1
|
Cui J, Li H, Zhang T, Lin F, Chen M, Zhang G, Feng Z. Research progress on the mechanism of curcumin anti-oxidative stress based on signaling pathway. Front Pharmacol 2025; 16:1548073. [PMID: 40260389 PMCID: PMC12009910 DOI: 10.3389/fphar.2025.1548073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Oxidative stress refers to an imbalance between oxidative capacity and antioxidant capacity, leading to oxidative damage to proteins, lipids, and DNA, which can result in cell senescence or death. It is closely associated with the occurrence and development of various diseases, including cardiovascular diseases, nephropathy, malignant tumors, neurodegenerative diseases, hypertension, diabetes, and inflammatory diseases. Curcumin is a natural polyphenol compound of β-diketone, which has a wide range of pharmacological activities such as anti-inflammatory, antibacterial, anti-oxidative stress, anti-tumor, anti-fibrosis, and hypolipidemic, demonstrating broad research and development value. It has a wide range of biological targets and can bind to various endogenous biomolecules. Additionally, it maintains the redox balance primarily by scavenging ROS, enhancing the activity of antioxidant enzymes, inhibiting lipid peroxidation, and chelating metal ions. This paper systematically describes the antioxidative stress mechanisms of curcumin from the perspective of signaling pathways, focusing on the Keap1-Nrf2/ARE, NF-κB, NOX, MAPK and other pathways. The study also discusses potential pathway targets and the complex crosstalk among these pathways, aiming to provide insights for further research on curcumin's antioxidant mechanisms and its clinical applications.
Collapse
Affiliation(s)
- Jie Cui
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haonan Li
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianyi Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fengli Lin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Meiyun Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Guimin Zhang
- Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Zhong Feng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
- Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| |
Collapse
|
2
|
He Y, Zhao Y, Lv RJ, Dong N, Wang X, Yu Q, Yue HM. Curcumin activates the Wnt/β-catenin signaling pathway to alleviate hippocampal neurogenesis abnormalities caused by intermittent hypoxia: A study based on network pharmacology and experimental verification. Int Immunopharmacol 2024; 143:113299. [PMID: 39362017 DOI: 10.1016/j.intimp.2024.113299] [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: 06/09/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
The purpose of this work was to investigate how curcumin (Cur) might enhance cognitive function and to gain a better understanding of the molecular mechanisms behind Cur's impacts on neurogenesis deficits brought on by intermittent hypoxia (IH). Using network pharmacology, we explored possible targets for Cur's obstructive sleep apnea (OSA) therapy. We established an IH model using C57BL/6 mice and c17.2 cells, and we assessed the influence of Cur on treatment outcomes as well as the effect of IH on cognitive function. Hippocampal damage and neurogenesis, as well as expression of core targets, were then examined. Network pharmacology analysis revealed that Cur has the potential for multi-target, multi-pathway therapy, with CTNNB1 and MYC as core target genes. The Morris water maze test showed that Cur (100 mg/kg, intragastrically) significantly improved cognitive dysfunction induced by IH. The hematoxylin and eosin (H&E) and Nissl staining indicated that Cur could alleviate damage to the hippocampus caused by IH. Immunohistochemistry, immunofluorescence, and western blotting results showed that Cur might promote neurogenesis and upregulate the expression of β-catenin and c-myc. In vitro, Cur (0.5 μM) has a protective effect on IH-induced neural stem cells (NSCs) injury and apoptosis and can restore the Wnt/β-catenin. Cur significantly increased the neurogenesis via the Wnt/β-catenin pathway, providing the scientific groundwork for the development of new treatment strategies for neurological damage linked to OSA.
Collapse
Affiliation(s)
- Yao He
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Yan Zhao
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Ren-Jun Lv
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Na Dong
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Xiao Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Qin Yu
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China; Department of Respiratory and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Hong-Mei Yue
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China; Department of Respiratory and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China.
| |
Collapse
|
3
|
He Y, Zhao Y, Lv RJ, Dong N, Wang X, Yu Q, Yue HM. Curcumin triggers the Wnt/β-catenin pathway and shields neurons from injury caused by intermittent hypoxia. Tissue Cell 2024; 91:102587. [PMID: 39454474 DOI: 10.1016/j.tice.2024.102587] [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: 05/10/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
The objective of this study was to explore the molecular basis through which Curcumin (Cur) mitigates neuronal damage caused by obstructive sleep apnea (OSA). HT22 was used to simulate intermittent hypoxia (IH) injury and explore the effect of Cur on these cells. We evaluated the cell viability, cytotoxicity, apoptosis, proliferation, and Wnt/β-catenin (WβC) pathway. IWR-1 was used to block the pathway and investigate the protective mechanism of Cur. We constructed an in vivo model of IH to validate the results of the cellular experiments. IH accelerated apoptosis and cytotoxicity, suppressed proliferation, and decreased the activity of the WβC pathway. Cur can significantly improve cell viability, reduce apoptosis rate and cell toxicity, promote cell proliferation, and up-regulate the WβC. After blocking the WβC pathway, the proliferative effect of Cur was observably weakened. In vivo, IH caused hippocampal damage and inhibited WβC pathway activity in mice, which was ameliorated by Cur treatment. This implies that Cur could be a novel treatment option for neurological impairment brought on by OSA.
Collapse
Affiliation(s)
- Yao He
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yan Zhao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Ren-Jun Lv
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Na Dong
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xiao Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Qin Yu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Respiratory and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Hong-Mei Yue
- The First Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Respiratory and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
4
|
Burtscher J, Citherlet T, Camacho-Cardenosa A, Camacho-Cardenosa M, Raberin A, Krumm B, Hohenauer E, Egg M, Lichtblau M, Müller J, Rybnikova EA, Gatterer H, Debevec T, Baillieul S, Manferdelli G, Behrendt T, Schega L, Ehrenreich H, Millet GP, Gassmann M, Schwarzer C, Glazachev O, Girard O, Lalande S, Hamlin M, Samaja M, Hüfner K, Burtscher M, Panza G, Mallet RT. Mechanisms underlying the health benefits of intermittent hypoxia conditioning. J Physiol 2024; 602:5757-5783. [PMID: 37860950 DOI: 10.1113/jp285230] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
Intermittent hypoxia (IH) is commonly associated with pathological conditions, particularly obstructive sleep apnoea. However, IH is also increasingly used to enhance health and performance and is emerging as a potent non-pharmacological intervention against numerous diseases. Whether IH is detrimental or beneficial for health is largely determined by the intensity, duration, number and frequency of the hypoxic exposures and by the specific responses they engender. Adaptive responses to hypoxia protect from future hypoxic or ischaemic insults, improve cellular resilience and functions, and boost mental and physical performance. The cellular and systemic mechanisms producing these benefits are highly complex, and the failure of different components can shift long-term adaptation to maladaptation and the development of pathologies. Rather than discussing in detail the well-characterized individual responses and adaptations to IH, we here aim to summarize and integrate hypoxia-activated mechanisms into a holistic picture of the body's adaptive responses to hypoxia and specifically IH, and demonstrate how these mechanisms might be mobilized for their health benefits while minimizing the risks of hypoxia exposure.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Tom Citherlet
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Alba Camacho-Cardenosa
- Department of Physical Education and Sports, Faculty of Sports Science, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
| | - Marta Camacho-Cardenosa
- Clinical Management Unit of Endocrinology and Nutrition - GC17, Maimónides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, Córdoba, Spain
| | - Antoine Raberin
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Bastien Krumm
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Erich Hohenauer
- Rehabilitation and Exercise Science Laboratory (RES lab), Department of Business Economics, Health and Social Care, University of Applied Sciences and Arts of Southern Switzerland, Landquart, Switzerland
- International University of Applied Sciences THIM, Landquart, Switzerland
- Department of Neurosciences and Movement Science, University of Fribourg, Fribourg, Switzerland
| | - Margit Egg
- Institute of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Mona Lichtblau
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Julian Müller
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Elena A Rybnikova
- Pavlov Institute of Physiology, Russian Academy of Sciences, St Petersburg, Russia
| | - Hannes Gatterer
- Institute of Mountain Emergency Medicine, Eurac Research, Bolzano, Italy
- Institute for Sports Medicine, Alpine Medicine and Health Tourism (ISAG), UMIT TIROL-Private University for Health Sciences and Health Technology, Hall in Tirol, Austria
| | - Tadej Debevec
- Faculty of Sport, University of Ljubljana, Ljubljana, Slovenia
- Department of Automatics, Biocybernetics and Robotics, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Sebastien Baillieul
- Service Universitaire de Pneumologie Physiologie, University of Grenoble Alpes, Inserm, Grenoble, France
| | | | - Tom Behrendt
- Chair Health and Physical Activity, Department of Sport Science, Institute III, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Lutz Schega
- Chair Health and Physical Activity, Department of Sport Science, Institute III, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Hannelore Ehrenreich
- Clinical Neuroscience, University Medical Center and Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
- Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru
| | - Christoph Schwarzer
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Oleg Glazachev
- Department of Normal Physiology, N.V. Sklifosovsky Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Olivier Girard
- School of Human Sciences (Exercise and Sport Science), The University of Western Australia, Crawley, Western Australia, Australia
| | - Sophie Lalande
- Department of Kinesiology and Health Education, University of Texas at Austin, Austin, TX, USA
| | - Michael Hamlin
- Department of Tourism, Sport and Society, Lincoln University, Christchurch, New Zealand
| | - Michele Samaja
- Department of Health Science, University of Milan, Milan, Italy
| | - Katharina Hüfner
- Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, University Hospital for Psychiatry II, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Gino Panza
- The Department of Health Care Sciences, Program of Occupational Therapy, Wayne State University, Detroit, MI, USA
- John D. Dingell VA Medical Center Detroit, Detroit, MI, USA
| | - Robert T Mallet
- Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
5
|
Wu L, Zhao P, Wu P, Jiang W, Liu Y, Ren H, Jin X, Zhou X, Feng L. Curcumin attenuates ochratoxin A and hypoxia co-induced liver injury in grass carp (Ctenopharyngodon idella) by dual targeting endoplasmic reticulum stress and apoptosis via reducing ROS content. J Anim Sci Biotechnol 2024; 15:131. [PMID: 39363374 PMCID: PMC11451059 DOI: 10.1186/s40104-024-01089-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/14/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Ochratoxin A (OTA) is a toxin widely found in aquafeed ingredients, and hypoxia is a common problem in fish farming. In practice, aquatic animals tend to be more sensitive to hypoxia while feeds are contaminated with OTA, but no studies exist in this area. This research investigated the multiple biotoxicities of OTA and hypoxia combined on the liver of grass carp and explored the mitigating effect of curcumin (CUR). METHODS A total of 720 healthy juvenile grass carp (11.06 ± 0.05 g) were selected and assigned randomly to 4 experimental groups: control group (without OTA and CUR), 1.2 mg/kg OTA group, 400 mg/kg CUR group, and 1.2 mg/kg OTA + 400 mg/kg CUR group with three replicates each for 60 d. Subsequently, 32 fish were selected, divided into normoxia (18 fish) and hypoxia (18 fish) groups, and subjected to hypoxia stress for 96 h. RESULTS CUR can attenuate histopathological damage caused by coming to OTA and hypoxia by reducing vacuolation and nuclear excursion. The alleviation of this damage was associated with the attenuation of apoptosis in the mitochondrial pathway by decreasing the expression of the pro-apoptotic proteins Caspase 3, 8, 9, Bax, and Apaf1 while increasing the expression of the anti-apoptotic protein Bcl-2, and attenuation of endoplasmic reticulum stress (ERS) by reducing Grp78 expression and chop levels. This may be attributed to the fact that the addition of CUR increased the levels of catalase (CAT) and glutathione reductase (GSH), increased antioxidant capacity, and ensured the proper functioning of respiratory chain complexes I and II, which in turn reduced the high production of reactive oxygen species (ROS), thus alleviating apoptosis and ERS. CONCLUSIONS In conclusion, our data demonstrate the effectiveness of CUR in attenuating liver injury caused by the combination of OTA and hypoxia. This study confirms the feasibility and efficacy of adding natural products to mitigate toxic damage to aquatic animals.
Collapse
Affiliation(s)
- Liangqin Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Piao Zhao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Weidan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Hongmei Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Xiaowan Jin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Chengdu, Sichuan, 611130, China
| | - Xiaoqiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China.
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Chengdu, Sichuan, 611130, China.
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China.
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
6
|
Yu Z, Teng Y, Yang H, Wang Y, Li X, Feng L, Xu W, Hao Y, Li Y. Inhibiting H2AX Can Ameliorate Myocardial Ischemia/Reperfusion Injury by Regulating P53/JNK Signaling Pathway. Cardiol Res Pract 2024; 2024:1905996. [PMID: 39257436 PMCID: PMC11387088 DOI: 10.1155/2024/1905996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/13/2024] [Indexed: 09/12/2024] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury is a significant area of focus in cardiovascular disease research. I/R injury can increase intracellular oxidative stress, leading to DNA damage. H2AX plays a crucial role in DNA repair. This study utilized mouse and cell models of myocardial I/R to investigate the impact of H2AX on cardiomyocytes during I/R. This study initially assessed the expression of H2AX in MI/R mice compared to a sham surgery group. Subsequently, cardiac function, infarct area, and mitochondrial damage were evaluated after inhibiting H2AX in MI/R mice and a negative control group. Furthermore, the study delved into the molecular mechanisms by analyzing the expression of H2AX, P53, p-JNK, SHP2, p-SHP2, p-RAS, parkin, Drp1, Cyt-C, Caspase-3, and Caspase-8 in cardiomyocytes following the addition of JNK or P53 agonists. The results from western blotting in vivo indicated significantly higher H2AX expression in the MI/R group compared to the sham group. Inhibiting H2AX improved cardiac function, reduced myocardial infarct area, and mitigated mitochondrial damage in the MI/R group. In vitro experiments demonstrated that inhibiting H2AX could attenuate mitochondrial damage and apoptosis in myocardial cells by modulating the P53 and JNK signaling pathways. These findings suggested that inhibiting H2AX may alleviate myocardial I/R injury through the regulation of the P53/JNK pathway, highlighting H2AX as a potential target for the treatment of myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Ziyang Yu
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Yirong Teng
- Department of General Practice The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Hongbo Yang
- Department of Cardiology Fuwai Yunnan Hospital Chinese Academy of Medical Sciences, Kunming, Yunnan, China
| | - Yudi Wang
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Xichen Li
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Lei Feng
- Department of Laboratory Yan'an Hospital of Kunming City, Kunming, Yunnan, China
| | - Wenbo Xu
- Department of Laboratory The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Yinglu Hao
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Yanping Li
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| |
Collapse
|
7
|
Mialet-Perez J, Belaidi E. Interplay between hypoxia inducible Factor-1 and mitochondria in cardiac diseases. Free Radic Biol Med 2024; 221:13-22. [PMID: 38697490 DOI: 10.1016/j.freeradbiomed.2024.04.239] [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: 02/14/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Ischemic heart diseases and cardiomyopathies are characterized by hypoxia, energy starvation and mitochondrial dysfunction. HIF-1 acts as a cellular oxygen sensor, tuning the balance of metabolic and oxidative stress pathways to provide ATP and sustain cell survival. Acting on mitochondria, HIF-1 regulates different processes such as energy substrate utilization, oxidative phosphorylation and mitochondrial dynamics. In turn, mitochondrial homeostasis modifications impact HIF-1 activity. This underlies that HIF-1 and mitochondria are tightly interconnected to maintain cell homeostasis. Despite many evidences linking HIF-1 and mitochondria, the mechanistic insights are far from being understood, particularly in the context of cardiac diseases. Here, we explore the current understanding of how HIF-1, reactive oxygen species and cell metabolism are interconnected, with a specific focus on mitochondrial function and dynamics. We also discuss the divergent roles of HIF in acute and chronic cardiac diseases in order to highlight that HIF-1, mitochondria and oxidative stress interaction deserves to be deeply investigated. While the strategies aiming at stabilizing HIF-1 have provided beneficial effects in acute ischemic injury, some deleterious effects were observed during prolonged HIF-1 activation. Thus, deciphering the link between HIF-1 and mitochondria will help to optimize HIF-1 modulation and provide new therapeutic perspectives for the treatment of cardiovascular pathologies.
Collapse
Affiliation(s)
- Jeanne Mialet-Perez
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe MitoLab, SFR ICAT, Angers, France
| | - Elise Belaidi
- Univ. Lyon 1, Laboratory of Tissue Biology and Therapeutic Engineering, CNRS, LBTI UMR 5305, 69367, Lyon, France.
| |
Collapse
|
8
|
Luo X, Liu P, Ye X, He J, Lai Y, Lv Y, Wu X, Liu Y, Zhang Q, Yang H, Wei W, Deng C, Kuang S, Wu S, Xue Y, Rao F. Curcumin improves atrial fibrillation susceptibility by regulating tsRNA expression in aging mouse atrium. PeerJ 2024; 12:e17495. [PMID: 39076782 PMCID: PMC11285363 DOI: 10.7717/peerj.17495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 05/09/2024] [Indexed: 07/31/2024] Open
Abstract
Age is an independent risk factor for atrial fibrillation (AF), and curcumin can delay aging related disease through reducing oxidative stress and inflammation. However, its target in aging-related AF remains unclear. Transfer RNA-derived small RNA (tsRNA) is a novel short non-coding RNA (sncRNA), and exerts a potential regulatory function in aging. This study was to explore the therapeutic targets of curcumin in atrium of aged mice by PANDORA-seq. Aged mice (18 month) were treated with curcumin (100 mg/kg). Rapid transjugular atrial pacing was performed to observe AF inducibility. SA-β-gal staining, reactive oxygen species (ROS) detection and qRT-PCR were used to assess the degree of aging and oxidative stress/inflammation levels. PANDORA-seq was performed to reveal the differentially expressed sncRNAs in the atrium of mice. The results showed that curcumin reduced the susceptibility AF of aged mice by improving aging-related atrial fibrosis. Compared to young mice (5 month) group, aged mice yielded 473 significantly altered tsRNA sequences, while 947 tsRNA sequences were significantly altered after treated with curcumin. Enrichment analysis revealed that the target genes were mainly related to DNA damage and protein modification. Compared with the 5 month group, the expression levels of mature-mt_tRNA-Val-TAC_CCA_end, mature-mt_tRNA-Glu-TTC_CCA_end, and mature-tRNA-Asp-GTC_CCA_end were up-regulated in the 18 month group, while the expression of mature-mt_tRNA-Thr-TGT_5_end was down-regulated. This trend was reversed in the 18 month + curcumin group. Increased cellular ROS levels, inflammation expression and senescence in aged mice atrium were improved by the down-regulation of mature-mt_tRNA-Val-TAC_CCA_end. In conclusion, our findings identified mature-mt_tRNA-Val-TAC_CCA_end participated in the mechanism of aging-related atrial fibrosis, providing new intervention target of aging-related AF.
Collapse
Affiliation(s)
- Xueshan Luo
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- South China University of Technology, Guangzhou, Guangdong, China
| | - Panyue Liu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- South China University of Technology, Guangzhou, Guangdong, China
| | - Xingdong Ye
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Jintao He
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- South China University of Technology, Guangzhou, Guangdong, China
| | - Yingyu Lai
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Yidong Lv
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Xiongbin Wu
- Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Yang Liu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Qianhuan Zhang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Hui Yang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Wei Wei
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Chunyu Deng
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- South China University of Technology, Guangzhou, Guangdong, China
| | - Sujuan Kuang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Shulin Wu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Yumei Xue
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- South China University of Technology, Guangzhou, Guangdong, China
| | - Fang Rao
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Marupudi N, Xiong MP. Genetic Targets and Applications of Iron Chelators for Neurodegeneration with Brain Iron Accumulation. ACS BIO & MED CHEM AU 2024; 4:119-130. [PMID: 38911909 PMCID: PMC11191567 DOI: 10.1021/acsbiomedchemau.3c00066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 06/25/2024]
Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a group of neurodegenerative diseases that are typically caused by a monogenetic mutation, leading to development of disordered movement symptoms such as dystonia, hyperreflexia, etc. Brain iron accumulation can be diagnosed through MRI imaging and is hypothesized to be the cause of oxidative stress, leading to the degeneration of brain tissue. There are four main types of NBIA: pantothenate kinase-associated neurodegeneration (PKAN), PLA2G6-associated neurodegeneration (PLAN), mitochondrial membrane protein-associated neurodegeneration (MKAN), and beta-propeller protein-associated neurodegeneration (BPAN). There are no causative therapies for these diseases, but iron chelators have been shown to have potential toward treating NBIA. Three chelators are investigated in this Review: deferoxamine (DFO), desferasirox (DFS), and deferiprone (DFP). DFO has been investigated to treat neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD); however, dose-related toxicity in these studies, as well as in PKAN studies, have shown that the drug still requires more development before it can be applied toward NBIA cases. Iron chelation therapies other than the ones currently in clinical use have not yet reached clinical studies, but they may possess characteristics that would allow them to access the brain in ways that current chelators cannot. Intranasal formulations are an attractive dosage form to study for chelation therapy, as this method of delivery can bypass the blood-brain barrier and access the CNS. Gene therapy differs from iron chelation therapy as it is a causal treatment of the disease, whereas iron chelators only target the disease progression of NBIA. Because the pathophysiology of NBIA diseases is still unclear, future courses of action should be focused on causative treatment; however, iron chelation therapy is the current best course of action.
Collapse
Affiliation(s)
- Neharika Marupudi
- Department of Pharmaceutical
& Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602-2352, United States
| | - May P. Xiong
- Department of Pharmaceutical
& Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602-2352, United States
| |
Collapse
|
10
|
Wang M, Wen W, Chen Y, Yishajiang S, Li Y, Li Z, Zhang X. TRPC5 channel participates in myocardial injury in chronic intermittent hypoxia. Clinics (Sao Paulo) 2024; 79:100368. [PMID: 38703717 PMCID: PMC11087918 DOI: 10.1016/j.clinsp.2024.100368] [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: 12/12/2023] [Revised: 03/25/2024] [Accepted: 04/14/2024] [Indexed: 05/06/2024] Open
Abstract
OBJECTIVE The purpose of this study is to develop an animal model of Chronic Intermittent Hypoxia (CIH) and investigate the role of the TRPC5 channel in cardiac damage in OSAHS rats. METHODS Twelve male Sprague Dawley rats were randomly divided into the CIH group and the Normoxic Control (NC) group. Changes in structure, function, and pathology of heart tissue were observed through echocardiography, transmission electron microscopy, HE-staining, and TUNEL staining. RESULTS The Interventricular Septum thickness at diastole (IVSd) and End-Diastolic Volume (EDV) of rats in the CIH group significantly increased, whereas the LV ejection fraction and LV fraction shortening significantly decreased. TEM showed that the myofilaments in the CIH group were loosely arranged, the sarcomere length varied, the cell matrix dissolved, the mitochondrial cristae were partly flocculent, the mitochondrial outer membrane dissolved and disappeared, and some mitochondria were swollen and vacuolated. The histopathological examination showed that the cardiomyocytes in the CIH group were swollen with granular degeneration, some of the myocardial fibers were broken and disorganized, and most of the nuclei were vacuolar and hypochromic. CONCLUSION CIH promoted oxidative stress, the influx of Ca2+, and the activation of the CaN/NFATc signaling pathway, which led to pathological changes in the morphology and ultrastructure of cardiomyocytes, the increase of myocardial apoptosis, and the decrease of myocardial contractility. These changes may be associated with the upregulation of TRPC5.
Collapse
Affiliation(s)
- Mengmeng Wang
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, China
| | - Wen Wen
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, China
| | - Yulan Chen
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, China.
| | - Sharezati Yishajiang
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, China
| | - Yu Li
- Second Department of General Internal Medicine, The First Affiliated Hospital of Xinjiang Medical University, China
| | - Zhiqiang Li
- Laboratory Animal Center, Xinjiang Medical University, China
| | - Xiangyang Zhang
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, China
| |
Collapse
|
11
|
Chen Z, Liu T, Yuan H, Sun H, Liu S, Zhang S, Liu L, Jiang S, Tang Y, Liu Z. Bioinformatics integration reveals key genes associated with mitophagy in myocardial ischemia-reperfusion injury. BMC Cardiovasc Disord 2024; 24:183. [PMID: 38539069 PMCID: PMC10967080 DOI: 10.1186/s12872-024-03834-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/09/2024] [Indexed: 11/14/2024] Open
Abstract
BACKGROUND Myocardial ischemia is a prevalent cardiovascular disorder associated with significant morbidity and mortality. While prompt restoration of blood flow is essential for improving patient outcomes, the subsequent reperfusion process can result in myocardial ischemia-reperfusion injury (MIRI). Mitophagy, a specialized autophagic mechanism, has consistently been implicated in various cardiovascular disorders. However, the specific connection between ischemia-reperfusion and mitophagy remains elusive. This study aims to elucidate and validate central mitophagy-related genes associated with MIRI through comprehensive bioinformatics analysis. METHODS We acquired the microarray expression profile dataset (GSE108940) from the Gene Expression Omnibus (GEO) and identified differentially expressed genes (DEGs) using GEO2R. Subsequently, these DEGs were cross-referenced with the mitophagy database, and differential nucleotide sequence analysis was performed through enrichment analysis. Protein-protein interaction (PPI) network analysis was employed to identify hub genes, followed by clustering of these hub genes using cytoHubba and MCODE within Cytoscape software. Gene set enrichment analysis (GSEA) was conducted on central genes. Additionally, Western blotting, immunofluorescence, and quantitative polymerase chain reaction (qPCR) analyses were conducted to validate the expression patterns of pivotal genes in MIRI rat model and H9C2 cardiomyocytes. RESULTS A total of 2719 DEGs and 61 mitophagy-DEGs were identified, followed by enrichment analyses and the construction of a PPI network. HSP90AA1, RPS27A, EEF2, EIF4A1, EIF2S1, HIF-1α, and BNIP3 emerged as the seven hub genes identified by cytoHubba and MCODE of Cytoscape software. Functional clustering analysis of HIF-1α and BNIP3 yielded a score of 9.647, as determined by Cytoscape (MCODE). In our MIRI rat model, Western blot and immunofluorescence analyses confirmed a significant elevation in the expression of HIF-1α and BNIP3, accompanied by a notable increase in the ratio of LC3II to LC3I. Subsequently, qPCR confirmed a significant upregulation of HIF-1α, BNIP3, and LC3 mRNA in the MIRI group. Activation of the HIF-1α/BNIP3 pathway mediates the regulation of the degree of Mitophagy, thereby effectively reducing apoptosis in rat H9C2 cardiomyocytes. CONCLUSIONS This study has identified seven central genes among mitophagy-related DEGs that may play a pivotal role in MIRI, suggesting a correlation between the HIF-1α/BNIP3 pathway of mitophagy and the pathogenesis of MIRI. The findings highlight the potential importance of mitophagy in MIRI and provide valuable insights into underlying mechanisms and potential therapeutic targets for further exploration in future studies.
Collapse
Affiliation(s)
- Zhian Chen
- Department of Clinical Medicine, Changchun University of Chinese Medicine, No. 1035, Boshuo Road, Nanguan District, Changchun, 130,117, Jilin Province, China
| | - Tianying Liu
- Department of Clinical Medicine, Changchun University of Chinese Medicine, No. 1035, Boshuo Road, Nanguan District, Changchun, 130,117, Jilin Province, China
| | - Hao Yuan
- Department of Clinical Medicine, Changchun University of Chinese Medicine, No. 1035, Boshuo Road, Nanguan District, Changchun, 130,117, Jilin Province, China
| | - Han Sun
- Department of Clinical Medicine, Changchun University of Chinese Medicine, No. 1035, Boshuo Road, Nanguan District, Changchun, 130,117, Jilin Province, China
| | - Sitong Liu
- Department of Clinical Medicine, Changchun University of Chinese Medicine, No. 1035, Boshuo Road, Nanguan District, Changchun, 130,117, Jilin Province, China
| | - Shuai Zhang
- Department of Clinical Medicine, Changchun University of Chinese Medicine, No. 1035, Boshuo Road, Nanguan District, Changchun, 130,117, Jilin Province, China
| | - Li Liu
- Department of Clinical Medicine, Changchun University of Chinese Medicine, No. 1035, Boshuo Road, Nanguan District, Changchun, 130,117, Jilin Province, China
| | - Shuang Jiang
- Department of Clinical Medicine, Changchun University of Chinese Medicine, No. 1035, Boshuo Road, Nanguan District, Changchun, 130,117, Jilin Province, China
| | - Yong Tang
- Department of Clinical Medicine, Changchun University of Chinese Medicine, No. 1035, Boshuo Road, Nanguan District, Changchun, 130,117, Jilin Province, China.
| | - Zhi Liu
- Department of Clinical Medicine, Changchun University of Chinese Medicine, No. 1035, Boshuo Road, Nanguan District, Changchun, 130,117, Jilin Province, China.
| |
Collapse
|
12
|
Liu W, Zhu Q, Li X, Wang Y, Zhao C, Ma C. Effects of obstructive sleep apnea on myocardial injury and dysfunction: a review focused on the molecular mechanisms of intermittent hypoxia. Sleep Breath 2024; 28:41-51. [PMID: 37548920 DOI: 10.1007/s11325-023-02893-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 06/08/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
Obstructive sleep apnea (OSA) is characterized by intermittent hypoxia (IH) and is strongly associated with adverse cardiovascular outcomes. Myocardial injury and dysfunction have been commonly observed in clinical practice, particularly in patients with severe OSA. However, the underlying mechanisms remain obscure. In this review, we summarized the molecular mechanisms by which IH impact on myocardial injury and dysfunction. In brief, IH-induced cardiomyocyte death proceeds through the regulation of multiple biological processes, including differentially expressed transcription factors, alternative epigenetic programs, and altered post-translational modification. Besides cell death, various cardiomyocyte injuries, such as endoplasmic reticulum stress, occurs with IH. In addition to the direct effects on cardiomyocytes, IH has been found to deteriorate myocardial blood and energy supply by affecting the microvascular structure and disrupting glucose and lipid metabolism. For better diagnosis and treatment of OSA, further studies on the molecular mechanisms of IH-induced myocardial injury and dysfunction are essential.
Collapse
Affiliation(s)
- Wen Liu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Qing Zhu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Xinxin Li
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Yonghuai Wang
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Cuiting Zhao
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China.
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China.
| |
Collapse
|
13
|
Yang C, Zhu Q, Chen Y, Ji K, Li S, Wu Q, Pan Q, Li J. Review of the Protective Mechanism of Curcumin on Cardiovascular Disease. Drug Des Devel Ther 2024; 18:165-192. [PMID: 38312990 PMCID: PMC10838105 DOI: 10.2147/dddt.s445555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/16/2024] [Indexed: 02/06/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the most common cause of death worldwide and has been the focus of research in the medical community. Curcumin is a polyphenolic compound extracted from the root of turmeric. Curcumin has been shown to have a variety of pharmacological properties over the past decades. Curcumin can significantly protect cardiomyocyte injury after ischemia and hypoxia, inhibit myocardial hypertrophy and fibrosis, improve ventricular remodeling, reduce drug-induced myocardial injury, improve diabetic cardiomyopathy(DCM), alleviate vascular endothelial dysfunction, inhibit foam cell formation, and reduce vascular smooth muscle cells(VSMCs) proliferation. Clinical studies have shown that curcumin has a protective effect on blood vessels. Toxicological studies have shown that curcumin is safe. But high doses of curcumin also have some side effects, such as liver damage and defects in embryonic heart development. This article reviews the mechanism of curcumin intervention on CVDs in recent years, in order to provide reference for the development of new drugs in the future.
Collapse
Affiliation(s)
- Chunkun Yang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qinwei Zhu
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Yanbo Chen
- Department of Arrhythmia, Weifang People's Hospital, Weifang, Shandong, People's Republic of China
| | - Kui Ji
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Shuanghong Li
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Qian Wu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qingquan Pan
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
14
|
Arnaud C, Billoir E, de Melo Junior AF, Pereira SA, O'Halloran KD, Monteiro EC. Chronic intermittent hypoxia-induced cardiovascular and renal dysfunction: from adaptation to maladaptation. J Physiol 2023; 601:5553-5577. [PMID: 37882783 DOI: 10.1113/jp284166] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023] Open
Abstract
Chronic intermittent hypoxia (CIH) is the dominant pathological feature of human obstructive sleep apnoea (OSA), which is highly prevalent and associated with cardiovascular and renal diseases. CIH causes hypertension, centred on sympathetic nervous overactivity, which persists following removal of the CIH stimulus. Molecular mechanisms contributing to CIH-induced hypertension have been carefully delineated. However, there is a dearth of knowledge on the efficacy of interventions to ameliorate high blood pressure in established disease. CIH causes endothelial dysfunction, aberrant structural remodelling of vessels and accelerates atherosclerotic processes. Pro-inflammatory and pro-oxidant pathways converge on disrupted nitric oxide signalling driving vascular dysfunction. In addition, CIH has adverse effects on the myocardium, manifesting atrial fibrillation, and cardiac remodelling progressing to contractile dysfunction. Sympatho-vagal imbalance, oxidative stress, inflammation, dysregulated HIF-1α transcriptional responses and resultant pro-apoptotic ER stress, calcium dysregulation, and mitochondrial dysfunction conspire to drive myocardial injury and failure. CIH elaborates direct and indirect effects in the kidney that initially contribute to the development of hypertension and later to chronic kidney disease. CIH-induced morphological damage of the kidney is dependent on TLR4/NF-κB/NLRP3/caspase-1 inflammasome activation and associated pyroptosis. Emerging potential therapies related to the gut-kidney axis and blockade of aryl hydrocarbon receptors (AhR) are promising. Cardiorenal outcomes in response to intermittent hypoxia present along a continuum from adaptation to maladaptation and are dependent on the intensity and duration of exposure to intermittent hypoxia. This heterogeneity of OSA is relevant to therapeutic treatment options and we argue the need for better stratification of OSA phenotypes.
Collapse
Affiliation(s)
- Claire Arnaud
- Université Grenoble-Alpes INSERM U1300, Laboratoire HP2, Grenoble, France
| | - Emma Billoir
- Université Grenoble-Alpes INSERM U1300, Laboratoire HP2, Grenoble, France
| | | | - Sofia A Pereira
- iNOVA4Health, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Emilia C Monteiro
- iNOVA4Health, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| |
Collapse
|
15
|
Bi M, Li D, Zhang J. Role of curcumin in ischemia and reperfusion injury. Front Pharmacol 2023; 14:1057144. [PMID: 37021057 PMCID: PMC10067738 DOI: 10.3389/fphar.2023.1057144] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 02/23/2023] [Indexed: 03/22/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) is an inevitable pathological process after organic transplantations. Although traditional treatments restore the blood supply of ischemic organs, the damage caused by IRI is always ignored. Therefore, the ideal and effective therapeutic strategy to mitigate IRI is warrented. Curcumin is a type of polyphenols, processing such properties as anti-oxidative stress, anti-inflammation and anti-apoptosis. However, although many researches have been confirmed that curcumin can exert great effects on the mitigation of IRI, there are still some controversies about its underlying mechanisms among these researches. Thus, this review is to summarize the protective role of curcumin against IRI as well as the controversies of current researches, so as to clarify its underlying mechanisms clearly and provide clinicians a novel idea of the therapy for IRI.
Collapse
Affiliation(s)
- Minglei Bi
- Department of Plastic Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Danyi Li
- Department of Ophthalmology, Jiading District Central Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jin Zhang
- Department of Plastic Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- *Correspondence: Jin Zhang,
| |
Collapse
|
16
|
Devaux CA, Raoult D. The impact of COVID-19 on populations living at high altitude: Role of hypoxia-inducible factors (HIFs) signaling pathway in SARS-CoV-2 infection and replication. Front Physiol 2022; 13:960308. [PMID: 36091390 PMCID: PMC9454615 DOI: 10.3389/fphys.2022.960308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Cases of coronavirus disease 2019 (COVID-19) have been reported worldwide. However, one epidemiological report has claimed a lower incidence of the disease in people living at high altitude (>2,500 m), proposing the hypothesis that adaptation to hypoxia may prove to be advantageous with respect to SARS-CoV-2 infection. This publication was initially greeted with skepticism, because social, genetic, or environmental parametric variables could underlie a difference in susceptibility to the virus for people living in chronic hypobaric hypoxia atmospheres. Moreover, in some patients positive for SARS-CoV-2, early post-infection ‘happy hypoxia” requires immediate ventilation, since it is associated with poor clinical outcome. If, however, we accept to consider the hypothesis according to which the adaptation to hypoxia may prove to be advantageous with respect to SARS-CoV-2 infection, identification of the molecular rational behind it is needed. Among several possibilities, HIF-1 regulation appears to be a molecular hub from which different signaling pathways linking hypoxia and COVID-19 are controlled. Interestingly, HIF-1α was reported to inhibit the infection of lung cells by SARS-CoV-2 by reducing ACE2 viral receptor expression. Moreover, an association of the rs11549465 variant of HIF-1α with COVID-19 susceptibility was recently discovered. Here, we review the evidence for a link between HIF-1α, ACE2 and AT1R expression, and the incidence/severity of COVID-19. We highlight the central role played by the HIF-1α signaling pathway in the pathophysiology of COVID-19.
Collapse
Affiliation(s)
- Christian Albert Devaux
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
- Centre National de la Recherche Scientifique, Marseille, France
- *Correspondence: Christian Albert Devaux,
| | - Didier Raoult
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
17
|
Intermittent Hypoxia-Induced Cardiomyocyte Death Is Mediated by HIF-1 Dependent MAM Disruption. Antioxidants (Basel) 2022; 11:antiox11081462. [PMID: 36009181 PMCID: PMC9405320 DOI: 10.3390/antiox11081462] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 12/04/2022] Open
Abstract
Rationale: Intermittent hypoxia (IH) is one of the main features of sleep-disordered breathing (SDB). Recent findings indicate that hypoxia inducible factor-1 (HIF-1) promotes cardiomyocytes apoptosis during chronic IH, but the mechanisms involved remain to be elucidated. Here, we hypothesize that IH-induced ER stress is associated with mitochondria-associated ER membrane (MAM) alteration and mitochondrial dysfunction, through HIF-1 activation. Methods: Right atrial appendage biopsies from patients with and without SDB were used to determine HIF-1α, Grp78 and CHOP expressions. Wild-type and HIF-1α+/− mice were exposed to normoxia (N) or IH (21–5% O2, 60 cycles/h, 8 h/day) for 21 days. Expressions of HIF-1α, Grp78 and CHOP, and apoptosis, were measured by Western blot and immunochemistry. In isolated cardiomyocytes, we examined structural integrity of MAM by proximity ligation assay and their function by measuring ER-to-mitochondria Ca2+ transfer by confocal microscopy. Finally, we measured mitochondrial respiration using oxygraphy and calcium retention capacity (CRC) by spectrofluorometry. MAM structure was also investigated in H9C2 cells incubated with 1 mM CoCl2, a potent HIF-1α inducer. Results: In human atrial biopsies and mice, IH induced HIF-1 activation, ER stress and apoptosis. IH disrupted MAM, altered Ca2+ homeostasis, mitochondrial respiration and CRC. Importantly, IH had no effect in HIF-1α+/− mice. Similar to what observed under IH, HIF-1α overexpression was associated with MAM alteration in H9C2. Conclusion: IH-induced ER stress, MAM alterations and mitochondrial dysfunction were mediated by HIF-1; all these intermediate mechanisms ultimately inducing cardiomyocyte apoptosis. This suggests that HIF-1 modulation might limit the deleterious cardiac effects of SDB.
Collapse
|
18
|
Zhang J, Xie B, Tang Y, Zhou B, Wang Q, Ge Q, Zhou Y, Gu T. Downregulation of miR-34c-5p alleviates chronic intermittent hypoxia-induced myocardial damage by targeting sirtuin 1. J Biochem Mol Toxicol 2022; 36:e23164. [PMID: 35848756 DOI: 10.1002/jbt.23164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/28/2022] [Accepted: 07/01/2022] [Indexed: 11/09/2022]
Abstract
Numerous microRNAs (miRs) are abnormally expressed in response to hypoxia-induced myocardial damage. Herein, miR-34c-5p as a potential pharmaco-target was investigated in a mouse model of chronic intermittent hypoxia (CIH)-induced myocardial damage. A mouse model of myocardial damage was established using CIH with 7% or 21% O2 alternating 60 s for 12 h/day, 21% O2 for 12 h/day. AntagomiR-34c-5p (20 nM/0.1 ml; once a week for 12 weeks) was used as a miR-34c-5p inhibitor in a mouse model with tail-vein injection. In another experiment, mice were administrated with Sirt1 activator SRT1720 (50 mg/kg/day) by intraperitoneal injection. Gene Expression Omnibus database showed a significant upregulation of miR-34c-5p expression in the ischemic myocardium of male mice. In CIH-stimulated mice, miR-34c-5p expression was also significantly increased compared with normal mice. Treatment of antagomiR-34c-5p significantly restrained CIH-triggered myocardial apoptosis. After administration of antagomiR-34c-5p or Sirt1 activator SRT1720, cardiac hypertrophy and oxidative stress were attenuated in CIH-stimulated mice. We also found sirtuin 1 (Sirt1) as a direct target of miR-34c-5p, which was able to mediate Sirt1 protein expression in cardiomyocytes. AntagomiR-34c-5p injection markedly elevated Sirt1 protein expression in CIH-stimulated mice. AntagomiR-34c-5p or Sirt1 activator SRT1720 administration exhibited the antioxidative activity and cardioprotective roles in CIH-stimulated mice.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Bo Xie
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Yanrong Tang
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Bo Zhou
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Qiong Wang
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Qing Ge
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Yufei Zhou
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Tongqing Gu
- School of Foreign Languages, Chengdu University of Information Technology, Chengdu, Sichuan, China
| |
Collapse
|
19
|
Naryzhnaya NV, Maslov LN, Derkachev IA, Ma H, Zhang Y, Prasad NR, Singh N, Fu F, Pei JM, Sarybaev A, Sydykov A. The effect of adaptation to hypoxia on cardiac tolerance to ischemia/reperfusion. J Biomed Res 2022:1-25. [PMID: 37183617 PMCID: PMC10387748 DOI: 10.7555/jbr.36.20220125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The acute myocardial infarction (AMI) and sudden cardiac death (SCD), both associated with acute cardiac ischemia, are one of the leading causes of adult death in economically developed countries. The development of new approaches for the treatment and prevention of AMI and SCD remains the highest priority for medicine. A study on the cardiovascular effects of chronic hypoxia (CH) may contribute to the development of these methods. Chronic hypoxia exerts both positive and adverse effects. The positive effects are the infarct-reducing, vasoprotective, and antiarrhythmic effects, which can lead to the improvement of cardiac contractility in reperfusion. The adverse effects are pulmonary hypertension and right ventricular hypertrophy. This review presents a comprehensive overview of how CH enhances cardiac tolerance to ischemia/reperfusion. It is an in-depth analysis of the published data on the underlying mechanisms, which can lead to future development of the cardioprotective effect of CH. A better understanding of the CH-activated protective signaling pathways may contribute to new therapeutic approaches in an increase of cardiac tolerance to ischemia/reperfusion.
Collapse
|
20
|
Balbaa M, El-Zeftawy M, Abdulmalek SA. Therapeutic Screening of Herbal Remedies for the Management of Diabetes. Molecules 2021; 26:6836. [PMID: 34833928 PMCID: PMC8618521 DOI: 10.3390/molecules26226836] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 02/07/2023] Open
Abstract
The study of diabetes mellitus (DM) patterns illustrates increasingly important facts. Most importantly, they include oxidative stress, inflammation, and cellular death. Up to now, there is a shortage of drug therapies for DM, and the discovery and the development of novel therapeutics for this disease are crucial. Medicinal plants are being used more and more as an alternative and natural cure for the disease. Consequently, the objective of this review was to examine the latest results on the effectiveness and protection of natural plants in the management of DM as adjuvant drugs for diabetes and its complex concomitant diseases.
Collapse
Affiliation(s)
- Mahmoud Balbaa
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21511, Egypt;
| | - Marwa El-Zeftawy
- Biochemistry Department, Faculty of Veterinary Medicine, New Valley University, New Valley 72511, Egypt;
| | - Shaymaa A. Abdulmalek
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21511, Egypt;
- Center of Excellency for Preclinical Study (CE-PCS), Pharmaceutical and Fermentation Industries Development Centre, The City of Scientific Research and Technological Applications, Alexandria 21511, Egypt
| |
Collapse
|
21
|
Liu YP, Cai Y, Lei YD, Yuan XY, Wang Y, Yi S, Li XY, Huang L, Long DX, Zhang ZH. Circular RNA expression profiles in human bronchial epithelial cells treated with beryllium sulfate. Toxicol Res (Camb) 2021; 10:1013-1021. [PMID: 34733486 DOI: 10.1093/toxres/tfab086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/21/2021] [Accepted: 08/05/2021] [Indexed: 11/13/2022] Open
Abstract
Circular RNAs (circRNAs), is a novel type of endogenous non-coding RNAs (ncRNAs) participated in the pathogenesis of many diseases. Beryllium is one of the carcinogenesis elements. However, the mechanism and function of circRNAs in human bronchial epithelial cells (16HBE) induced by beryllium sulfate (BeSO4) was rarely reported. Therefore, the high-throughput RNA sequencing analysis was performed to detect the circRNA profiles between control groups and BeSO4-induced groups. Furthermore, circRNA-miRNA-mRNA network, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and PPI network analysis were used for bioinformatics analysis. CircRNA sequencing analysis revealed that 36 circRNAs were up-regulated and 35 circRNAs were down-regulated in the BeSO4-exposed groups. The selected circRNAs were verified by real-time fluorescent quantitative PCR (qRT-PCR). Hsa_circ_0004214 and hsa_circ_0003586 were validated to be up-regulated, hsa_circ_0047958, hsa_circ_0001944, and hsa_circ_0008982 were down-regulated. The circRNA-miRNA-mRNA network annotated the key signaling pathway including cellular senescence, TNF signaling pathway, NF-kappa B signaling pathway, HIF-1 signaling pathway, and Hippo signaling pathway. The PPI network indicated the most circRNAs might participate in the BeSO4 toxicity by acting as a sponge for the miR-663b through JAK-STAT signaling pathway. In summary, our study suggests that circRNAs may play roles in the mechanism of beryllium toxicity.
Collapse
Affiliation(s)
- Yan-Ping Liu
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Ying Cai
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Yuan-di Lei
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiao-Yan Yuan
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Ye Wang
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Shan Yi
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Xun-Ya Li
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Lian Huang
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Ding-Xin Long
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhao-Hui Zhang
- Department of Preventive Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
22
|
Mauerhofer C, Grumet L, Schemmer P, Leber B, Stiegler P. Combating Ischemia-Reperfusion Injury with Micronutrients and Natural Compounds during Solid Organ Transplantation: Data of Clinical Trials and Lessons of Preclinical Findings. Int J Mol Sci 2021; 22:10675. [PMID: 34639016 PMCID: PMC8508760 DOI: 10.3390/ijms221910675] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Although extended donor criteria grafts bear a higher risk of complications such as graft dysfunction, the exceeding demand requires to extent the pool of potential donors. The risk of complications is highly associated with ischemia-reperfusion injury, a condition characterized by high loads of oxidative stress exceeding antioxidative defense mechanisms. The antioxidative properties, along with other beneficial effects like anti-inflammatory, antiapoptotic or antiarrhythmic effects of several micronutrients and natural compounds, have recently emerged increasing research interest resulting in various preclinical and clinical studies. Preclinical studies reported about ameliorated oxidative stress and inflammatory status, resulting in improved graft survival. Although the majority of clinical studies confirmed these results, reporting about improved recovery and superior organ function, others failed to do so. Yet, only a limited number of micronutrients and natural compounds have been investigated in a (large) clinical trial. Despite some ambiguous clinical results and modest clinical data availability, the vast majority of convincing animal and in vitro data, along with low cost and easy availability, encourage the conductance of future clinical trials. These should implement insights gained from animal data.
Collapse
Affiliation(s)
- Christina Mauerhofer
- Department of Science and Product Development, pro medico HandelsGmbH, Liebenauer Tangente 6, 8041 Graz, Austria; (C.M.); (L.G.)
| | - Lukas Grumet
- Department of Science and Product Development, pro medico HandelsGmbH, Liebenauer Tangente 6, 8041 Graz, Austria; (C.M.); (L.G.)
| | - Peter Schemmer
- Division of Transplant Surgery, Department of Surgery, Medical University, 8036 Graz, Austria; (P.S.); (B.L.)
| | - Bettina Leber
- Division of Transplant Surgery, Department of Surgery, Medical University, 8036 Graz, Austria; (P.S.); (B.L.)
| | - Philipp Stiegler
- Division of Transplant Surgery, Department of Surgery, Medical University, 8036 Graz, Austria; (P.S.); (B.L.)
| |
Collapse
|
23
|
MKRN1 Ubiquitylates p21 to Protect against Intermittent Hypoxia-Induced Myocardial Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9360339. [PMID: 34504644 PMCID: PMC8423574 DOI: 10.1155/2021/9360339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/09/2021] [Indexed: 12/16/2022]
Abstract
Although chronic intermittent hypoxia- (IH-) induced myocardial apoptosis is an established pathophysiological process resulting in a poor prognosis for patients with obstructive sleep apnea syndrome, its underlying mechanism remains unclear. This study is aimed at exploring the role of makorin ring finger protein 1 (MKRN1) in IH-induced myocardial apoptosis and elucidating its molecular activity. First, the GSE2271 dataset was downloaded from the Gene Expression Omnibus database to identify the differentially expressed genes. Then, an SD rat model of IH, together with rat cardiomyocyte H9C2 and human cardiomyocyte AC16 IH models, was constructed. TUNEL, Western blot, and immunohistochemistry assays were used to detect cell apoptosis. Dihydroethidium staining was conducted to analyze the concentration of reactive oxygen species. In addition, RT-qPCR, Western blot, and immunohistochemistry were performed to measure the expression levels of MKRN1 and p21. The direct interaction between MKRN1 and p21 was determined using coimmunoprecipitation and ubiquitination analysis. MKRN1 expression was found to be downregulated in IH rat myocardial tissues as well as in H9C2 and AC16 cells. Upregulated expression of MKRN1 in H9C2 and AC16 cells alleviated the IH-induced reactive oxygen species production and cell apoptosis. Mechanistically, MKRN1 promoted p21 protein ubiquitination and the proteasome pathway degradation to negatively regulate p21 expression. Thus, MKRN1 regulates p21 ubiquitination to prevent IH-induced myocardial apoptosis.
Collapse
|
24
|
Pourbagher-Shahri AM, Farkhondeh T, Ashrafizadeh M, Talebi M, Samargahndian S. Curcumin and cardiovascular diseases: Focus on cellular targets and cascades. Biomed Pharmacother 2021; 136:111214. [PMID: 33450488 DOI: 10.1016/j.biopha.2020.111214] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases (CVDs) are one of the leading causes of the most considerable mortality globally, and it has been tried to find the molecular mechanisms and design new drugs that triggered the molecular target. Curcumin is the main ingredient of Curcuma longa (turmeric) that has been used in traditional medicine for treating several diseases for years. Numerous investigations have indicated the beneficial effect of Curcumin in modulating multiple signaling pathways involved in oxidative stress, inflammation, apoptosis, and proliferation. The cardiovascular protective effects of Curcumin against CVDs have been indicated in several studies. In the current review study, we provided novel information on Curcumin's protective effects against various CVDs and potential molecular signaling targets of Curcumin. Nonetheless, more studies should be performed to discover the exact molecular target of Curcumin against CVDs.
Collapse
Affiliation(s)
| | - Tahereh Farkhondeh
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), Birjand, Iran; Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Marjan Talebi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, 19968 35115, Iran
| | - Saeed Samargahndian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
25
|
Gaspar LS, Sousa C, Álvaro AR, Cavadas C, Mendes AF. Common risk factors and therapeutic targets in obstructive sleep apnea and osteoarthritis: An unexpectable link? Pharmacol Res 2020; 164:105369. [PMID: 33352231 DOI: 10.1016/j.phrs.2020.105369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/11/2020] [Accepted: 12/09/2020] [Indexed: 10/22/2022]
Abstract
Osteoarthritis (OA) and Obstructive Sleep Apnea (OSA) are two highly prevalent chronic diseases for which effective therapies are urgently needed. Recent epidemiologic studies, although scarce, suggest that the concomitant occurrence of OA and OSA is associated with more severe manifestations of both diseases. Moreover, OA and OSA share risk factors, such as aging and metabolic disturbances, and co-morbidities, including cardiovascular and metabolic diseases, sleep deprivation and depression. Whether this coincidental occurrence is fortuitous or involves cause-effect relationships is unknown. This review aims at collating and integrating present knowledge on both diseases by providing a brief overview of their epidemiology and pathophysiology, analyzing current evidences relating OA and OSA and discussing potential common mechanisms by which they can aggravate each other. Such mechanisms constitute potential therapeutic targets whose pharmacological modulation may provide more efficient ways of reducing the consequences of OA and OSA and, thus, lessen the huge individual and social burden that they impose.
Collapse
Affiliation(s)
- Laetitia S Gaspar
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal; PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Cátia Sousa
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Ana Rita Álvaro
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Cláudia Cavadas
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| | - Alexandrina Ferreira Mendes
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
26
|
Soni VK, Mehta A, Ratre YK, Tiwari AK, Amit A, Singh RP, Sonkar SC, Chaturvedi N, Shukla D, Vishvakarma NK. Curcumin, a traditional spice component, can hold the promise against COVID-19? Eur J Pharmacol 2020; 886:173551. [PMID: 32931783 PMCID: PMC7832734 DOI: 10.1016/j.ejphar.2020.173551] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/17/2020] [Accepted: 09/10/2020] [Indexed: 12/16/2022]
Abstract
The severity of the recent pandemic and the absence of any specific medication impelled the identification of existing drugs with potential in the treatment of Coronavirus disease-2019 (COVID-19), caused by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). Curcumin, known for its pharmacological abilities especially as an anti-inflammatory agent, can be hypothesized as a potential candidate in the therapeutic regimen. COVID-19 has an assorted range of pathophysiological consequences, including pulmonary damage, elevated inflammatory response, coagulopathy, and multi-organ damage. This review summarizes the several evidences for the pharmacological benefits of curcumin in COVID-19-associated clinical manifestations. Curcumin can be appraised to hinder cellular entry, replication of SARS-CoV-2, and to prevent and repair COVID-19-associated damage of pneumocytes, renal cells, cardiomyocytes, hematopoietic stem cells, etc. The modulation and protective effect of curcumin on cytokine storm-related disorders are also discussed. Collectively, this review provides grounds for its clinical evaluation in the therapeutic management of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Vivek Kumar Soni
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, 495009, India
| | - Arundhati Mehta
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, 495009, India
| | - Yashwant Kumar Ratre
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, 495009, India
| | - Atul Kumar Tiwari
- Department of Zoology, Bhanwar Singh Porte Government Science College, Pendra, Chhattisgarh, India
| | - Ajay Amit
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, 495009, India
| | - Rajat Pratap Singh
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, 495009, India
| | - Subash Chandra Sonkar
- Multidisciplinary Research Unit, Maulana Azad Medical College, University of Delhi, New Delhi, India
| | - Navaneet Chaturvedi
- Department of Molecular and Cell Biology, Henry Welcome Building, University of Leicester, Leicester, LE26AW, UK; School of Biochemical Engineering, Indian Institute of Technology-Banaras Hindu University (IIT-BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Dhananjay Shukla
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, 495009, India.
| | - Naveen Kumar Vishvakarma
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, 495009, India.
| |
Collapse
|
27
|
Bourdier G, Détrait M, Bouyon S, Lemarié E, Brasseur S, Doutreleau S, Pépin J, Godin‐Ribuot D, Belaidi E, Arnaud C. Intermittent Hypoxia Triggers Early Cardiac Remodeling and Contractile Dysfunction in the Time-Course of Ischemic Cardiomyopathy in Rats. J Am Heart Assoc 2020; 9:e016369. [PMID: 32805159 PMCID: PMC7660805 DOI: 10.1161/jaha.120.016369] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Sleep-disordered breathing is associated with a poor prognosis (mortality) in patients with ischemic cardiomyopathy. The understanding of mechanisms linking intermittent hypoxia (IH), the key feature of sleep-disordered breathing, to ischemic cardiomyopathy progression is crucial for identifying specific actionable therapeutic targets. The aims of the present study were (1) to evaluate the impact of IH on the time course evolution of cardiac remodeling and contractile dysfunction in a rat model of ischemic cardiomyopathy; and (2) to determine the impact of IH on sympathetic activity, hypoxia inducible factor-1 activation, and endoplasmic reticulum stress in the time course of ischemic cardiomyopathy progression. METHODS AND RESULTS Ischemic cardiomyopathy was induced by a permanent ligature of the left coronary artery in male Wistar rats (rats with myocardial infarction). Rats with myocardial infarction were then exposed to either IH or normoxia for up to 12 weeks. Cardiac remodeling and function were analyzed by Sirius red and wheat germ agglutinin staining, ultrasonography, and cardiac catheterization. Sympathetic activity was evaluated by spectral analysis of blood pressure variability. Hypoxia-inducible factor-1α activation and burden of endoplasmic reticulum stress were characterized by Western blots. Long-term IH exposure precipitated cardiac remodeling (hypertrophy and interstitial fibrosis) and contractile dysfunction during the time course evolution of ischemic cardiomyopathy in rodents. Among associated mechanisms, we identified the early occurrence and persistence of sympathetic activation, associated with sustained hypoxia-inducible factor-1α expression and a delayed pro-apoptotic endoplasmic reticulum stress. CONCLUSIONS Our data provide the demonstration of the deleterious impact of IH on post-myocardial infarction remodeling and contractile dysfunction. Further studies are needed to evaluate whether targeting sympathetic nervous system or HIF-1 overactivities could limit these effects and improve management of coexisting ischemic cardiomyopathy and sleep-disordered breathing.
Collapse
Affiliation(s)
| | - Maximin Détrait
- Univ. Grenoble AlpesINSERMCHU Grenoble AlpesHP2GrenobleFrance
| | - Sophie Bouyon
- Univ. Grenoble AlpesINSERMCHU Grenoble AlpesHP2GrenobleFrance
| | - Emeline Lemarié
- Univ. Grenoble AlpesINSERMCHU Grenoble AlpesHP2GrenobleFrance
| | | | | | | | | | - Elise Belaidi
- Univ. Grenoble AlpesINSERMCHU Grenoble AlpesHP2GrenobleFrance
| | - Claire Arnaud
- Univ. Grenoble AlpesINSERMCHU Grenoble AlpesHP2GrenobleFrance
| |
Collapse
|