1
|
Gianopoulos I, Mantzoros CS, Daskalopoulou SS. Adiponectin and Adiponectin Receptors in Atherosclerosis. Endocr Rev 2025; 46:1-25. [PMID: 39106421 PMCID: PMC11720176 DOI: 10.1210/endrev/bnae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/14/2024] [Accepted: 08/02/2024] [Indexed: 08/09/2024]
Abstract
Adiponectin is an abundantly secreted hormone that communicates information between the adipose tissue, and the immune and cardiovascular systems. In metabolically healthy individuals, adiponectin is usually found at high levels and helps improve insulin responsiveness of peripheral tissues, glucose tolerance, and fatty acid oxidation. Beyond its metabolic functions in insulin-sensitive tissues, adiponectin plays a prominent role in attenuating the development of atherosclerotic plaques, partially through regulating macrophage-mediated responses. In this context, adiponectin binds to its receptors, adiponectin receptor 1 (AdipoR1) and AdipoR2 on the cell surface of macrophages to activate a downstream signaling cascade and induce specific atheroprotective functions. Notably, macrophages modulate the stability of the plaque through their ability to switch between proinflammatory responders, and anti-inflammatory proresolving mediators. Traditionally, the extremes of the macrophage polarization spectrum span from M1 proinflammatory and M2 anti-inflammatory phenotypes. Previous evidence has demonstrated that the adiponectin-AdipoR pathway influences M1-M2 macrophage polarization; adiponectin promotes a shift toward an M2-like state, whereas AdipoR1- and AdipoR2-specific contributions are more nuanced. To explore these concepts in depth, we discuss in this review the effect of adiponectin and AdipoR1/R2 on 1) metabolic and immune responses, and 2) M1-M2 macrophage polarization, including their ability to attenuate atherosclerotic plaque inflammation, and their potential as therapeutic targets for clinical applications.
Collapse
Affiliation(s)
- Ioanna Gianopoulos
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Section of Endocrinology, Diabetes and Metabolism, Boston VA Healthcare System, Boston, MA 02130, USA
| | - Stella S Daskalopoulou
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec H4A 3J1, Canada
- Division of Internal Medicine, Department of Medicine, Faculty of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
2
|
Rehman IU, Park JS, Choe K, Park HY, Park TJ, Kim MO. Overview of a novel osmotin abolishes abnormal metabolic-associated adiponectin mechanism in Alzheimer's disease: Peripheral and CNS insights. Ageing Res Rev 2024; 100:102447. [PMID: 39111409 DOI: 10.1016/j.arr.2024.102447] [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: 06/11/2024] [Revised: 07/19/2024] [Accepted: 08/03/2024] [Indexed: 08/16/2024]
Abstract
Alzheimer's disease (AD) is a degenerative brain disease that affects millions of people worldwide. It is caused by abnormalities in cholinergic neurons, oxidative stress, and inflammatory cascades. The illness is accompanied by personality changes, memory issues, and dementia. Metabolic signaling pathways help with fundamental processes like DNA replication and RNA transcription. Being adaptable is essential for both surviving and treating illness. The body's metabolic signaling depends on adipokines, including adiponectin (APN) and other adipokines secreted by adipose tissues. Energy homeostasis is balanced by adipokines, and nutrients. Overconsumption of nutrients messes with irregular signaling of adipokines, such as APN in both peripheral and brain which leads to neurodegeneration, such as AD. Despite the failure of traditional treatments like memantine and cholinesterase inhibitors, natural plant bioactive substances like Osmotin (OSM) have been given a focus as potential therapeutics due to their antioxidant properties, better blood brain barrier (BBB) permeability, excellent cell viability, and especially nanoparticle approaches. The review highlights the published preclinical literature regarding the role of OSM in AD pathology while there is a need for more research to investigate the hidden therapeutic potential of OSM which may open a new gateway and further strengthen its healing role in the pathogenesis of neurodegeneration, especially AD.
Collapse
Affiliation(s)
- Inayat Ur Rehman
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea.
| | - Jun Sung Park
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea.
| | - Kyonghwan Choe
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht 6229 ER, the Netherlands.
| | - Hyun Young Park
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht 6229 ER, the Netherlands; Department of Pediatrics, Maastricht University Medical Center (MUMC+), Maastricht 6202 AZ, the Netherlands.
| | - Tae Ju Park
- Haemato-oncology/Systems Medicine Group, Paul O'Gorman Leukemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences (MVLS), University of Glasgow, Glasgow G12 0ZD, United Kingdom.
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Alz-Dementia Korea Co., Jinju 52828, Republic of Korea.
| |
Collapse
|
3
|
Sun K, Yuan R, He J, Zhuo Y, Yang M, Hao E, Hou X, Yao C, Yang S, Gao H. Sugarcane leaf polysaccharide exerts a therapeutic effect on cardiovascular diseases through necroptosis. Heliyon 2023; 9:e21889. [PMID: 38027563 PMCID: PMC10658330 DOI: 10.1016/j.heliyon.2023.e21889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 10/06/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Background Necroptosis, a novel form of programmed cell death wherein the necrotic morphology is characterized by swelling of the cells, rupture of the plasma membrane, and dysfunction of the organelle, has been always observed in cardiovascular diseases. Sugarcane leaf polysaccharide (SLP) are primary components present in sugarcane leaves that exert cardiovascular protective effects. However, the positive effect of SLP and underlying mechanisms in myocardial ischemia-reperfusion (MI/R) remain unexplored. Aim In this study, the protective effects of SLP on MI/R injury were investigated under in vitro and in vivo conditions. Methods The protective effects of SLP on MI/R injury were assessed using tertiary butyl hydrogen peroxide (TBHP)-stimulated-H9c2 cells in the in vitro assay and using Sprague Dawley rats in the in vivo assay. Results In vitro, SLP significantly reversed TBHP-induced H9c2 cell death by inhibiting necroptosis and oxidative stress. SLP exerted antioxidant activity through the Nrf2/HO-1 pathway. SLP suppressed necroptosis by decreasing phosphorylation of RIP1, RIP3, and MLKL in TBHP-stimulated H9c2 cells. In vivo, SLP attenuated MI/R injury by decreasing the myocardial infarct area; increasing myeloperoxidase and superoxide dismutase levels; and reducing malondialdehyde, interleukin-6, and tumor necrosis factor-α levels.
Collapse
Affiliation(s)
- Kaili Sun
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Renyikun Yuan
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Jia He
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Youqiong Zhuo
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Ming Yang
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Erwei Hao
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica/Guangxi University of Chinese Medicine, Nanning, Guangxi, 530001, China
- Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues/Guangxi University of Chinese Medicine, Nanning, Guangxi, 530200, China
| | - Xiaotao Hou
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica/Guangxi University of Chinese Medicine, Nanning, Guangxi, 530001, China
- Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues/Guangxi University of Chinese Medicine, Nanning, Guangxi, 530200, China
| | - Chun Yao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Shilin Yang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
| |
Collapse
|
4
|
Cohen KE, Katunaric B, Schulz ME, SenthilKumar G, Young MS, Mace JE, Freed JK. Role of Adiponectin Receptor 1 in Promoting Nitric Oxide-Mediated Flow-Induced Dilation in the Human Microvasculature. Front Pharmacol 2022; 13:875900. [PMID: 35444544 PMCID: PMC9014203 DOI: 10.3389/fphar.2022.875900] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic administration of exogenous adiponectin restores nitric oxide (NO) as the mediator of flow-induced dilation (FID) in arterioles collected from patients with coronary artery disease (CAD). Here we hypothesize that this effect as well as NO signaling during flow during health relies on activation of Adiponectin Receptor 1 (AdipoR1). We further posit that osmotin, a plant-derived protein and AdipoR1 activator, is capable of eliciting similar effects as adiponectin. Human arterioles (80-200 μm) collected from discarded surgical adipose specimens were cannulated, pressurized, and pre-constricted with endothelin-1 (ET-1). Changes in vessel internal diameters were measured during flow using videomicroscopy. Immunofluorescence was utilized to compare expression of AdipoR1 during both health and disease. Administration of exogenous adiponectin failed to restore NO-mediated FID in CAD arterioles treated with siRNA against AdipoR1 (siAdipoR1), compared to vessels treated with negative control siRNA. Osmotin treatment of arterioles from patients with CAD resulted in a partial restoration of NO as the mediator of FID, which was inhibited in arterioles with decreased expression of AdipoR1. Together these data highlight the critical role of AdipoR1 in adiponectin-induced NO signaling during shear. Further, osmotin may serve as a potential therapy to prevent microvascular endothelial dysfunction as well as restore endothelial homeostasis in patients with cardiovascular disease.
Collapse
Affiliation(s)
- Katie E. Cohen
- Department of Medicine-Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Boran Katunaric
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Mary E. Schulz
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Gopika SenthilKumar
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Micaela S. Young
- Department of Medicine-Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - James E. Mace
- Department of Surgery-Division of Adult Cardiothoracic Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Julie K. Freed
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
5
|
Chen Z, Wu J, Li S, Liu C, Ren Y. Inhibition of Myocardial Cell Apoptosis Is Important Mechanism for Ginsenoside in the Limitation of Myocardial Ischemia/Reperfusion Injury. Front Pharmacol 2022; 13:806216. [PMID: 35300297 PMCID: PMC8921549 DOI: 10.3389/fphar.2022.806216] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/09/2022] [Indexed: 12/25/2022] Open
Abstract
Ischemic heart disease has a high mortality, and the recommended therapy is reperfusion. Nevertheless, the restoration of blood flow to ischemic tissue leads to further damage, namely, myocardial ischemia/reperfusion injury (MIRI). Apoptosis is an essential pathogenic factor in MIRI, and ginsenosides are effective in inhibiting apoptosis and alleviating MIRI. Here, we reviewed published studies on the anti-apoptotic effects of ginsenosides and their mechanisms of action in improving MIRI. Each ginsenoside can regulate multiple pathways to protect the myocardium. Overall, the involved apoptotic pathways include the death receptor signaling pathway, mitochondria signaling pathway, PI3K/Akt signaling pathway, NF-κB signaling pathway, and MAPK signaling pathway. Ginsenosides, with diverse chemical structures, regulate different apoptotic pathways to relieve MIRI. Summarizing the effects and mechanisms of ginsenosides contributes to further mechanism research studies and structure-function relationship research studies, which can help the development of new drugs. Therefore, we expect that this review will highlight the importance of ginsenosides in improving MIRI via anti-apoptosis and provide references and suggestions for further research in this field.
Collapse
Affiliation(s)
- Zhihan Chen
- School of Acupuncture Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingping Wu
- Department of Medical Cosmetology, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sijing Li
- School of Acupuncture Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Caijiao Liu
- School of Acupuncture Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yulan Ren
- School of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
6
|
Jochum F, Abdellatif M, Adel A, Alhammadi A, Alnemri A, Alohali E, AlSarraf K, Al Said K, Elzalabany M, Isa HMA, Kalyanasundaram S, Reheim NA, Saadah O. Burden of Early Life Obesity and Its Relationship with Protein Intake in Infancy: The Middle East Expert Consensus. Pediatr Gastroenterol Hepatol Nutr 2022; 25:93-108. [PMID: 35360379 PMCID: PMC8958054 DOI: 10.5223/pghn.2022.25.2.93] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 12/13/2021] [Accepted: 02/20/2022] [Indexed: 02/06/2023] Open
Abstract
Adequate nutrition in early life is proposed to shape a child's future health by launching the growth trajectory in the proper direction, which helps to avoid negative metabolic programming effects. Protein intake during infancy and early childhood is of great importance, as it plays a key role in infant metabolic programming and the future risk of obesity. Breastfeeding provides the best nutrition in early life, with many benefits tailored for the baby, including the appropriate quantity and quality of proteins. Considering the high prevalence of childhood, and subsequent adult, obesity in the region, a virtual Middle East expert consensus meeting was held to discuss an effective approach for managing childhood obesity. Leading pediatric experts from Bahrain, Egypt, Kuwait, Oman, Qatar, Saudi Arabia, and the United Arab Emirates participated in the meeting. The experts discussed, debated, and agreed on certain directions, including the importance of educating parents, endorsing breastfeeding, and ensuring optimum quantity and quality intake of proteins in early life. This expert consensus may serve as the starting point for healthcare professionals in the region who are interested in shaping a healthy future for the generations to come.
Collapse
Affiliation(s)
- Frank Jochum
- Department of Pediatrics, Evangelisches Waldkrankenhaus Spandau, Berlin, Germany.,Pediatric Medicine, Brandenburg Medical School (MHB) Theodor Fontane, Neuruppin, Germany
| | | | - Ashraf Adel
- Department of General Pediatrics, Sidra Medicine, Doha, Qatar
| | - Ahmed Alhammadi
- Department of General Pediatrics, Sidra Medicine, Doha, Qatar
| | | | - Eman Alohali
- Dietetics Department, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Khaled AlSarraf
- Department of Pediatrics-Pediatric Gastroenterology, Amiri Hospital, Sharq, Kuwait
| | - Khoula Al Said
- Department of Child Health, Royal Hospital, Muscat, Oman
| | - Mahmoud Elzalabany
- Pediatrics Department, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Hasan M A Isa
- Pediatric Department, Salmaniya Medical Complex/Arabian Gulf University, Manama, Bahrain
| | | | | | - Omar Saadah
- Pediatrics Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
7
|
Socioeconomic status, BMI, and brain development in children. Transl Psychiatry 2022; 12:33. [PMID: 35075111 PMCID: PMC8786961 DOI: 10.1038/s41398-022-01779-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 11/10/2022] Open
Abstract
Low socioeconomic status (SES) in childhood is associated with deficits in executive function and changes in cortical morphology. Furthermore, rates of childhood obesity are greater among low SES children and childhood obesity is also associated with cortical alterations and impaired neurocognition, specifically in the domain of executive function. To investigate the influence of BMI on the relationships between SES and both neurocognition and brain morphology, we used data from the Adolescent Brain Cognitive Development (ABCD) study to construct multiple linear regression models and conduct mediation analyses. Overall, SES as measured by household income, highest level of parental education, and area deprivation, was associated with lower BMI, greater total and prefrontal cortical volume, and better performance on assessments of executive function. Mediation analysis indicated that BMI had a significant indirect effect on associations between area deprivation and both total and prefrontal cortical volumes. BMI also played a mediating role in the associations between area deprivation and composite neurocognitive scores, which were driven by performance on tasks of working memory and cognitive flexibility, but not cognitive control. These findings suggest that BMI should be considered in future studies investigating the relationship between low SES and poor neurodevelopmental outcomes.
Collapse
|
8
|
Zhou YX, Zhang H, Peng C. Effects of Puerarin on the Prevention and Treatment of Cardiovascular Diseases. Front Pharmacol 2021; 12:771793. [PMID: 34950032 PMCID: PMC8689134 DOI: 10.3389/fphar.2021.771793] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Puerarin, an isoflavone glycoside derived from Pueraria lobata (Willd.) Ohwi, has been identified as a pharmacologically active component with diverse benefits. A large number of experimental and clinical studies have demonstrated that puerarin is widely used in the treatment of a variety of diseases. Among them, cardiovascular diseases (CVDs) are the leading cause of death in the world, and therefore remain one of the most prominent global public health concerns. In this review, we systematically analyze the preclinical investigations of puerarin in CVDs, such as atherosclerosis, cardiac hypertrophy, heart failure, diabetic cardiovascular complications, myocardial infarction, stroke and hypertension. In addition, the potential molecular targets of puerarin are also discussed. Furthermore, we summarize the clinical trails of puerarin in the treatment of CVDs. Finally, the therapeutic effects of puerarin derivatives and its drug delivery systems are overviewed.
Collapse
Affiliation(s)
- Yan-Xi Zhou
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Library, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Zhang
- Institute of Interdisciplinary Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Peng
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
9
|
Wu Q, Liu F, Shen T, Zhang W. Multiple pathways are responsible to the inhibitory effect of butorphanol on OGD/R-induced apoptosis in AC16 cardiomyocytes. J Appl Toxicol 2021; 42:830-840. [PMID: 34708435 DOI: 10.1002/jat.4260] [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: 03/29/2021] [Revised: 09/28/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022]
Abstract
Ischemic heart disease is the leading cause of cardiovascular mortality, which is related to cardiac myocyte apoptosis. Butorphanol is an opioid receptor agonist with potential cardioprotective function. The purpose of this work is to explore the function and mechanism of butorphanol in oxygen and glucose deprivation/reperfusion (OGD/R)-induced cardiomyocyte apoptosis. The overlapping targets of ischemic heart disease and butorphanol were analyzed according to GeneCards, ParmMapper, Cytoscape, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Human cardiomyocyte AC16 cells were incubated with butorphanol and then stimulated with OGD/R. Cell injury was investigated by Cell Counting Kit-8, lactate dehydrogenase (LDH) assay kit, TUNEL staining, caspase-3 activity assay kit, and Western blotting. The proteins in signaling pathways were measured using Western blotting. A total of 93 overlapping targets of ischemic heart disease and butorphanol were obtained. Pathway analysis exhibited that these targets might be involved in multiple signaling pathways. Butorphanol alone showed little cytotoxicity to cardiomyocytes, and it protected against OGD/R-induced viability inhibition, LDH release, cell apoptosis, and increase of caspase-3 activity and expression levels of cleaved caspase-3 and Bim. Butorphanol promoted the activation of the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/forkhead box O (FoxO) and hypoxia-inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF) pathways and attenuated the activation of the mitogen-activated protein kinase (MAPK) signaling in OGD/R-treated cardiomyocytes. In conclusion, butorphanol prevents OGD/R-induced cardiomyocyte apoptosis through activating the PI3K/Akt/FoxO and HIF-1α/VEGF pathways and inactivating the MAPK pathway.
Collapse
Affiliation(s)
- Qiaoling Wu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Feifei Liu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Tu Shen
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Wei Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
10
|
Sharma A, Mah M, Ritchie RH, De Blasio MJ. The adiponectin signalling pathway - A therapeutic target for the cardiac complications of type 2 diabetes? Pharmacol Ther 2021; 232:108008. [PMID: 34610378 DOI: 10.1016/j.pharmthera.2021.108008] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/17/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022]
Abstract
Diabetes is associated with an increased risk of heart failure (HF). This is commonly termed diabetic cardiomyopathy and is often characterised by increased cardiac fibrosis, pathological hypertrophy, increased oxidative and endoplasmic reticulum stress as well as diastolic dysfunction. Adiponectin is a cardioprotective adipokine that is downregulated in settings of type 2 diabetes (T2D) and obesity. Furthermore, both adiponectin receptors (AdipoR1 and R2) are also downregulated in these settings which further results in impaired cardiac adiponectin signalling and reduced cardioprotection. In many cardiac pathologies, adiponectin signalling has been shown to protect against cardiac remodelling and lipotoxicity, however its cardioprotective actions in T2D-induced cardiomyopathy remain unresolved. Diabetic cardiomyopathy has historically lacked effective treatment options. In this review, we summarise the current evidence for links between the suppressed adiponectin signalling pathway and cardiac dysfunction, in diabetes. We describe adiponectin receptor-mediated signalling pathways that are normally associated with cardioprotection, as well as current and potential future therapeutic approaches that could target this pathway as possible interventions for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Abhipree Sharma
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Michael Mah
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; Department of Pharmacology, Monash University, Clayton, VIC 3800, Australia; Department of Medicine, Monash University, Clayton, VIC 3800, Australia
| | - Miles J De Blasio
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; Department of Pharmacology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
11
|
Shen YJ, Shen YC, Lee WS, Yang KT. Methyl palmitate protects heart against ischemia/reperfusion-induced injury through G-protein coupled receptor 40-mediated activation of the PI3K/AKT pathway. Eur J Pharmacol 2021; 905:174183. [PMID: 34015318 DOI: 10.1016/j.ejphar.2021.174183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/08/2021] [Accepted: 05/12/2021] [Indexed: 10/21/2022]
Abstract
This study aimed to investigate whether methyl palmitate (MP) exerts cardioprotective effect against the ischemia/reperfusion (I/R) injury and its mechanisms underlying. The cultured adult cardiomyocytes were treated with vehicle or lactic acid ischemic buffer (pH 6.8) during hypoxia/reoxygenation. In addition, the cardioprotective effect of MP was evaluated using the ex vivo heart model of I/R injury. Here, we found that MP significantly reduced the I/R-induced cardiomyocyte death. Treatment with GW1100 (a GPR40-antagonist) or wortmannin (a phosphatidylinositol 3-kinase, PI3K, specific inhibitor) significantly attenuated the level of phospho-AKT (p-AKT) and abolished the MP-induced cardioprotection against the I/R-induced injury. Using the ex vivo I/R model, we also demonstrated that pretreatment with MP significantly reduced the size of myocardial infarction and the levels of cleaved-caspase 3 and MDA, and increased the protein levels of GPR40 and p-AKT induced by I/R. The cardioprotective effect of MP was evaluated also using the in vivo heart model of I/R injury. We demonstrated that post-ischemic treatment with MP significantly attenuated the size of myocardial infarction and the serum level of CK-MB induced by in vivo I/R model. Taken together, our data suggest that MP could provide significant cardioprotection against the I/R injury, and the underlying mechanisms by which MP prevented the cardiomyocyte death might be mediated through the GPR40-activated PI3K/AKT signaling pathways. These findings suggest the potential applications of MP in the treatment of I/R-induced heart injury.
Collapse
Affiliation(s)
- Yan-Jhih Shen
- Ph.D. Program in Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Yan-Cheng Shen
- Ph.D. Program in Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Sen Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kun-Ta Yang
- Department of Physiology, School of Medicine, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
12
|
Geetha RG, Krishnankutty Nair Chandrika S, Saraswathy GG, Nair Sivakumari A, Sakuntala M. ROS Dependent Antifungal and Anticancer Modulations of Piper colubrinum Osmotin. Molecules 2021; 26:molecules26082239. [PMID: 33924432 PMCID: PMC8070354 DOI: 10.3390/molecules26082239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 11/16/2022] Open
Abstract
Osmotin, a plant defense protein, has functional similarity to adiponectin, an insulin sensitizingsensitising hormone secreted by adipocytes. We speculated that Piper colubrinum Osmotin (PcOSM) could have functional roles in obesity-related cancers, especially breast cancer. Immunofluorescence assays, flow cytometry, cell cycle analysis and a senescence assay were employed to delineate the activity in MDAMB231 breast cancer cell line. PcOSM pre-treated P. nigrum leaves showed significant reduction in disease symptoms correlated with high ROS production. In silico analysis predicted that PcOSM has higher binding efficiency with adiponectin receptor compared to adiponectin. PcOSM was effectively taken up by MDAMB231 cancer cells which resulted in marked increase in intracellular ROS levels leading to senescence and cell cycle arrest in G2/M stage. This study provides evidence on the ROS mediated direct inhibitory activity of the plant derived osmotin protein on the phytopathogen Phytophthora capsici, and the additional functional roles of this plant defense protein on cancer cells through inducing ROS associated senescence. The strong leads produced from this study could be pursued further to obtain more insights into the therapeutic potential of osmotin in human cancers.
Collapse
Affiliation(s)
- Rajeswari Gopal Geetha
- Plant Disease Biology Laboratory, Rajiv Gandhi Centre for Biotechnology, Jagathy, Thycaud P.O., Thiruvananthapuram 695014, Kerala, India; (R.G.G.); (G.G.S.)
| | | | - Gayathri G. Saraswathy
- Plant Disease Biology Laboratory, Rajiv Gandhi Centre for Biotechnology, Jagathy, Thycaud P.O., Thiruvananthapuram 695014, Kerala, India; (R.G.G.); (G.G.S.)
| | - Asha Nair Sivakumari
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thycaud P.O., Thiruvananthapuram 695014, Kerala, India;
| | - Manjula Sakuntala
- Plant Disease Biology Laboratory, Rajiv Gandhi Centre for Biotechnology, Jagathy, Thycaud P.O., Thiruvananthapuram 695014, Kerala, India; (R.G.G.); (G.G.S.)
- Correspondence:
| |
Collapse
|
13
|
Li Y, Liu M, Yi J, Song X, Zheng X, Liu D, Wang S, Chu C, Yang J. Exogenous hydrogen sulfide inhibits apoptosis by regulating endoplasmic reticulum stress-autophagy axis and improves myocardial reconstruction after acute myocardial infarction. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1325-1336. [PMID: 33210714 DOI: 10.1093/abbs/gmaa133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
During acute myocardial infarction, endoplasmic reticulum (ER) stress-induced autophagy and apoptosis have been shown as important pathogeneses of myocardial reconstruction. Importantly, hydrogen sulfide (H2S), as a third endogenous gas signaling molecule, exerts strong cytoprotective effect on anti-ER stress, autophagy regulation and antiapoptosis. Here, we showed that H2S treatment inhibits apoptosis by regulating ER stress-autophagy axis and improves myocardial reconstruction after acute myocardial infarction. We found that H2S intervention improved left ventricle function, reduced glycogen deposition in myocardial tissue mesenchyme, and inhibited apoptosis. Moreover, the expressions of fibrosis indicators (Col3a1 and Col1a2), ER stress-related proteins (CHOP and BIP/ERP78), autophagy-related proteins (Beclin and ATG5), apoptosis protein (Bax), as well as fibrosis protein Col4a3bp were all decreased after treatment with H2S. H2S administration also maintained MMP/TIMP balance. Mechanistically, H2S activated the PI3K/AKT signaling pathway. In addition, H2S treatment also reduced the expressions of ER stress-related proteins, autophagy-related proteins, and apoptins in in vitro experiments. Interestingly, activation of ER stress-autophagy axis could reverse the inhibitory effect of H2S on myocardial apoptosis. Altogether, these results suggested that exogenous H2S suppresses myocardial apoptosis by blocking ER stress-autophagy axis, which in turn reverses cardiac remodeling after myocardial infarction.
Collapse
Affiliation(s)
- Yaling Li
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Maojun Liu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Jiali Yi
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Xiong Song
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Xia Zheng
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Da Liu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Sen Wang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Chun Chu
- Department of Pharmacy, The Second Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| |
Collapse
|
14
|
Long non-coding RNA plasmacytoma variant translocation 1 linked to hypoxia-induced cardiomyocyte injury of H9c2 cells by targeting miR-135a-5p/forkhead box O1 axis. Chin Med J (Engl) 2020; 133:2953-2962. [PMID: 33093283 PMCID: PMC7752684 DOI: 10.1097/cm9.0000000000001147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Myocardial infarction occurs due to insufficient (ischemia) blood supply to heart for long time; plasmacytoma variant translocation 1 (PVT1) is a long non-coding RNAs (lncRNAs) involved in the pathogenesis of various diseases, including heart disease; However, few studies have explored its role. The present study evaluated the effects of lncRNA PVT1 on hypoxic rat H9c2 cells. Methods Hypoxic injury was examined by measuring cell viability and apoptosis by using cell counting kit-8 activity and flow cytometry assays. Gene expressions after hypoxia were estimated by quantitative real time polymerase chain reaction and the signaling pathway were explored by Western blot analysis. RNA immunoprecipitation and luciferase reporter assays were applied to examine the interactions among genes. Data were analyzed using t-test with one-way or two-way analysis of variance. Results The lncRNA PVT1 is up-regulated in hypoxia-stressed H9c2 cells and knockdown of PVT1 mitigates hypoxia-induced injury in H9c2 cells. PVT1 acts as a sponge for miR-135a-5p and knockdown of PVT1 attenuated the increased hypoxia-induced injury by up-regulating miR-135a-5p. Forkhead box O1 (FOXO1) was identified as a target of miR-135a-5p, and the expression was negatively regulated by miR-135a-5p. The exploration of the underlying mechanism demonstrated that knockdown of FOXO1 reversed PVT1/miR-135a-5p mediated hypoxia-induced injury in H9c2 cells. Conclusions PVT1 plays a crucial role in hypoxia-injured H9c2 cells through sponging miR-135a-5p and then positively regulating FOXO1.
Collapse
|
15
|
Carballo MCS, Pinto LCS, Brito MVH. The role of adiponectin in ischemia-reperfusion syndrome: a literature review. EINSTEIN-SAO PAULO 2020; 18:eRW5160. [PMID: 32876087 PMCID: PMC7444600 DOI: 10.31744/einstein_journal/2020rw5160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 12/03/2019] [Indexed: 02/05/2023] Open
Abstract
Adiponectin, among other diverse adipokines, is produced in greater quantity and has an effect on the adipose tissue and other tissues in the body. Adiponectin plays three main roles: regulatory metabolic and sensitizing function of insulin in the liver and muscles; it acts as an anti-inflammatory cytokine and in vascular protection, besides important cardiac protection in the presence of ischemia-reperfusion syndrome. Since many situations resulting from traumatic accidents or pathologies are due to cell damage caused by ischemia-reperfusion syndrome, it is relevant to study new therapeutic alternatives that will contribute to reducing these lesions. The objective of this study is to carry out a literature review on the role of adiponectin in ischemia-reperfusion syndrome.
Collapse
|
16
|
Vahabzadeh G, Soltani H, Barati M, Golab F, Jafari-Sabet M, Safari S, Moazam A, Mohamadrezaei H. Noscapine protects the H9c2 cardiomyocytes of rats against oxygen-glucose deprivation/reperfusion injury. Mol Biol Rep 2020; 47:5711-5719. [PMID: 32648076 DOI: 10.1007/s11033-020-05549-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/23/2020] [Indexed: 11/29/2022]
Abstract
Noscapine is an antitumor alkaloid derived from Papaver somniferum plants. Our previous study has demonstrated that exposure of noscapine on primary murine fetal cortical neurons exposed to oxygen-glucose deprivation/reperfusion (OGD/R) has neuroprotective effects. In current study, the effects of noscapine on cardiomyocytes (H9c2 cells) damage caused by 120 minutes (min) of OGD/R were evaluated and we determined whether the addition of BD1047, sigma-one receptor antagonist, prevents the protective effects of noscapine in H9c2 cells through the production of nitric oxide (NO) and apoptosis. To initiate OGD, H9c2 cells was transferred to glucose-free DMEM, and placed in a humidified incubation chamber. Cell viability was assessed with noscapine (1-5 μM) in the presence or absence of BD1047, 24 hours (h) after OGD/R. Cell viability, NO production and apoptosis ratio were evaluated by the MTT assay, the Griess method and the quantitative real-time PCR. Noscapine considerably improved the survival of H9c2 cells compared to OGD/R. Also, noscapine was extremely capable of reducing the concentrations of NO and Bax/Bcl-2 ratio expression. While the BD1047 administration alone diminished cell viability and increased the Bax/Bcl-2 ratio and NO levels. The addition of noscapine in the presence of BD1047 did not increase the cell viability relative to noscapine alone. Noscapine exerted cardioprotective effects exposed to OGD/R-induced injury in H9c2 cells, at least partly via attenuation of NO production and Bax/Bcl-2 ratio, which indicates that the sigma-one receptor activation is involved in the protection by noscapine of H9c2 cells injured by OGD/R.
Collapse
Affiliation(s)
- Gelareh Vahabzadeh
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Science, Tehran, Iran.
| | - Hamidreza Soltani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Barati
- Department of Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | - Fereshteh Golab
- Cellular and Molecular Research Center, Iran University of Medical Science, Tehran, Iran
| | - Majid Jafari-Sabet
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Safari
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ashrafolsadat Moazam
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hananeh Mohamadrezaei
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| |
Collapse
|
17
|
Li L, Li ZB, Jia M, Chu HT. Therapeutic effects of KANK2 in myocardial infarction rats might be associated with the NF-κB p65 inhibition. Int Immunopharmacol 2020; 86:106687. [PMID: 32570033 DOI: 10.1016/j.intimp.2020.106687] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/05/2020] [Accepted: 06/05/2020] [Indexed: 01/07/2023]
Abstract
OBJECTIVE KN motif and ankyrin repeat domains 2 (KANK2) may inhibit the activation of (NF-kappaB) p65, which plays a role in myocardial injury. Thus, our study aims to discover the effect of KANK2 on myocardial infarction (MI) induced by ligating the left anterior descending coronary artery (LAD) through regulating NF-κB p65 in vivo. METHODS MI rats underwent LAD ligation were administered with intramyocardial injections of KANK2/Control activation plasmids. Six weeks after MI, pressure-volume (P/V) loops was used to investigate the cardiac function of rats, then the following detections were performed, including TTC staining, HE staining, immunofluorescence, Masson' s trichrome staining, ELISA assay, TUNEL staining, immunohistochemistry, qRT-PCR and Western blotting. RESULTS MI rats decreased in maximum pressure (pmax), ejection fraction (EF%), peak rate of pressure rise (dpdtmax) and decline (-dpdtmax) with increased end diastolic pressure (EDP), which was partially reversed by KANK2 overexpression. Besides, KANK2 CRISPR activation plasmids reduced infarct size with less collagen fiber proliferation and neutrophil infiltration in infarct tissues, as well as suppressed pro-inflammatory factors expressions in MI rats. Moreover, injection of KANK2 activation plasmid decreased collagen deposition, aggravated cardiomyocyte apoptosis, enhanced the capillary density, and increased the expressions of VEGF and bFGF in the infarct and peri-infarct regions of MI rats. KANK2 lowered myocardial NF-κB p65 expression in MI rats. CONCLUSION KANK2 may play its therapeutic role in MI through improving cardiac function, decreasing myocardial collagen deposition, reducing cardiomyocyte apoptosis, and increasing angiogenesis, which might be associated with the reduction of NF-κB p65 expression.
Collapse
Affiliation(s)
- Lin Li
- Department of Cardiovascular Medicine, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, PR China
| | - Zai-Bo Li
- Department of Cardiovascular Medicine, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, PR China
| | - Min Jia
- Department of Cardiovascular Medicine, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, PR China
| | - Hong-Tao Chu
- Department of Cardiovascular Medicine, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, PR China.
| |
Collapse
|
18
|
Yu ZP, Yu HQ, Li J, Li C, Hua X, Sheng XS. Troxerutin attenuates oxygen‑glucose deprivation and reoxygenation‑induced oxidative stress and inflammation by enhancing the PI3K/AKT/HIF‑1α signaling pathway in H9C2 cardiomyocytes. Mol Med Rep 2020; 22:1351-1361. [PMID: 32626962 PMCID: PMC7339651 DOI: 10.3892/mmr.2020.11207] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 08/16/2019] [Indexed: 01/04/2023] Open
Abstract
Myocardial ischemia-reperfusion (MI/R) injury is a complex pathological process that occurs when tissues are reperfused following a prolonged period of ischemia. Troxerutin has been reported to have cardioprotective functions. However, the underlying mechanism by which troxerutin protects against MI/R injury has not been fully elucidated. The aim of the present study was to explore whether troxerutin-mediated protection against oxygen-glucose deprivation/reoxygenation (OGD/R)-induced H9C2 cell injury was associated with the inhibition of oxidative stress and the inflammatory response by regulating the PI3K/AKT/hypoxia-inducible factor-1α (HIF-1α) signaling pathway. The results of the present study suggested that troxerutin pretreatment prevented the OGD/R-induced reduction in cell viability, and the increase in lactate dehydrogenase activity and apoptosis. Troxerutin reversed OGD/R-induced the inhibition of the PI3K/AKT/HIF-1α signaling pathway as demonstrated by the increased expression of PI3K and HIF-1α, and the increased ratio of phosphorylated AKT/AKT. LY294002, a selective PI3K inhibitor, inhibited the PI3K/AKT/HIF-1α signaling pathway and further attenuated the protective effect of troxerutin against OGD/R-induced H9C2 cell damage. Furthermore, small interfering (si)RNA-mediated knockdown of HIF-1α reduced troxerutin-induced protection against OGD/R injury. Troxerutin pretreatment alleviated OGD/R-induced oxidative stress, as demonstrated by the reduced generation of reactive oxygen species and malonaldehyde content, and the increased activities of superoxide dismutase and glutathione peroxidase, which were reduced by HIF-1α-siRNA. Troxerutin-induced decreases in the levels of interleukin (IL)-1β, IL-6 and tumor necrosis factor-α in OGD/R conditions were also reduced by HIF-1α-siRNA. The results from the present study indicated that troxerutin aggravated OGD/R-induced H9C2 cell injury by inhibiting oxidative stress and the inflammatory response. The primary underlying protective mechanism of troxerutin was mediated by the activation of the PI3K/AKT/HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Zhang-Ping Yu
- Department of Cardiology, Jinhua People's Hospital, Jinhua, Zhejiang 321000, P.R. China
| | - Han-Qiao Yu
- Department of Cardiology, Jinhua People's Hospital, Jinhua, Zhejiang 321000, P.R. China
| | - Jun Li
- Department of Cardiology, Jinhua People's Hospital, Jinhua, Zhejiang 321000, P.R. China
| | - Chao Li
- Department of Cardiology, Jinhua People's Hospital, Jinhua, Zhejiang 321000, P.R. China
| | - Xian Hua
- Department of Cardiology, Jinhua People's Hospital, Jinhua, Zhejiang 321000, P.R. China
| | - Xiao-Sheng Sheng
- Department of Cardiology, Jinhua People's Hospital, Jinhua, Zhejiang 321000, P.R. China
| |
Collapse
|
19
|
Wenxin Granule Ameliorates Hypoxia/Reoxygenation-Induced Oxidative Stress in Mitochondria via the PKC- δ/NOX2/ROS Pathway in H9c2 Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3245483. [PMID: 32566078 PMCID: PMC7260629 DOI: 10.1155/2020/3245483] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/24/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Myocardial infarction and following reperfusion therapy-induced myocardial ischemia/reperfusion (I/R) injury have been recognized as an important subject of cardiovascular disease with high mortality. As the antiarrhythmic agent, Wenxin Granule (WXG) is widely used to arrhythmia and heart failure. In our pilot study, we found the antioxidative potential of WXG in the treatment of myocardial I/R. This study is aimed at investigating whether WXG could treat cardiomyocyte hypoxia/reoxygenation (H/R) injury by inhibiting oxidative stress in mitochondria. The H9c2 cardiomyocyte cell line was subject to H/R stimuli to mimic I/R injury in vitro. WXG was added to the culture medium 24 h before H/R exposing as pretreatment. Protein kinase C-δ (PKC-δ) inhibitor rottlerin or PKC-δ lentivirus vectors were conducted on H9c2 cells to downregulate or overexpress PKC-δ protein. Then, the cell viability, oxidative stress levels, intracellular and mitochondrial ROS levels, mitochondrial function, and apoptosis index were analyzed. In addition, PKC-δ protein expression in each group was verified by western blot analysis. Compared with the control group, the PKC-δ protein level was significantly increased in the H/R group, which was remarkably improved by WXG or rottlerin. PKC-δ lentivirus vector-mediated PKC-δ overexpression was not reduced by WXG. WXG significantly improved H/R-induced cell injury, lower levels of SOD and GSH/GSSG ratio, higher levels of MDA, intracellular and mitochondrial ROS content, mitochondrial membrane potential and ATP loss, mitochondrial permeability transition pore opening, NOX2 activation, cytochrome C release, Bax/Bcl-2 ratio and cleaved caspase-3 increasing, and cell apoptosis. Similar findings were obtained from rottlerin treatment. However, the protective effects of WXG were abolished by PKC-δ overexpression, indicating that PKC-δ was a potential target of WXG treatment. Our findings demonstrated a novel mechanism by which WXG attenuated oxidative stress and mitochondrial dysfunction of H9c2 cells induced by H/R stimulation via inhibitory regulation of PKC-δ/NOX2/ROS signaling.
Collapse
|
20
|
Xu Y, Tang C, Tan S, Duan J, Tian H, Yang Y. Cardioprotective effect of isorhamnetin against myocardial ischemia reperfusion (I/R) injury in isolated rat heart through attenuation of apoptosis. J Cell Mol Med 2020; 24:6253-6262. [PMID: 32307912 PMCID: PMC7294129 DOI: 10.1111/jcmm.15267] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/19/2020] [Accepted: 03/26/2020] [Indexed: 12/16/2022] Open
Abstract
In this study, we investigated the effects of isorhamnetin on myocardial ischaemia reperfusion (I/R) injury in Langendorff‐perfused rat hearts. Isorhamnetin treatment (5, 10 and 20 μg/mL) significantly alleviated cardiac morphological injury, reduced myocardial infarct size, decreased the levels of marker enzymes (LDH and CK) and improved the haemodynamic parameters, reflected by the elevated levels of the left ventricular developed pressure (LVDP), coronary flow (CF) and the maximum up/down velocity of left ventricular pressure (+dp/dtmax). Moreover, isorhamnetin reperfusion inhibited apoptosis of cardiomyocytes in the rats subjected to cardiac I/R in a dose‐dependent manner concomitant with decreased protein expression of Bax and cleaved‐caspase‐3, as well as increased protein expression of Bcl‐2. In addition, I/R‐induced oxidative stress was manifestly mitigated by isorhamnetin treatment, as showed by the decreased malondialdehyde (MDA) level and increased antioxidant enzymes activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH‐Px). These results indicated that isorhamnetin exerts a protective effect against I/R‐induced myocardial injury through the attenuation of apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Yan Xu
- Department of Geriatrics, the Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Chun Tang
- Department of Nephrology, Center of Nephrology and Urology, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, PR China
| | - Shengyu Tan
- Department of Geriatrics, the Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Juan Duan
- Department of Geriatrics, the Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Hongmei Tian
- Department of Geriatrics, the Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Yu Yang
- Department of Geriatrics, the Second Xiangya Hospital, Central South University, Changsha, PR China
| |
Collapse
|
21
|
Abou-Samra M, Selvais CM, Dubuisson N, Brichard SM. Adiponectin and Its Mimics on Skeletal Muscle: Insulin Sensitizers, Fat Burners, Exercise Mimickers, Muscling Pills … or Everything Together? Int J Mol Sci 2020; 21:ijms21072620. [PMID: 32283840 PMCID: PMC7178193 DOI: 10.3390/ijms21072620] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 02/07/2023] Open
Abstract
Adiponectin (ApN) is a hormone abundantly secreted by adipocytes and it is known to be tightly linked to the metabolic syndrome. It promotes insulin-sensitizing, fat-burning, and anti-atherosclerotic actions, thereby effectively counteracting several metabolic disorders, including type 2 diabetes, obesity, and cardiovascular diseases. ApN is also known today to possess powerful anti-inflammatory/oxidative and pro-myogenic effects on skeletal muscles exposed to acute or chronic inflammation and injury, mainly through AdipoR1 (ApN specific muscle receptor) and AMP-activated protein kinase (AMPK) pathway, but also via T-cadherin. In this review, we will report all the beneficial and protective properties that ApN can exert, specifically on the skeletal muscle as a target tissue. We will highlight its effects and mechanisms of action, first in healthy skeletal muscle including exercised muscle, and second in diseased muscle from a variety of pathological conditions. In the end, we will go over some of AdipoRs agonists that can be easily produced and administered, and which can greatly mimic ApN. These interesting and newly identified molecules could pave the way towards future therapeutic approaches to potentially prevent or combat not only skeletal muscle disorders but also a plethora of other diseases with sterile inflammation or metabolic dysfunction.
Collapse
|
22
|
Niedziela M, Wojciechowska M, Zarębiński M, Cudnoch-Jędrzejewska A, Mazurek T. Adiponectin promotes ischemic heart preconditioning- PRO and CON. Cytokine 2020; 127:154981. [PMID: 31911263 DOI: 10.1016/j.cyto.2019.154981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 12/07/2019] [Accepted: 12/27/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Magdalena Niedziela
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Małgorzata Wojciechowska
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland; Independent Public Specialist Western Hospital John Paul II in Grodzisk Mazowiecki, Poland.
| | - Maciej Zarębiński
- Independent Public Specialist Western Hospital John Paul II in Grodzisk Mazowiecki, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Mazurek
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Poland
| |
Collapse
|
23
|
Li P, Lin N, Guo M, Huang H, Yu T, Zhang L. REDD1 knockdown protects H9c2 cells against myocardial ischemia/reperfusion injury through Akt/mTORC1/Nrf2 pathway-ameliorated oxidative stress: An in vitro study. Biochem Biophys Res Commun 2019; 519:179-185. [DOI: 10.1016/j.bbrc.2019.08.095] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 08/16/2019] [Indexed: 01/06/2023]
|
24
|
Liu B, Liu J, Wang J, Sun F, Jiang S, Hu F, Wang D, Liu D, Liu C, Yan H. Adiponectin Protects Against Cerebral Ischemic Injury Through AdipoR1/AMPK Pathways. Front Pharmacol 2019; 10:597. [PMID: 31231213 PMCID: PMC6558395 DOI: 10.3389/fphar.2019.00597] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 05/09/2019] [Indexed: 12/11/2022] Open
Abstract
Excitotoxicity induced by excessive N-methyl-D-aspartate (NMDA) receptor activation underlies the pathology of ischemic injury. Adiponectin (APN) is an adipocyte-derived protein hormone that modulates a number of metabolic processes. APN exerts a wide range of biological functions in the central nervous system. However, the role of APN and its receptors in cerebral ischemia/reperfusion (I/R)-induced injury and the related mechanisms remain to be clarified. Here, we found that APN and APN receptor agonist AdipoRon (APR) were protective against excitotoxicity induced by oxygen and glucose deprivation/reperfusion (OGD/R) and NMDA in primary neurons. Adiponectin receptor 1 (AdipoR1) knockdown reversed the protection conferred by either APN or APR. Moreover, the protective effects offered by both APN and APR were compromised by compound C, an inhibitor of amp-activated protein kinase (AMPK) phosphorylation. Both APN and APR protected the dissipation of the ΔΨm caused by OGD/R. They also up-regulated the PGC-1α expression, which was reversed by compound C. Furthermore, both APN and APR ameliorated but APN knockout aggravated the infarct volume and neurological deficient induced by transient middle cerebral artery occlusion (tMCAO) in vivo. Taken together, these findings show that APN and APR protect against ischemic injury in vitro and in vivo. The protective mechanism is mainly related to AdipoR1-dependent AMPK phosphorylation and PGC-1α up-regulation.
Collapse
Affiliation(s)
- Bin Liu
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Jing Liu
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Jiangong Wang
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Fengjiao Sun
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Shujun Jiang
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Fengai Hu
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Dan Wang
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Dunjiang Liu
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Cuilan Liu
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Haijing Yan
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
25
|
Liu S, He Y, Shi J, Liu L, Ma H, He L, Guo Y. STAT1-avtiviated LINC00961 regulates myocardial infarction by the PI3K/AKT/GSK3β signaling pathway. J Cell Biochem 2019; 120:13226-13236. [PMID: 30887575 DOI: 10.1002/jcb.28596] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/02/2019] [Accepted: 02/14/2019] [Indexed: 02/05/2023]
Abstract
Myocardial infarction (MI) remains a severe cardiac disease because of its high incidence and mortality worldwide. A growing body of recent investigations have confirmed that LINC00961 acts as a tumor suppressor in diverse malignancies. However, the biological significance of LINC00961 and its molecular mechanism in MI are still elusive. Hypoxia is the leading cause of MI and induces myocardial injury. In this study, we found the upregulated expression of LINC00961 in cardiomyocytes H9c2 after hypoxia treatment. Knockdown of LINC00961 ameliorated hypoxia-induced cell injury by facilitating cell viability while repressing cell apoptosis. The significant increase of signal transducer and activator of transcription 1 (STAT1) expression and phosphorylation levels was observed in hypoxia-induced cells and proved to exacerbate hypoxia injury. In addition, STAT1 transcriptionally activated LINC00961 by binding to LINC00961 promoter. Furthermore, our results validated that suppressing LINC00961 contributed to the remarkable diminution in the phosphorylation levels of phosphoinositide 3-kinases (PI3K), AKT, and glycogen synthase kinase-3β (GSK3β). Inhibition of PI3K/AKT signaling or GSK3β pathway rescued the effects of LINC00961 knockdown on the hypoxia-induced injury of cardiomyocytes. Namely, we concluded that STAT1-avtiviated LINC00961 accelerated MI via the PI3K/AKT/GSK3β pathway, which may provide clues for the treatment of patients with MI.
Collapse
Affiliation(s)
- Shengzhong Liu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Cardiac Surgery Center, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Ying He
- Psychosomatic Medicine Center, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Jun Shi
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lulu Liu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Ma
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li He
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingqiang Guo
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
26
|
Zhao L, Zhang JH, Sherchan P, Krafft PR, Zhao W, Wang S, Chen S, Guo Z, Tang J. Administration of rCTRP9 Attenuates Neuronal Apoptosis Through AdipoR1/PI3K/Akt Signaling Pathway after ICH in Mice. Cell Transplant 2019; 28:756-766. [PMID: 30642187 PMCID: PMC6686438 DOI: 10.1177/0963689718822809] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Targeting neuronal apoptosis after intracerebral hemorrhage (ICH) may be an important therapeutic strategy for ICH patients. Emerging evidence indicates that C1q/TNF-Related Protein 9 (CTRP9), a newly discovered adiponectin receptor agonist, exerts neuroprotection in cerebrovascular disease. The aim of this study was to investigate the anti-apoptotic role of CTRP9 after experimental ICH and to explore the underlying molecular mechanisms. ICH was induced in mice via intrastriatal injection of bacterial collagenase. Recombinant CTRP9 (rCTRP9) was administrated intranasally at 1 h after ICH. To elucidate the underlying mechanisms, adiponectin receptor1 small interfering ribonucleic acid (AdipoR1 siRNA) and selective PI3 K inhibitor LY294002 were administered prior to rCTRP9 treatment. Western blots, neurofunctional assessments, immunofluorescence staining, and Fluoro-Jade C (FJC) staining experiments were performed. Administration of rCTRP9 significantly improved both short- and long-term neurofunctional behavior after ICH. RCTRP9 treatment significantly increased the expression of AdipoR1, PI3 K, p-Akt, and Bcl-2, while at the same time was found to decrease the expression of Bax in the brain, which was reversed by inhibition of AdipoR1 and PI3 K. The neuroprotective effect of rCTRP9 after ICH was mediated by attenuation of neuronal apoptosis via the AdipoR1/PI3K/Akt signaling pathway; therefore, rCTRP9 should be further evaluated as a potential therapeutic agent for ICH patients.
Collapse
Affiliation(s)
- Lianhua Zhao
- 1 Department of Neurology, Tianjin TEDA Hospital, Tianjin, China.,2 Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- 2 Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Prativa Sherchan
- 2 Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Paul R Krafft
- 3 Department of Neurological Surgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Wei Zhao
- 1 Department of Neurology, Tianjin TEDA Hospital, Tianjin, China
| | - Sa Wang
- 4 Department of Neurology, Affilicated Wenling Hospital of Wenzhou Medical University, Wenling, Zhejiang, China
| | - Shengpan Chen
- 2 Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,5 Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, China
| | - Zaiyu Guo
- 1 Department of Neurology, Tianjin TEDA Hospital, Tianjin, China
| | - Jiping Tang
- 2 Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
27
|
Liu Y, Vu V, Sweeney G. Examining the Potential of Developing and Implementing Use of Adiponectin-Targeted Therapeutics for Metabolic and Cardiovascular Diseases. Front Endocrinol (Lausanne) 2019; 10:842. [PMID: 31920962 PMCID: PMC6918867 DOI: 10.3389/fendo.2019.00842] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiometabolic diseases encompass those affecting the heart and vasculature as well as other metabolic problems, such as insulin resistance, diabetes, and non-alcoholic fatty liver disease. These diseases tend to have common risk factors, one of which is impaired adiponectin action. This may be due to reduced bioavailability of the hormone or resistance to its effects on target tissues. A strong negative correlation between adiponectin levels and cardiometabolic diseases has been well-documented and research shown that adiponectin has cardioprotective, insulin sensitizing and direct beneficial metabolic effects. Thus, therapeutic approaches to enhance adiponectin action are widely considered to be desirable. The complexity of adiponectin structure and function has so far made progress in this area less than ideal. In this article we will review the effects and mechanism of action of adiponectin on cardiometabolic tissues, identify scenarios where enhancing adiponectin action would be of clinical value and finally discuss approaches via which this can be achieved.
Collapse
Affiliation(s)
- Ying Liu
- Metabolic Disease Research Division, iCarbonX Co. Ltd., Shenzhen, China
- *Correspondence: Ying Liu
| | - Vivian Vu
- Department of Biology, York University, Toronto, ON, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
- Gary Sweeney
| |
Collapse
|
28
|
Cardioprotective Effects of Puerarin-V on Isoproterenol-Induced Myocardial Infarction Mice Is Associated with Regulation of PPAR-Υ/NF-κB Pathway. Molecules 2018; 23:molecules23123322. [PMID: 30558188 PMCID: PMC6321361 DOI: 10.3390/molecules23123322] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/10/2018] [Accepted: 12/13/2018] [Indexed: 01/10/2023] Open
Abstract
Puerarin is a well-known traditional Chinese medicine which has been used for the treatment of cardiovascular diseases. Recently, a new advantageous crystal form of puerarin, puerarin-V, has been developed. However, the cardioprotective effects of puerarin-V on myocardial infarction (MI) heart failure are still unclear. In this research, we aim to evaluate the cardioprotective effects of puerarin-V on the isoproterenol (ISO)-induced MI mice and elucidate the underlying mechanisms. To induce MI in C57BL/6 mice, ISO was administered at 40 mg/kg subcutaneously every 12 h for three times in total. The mice were randomly divided into nine groups: (1) control; (2) ISO; (3) ISO + puerarin injection; (4⁻9) ISO + puerarin-V at different doses and timings. After treatment, cardiac function was evaluated by electrocardiogram (ECG), biochemical and histochemical analysis. In vitro inflammatory responses and apoptosis were evaluated in human coronary artery endothelial cells (HCAECs) challenged by lipopolysaccharide (LPS). LPS-induced PPAR-Υ/NF-κB and subsequently activation of cytokines were assessed by the western blot and real-time polymerase chain reaction (PCR). Administration of puerarin-V significantly inhibits the typical ST segment depression compared with that in MI mice. Further, puerarin-V treatment significantly improves ventricular wall infarction, decreases the incidence of mortality, and inhibits the levels of myocardial injury markers. Moreover, puerarin-V treatment reduces the inflammatory milieu in the heart of MI mice, thereby blocking the upregulation of proinflammatory cytokines (TNF-α, IL-1β and IL-6). The beneficial effects of puerarin-V might be associated with the normalization in gene expression of PPAR-Υ and PPAR-Υ/NF-κB /ΙκB-α/ΙΚΚα/β phosphorylation. In the in vitro experiment, treatment with puerarin-V (0.3, 1 and 3 μM) significantly reduces cell death and suppresses the inflammation cytokines expression. Likewise, puerarin-V exhibits similar mechanisms. The cardioprotective effects of puerarin-V treatment on MI mice in the pre + post-ISO group seem to be more prominent compared to those in the post-ISO group. Puerarin-V exerts cardioprotective effects against ISO-induced MI in mice, which may be related to the activation of PPAR-γ and the inhibition of NF-κB signaling in vivo and in vitro. Taken together, our research provides a new therapeutic option for the treatment of MI in clinic.
Collapse
|
29
|
6-Gingerol Protects Heart by Suppressing Myocardial Ischemia/Reperfusion Induced Inflammation via the PI3K/Akt-Dependent Mechanism in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:6209679. [PMID: 30519268 PMCID: PMC6241357 DOI: 10.1155/2018/6209679] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/19/2018] [Accepted: 10/25/2018] [Indexed: 01/04/2023]
Abstract
Our previous study has demonstrated that 6-Gingerol (6-G) could alleviate myocardial ischemia/reperfusion injury (MIRI). However, the molecular mechanism underlying the process of myocardial ischemia/reperfusion (I/R) injury alleviation by 6-G remains unelucidated. The objective of the present study is to further investigate the potential mechanism for 6-G to alleviate MIRI in rats. Thirty-two Sprague-Dawley rats were randomly divided into four groups: the Sham group, the I/R group, the 6-G + I/R group, and the LY294002 (LY) + 6-G + I/R group. For the rats in each of the groups, data were collected for cardiogram, cardiac function, area of myocardial infarction, myocardial pathology, myocardial enzyme, marker of inflammatory response, and PI3K/Akt signaling pathway. We found that the pretreatment of 6-G with 6 mg/kg could shrink the ST section of cardiogram, improve the cardiac function, reduce the area of myocardial infarction and the degree of cardiac pathological injury, lower the level of myocardial enzyme, and inhibit the inflammatory response. In addition, our results also indicated that 6-G could upregulate the expression of PI3K and p-Akt and that LY294002, a blocking agent of PI3K/Akt signaling pathway, could nullify the protecting role of 6-G. Our experimental results showed that 6-G could inhibit I/R-induced inflammatory response through the activation of the PI3K/Akt signaling pathway.
Collapse
|
30
|
Trotta MC, Maisto R, Alessio N, Hermenean A, D'Amico M, Di Filippo C. The Melanocortin MC5R as a New Target for Treatment of High Glucose-Induced Hypertrophy of the Cardiac H9c2 Cells. Front Physiol 2018; 9:1475. [PMID: 30416452 PMCID: PMC6212602 DOI: 10.3389/fphys.2018.01475] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/28/2018] [Indexed: 12/15/2022] Open
Abstract
The study explored the anti-hypertrophic effect of the melanocortin MC5R stimulation in H9c2 cardiac myocytes exposed to high glucose. This has been done by using α-MSH and selective MC5R agonists and assessing the expression of GLUT4 and GLUT1 transporters, miR-133 and urotensin receptor levels as a marker of cardiac hypertrophy. The study shows for the first time an up-regulation of MC5R expression levels in H9c2 cardiomyocytes exposed to high glucose medium (33 mM D-glucose) for 48 h, compared to cells grown in normal glucose medium (5.5 mM D-glucose). Moreover, H9c2 cells exposed to high glucose showed a significant reduction in cell viability (-40%), a significant increase in total protein per cell number (+109%), and an increase of the urotensin receptor expression levels as an evidence of cells hypertrophy. The pharmacological stimulation of MC5R with α-MSH (90 pM)of the high glucose exposed H9c2 cells increased the cell survival (+50,8%) and reduced the total protein per cell number (-28,2%) with respect to high glucose alone, confirming a reduction of the hypertrophic state as per cell area measurement. Similarly, PG-901 (selective agonist, 10-10 M) significantly increased cell viability (+61,0 %) and reduced total protein per cell number (-40,2%), compared to cells exposed to high glucose alone. Interestingly, the MC5R agonist reduced the GLUT1/GLUT4 glucose transporters ratio on the cell membranes exhibited by the hypertrophic H9c2 cells and increased the intracellular PI3K activity, mediated by a decrease of the levels of the miRNA miR-133a. The beneficial effects of MC5R agonism on the cardiac hypertrophy caused by high glucose was also observed also by echocardiographic evaluations of rats made diabetics with streptozotocin (65 mg/kg i.p.). Therefore, the melanocortin MC5R could be a new target for the treatment of high glucose-induced hypertrophy of the cardiac H9c2 cells.
Collapse
Affiliation(s)
- Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Rosa Maisto
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Nicola Alessio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Anca Hermenean
- Institute of Life Sciences, "Vasile Goldis" Western University of Arad, Arad, Romania
| | - Michele D'Amico
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Clara Di Filippo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
31
|
Zhang Y, Qiao B, Gao F, Wang H, Miao S, Zhao H. Melatonin protects H9c2 cells against ischemia/reperfusion‑induced apoptosis and oxidative stress via activation of the Nrf2 signaling pathway. Mol Med Rep 2018; 18:3497-3505. [PMID: 30066862 DOI: 10.3892/mmr.2018.9315] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 06/29/2018] [Indexed: 11/05/2022] Open
Abstract
Melatonin can protect against cardiac ischemia/reperfusion (I/R) injury in models in vitro and in vivo by regulating oxidative stress and apoptosis; however, the precise molecular mechanisms involved remain unclear. Nuclear factor erythroid 2‑related factor 2 (Nrf2) is a transcription factor, which has been associated with the regulation of oxidative stress by translocating to the nucleus. Therefore, the present study investigated whether activation of the Nrf2 signaling pathway may be responsible for the protective effects of melatonin on I/R‑injured cardiomyocytes. In the present study, H9c2 cells were subjected to simulated I/R (SIR) injury and pretreated with melatonin and/or Nrf2 small interfering RNA (siRNA). Cell viability was detected via Cell Counting kit‑8 assay, apoptosis was examined by caspase‑3 cleavage and activity analysis; oxidative stress levels were determined by specific activity analysis assays. In the present study, it was observed that SIR induced significant increases in apoptosis and oxidative stress, and enhanced Nrf2 expression within H9c2 cells. Pretreatment with melatonin partially reversed these alterations and promoted Nrf2 nuclear translocation. Transfection with Nrf2 siRNA inhibited the protective effects of melatonin on SIR‑induced H9c2 cells. These results indicated that melatonin may protect H9c2 cells against I/R injury by reducing apoptosis and oxidative stress; this effect may be mediated via activation of the Nrf2 signaling pathway. Collectively, the results of the present study may suggest melatonin as a potential therapeutic agent against cardiac I/R injury.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, Heze Municipal Hospital, Heze, Shandong 274031, P.R. China
| | - Baoguang Qiao
- Department of Anesthesiology, Heze Municipal Hospital, Heze, Shandong 274031, P.R. China
| | - Fei Gao
- Department of Anesthesiology, Heze Municipal Hospital, Heze, Shandong 274031, P.R. China
| | - Haifeng Wang
- Department of Anesthesiology, Heze Municipal Hospital, Heze, Shandong 274031, P.R. China
| | - Shaohua Miao
- Department of Anesthesiology, Heze Municipal Hospital, Heze, Shandong 274031, P.R. China
| | - Huan Zhao
- Department of Anesthesiology, Heze Municipal Hospital, Heze, Shandong 274031, P.R. China
| |
Collapse
|
32
|
Takahashi Y, Watanabe R, Sato Y, Ozawa N, Kojima M, Watanabe-Kominato K, Shirai R, Sato K, Hirano T, Watanabe T. Novel phytopeptide osmotin mimics preventive effects of adiponectin on vascular inflammation and atherosclerosis. Metabolism 2018; 83:128-138. [PMID: 29410350 DOI: 10.1016/j.metabol.2018.01.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 12/08/2017] [Accepted: 01/17/2018] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The novel phytohormone, osmotin, has been reported to act like mammalian adiponectin through PHO36/AdipoR1 in various in vitro and in vivo models. However, there have been no reports regarding the precise effects of osmotin on atherosclerosis. METHODS We assessed the atheroprotective effects of osmotin on inflammatory molecules in human umbilical vein endothelial cells (HUVECs), human leukemic monocyte (THP-1) adhesion, inflammatory responses, and foam cell formation in THP-1-derived macrophages, and the migration, proliferation, and extracellular matrix expression in human aortic smooth muscle cells (HASMCs). We examined whether 4-week infusion of osmotin could suppress the development of aortic atherosclerotic lesions in apolipoprotein E-deficient (ApoE-/-) mice. RESULTS AdipoR1 was abundantly expressed in HUVECs, HASMCs, THP-1, and derived macrophages. Osmotin suppressed lipopolysaccharide-induced upregulation of tumor necrosis factor-α (TNF-α), monocyte chemotactic protein-1, vascular cell adhesion molecule-1, intercellular adhesion molecule-1, and E-selectin in HUVECs, and TNF-α-induced THP-1-HUVEC adhesion. In THP-1-derived macrophages, osmotin suppressed the inflammatory M1 phenotype, lipopolysaccharide-induced secretion of interleukin-6 and TNF-α, and oxidized low-density lipoprotein-induced foam cell formation associated with CD36 and acyl-CoA:cholesterol acyltransferase 1 downregulation and ATP-binding cassette transporter A1 upregulation. In HASMCs, osmotin suppressed angiotensin II-induced migration, proliferation, collagen-1 and fibronectin expression, and matrix metalloproteinase-2 activity without inducing apoptosis. Infusion of osmotin into ApoE-/- mice prevented the development of aortic atherosclerotic lesions with reductions of intraplaque pentraxin-3 expression, fasting plasma glucose, and insulin resistance. CONCLUSIONS This study provided the first evidence that osmotin exerts preventive effects on vascular inflammation and atherosclerosis, which may facilitate the development of new therapeutic modalities for combating atherosclerosis and related diseases.
Collapse
Affiliation(s)
- Yui Takahashi
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Rena Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Yuki Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Nana Ozawa
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Miho Kojima
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Kaho Watanabe-Kominato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Remina Shirai
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Kengo Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Tsutomu Hirano
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan
| | - Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan.
| |
Collapse
|