14/2022]
Abstract
Oxidative stress is the central cause of angiotensin II (Ang II)-induced myocardial injury, and nuclear factor erythroid 2-related factor (Nrf2) is the core molecule of the anti-oxidant defense system. We have previously demonstrated that sulforaphane (SFN) can prevent Ang II-induced myocardial injury by activating Nrf2; however, the underlying molecular mechanism is still unclear. This study aimed to evaluate whether SFN prevents Ang II-induced cardiomyocyte apoptosis through acetylation modification of <i>Nrf2</i>. Wild-type and <i>Nrf2</i> knockdown embryonic rat cardiomyocytes (H9C2) were exposed to Ang II to induce apoptosis, oxidative stress, and inflammatory responses. SFN treatment significantly reduced Ang II-induced cardiomyocyte apoptosis, inflammation and oxidative stress. Activation of Nrf2 played a critical role in preventing cardiomyocyte apoptosis. After Nrf2 was knockdown, the anti-inflammatory, antioxidant stress of SFN were eliminated. Furthermore, Nrf2 activation by SFN was closely related to the decreased activity of histone deacetylases (HDACs) and increased histone-3 (H3) acetylation levels in <i>Nrf2</i> promoter region. These findings confirm that Nrf2 plays a key role in SFN preventing Ang II-induced cardiomyocyte apoptosis. SFN activates Nrf2 by inhibiting HDACs expression and activation.
Collapse
|
27
|
Mao Y, Han CY, Hao L, Lee Y, Son JB, Choi H, Lee MR, Yang JD, Hong SK, Suh KS, Yu HC, Kim ND, Bae EJ, Park BH. p21-activated kinase 4 inhibition protects against liver ischemia/reperfusion injury: Role of nuclear factor erythroid 2-related factor 2 phosphorylation. Hepatology 2022; 76:345-356. [PMID: 35108418 DOI: 10.1002/hep.32384] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND AIMS p21-activated kinase 4 (PAK4), an oncogenic protein, has emerged as a promising target for anticancer drug development. Its role in oxidative stress conditions, however, remains elusive. We investigated the effects of PAK4 signaling on hepatic ischemia/reperfusion (I/R) injury. APPROACH AND RESULTS Hepatocyte- and myeloid-specific Pak4 knockout (KO) mice and their littermate controls were subjected to a partial hepatic I/R (HIR) injury. We manipulated the catalytic activity of PAK4, either through genetic engineering (gene knockout, overexpression of wild-type [WT] or dominant-negative kinase) or pharmacological inhibitor, coupled with a readout of nuclear factor erythroid 2-related factor 2 (Nrf2) activity, to test the potential function of PAK4 on HIR injury. PAK4 expression was markedly up-regulated in liver during HIR injury in mice and humans. Deletion of PAK4 in hepatocytes, but not in myeloid cells, ameliorated liver damages, as demonstrated in the decrease in hepatocellular necrosis and inflammatory responses. Conversely, the forced expression of WT PAK4 aggravated the pathological changes. PAK4 directly phosphorylated Nrf2 at T369, and it led to its nuclear export and proteasomal degradation, all of which impaired antioxidant responses in hepatocytes. Nrf2 silencing in liver abolished the protective effects of PAK4 deficiency. A PAK4 inhibitor protected mice from HIR injury. CONCLUSIONS PAK4 phosphorylates Nrf2 and suppresses its transcriptional activity. Genetic or pharmacological suppression of PAK4 alleviates HIR injury. Thus, PAK4 inhibition may represent a promising intervention against I/R-induced liver injury.
Collapse
Affiliation(s)
- Yuancheng Mao
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Chang Yeob Han
- School of Pharmacy, Jeonbuk National University, Jeonju, Republic of Korea
| | - Lihua Hao
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | | | | | | | - Mi Rin Lee
- Department of Surgery, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Jae Do Yang
- Department of Surgery, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Suk Kyun Hong
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Suk Suh
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hee Chul Yu
- Department of Surgery, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | | | - Eun Ju Bae
- School of Pharmacy, Jeonbuk National University, Jeonju, Republic of Korea
| | - Byung-Hyun Park
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| |
Collapse
|
28
|
Li J, Cheng Y, Li R, Wu X, Zheng C, Shiu PHT, Chan JCK, Rangsinth P, Liu C, Leung SWS, Lee SMY, Zhang C, Fu C, Zhang J, Cheung TMY, Leung GPH. Protective Effects of Amauroderma rugosum on Doxorubicin-Induced Cardiotoxicity through Suppressing Oxidative Stress, Mitochondrial Dysfunction, Apoptosis, and Activating Akt/mTOR and Nrf2/HO-1 Signaling Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9266178. [PMID: 35693699 PMCID: PMC9177334 DOI: 10.1155/2022/9266178] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/12/2022] [Accepted: 05/14/2022] [Indexed: 12/15/2022]
Abstract
Clinical outcomes for doxorubicin (Dox) are limited by its cardiotoxicity but a combination of Dox and agents with cardioprotective activities is an effective strategy to improve its therapeutic outcome. Natural products provide abundant resources to search for novel cardioprotective agents. Ganoderma lucidum (GL) is the most well-known edible mushroom within the Ganodermataceae family. It is commonly used in traditional Chinese medicine or as a healthcare product. Amauroderma rugosum (AR) is another genus of mushroom from the Ganodermataceae family, but its pharmacological activity and medicinal value have rarely been reported. In the present study, the cardioprotective effects of the AR water extract against Dox-induced cardiotoxicity were studied in vitro and in vivo. Results showed that both the AR and GL extracts could potentiate the anticancer effect of Dox. The AR extract significantly decreased the oxidative stress, mitochondrial dysfunction, and apoptosis seen in Dox-treated H9c2 rat cardiomyocytes. However, knockdown of Nrf2 by siRNA abolished the protective effects of AR in these cells. In addition, Dox upregulated the expression of proapoptotic proteins and downregulated the Akt/mTOR and Nrf2/HO-1 signaling pathways, and these effects could be reversed by the AR extract. Consistently, the AR extract significantly prolonged survival time, reversed weight loss, and reduced cardiac dysfunction in Dox-treated mice. In addition, oxidative stress and apoptosis were suppressed, while Nrf2 and HO-1 expressions were elevated in the heart tissues of Dox-treated mice after treatment with the AR extract. However, the GL extract had less cardioprotective effect against Dox in both the cell and animal models. In conclusion, the AR water extract demonstrated a remarkable cardioprotective effect against Dox-induced cardiotoxicity. One of the possible mechanisms for this effect was the upregulation of the mTOR/Akt and Nrf2/HO-1-dependent pathways, which may reduce oxidative stress, mitochondrial dysfunction, and cardiomyocyte apoptosis. These findings suggested that AR may be beneficial for the heart, especially in patients receiving Dox-based chemotherapy.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Yanfen Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Renkai Li
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiaoping Wu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chengwen Zheng
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Polly Ho-Ting Shiu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jacqueline Cho-Ki Chan
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Panthakarn Rangsinth
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Conghui Liu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Susan Wai-Sum Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Chen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
29
|
Role of Oxidative Stress in Diabetic Cardiomyopathy. Antioxidants (Basel) 2022; 11:antiox11040784. [PMID: 35453469 PMCID: PMC9030255 DOI: 10.3390/antiox11040784] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/18/2022] [Accepted: 04/12/2022] [Indexed: 02/04/2023] Open
Abstract
Type 2 diabetes is a redox disease. Oxidative stress and chronic inflammation induce a switch of metabolic homeostatic set points, leading to glucose intolerance. Several diabetes-specific mechanisms contribute to prominent oxidative distress in the heart, resulting in the development of diabetic cardiomyopathy. Mitochondrial overproduction of reactive oxygen species in diabetic subjects is not only caused by intracellular hyperglycemia in the microvasculature but is also the result of increased fatty oxidation and lipotoxicity in cardiomyocytes. Mitochondrial overproduction of superoxide anion radicals induces, via inhibition of glyceraldehyde 3-phosphate dehydrogenase, an increased polyol pathway flux, increased formation of advanced glycation end-products (AGE) and activation of the receptor for AGE (RAGE), activation of protein kinase C isoforms, and an increased hexosamine pathway flux. These pathways not only directly contribute to diabetic cardiomyopathy but are themselves a source of additional reactive oxygen species. Reactive oxygen species and oxidative distress lead to cell dysfunction and cellular injury not only via protein oxidation, lipid peroxidation, DNA damage, and oxidative changes in microRNAs but also via activation of stress-sensitive pathways and redox regulation. Investigations in animal models of diabetic cardiomyopathy have consistently demonstrated that increased expression of the primary antioxidant enzymes attenuates myocardial pathology and improves cardiac function.
Collapse
|
30
|
Ma K, Bai Y, Li J, Ren Z, Li J, Zhang J, Shan A. Lactobacillus rhamnosus GG ameliorates deoxynivalenol-induced kidney oxidative damage and mitochondrial injury in weaned piglets. Food Funct 2022; 13:3905-3916. [PMID: 35285834 DOI: 10.1039/d2fo00185c] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Deoxynivalenol (DON) is a common mycotoxin that pollutes food crops and adversely affects the health of animals, even humans. Lactobacillus rhamnosus GG (LGG) can alleviate intestinal injury, and anti-inflammatory and antioxidant effects. However, the potential of LGG in alleviating kidney injury induced by DON in piglets remains to be studied. The objective of this study was to investigate the adverse effect of DON on kidney injury and the protective ability of LGG. A total of twenty-seven weaned piglets were divided into three groups: CON group, DON group (3.11 mg kg-1 feed) and LGG + DON group (LGG powder 1 g kg-1 + DON 3.15 mg kg-1 feed). DON increased the MDA content, and decreased antioxidant enzyme activity (GSH-Px) and total antioxidant capacity (P < 0.05). Meanwhile, DON activated the Nrf2 antioxidant pathway. However, LGG supplementation alleviated the damage of DON to the kidney antioxidant system of piglets. Notably, DON significantly reduced the Sirt3 expression (P < 0.05), which was alleviated by LGG addition. The expression of mitochondrial biogenesis related factors such as VDAC1 and Cyt C was up-regulated by DON (P < 0.05), and LGG could improve mitochondrial ultrastructural abnormalities and mitochondrial dysfunction. In addition, LGG mitigated DON-induced mitochondrial fusion inhibition, and prevented DON-mediated mitochondrial autophagy. In conclusion, LGG play a protective role in DON-induced kidney toxicity, and dietary intervention may be a strategy to reduce mycotoxins.
Collapse
Affiliation(s)
- Kaidi Ma
- Institute of Animal Nutrition, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, P. R. China.
| | - Yongsong Bai
- Institute of Animal Nutrition, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, P. R. China.
| | - Jibo Li
- Institute of Animal Nutrition, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, P. R. China.
| | - Zhongshuai Ren
- College of Animal Science, Jilin University, Changchun 130062, P. R. China.
| | - Jianping Li
- Institute of Animal Nutrition, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, P. R. China.
| | - Jing Zhang
- College of Animal Science, Jilin University, Changchun 130062, P. R. China.
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, P. R. China.
| |
Collapse
|
31
|
Gembillo G, Visconti L, Giuffrida AE, Labbozzetta V, Peritore L, Lipari A, Calabrese V, Piccoli GB, Torreggiani M, Siligato R, Santoro D. Role of Zinc in Diabetic Kidney Disease. Nutrients 2022; 14:1353. [PMID: 35405968 PMCID: PMC9003285 DOI: 10.3390/nu14071353] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/19/2022] [Accepted: 03/22/2022] [Indexed: 12/23/2022] Open
Abstract
Diabetic Kidney Disease (DKD) represents the most common cause of Chronic Kidney Disease (CKD) in developed countries. Approximately 30% to 40% of diabetes mellitus (DM) subjects develop DKD, and its presence significantly increases the risk for morbidity and mortality. In this context, Zinc seems to have a potential role in kidney and body homeostasis in diabetic individuals as well as in patients at a high risk of developing this condition. This essential element has functions that may counteract diabetes-related risk factors and complications, which include stabilization of insulin hexamers and pancreatic insulin storage and improved glycemic control. In our review, we analyzed the current knowledge on the role of zinc in the management of renal impairment in course of DM. Several studies underline the critical role of zinc in reducing oxidative stress levels, which is considered the common denominator of the mechanisms responsible for the progression of kidney disease. Reaching and maintaining a proper serum zinc level could represent a valuable target to reduce symptoms related to DM complications and contrast the progression of kidney impairment in patients with the high risk of developing end-stage renal disease. In conclusion, analyzing the beneficial role of zinc in this review would advance our knowledge on the possible strategies of DM and DKD treatment.
Collapse
Affiliation(s)
- Guido Gembillo
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.E.G.); (V.L.); (L.P.); (A.L.); (V.C.)
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy;
| | - Luca Visconti
- Unit of Nephrology and Dialysis, Ospedali Riuniti Villa Sofia Cervello, University of Palermo, 90146 Palermo, Italy;
| | - Alfio Edoardo Giuffrida
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.E.G.); (V.L.); (L.P.); (A.L.); (V.C.)
| | - Vincenzo Labbozzetta
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.E.G.); (V.L.); (L.P.); (A.L.); (V.C.)
| | - Luigi Peritore
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.E.G.); (V.L.); (L.P.); (A.L.); (V.C.)
| | - Antonella Lipari
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.E.G.); (V.L.); (L.P.); (A.L.); (V.C.)
| | - Vincenzo Calabrese
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.E.G.); (V.L.); (L.P.); (A.L.); (V.C.)
| | - Giorgina Barbara Piccoli
- Néphrologie Et Dialyse, Centre Hospitalier Le Mans, 194 Avenue Rubillard, 72000 Le Mans, France; (G.B.P.); (M.T.)
| | - Massimo Torreggiani
- Néphrologie Et Dialyse, Centre Hospitalier Le Mans, 194 Avenue Rubillard, 72000 Le Mans, France; (G.B.P.); (M.T.)
| | - Rossella Siligato
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy;
- Unit of Nephrology, Azienda Ospedaliera Universitaria Sant’Anna, 44124 Ferrara, Italy
| | - Domenico Santoro
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.E.G.); (V.L.); (L.P.); (A.L.); (V.C.)
| |
Collapse
|
32
|
Mansouri A, Reiner Ž, Ruscica M, Tedeschi-Reiner E, Radbakhsh S, Bagheri Ekta M, Sahebkar A. Antioxidant Effects of Statins by Modulating Nrf2 and Nrf2/HO-1 Signaling in Different Diseases. J Clin Med 2022; 11:1313. [PMID: 35268403 PMCID: PMC8911353 DOI: 10.3390/jcm11051313] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/14/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
Statins are competitive inhibitors of hydroxymethylglutaryl-CoA (HMG-CoA) reductase and have been used to treat elevated low-density lipoprotein cholesterol (LDL-C) for almost four decades. Antioxidant and anti-inflammatory properties which are independent of the lipid-lowering effects of statins, i.e., their pleiotropic effects, might be beneficial in the prevention or treatment of many diseases. This review discusses the antioxidant effects of statins achieved by modulating the nuclear factor erythroid 2 related factor 2/ heme oxygenase-1 (Nrf2/HO-1) pathway in different organs and diseases. Nrf2 and other proteins involved in the Nrf2/HO-1 signaling pathway have a crucial role in cellular responses to oxidative stress, which is a risk factor for ASCVD. Statins can significantly increase the DNA-binding activity of Nrf2 and induce the expression of its target genes, such as HO-1 and glutathione peroxidase) GPx, (thus protecting the cells against oxidative stress. Antioxidant and anti-inflammatory properties of statins, which are independent of their lipid-lowering effects, could be partly explained by the modulation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Atena Mansouri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Željko Reiner
- Department of Internal Medicine, School of Medicine, University Hospital Center Zagreb, University of Zagreb, 10000 Zagreb, Croatia;
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20100 Milan, Italy;
| | - Eugenia Tedeschi-Reiner
- University Hospital Center Sestre Milosrdnice, University of Osijek, Vinogradska Cesta 29, 10000 Zagreb, Croatia;
| | - Shabnam Radbakhsh
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
- Department of Medical Biotechnology and Nanotechnology, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Mariam Bagheri Ekta
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, A.P. Avtsyn Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia;
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| |
Collapse
|
33
|
Gutiérrez-Cuevas J, Galicia-Moreno M, Monroy-Ramírez HC, Sandoval-Rodriguez A, García-Bañuelos J, Santos A, Armendariz-Borunda J. The Role of NRF2 in Obesity-Associated Cardiovascular Risk Factors. Antioxidants (Basel) 2022; 11:235. [PMID: 35204118 PMCID: PMC8868420 DOI: 10.3390/antiox11020235] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
The raising prevalence of obesity is associated with an increased risk for cardiovascular diseases (CVDs), particularly coronary artery disease (CAD), and heart failure, including atrial fibrillation, ventricular arrhythmias and sudden death. Obesity contributes directly to incident cardiovascular risk factors, including hyperglycemia or diabetes, dyslipidemia, and hypertension, which are involved in atherosclerosis, including structural and functional cardiac alterations, which lead to cardiac dysfunction. CVDs are the main cause of morbidity and mortality worldwide. In obesity, visceral and epicardial adipose tissue generate inflammatory cytokines and reactive oxygen species (ROS), which induce oxidative stress and contribute to the pathogenesis of CVDs. Nuclear factor erythroid 2-related factor 2 (NRF2; encoded by Nfe2l2 gene) protects against oxidative stress and electrophilic stress. NRF2 participates in the regulation of cell inflammatory responses and lipid metabolism, including the expression of over 1000 genes in the cell under normal and stressed environments. NRF2 is downregulated in diabetes, hypertension, and inflammation. Nfe2l2 knockout mice develop structural and functional cardiac alterations, and NRF2 deficiency in macrophages increases atherosclerosis. Given the endothelial and cardiac protective effects of NRF2 in experimental models, its activation using pharmacological or natural products is a promising therapeutic approach for obesity and CVDs. This review provides a comprehensive summary of the current knowledge on the role of NRF2 in obesity-associated cardiovascular risk factors.
Collapse
Affiliation(s)
- Jorge Gutiérrez-Cuevas
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara 44340, JAL, Mexico; (M.G.-M.); (H.C.M.-R.); (A.S.-R.); (J.G.-B.)
| | - Marina Galicia-Moreno
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara 44340, JAL, Mexico; (M.G.-M.); (H.C.M.-R.); (A.S.-R.); (J.G.-B.)
| | - Hugo Christian Monroy-Ramírez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara 44340, JAL, Mexico; (M.G.-M.); (H.C.M.-R.); (A.S.-R.); (J.G.-B.)
| | - Ana Sandoval-Rodriguez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara 44340, JAL, Mexico; (M.G.-M.); (H.C.M.-R.); (A.S.-R.); (J.G.-B.)
| | - Jesús García-Bañuelos
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara 44340, JAL, Mexico; (M.G.-M.); (H.C.M.-R.); (A.S.-R.); (J.G.-B.)
| | - Arturo Santos
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Campus Guadalajara, Zapopan 45201, JAL, Mexico;
| | - Juan Armendariz-Borunda
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara 44340, JAL, Mexico; (M.G.-M.); (H.C.M.-R.); (A.S.-R.); (J.G.-B.)
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Campus Guadalajara, Zapopan 45201, JAL, Mexico;
| |
Collapse
|
34
|
Ma J, Liu J, Chen Y, Yu H, Xiang L. Myricetin Improves Impaired Nerve Functions in Experimental Diabetic Rats. Front Endocrinol (Lausanne) 2022; 13:915603. [PMID: 35928887 PMCID: PMC9343592 DOI: 10.3389/fendo.2022.915603] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic peripheral neuropathy (DPN) is considered as one of the most important complications of diabetes mellitus. At present, effective treatments that might improve the damaged neurological function in DPN are sorely needed. As myricetin has been proved to possess excellent neuroprotective and antioxidant effects, it might have therapeutic potential for DPN. Therefore, the purpose of our study was to detect the potential beneficial effect of myricetin on DPN. A single dose of 50 mg/kg of streptozotocin was applied in rats for the establishment of diabetic models. Different doses of myricetin (0.5 mg/kg/day, 1.0 mg/kg/day, and 2.0 mg/kg/day) were intraperitoneally injected for 2 weeks from the 21st day after streptozotocin injection. After the final myricetin injection, behavioral, electrophysiological, biochemical, and protein analyses were performed. In the present study, myricetin significantly ameliorated diabetes-induced impairment in sensation, nerve conduction velocities, and nerve blood flow. In addition, myricetin significantly reduced the generation of advanced glycation end-products (AGEs) and reactive oxygen species (ROS), and elevated Na+, K+-ATPase activity and antioxidant activities in nerves in diabetic animals. Additional studies revealed that myricetin significantly raised the hydrogen sulfide (H2S) levels, and elevated the expression level of heme oxygenase-1 (HO-1) as well as nuclear factor-E2-related factor-2 (Nrf2) in diabetic rats. In addition, myricetin has the capability of decreasing plasma glucose under diabetic conditions. The findings in our present study collectively indicated that myricetin could restore the impaired motor and sensory functions under diabetic conditions. The Nrf2-dependent antioxidant action and the capability of decreasing plasma glucose might be the underlying mechanisms for the beneficial effect of myricetin on impaired neural functions. Our study showed the therapeutic potential of myricetin in the management of DPN.
Collapse
Affiliation(s)
| | | | | | - Hailong Yu
- *Correspondence: Hailong Yu, ; Liangbi Xiang,
| | | |
Collapse
|
35
|
Zoccarato A, Nabeebaccus AA, Oexner RR, Santos CXC, Shah AM. The nexus between redox state and intermediary metabolism. FEBS J 2021; 289:5440-5462. [PMID: 34496138 DOI: 10.1111/febs.16191] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/20/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022]
Abstract
Reactive oxygen species (ROS) are not just a by-product of cellular metabolic processes but act as signalling molecules that regulate both physiological and pathophysiological processes. A close connection exists in cells between redox homeostasis and cellular metabolism. In this review, we describe how intracellular redox state and glycolytic intermediary metabolism are closely coupled. On the one hand, ROS signalling can control glycolytic intermediary metabolism by direct regulation of the activity of key metabolic enzymes and indirect regulation via redox-sensitive transcription factors. On the other hand, metabolic adaptation and reprogramming in response to physiological or pathological stimuli regulate intracellular redox balance, through mechanisms such as the generation of reducing equivalents. We also discuss the impact of these intermediary metabolism-redox circuits in physiological and disease settings across different tissues. A better understanding of the mechanisms regulating these intermediary metabolism-redox circuits will be crucial to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Anna Zoccarato
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Adam A Nabeebaccus
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Rafael R Oexner
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Celio X C Santos
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| |
Collapse
|
36
|
Shilovsky GA, Putyatina TS, Morgunova GV, Seliverstov AV, Ashapkin VV, Sorokina EV, Markov AV, Skulachev VP. A Crosstalk between the Biorhythms and Gatekeepers of Longevity: Dual Role of Glycogen Synthase Kinase-3. BIOCHEMISTRY (MOSCOW) 2021; 86:433-448. [PMID: 33941065 PMCID: PMC8033555 DOI: 10.1134/s0006297921040052] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This review discusses genetic and molecular pathways that link circadian timing with metabolism, resulting in the emergence of positive and negative regulatory feedback loops. The Nrf2 pathway is believed to be a component of the anti-aging program responsible for the healthspan and longevity. Nrf2 enables stress adaptation by activating cell antioxidant defense and other metabolic processes via control of expression of over 200 target genes in response to various types of stress. The GSK3 system represents a “regulating valve” that controls fine oscillations in the Nrf2 level, unlike Keap1, which prevents significant changes in the Nrf2 content in the absence of oxidative stress and which is inactivated by the oxidative stress. Furthermore, GSK3 modifies core circadian clock proteins (Bmal1, Clock, Per, Cry, and Rev-erbα). Phosphorylation by GSK3 leads to the inactivation and degradation of circadian rhythm-activating proteins (Bmal1 and Clock) and vice versa to the activation and nuclear translocation of proteins suppressing circadian rhythms (Per and Rev-erbα) with the exception of Cry protein, which is likely to be implicated in the fine tuning of biological clock. Functionally, GSK3 appears to be one of the hubs in the cross-regulation of circadian rhythms and antioxidant defense. Here, we present the data on the crosstalk between the most powerful cell antioxidant mechanism, the Nrf2 system, and the biorhythm-regulating system in mammals, including the impact of GSK3 overexpression and knockout on the Nrf2 signaling. Understanding the interactions between the regulatory cascades linking homeostasis maintenance and cell response to oxidative stress will help in elucidating molecular mechanisms that underlie aging and longevity.
Collapse
Affiliation(s)
- Gregory A Shilovsky
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia. .,Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.,Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, 127051, Russia
| | - Tatyana S Putyatina
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Galina V Morgunova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Alexander V Seliverstov
- Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, 127051, Russia
| | - Vasily V Ashapkin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Elena V Sorokina
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Alexander V Markov
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Vladimir P Skulachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
37
|
Chen S, Yin Q, Hu H, Chen Q, Huang Q, Zhong M. AOPPs induce HTR-8/SVneo cell apoptosis by downregulating the Nrf-2/ARE/HO-1 anti-oxidative pathway: Potential implications for preeclampsia. Placenta 2021; 112:1-8. [PMID: 34237527 DOI: 10.1016/j.placenta.2021.06.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/29/2021] [Accepted: 06/14/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Advanced oxidation protein products (AOPPs), which are novel markers of oxidant-mediated protein damage, are prevalent in numerous diseases. We previously demonstrated that AOPPs act as a new class of pathogenic mediators in preeclampsia by causing trophoblast damage and dysfunction. Herein, we explored whether AOPPs could regulate the Nrf-2/ARE/HO-1 anti-oxidative pathway to facilitate the progression of preeclampsia. METHODS To investigate the pathophysiology of preeclampsia, we evaluated the effects of AOPPs on trophoblast damage, apoptotic proteins, and Nrf-2/ARE/HO-1 anti-oxidative pathway expression, as well as their underlying mechanisms. RESULTS AOPPs directly increased the expression of apoptotic proteins and significantly inhibited the expression of Nrf-2/ARE/HO-1 pathway in trophoblasts. Nrf-2 silencing aggravated the AOPPs-induced cell apoptosis in vitro by activating p53 and caspase cascade, whereas Nrf-2 overexpression had the opposite effect. Moreover, Nrf-2 exerted cytoprotective effects by increasing HO-1. DISCUSSION These findings suggest that AOPPs induce trophoblast apoptosis by triggering p53 and caspase activation via inhibition of the Nrf-2/ARE/HO-1 anti-oxidative pathway. Hence, Nrf-2/ARE/HO-1 pathway activation plays a protective role in AOPPs-induced cell apoptosis; thus, holding potential as a therapeutic target against preeclampsia.
Collapse
Affiliation(s)
- Shuying Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Yin
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haoyue Hu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qitao Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Byrne NJ, Rajasekaran NS, Abel ED, Bugger H. Therapeutic potential of targeting oxidative stress in diabetic cardiomyopathy. Free Radic Biol Med 2021; 169:317-342. [PMID: 33910093 PMCID: PMC8285002 DOI: 10.1016/j.freeradbiomed.2021.03.046] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/24/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Even in the absence of coronary artery disease and hypertension, diabetes mellitus (DM) may increase the risk for heart failure development. This risk evolves from functional and structural alterations induced by diabetes in the heart, a cardiac entity termed diabetic cardiomyopathy (DbCM). Oxidative stress, defined as the imbalance of reactive oxygen species (ROS) has been increasingly proposed to contribute to the development of DbCM. There are several sources of ROS production including the mitochondria, NAD(P)H oxidase, xanthine oxidase, and uncoupled nitric oxide synthase. Overproduction of ROS in DbCM is thought to be counterbalanced by elevated antioxidant defense enzymes such as catalase and superoxide dismutase. Excess ROS in the cardiomyocyte results in further ROS production, mitochondrial DNA damage, lipid peroxidation, post-translational modifications of proteins and ultimately cell death and cardiac dysfunction. Furthermore, ROS modulates transcription factors responsible for expression of antioxidant enzymes. Lastly, evidence exists that several pharmacological agents may convey cardiovascular benefit by antioxidant mechanisms. As such, increasing our understanding of the pathways that lead to increased ROS production and impaired antioxidant defense may enable the development of therapeutic strategies against the progression of DbCM. Herein, we review the current knowledge about causes and consequences of ROS in DbCM, as well as the therapeutic potential and strategies of targeting oxidative stress in the diabetic heart.
Collapse
Affiliation(s)
- Nikole J Byrne
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Namakkal S Rajasekaran
- Cardiac Aging & Redox Signaling Laboratory, Molecular and Cellular Pathology, Department of Pathology, Birmingham, AL, USA; Division of Cardiovascular Medicine, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Heiko Bugger
- Division of Cardiology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
39
|
Barteková M, Adameová A, Görbe A, Ferenczyová K, Pecháňová O, Lazou A, Dhalla NS, Ferdinandy P, Giricz Z. Natural and synthetic antioxidants targeting cardiac oxidative stress and redox signaling in cardiometabolic diseases. Free Radic Biol Med 2021; 169:446-477. [PMID: 33905865 DOI: 10.1016/j.freeradbiomed.2021.03.045] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022]
Abstract
Cardiometabolic diseases (CMDs) are metabolic diseases (e.g., obesity, diabetes, atherosclerosis, rare genetic metabolic diseases, etc.) associated with cardiac pathologies. Pathophysiology of most CMDs involves increased production of reactive oxygen species and impaired antioxidant defense systems, resulting in cardiac oxidative stress (OxS). To alleviate OxS, various antioxidants have been investigated in several diseases with conflicting results. Here we review the effect of CMDs on cardiac redox homeostasis, the role of OxS in cardiac pathologies, as well as experimental and clinical data on the therapeutic potential of natural antioxidants (including resveratrol, quercetin, curcumin, vitamins A, C, and E, coenzyme Q10, etc.), synthetic antioxidants (including N-acetylcysteine, SOD mimetics, mitoTEMPO, SkQ1, etc.), and promoters of antioxidant enzymes in CMDs. As no antioxidant indicated for the prevention and/or treatment of CMDs has reached the market despite the large number of preclinical and clinical studies, a sizeable translational gap is evident in this field. Thus, we also highlight potential underlying factors that may contribute to the failure of translation of antioxidant therapies in CMDs.
Collapse
Affiliation(s)
- Monika Barteková
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 81372 Bratislava, Slovakia.
| | - Adriana Adameová
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, 83232 Bratislava, Slovakia
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| | - Kristína Ferenczyová
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Oľga Pecháňová
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, 81371 Bratislava, Slovakia
| | - Antigone Lazou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, And Department of Physiology & Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; Pharmahungary Group, 6722 Szeged, Hungary
| |
Collapse
|
40
|
McKenna C, Keogh K, Porter RK, Waters SM, Cormican P, Kenny DA. An examination of skeletal muscle and hepatic tissue transcriptomes from beef cattle divergent for residual feed intake. Sci Rep 2021; 11:8942. [PMID: 33903612 PMCID: PMC8076192 DOI: 10.1038/s41598-021-87842-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/08/2021] [Indexed: 12/27/2022] Open
Abstract
The selection of cattle with enhanced feed efficiency is of importance with regard to reducing feed costs in the beef industry. Global transcriptome profiling was undertaken on liver and skeletal muscle biopsies from Simmental heifers and bulls divergent for residual feed intake (RFI), a widely acknowledged feed efficiency phenotype, in order to identify genes that may be associated with this trait. We identified 5 genes (adj. p < 0.1) to be differentially expressed in skeletal muscle between high and low RFI heifers with all transcripts involved in oxidative phosphorylation and mitochondrial homeostasis. A total of 11 genes (adj. p < 0. 1) were differentially expressed in liver tissue between high and low RFI bulls with differentially expressed genes related to amino and nucleotide metabolism as well as endoplasmic reticulum protein processing. No genes were identified as differentially expressed in either heifer liver or bull muscle analyses. Results from this study show that the molecular control of RFI in young cattle is modified according to gender, which may be attributable to differences in physiological maturity between heifers and bulls of the same age. Despite this we have highlighted a number of genes that may hold potential as molecular biomarkers for RFI cattle.
Collapse
Affiliation(s)
- Clare McKenna
- Animal and Bioscience Research Department, Teagasc Grange, Dunsany, C15 PW93, Co. Meath, Ireland.,School of Biochemistry & Immunology, Trinity College Dublin, Dublin 2, D02 R590, Ireland
| | - Kate Keogh
- Animal and Bioscience Research Department, Teagasc Grange, Dunsany, C15 PW93, Co. Meath, Ireland
| | - Richard K Porter
- School of Biochemistry & Immunology, Trinity College Dublin, Dublin 2, D02 R590, Ireland
| | - Sinead M Waters
- Animal and Bioscience Research Department, Teagasc Grange, Dunsany, C15 PW93, Co. Meath, Ireland
| | - Paul Cormican
- Animal and Bioscience Research Department, Teagasc Grange, Dunsany, C15 PW93, Co. Meath, Ireland
| | - David A Kenny
- Animal and Bioscience Research Department, Teagasc Grange, Dunsany, C15 PW93, Co. Meath, Ireland.
| |
Collapse
|
41
|
Bansod S, Chilvery S, Saifi MA, Das TJ, Tag H, Godugu C. Borneol protects against cerulein-induced oxidative stress and inflammation in acute pancreatitis mice model. ENVIRONMENTAL TOXICOLOGY 2021; 36:530-539. [PMID: 33166053 DOI: 10.1002/tox.23058] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 09/09/2020] [Accepted: 10/22/2020] [Indexed: 06/11/2023]
Abstract
Borneol is a commonly used flavouring substance in traditional Chinese medicine, which possesses several pharmacological activities including analgesic, antiinflammatory, and antioxidant properties. The aim of this study was to investigate the effects of borneol on cerulein-induced acute pancreatitis (AP) model. Swiss albino mice were pretreated with borneol (100 and 300 mg/kg) daily for 7 days, before six consecutive injections of cerulein (50 μg/kg/hr, intraperitoneally). The protective effect of borneol was studied by biochemical, enzyme linked immunosorbent assay, histological, immunoblotting, and immunohistochemical analysis. Oral administration of borneol significantly attenuated pancreatic damage by reducing amylase, lipase levels and histological changes. Borneol attenuated cerulein-induced oxidative-nitrosative stress by decreasing malondialdehyde, nitrite levels, and elevating reduced glutathione levels. Pancreatic inflammation was ameliorated by inhibiting myeloperoxidase activity and pro-inflammatory cytokine (Interleukins and TNF-α) levels. Furthermore, borneol administration significantly increased nuclear factor E2-related factor 2 (Nrf2), superoxide dismutase (SOD1) expression and reduced phospho-NF-κB p65 expression. Treatment with borneol significantly inhibited TNF-α, IL-1β, IL-6, and inducible nitric oxide synthase expression in cerulein-induced AP mouse model. Together, these results indicate that borneol which is currently used as US-FDA approved food adjuvant has the potential to attenuate cerulein-induced AP possibly by reducing the oxidative damage and pancreatic inflammation by modulating Nrf2/NF-κB pathway.
Collapse
Affiliation(s)
- Sapana Bansod
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Shrilekha Chilvery
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Mohd Aslam Saifi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Tridip Jyoti Das
- Department of Botany, Rajiv Gandhi University, Ron Hills, Doimukh, Arunachal Pradesh, India
| | - Hui Tag
- Department of Botany, Rajiv Gandhi University, Ron Hills, Doimukh, Arunachal Pradesh, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| |
Collapse
|
42
|
ALTamimi JZ, AlFaris NA, Alshammari GM, Alagal RI, Aljabryn DH, Aldera H, Alrfaei BM, Alkhateeb MA, Yahya MA. Ellagic acid protects against diabetic nephropathy in rats by regulating the transcription and activity of Nrf2. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
43
|
Abstract
Diabetic heart disease is a growing and important public health risk. Apart from the risk of coronary artery disease or hypertension, diabetes mellitus (DM) is a well-known risk factor for heart failure in the form of diabetic cardiomyopathy (DiaCM). Currently, DiaCM is defined as myocardial dysfunction in patients with DM in the absence of coronary artery disease and hypertension. The underlying pathomechanism of DiaCM is partially understood, but accumulating evidence suggests that metabolic derangements, oxidative stress, increased myocardial fibrosis and hypertrophy, inflammation, enhanced apoptosis, impaired intracellular calcium handling, activation of the renin-angiotensin-aldosterone system, mitochondrial dysfunction, and dysregulation of microRNAs, among other factors, are involved. Numerous animal models have been used to investigate the pathomechanisms of DiaCM. Despite some limitations, animal models for DiaCM have greatly advanced our understanding of pathomechanisms and have helped in the development of successful disease management strategies. In this review, we summarize the current pathomechanisms of DiaCM and provide animal models for DiaCM according to its pathomechanisms, which may contribute to broadening our understanding of the underlying mechanisms and facilitating the identification of possible new therapeutic targets.
Collapse
Affiliation(s)
- Wang-Soo Lee
- Division of Cardiology, Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
- Corresponding authors: Wang-Soo Lee https://orcid.org/0000-0002-8264-0866 Division of Cardiology, Department of Internal Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Dongjak-gu, Seoul 06973, Korea E-mail:
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
- Corresponding authors: Wang-Soo Lee https://orcid.org/0000-0002-8264-0866 Division of Cardiology, Department of Internal Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Dongjak-gu, Seoul 06973, Korea E-mail:
| |
Collapse
|
44
|
Hiromura M, Mori Y, Terasaki M, Kushima H, Saito T, Osaka N, Yashima H, Ohara M, Fukui T, Matsui T, Yamagishi SI. Glucose-dependent insulinotropic polypeptide inhibits cardiac hypertrophy and fibrosis in diabetic mice via suppression of TGF-β2. Diab Vasc Dis Res 2021; 18:1479164121999034. [PMID: 35012372 PMCID: PMC8755933 DOI: 10.1177/1479164121999034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Diabetic cardiomyopathy is associated with an increased risk for heart failure and death in patients with diabetes. We investigated here whether and how GIP attenuated cardiac hypertrophy and fibrosis in diabetic mice with obesity. Diabetic db/db mice at 7 weeks old were infused with vehicle or GIP (50 nmol/kg/day) for 6 weeks, and hearts were collected for histological and RT-PCR analyzes. Cardiomyocytes isolated from neonatal mice were incubated with or without 300 nM [D-Ala2]-GIP, 30 mM glucose, or 100 μg/mL advanced glycation end products (AGEs) for RT-PCR and lucigenin assays. Compared with non-diabetic mice, diabetic mice exhibited larger left ventricle wall thickness and cardiomyocyte sizes and more fibrotic areas in association with up-regulation of myosin heavy chain β (β-Mhc) and transforming growth factor-beta2 (Tgf-β2) mRNA levels, all of which were inhibited by GIP infusion. High glucose increased NADPH oxidase-driven superoxide generation and up-regulated β-Mhc, Tgf-β2, and receptor for AGEs mRNA levels in cardiomyocytes, and augmented the AGE-induced β-Mhc gene expression. [D-Ala2]-GIP attenuated all of the deleterious effects of high glucose and/or AGEs on cardiomyocytes. Our present findings suggest that GIP could inhibit cardiac hypertrophy and fibrosis in diabetic mice via suppression of TGF-β2.
Collapse
Affiliation(s)
- Munenori Hiromura
- Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Showa University School of Medicine, Shinagawa, Tokyo, Japan
| | - Yusaku Mori
- Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Anti-glycation Research Section, Showa University School of Medicine, Shinagawa, Tokyo, Japan
- Yusaku Mori, Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Anti-glycation Research Section, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan.
| | - Michishige Terasaki
- Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Showa University School of Medicine, Shinagawa, Tokyo, Japan
| | - Hideki Kushima
- Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Showa University School of Medicine, Shinagawa, Tokyo, Japan
| | - Tomomi Saito
- Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Showa University School of Medicine, Shinagawa, Tokyo, Japan
| | - Naoya Osaka
- Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Showa University School of Medicine, Shinagawa, Tokyo, Japan
| | - Hironori Yashima
- Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Showa University School of Medicine, Shinagawa, Tokyo, Japan
| | - Makoto Ohara
- Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Showa University School of Medicine, Shinagawa, Tokyo, Japan
| | - Tomoyasu Fukui
- Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Showa University School of Medicine, Shinagawa, Tokyo, Japan
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Sho-ichi Yamagishi
- Department of Medicine, Division of Diabetes, Metabolism and Endocrinology, Showa University School of Medicine, Shinagawa, Tokyo, Japan
| |
Collapse
|
45
|
Rahimi G, Heydari S, Rahimi B, Abedpoor N, Niktab I, Safaeinejad Z, Peymani M, Seyed Forootan F, Derakhshan Z, Esfahani MHN, Ghaedi K. A combination of herbal compound (SPTC) along with exercise or metformin more efficiently alleviated diabetic complications through down-regulation of stress oxidative pathway upon activating Nrf2-Keap1 axis in AGE rich diet-induced type 2 diabetic mice. Nutr Metab (Lond) 2021; 18:14. [PMID: 33468193 PMCID: PMC7816367 DOI: 10.1186/s12986-021-00543-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND SPTC is a mix of four herbal components (Salvia officinalis, Panax ginseng, Trigonella foenum-graeceum, and Cinnamomum zeylanicum) which might be prevented the development of AGE rich diet-induced diabetic complication and liver injury through activated the nuclear factor erythroid-2-related-factor-2 (Nrf2) pathway. Nrf2, as a master regulator of antioxidant response elements by activating cytoprotective genes expression, is decreased oxidative stress that associated with hyperglycemia and increases insulin sensitivity. the aim of this study was to assess whether the combination therapy of SPTC along with exercise or metformin moderate oxidative stress related liver injurie with more favorable effects in the treatment of AGE rich diet-induced type 2 diabetic mice. METHODS We induced diabetes in C57BL/6 mice by AGE using a diet supplementation and limitation of physical activity. After 16 weeks of intervention, AGE fed mice were compared to control mice. Diabetic mice were assigned into seven experimental groups (each group; n = 5): diabetic mice, diabetic mice treated with SPTC (130 mg/kg), diabetic mice treated with Salvia Officinalis (65 mg/kg), diabetic mice treated with metformin (300 mg/kg), diabetic mice with endurance exercise training, diabetic mice treated with SPTC + metformin (130/300 mg/kg), diabetic mice treated with SPTC + exercise training. RESULTS SPTC + exercise and SPTC + metformin reduced diabetic complications like gain weight, water and calorie intake, blood glucose, insulin, and GLUT4 content more efficiently than each treatment. These combinations improved oxidative stress hemostasis by activating the Nrf2 signaling pathway and attenuating keap1 protein more significantly. CONCLUSION Eventually, combined treatment of SPTC with exercise or metformin as a novel approach had more beneficial effects to prevent the development of diabetes and oxidative stress associated with hyperglycemia.
Collapse
Affiliation(s)
- Golbarg Rahimi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Avenue, Azadi Sq., Isfahan, 81746-73441 Iran
| | - Salime Heydari
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Avenue, Azadi Sq., Isfahan, 81746-73441 Iran
| | - Bahareh Rahimi
- Department of Medical Biotechnology, Faculty of Allied Medical Science, Iran University of Medical Science, Tehran, Iran
| | - Navid Abedpoor
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Avenue, Azadi Sq., Isfahan, 81746-73441 Iran
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Royan Street, Salman Street, Isfahan, 816513-1378 Iran
| | - Iman Niktab
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Royan Street, Salman Street, Isfahan, 816513-1378 Iran
| | - Zahra Safaeinejad
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Royan Street, Salman Street, Isfahan, 816513-1378 Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | | | - Zahra Derakhshan
- Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Hossein Nasr Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Royan Street, Salman Street, Isfahan, 816513-1378 Iran
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Avenue, Azadi Sq., Isfahan, 81746-73441 Iran
| |
Collapse
|
46
|
Fenofibrate Protects Cardiomyocytes from Hypoxia/Reperfusion- and High Glucose-Induced Detrimental Effects. PPAR Res 2021; 2021:8895376. [PMID: 33505452 PMCID: PMC7811426 DOI: 10.1155/2021/8895376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/03/2020] [Accepted: 12/22/2020] [Indexed: 01/11/2023] Open
Abstract
Lesions caused by high glucose (HG), hypoxia/reperfusion (H/R), and the coexistence of both conditions in cardiomyocytes are linked to an overproduction of reactive oxygen species (ROS), causing irreversible damage to macromolecules in the cardiomyocyte as well as its ultrastructure. Fenofibrate, a peroxisome proliferator-activated receptor alpha (PPARα) agonist, promotes beneficial activities counteracting cardiac injury. Therefore, the objective of this work was to determine the potential protective effect of fenofibrate in cardiomyocytes exposed to HG, H/R, and HG+H/R. Cardiomyocyte cultures were divided into four main groups: (1) control (CT), (2) HG (25 mM), (3) H/R, and (4) HG+H/R. Our results indicate that cell viability decreases in cardiomyocytes undergoing HG, H/R, and both conditions, while fenofibrate improves cell viability in every case. Fenofibrate also decreases ROS production as well as nicotinamide adenine dinucleotide phosphate oxidase (NADPH) subunit expression. Regarding the antioxidant defense, superoxide dismutase (SOD Cu2+/Zn2+ and SOD Mn2+), catalase, and the antioxidant capacity were decreased in HG, H/R, and HG+H/R-exposed cardiomyocytes, while fenofibrate increased those parameters. The expression of nuclear factor erythroid 2-related factor 2 (Nrf2) increased significantly in treated cells, while pathologies increased the expression of its inhibitor Keap1. Oxidative stress-induced mitochondrial damage was lower in fenofibrate-exposed cardiomyocytes. Endothelial nitric oxide synthase was also favored in cardiomyocytes treated with fenofibrate. Our results suggest that fenofibrate preserves the antioxidant status and the ultrastructure in cardiomyocytes undergoing HG, H/R, and HG+H/R preventing damage to essential macromolecules involved in the proper functioning of the cardiomyocyte.
Collapse
|
47
|
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
|
48
|
Wu P, Chen L, Cheng J, Pan Y, Guo X, Chu W, Zhang J, Liu X. MiRNAs-Modulation of Nrf2 Signaling Networks in Regulation Oxidative Stress of Chinese Perch Skeletal Muscle After Fasting Treatment. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2020; 22:620-630. [PMID: 32839828 DOI: 10.1007/s10126-020-09982-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/17/2020] [Indexed: 06/11/2023]
Abstract
Nrf2 is an important transcription factor involved in the antioxidant response and is widely expressed in animal tissues. The function of Nrf2 is regulated by its negative regulator Keap1 by inducing its cytoplasmic degradation. Recent studies have suggested that Nrf2 is also regulated post-transcriptionally via miRNAs. However, to date, how miRNAs regulate Nrf2 in fish skeletal muscles is unknown. In this study, the full-length cDNAs with 2398 bp of the Nrf2 was firstly cloned by SMART RACE amplification tools from Chinese perch. The Nrf2 gene structure and its 3'-UTR region for possible miRNA binding sites, as well as its spatial expression profile were assayed. Then, we employed TargetScan Fish tool MiRNAnome to predict putative sites for five miRNAs including miR-181a-5p, MiR-194a, MiR-216a, miR-459-5p, and miR-724. Using qRT-PCR assay, we found that Nrf2 mRNA levels have negative correlation with all five miRNAs expression in muscle of nutritionally deprived fish, and that ectopic expression of miR-181a-5p alone reduces Nrf2 mRNA levels. Luciferase reporter assay in a heterologous cell system revealed that each of the five miRNAs reduced Nrf2 expression, suggesting a direct regulatory mechanism. Moreover, the miR-181a-5p suppression using specific antagomir led to a significant increase in Nrf2 expression in vivo. At the same time, the expression levels of the antioxidant enzymes CAT, ZnSOD, GPx, GSTA, and GSTA genes increased significantly after injecting miR-181a-5p antagomir. Taken together, these findings provide evidence that miRNAs are involved in the Nrf2 signaling networks in regulation of oxidative stress in fish, at least in Chinese perch muscle.
Collapse
Affiliation(s)
- Ping Wu
- State Key laboratory of Chemo/Biosening and Chemomertrics, College of Biology, Hunan University, Changsha, 410082, People's Republic of China
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, 410022, China
| | - Lin Chen
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, 410022, China
| | - Jia Cheng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, 410022, China
| | - Yaxiong Pan
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, 410022, China
| | - Xinhong Guo
- State Key laboratory of Chemo/Biosening and Chemomertrics, College of Biology, Hunan University, Changsha, 410082, People's Republic of China
| | - Wuying Chu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, 410022, China
| | - Jianshe Zhang
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, 410022, China.
| | - Xuanming Liu
- State Key laboratory of Chemo/Biosening and Chemomertrics, College of Biology, Hunan University, Changsha, 410082, People's Republic of China.
| |
Collapse
|
49
|
Gatiatulina ER, Sheina EA, Nemereshina ON, Popova EV, Polyakova VS, Agletdinov EF, Sinitskii AI, Skalny AV, Nikonorov AA, Tinkov AA. Effect of Zn Supplementation on Trace Element Status in Rats with Diet-Induced Non-alcoholic Fatty Liver Disease. Biol Trace Elem Res 2020; 197:202-212. [PMID: 31832925 DOI: 10.1007/s12011-019-01985-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/13/2019] [Indexed: 12/19/2022]
Abstract
The present study aimed to assess the effect of Zn supplementation on trace element levels in the liver, serum, and hair of rats with dietary-induced non-alcoholic fatty liver disease (NAFLD). A total of 26 3-month-old female Wistar rats were divided into four groups: control, NAFLD, Zn-supplemented (227 mg/L zinc as Zn sulfate Zn(SO)4 dissolved in a drinking water), and NAFLD-Zn-supplemented. NAFLD was verified by histological assessment of liver samples. The serum was examined for routine biochemical parameters. Trace elements content was assessed using inductively coupled plasma mass spectrometry (ICP-MS). Zn treatment resulted in an improvement in liver weight and morphology. Dietary supplementation with Zn prevented NAFLD-induced decrease liver Co. The tendency to increase liver Fe in the Zn-treated group was observed. Zn treatment decreased hepatic Al and serum V levels. However, Zn administration did not affect NAFLD-induced I, Mn, and Se depletion in the liver. Hair Zn levels raised in Zn-supplemented groups. Conclusively, the results of the study indicate that Zn supplementation could have a beneficial effect in modulation of the altered trace element status and liver morphology. HIGHLIGHTS: •Zn treatment improved liver weight and morphology in rats with NAFLD. •Zn supplementation decreased liver Al in NAFLD. •Treatment by Zn prevented depletion of liver Co. •Zn decreased serum V and increased hair Zn levels. •No effect of Zn on NAFLD-induced hepatic I, Mn and Se depletion was observed.
Collapse
Affiliation(s)
- Eugenia R Gatiatulina
- All-Russian Research Institute of Medicinal and Aromatic Plants (VILAR), Grina St., 7, Moscow, 117216, Russia.
| | - Evgenia A Sheina
- Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, Moscow, 105064, Russia
| | - Olga N Nemereshina
- Orenburg State Medical University, Sovetskaya St., 6, Orenburg, 460000, Russia
| | - Elizaveta V Popova
- St. Joseph College of Health and Allied Sciences, St Joseph University in Tanzania, 11007, Dar es Salaam, Tanzania
| | | | | | - Anton I Sinitskii
- South Ural State Medical University, Vorovskogo St., 64, Chelyabinsk, 454092, Russia
| | - Anatoly V Skalny
- Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, Moscow, 105064, Russia
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl, 150000, Russia
- IM Sechenov First Moscow State Medical University, Trubetskaya St., 8-2, Moscow, 119991, Russia
| | - Alexandr A Nikonorov
- State Research Center of Dermatovenerology and Cosmetology, Korolenko St., 3-6, Moscow, 107076, Russia
| | - Alexey A Tinkov
- Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, Moscow, 105064, Russia
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl, 150000, Russia
- IM Sechenov First Moscow State Medical University, Trubetskaya St., 8-2, Moscow, 119991, Russia
| |
Collapse
|
50
|
Tan Y, Zhang Z, Zheng C, Wintergerst KA, Keller BB, Cai L. Mechanisms of diabetic cardiomyopathy and potential therapeutic strategies: preclinical and clinical evidence. Nat Rev Cardiol 2020; 17:585-607. [PMID: 32080423 PMCID: PMC7849055 DOI: 10.1038/s41569-020-0339-2] [Citation(s) in RCA: 480] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2020] [Indexed: 02/07/2023]
Abstract
The pathogenesis and clinical features of diabetic cardiomyopathy have been well-studied in the past decade, but effective approaches to prevent and treat this disease are limited. Diabetic cardiomyopathy occurs as a result of the dysregulated glucose and lipid metabolism associated with diabetes mellitus, which leads to increased oxidative stress and the activation of multiple inflammatory pathways that mediate cellular and extracellular injury, pathological cardiac remodelling, and diastolic and systolic dysfunction. Preclinical studies in animal models of diabetes have identified multiple intracellular pathways involved in the pathogenesis of diabetic cardiomyopathy and potential cardioprotective strategies to prevent and treat the disease, including antifibrotic agents, anti-inflammatory agents and antioxidants. Some of these interventions have been tested in clinical trials and have shown favourable initial results. In this Review, we discuss the mechanisms underlying the development of diabetic cardiomyopathy and heart failure in type 1 and type 2 diabetes mellitus, and we summarize the evidence from preclinical and clinical studies that might provide guidance for the development of targeted strategies. We also highlight some of the novel pharmacological therapeutic strategies for the treatment and prevention of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.
- Wendy Novak Diabetes Center, University of Louisville, Norton Children's Hospital, Louisville, KY, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
| | - Zhiguo Zhang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Chao Zheng
- The Second Affiliated Hospital Center of Chinese-American Research Institute for Diabetic Complications, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kupper A Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
- Wendy Novak Diabetes Center, University of Louisville, Norton Children's Hospital, Louisville, KY, USA
- Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Bradley B Keller
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.
- Wendy Novak Diabetes Center, University of Louisville, Norton Children's Hospital, Louisville, KY, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
- Department of Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|