1
|
Lisboa CD, Maciel de Souza JL, Gaspar CJ, Turck P, Ortiz VD, Teixeira Proença IC, Fernandes TRG, Fernandes E, Tasca S, Carraro CC, Belló-Klein A, Sander da Rosa Araujo A, Luz de Castro A. Melatonin effects on oxidative stress and on TLR4/NF-kβ inflammatory pathway in the right ventricle of rats with pulmonary arterial hypertension. Mol Cell Endocrinol 2024; 592:112330. [PMID: 39002930 DOI: 10.1016/j.mce.2024.112330] [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: 05/23/2024] [Revised: 06/19/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Pulmonary arterial hypertension (PAH) is characterised by an increase in mean pulmonary arterial pressure and a compromised the right ventricle (RV), together with progression to heart failure and premature death. Studies have evaluated the role of melatonin as a promising therapeutic strategy for PAH. The objective of this study was to evaluate melatonin's effects on oxidative stress and on the TLR4/NF-kβ inflammatory pathway in the RV of rats with PAH. Male Wistar rats were divided into the following groups: control, monocrotaline (MCT), and monocrotaline plus melatonin groups. These two last groups received one intraperitoneal injection of MCT (60 mg/kg) on the first day of experimental protocol. The monocrotaline plus melatonin group received 10 mg/kg/day of melatonin by gavage for 21 days. Echocardiographic analysis was performed, and the RV was collected for morphometric analysis oxidative stress and molecular evaluations. The main findings of the present study were that melatonin administration attenuated the reduction in RV function that was induced by monocrotaline, as assessed by TAPSE. In addition, melatonin prevented RV diastolic area reduction caused by PAH. Furthermore, animals treated with melatonin did not show an increase in ROS levels or in NF-kβ expression. In addition, the monocrotaline plus melatonin group showed a reduction in TLR4 expression when compared with control and monocrotaline groups. To our knowledge, this is the first study demonstrating a positive effect of melatonin on the TLR4/NF-kβ pathway in the RV of rats with PAH. In this sense, this study makes it possible to think of melatonin as a possible ally in mitigating RV alterations caused by PAH.
Collapse
Affiliation(s)
- Cristiane Dias Lisboa
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - José Luciano Maciel de Souza
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Custódio José Gaspar
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Patrick Turck
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Vanessa Duarte Ortiz
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Isabel Cristina Teixeira Proença
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Tânia Regina G Fernandes
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Elissa Fernandes
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Silvio Tasca
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Cristina Campos Carraro
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Adriane Belló-Klein
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Alex Sander da Rosa Araujo
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Alexandre Luz de Castro
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil.
| |
Collapse
|
2
|
Jing X, Chen Z, Zhang M, Luo C, Yang B, Lv Y, Li Y, Zeng L, Lin W. Melatonin mitigates the lipopolysaccharide-induced myocardial injury in rats by blocking the p53/xCT pathway-mediated ferroptosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03367-2. [PMID: 39153053 DOI: 10.1007/s00210-024-03367-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
This article examined the therapeutic effect of melatonin (MT) on the lipopolysaccharide (LPS)-induced myocardial injury, and the mechanisms involved. Septic rat model was constructed by exposing to lipopolysaccharide (LPS), and treated by MT, Ferrostatin-1 (Fer-1) and Erastin (Era). Hematoxylin-eosin staining was executed to appraise myocardial injury. H9c2 cells that exposed to LPS to induce in vitro sepsis cell model were treated by MT. p53 overexpression vectors were transfected into H9c2 cells. Inflammation- and ferroptosis-related indicators were examined by enzyme-linked immunosorbent assay. Expression of p53, xCT and GPX4 was scrutinized by quantitative real-time polymerase chain reaction and Western blot. MT relieved myocardial injury in septic rats. It decreased IL-6 and TNF-α, elevated GPX4 and GSH, and reduced MDA and Fe2+ in myocardial tissues of septic rats. LPS induced p53 elevation and xCT reduction in rats' myocardial tissues. Nevertheless, MT treatment declined p53 and increased xCT in myocardial tissues of septic rats. Interestingly, the relieving effect of MT on myocardial injury in septic rats was enhanced by Fer-1, but reversed by Era. The LPS-induced H9c2 cell damage was relieved by MT treatment. Besides, MT decreased LDH, IL-6 and TNF-α, elevated xCT, GPX4 and GSH, and reduced MDA and Fe2+ in the LPS-induced H9c2 cells. Conversely, these influences of MT on the LPS-induced H9c2 cells were reversed by p53 overexpression. MT is proposed to be a promising agent for treating the LPS-induced myocardial injury, as it relieves myocardial injury by hindering the p53/xCT-mediated ferroptosis in the LPS-induced septic rats.
Collapse
Affiliation(s)
- Xin Jing
- Department of Intensive Care Medicine, Cardio-Cerebrovascular Hospital, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zhida Chen
- Department of Intensive Care Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Mingdao Zhang
- Department of Intensive Care Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Caiqin Luo
- Department of Intensive Care Medicine, Cardio-Cerebrovascular Hospital, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Bo Yang
- Department of Intensive Care Medicine, Cardio-Cerebrovascular Hospital, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yanlan Lv
- Department of Intensive Care Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Yue Li
- Department of Intensive Care Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Lina Zeng
- Department of Intensive Care Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Wenji Lin
- Emergency Department, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China.
| |
Collapse
|
3
|
Wen R, Zhang TN, Yang N. [Recent research on pyroptosis in sepsis-induced myocardial depression]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:774-781. [PMID: 39014956 PMCID: PMC11562036 DOI: 10.7499/j.issn.1008-8830.2312039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/30/2024] [Indexed: 07/18/2024]
Abstract
Sepsis-induced myocardial depression (SIMD), a common complication of sepsis, is one of the main causes of death in patients with sepsis. The pathogenesis of SIMD is complicated, and the process of SIMD remains incompletely understood, with no single or definitive mechanism fully elucidated. Notably, pyroptosis, as a pro-inflammatory programmed cell death, is characterized by Gasdermin-mediated formation of pores on the cell membrane, cell swelling, and cell rupture accompanied by the release of large amounts of inflammatory factors and other cellular contents. Mechanistically, pyroptosis is mainly divided into the canonical pathway mediated by caspase-1 and the non-canonical pathway mediated by caspase-4/5/11. Pyroptosis has been confirmed to participate in various inflammation-associated diseases. In recent years, more and more studies have shown that pyroptosis is also involved in the occurrence and development of SIMD. This article reviews the molecular mechanisms of pyroptosis and its research progress in SIMD, aiming to provide novel strategies and targets for the treatment of SIMD.
Collapse
Affiliation(s)
- Ri Wen
- Department of Pediatric Intensive Care Unit, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Tie-Ning Zhang
- Department of Pediatric Intensive Care Unit, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Ni Yang
- Department of Pediatric Intensive Care Unit, Shengjing Hospital, China Medical University, Shenyang 110004, China
| |
Collapse
|
4
|
Li S, Gu X, Zhang M, Jiang Q, Xu T. Di (2-ethylhexyl) phthalate and polystyrene microplastics co-exposure caused oxidative stress to activate NF-κB/NLRP3 pathway aggravated pyroptosis and inflammation in mouse kidney. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 926:171817. [PMID: 38513858 DOI: 10.1016/j.scitotenv.2024.171817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
Polystyrene microplastic (PS-MPs) contamination has become a worldwide hotspot of concern, and its entry into organisms can cause oxidative stress resulting in multi-organ damage. The plasticizer di (2-ethylhexyl) phthalate (DEHP) is a common endocrine disruptor, these two environmental toxins often occur together, but their combined toxicity to the kidney and its mechanism of toxicity are unknown. Therefore, in this study, we established PS-MPS and/or DEHP-exposed mouse models. The results showed that alone exposure to both PS-MPs and DEHP caused inflammatory cell infiltration, cell membrane rupture, and content spillage in kidney tissues. There were also down-regulation of antioxidant enzyme levels, increased ROS content, activated of the NF-κB pathway, stimulated the levels of heat shock proteins (HSPs), pyroptosis, and inflammatory associated factors. Notably, the co-exposure group showed greater toxicity to kidney tissues, the cellular assay further validated these results. The introduction of the antioxidant n-acetylcysteine (NAC) and the NLRP3 inhibitor (MCC950) could mitigate the changes in the above measures. In summary, co-exposure of PS-MPs and DEHP induced oxidative stress that activated the NF-κB/NLRP3 pathway and aggravated kidney pyroptosis and inflammation, as well as that HSPs are also involved in this pathologic injury process. This study not only enriched the nephrotoxicity of plasticizers and microplastics, but also provided new insights into the toxicity mechanisms of multicomponent co-pollution in environmental.
Collapse
Affiliation(s)
- Shanshan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xuedie Gu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Muyue Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Qihang Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Tong Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China..
| |
Collapse
|
5
|
Fan Y, Guan B, Xu J, Zhang H, Yi L, Yang Z. Role of toll-like receptor-mediated pyroptosis in sepsis-induced cardiomyopathy. Biomed Pharmacother 2023; 167:115493. [PMID: 37734261 DOI: 10.1016/j.biopha.2023.115493] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023] Open
Abstract
Sepsis, a life-threatening dysregulated status of the host response to infection, can cause multiorgan dysfunction and mortality. Sepsis places a heavy burden on the cardiovascular system due to the pathological imbalance of hyperinflammation and immune suppression. Myocardial injury and cardiac dysfunction caused by the aberrant host responses to pathogens can lead to cardiomyopathy, one of the most critical complications of sepsis. However, many questions about the specific mechanisms and characteristics of this complication remain to be answered. The causes of sepsis-induced cardiac dysfunction include abnormal cardiac perfusion, myocardial inhibitory substances, autonomic dysfunction, mitochondrial dysfunction, and calcium homeostasis dysregulation. The fight between the host and pathogens acts as the trigger for sepsis-induced cardiomyopathy. Pyroptosis, a form of programmed cell death, plays a critical role in the progress of sepsis. Toll-like receptors (TLRs) act as pattern recognition receptors and participate in innate immune pathways that recognize damage-associated molecular patterns as well as pathogen-associated molecular patterns to mediate pyroptosis. Notably, pyroptosis is tightly associated with cardiac dysfunction in sepsis and septic shock. In line with these observations, induction of TLR-mediated pyroptosis may be a promising therapeutic approach to treat sepsis-induced cardiomyopathy. This review focuses on the potential roles of TLR-mediated pyroptosis in sepsis-induced cardiomyopathy, to shed light on this promising therapeutic approach, thus helping to prevent and control septic shock caused by cardiovascular disorders and improve the prognosis of sepsis patients.
Collapse
Affiliation(s)
- Yixuan Fan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Intensive Care Unit, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baoyi Guan
- Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Guangzhou University of Chinese Medicine
| | - Jianxing Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Intensive Care Unit, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - He Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Liang Yi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Intensive Care Unit, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Zhixu Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Intensive Care Unit, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
6
|
Zhang H, Liao J, Jin L, Lin Y. NLRP3 inflammasome involves in the pathophysiology of sepsis-induced myocardial dysfunction by multiple mechanisms. Biomed Pharmacother 2023; 167:115497. [PMID: 37741253 DOI: 10.1016/j.biopha.2023.115497] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/25/2023] Open
Abstract
Sepsis-induced myocardial dysfunction (SIMD) is one of the serious health-affecting problems worldwide. At present, the mechanisms of SIMD are still not clearly elucidated. The NOD-like receptor protein 3 (NLRP3) inflammasome has been assumed to be involved in the pathophysiology of SIMD by regulating multiple biological processes. NLRP3 inflammasome and its related signaling pathways might affect the regulation of inflammation, autophagy, apoptosis, and pyroptosis in SIMD. A few molecular specific inhibitors of NLRP3 inflammasome (e.g., Melatonin, Ulinastatin, Irisin, Nifuroxazide, and Ginsenoside Rg1, etc.) have been developed, which showed a promising anti-inflammatory effect in a cellular or animal model of SIMD. These experimental findings indicated that NLRP3 inflammasome could be a promising therapeutic target for SIMD treatment. However, the clinical translation of NLRP3 inhibitors for treating SIMD still requires robust in vivo and preclinical trials.
Collapse
Affiliation(s)
- Hongwei Zhang
- Department of Emergency Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China
| | - Jian Liao
- Department of Nephrology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, Zhejiang, China
| | - Litong Jin
- Department of Emergency Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China
| | - Yan Lin
- Department of Critical Care Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China.
| |
Collapse
|
7
|
Yang D, Yuan L, Chen G, Chen S, Ma X, Xing Y, Song J. Expression and role of melatonin membrane receptors in the hypothalamic-pituitary-testicular axis of Tibetan sheep in a plateau pastoral area. PLoS One 2023; 18:e0290775. [PMID: 37878614 PMCID: PMC10599587 DOI: 10.1371/journal.pone.0290775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/16/2023] [Indexed: 10/27/2023] Open
Abstract
MTNR1A and MTNR1B, two high-affinity MT membrane receptors found in mammals, mediate the activity of MT on the HPGA to regulate animal reproduction. Nevertheless, the expression patterns and function of the MTNR1A and MTNR1B genes in the HPTA of seasonal estrus sheep and perennial estrus sheep have not been elucidated. We studied the expression of MTNR1A and MTNR1B in the hypothalamic-pituitary-testicular axis (HPTA) of Tibetan sheep at different reproductive stages using histochemistry, enzyme linked immunosorbent assay (ELSIA), scanning electron microscopy, transmission electron microscopy, quantitative Real-time PCR (qRT-PCR), and Western blot (WB), and analyzed the relationship between their expression and reproductive hormone receptors. We also compared relevant characteristics between seasonal Tibetan sheep and non-seasonal Small Tail Han sheep in the same pastoral area. The results showed that MTNR1A and MTNR1B were expressed in all tissues of the Tibetan sheep HPTA, and both were co-expressed in the cytoplasm of epididymis basal and halo cells located at common sites of the epididymis basement membrane, forming an immune barrier. The qRT-PCR analysis showed that not only MTNR1A but also N-acetyltransferase (AANAT), hydroxyindole-oxygen- methyltransferase (HIOMT), androgen receptor (AR), and estrogen receptor α (ERα) mRNA expression was significantly upregulated in the testis and epididymis of Tibetan sheep during the breeding season, whereas no clear upregulation of these genes was observed in the tissues of Small Tail Han sheep. MTNR1A and MTNR1B are important regulators of the HPTA in sheep. MTNR1A mediates seasonal estrus regulation in Tibetan sheep. Both MTNR1A and MTNR1B may play important roles in formation of the blood-epididymal barrier. The results of this study should help advance research on the mechanism of reproductive regulation of the HPTA in male animals and provide reference data for improving the reproductive rate of seasonal breeding animals.
Collapse
Affiliation(s)
- Dapeng Yang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Ligang Yuan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, China
| | - Guojuan Chen
- Huangzhong District Animal Disease Control Center of Xining City, Xining City, Qinghai Province, China
| | - Shaoyu Chen
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Xiaojie Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yindi Xing
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Juanjuan Song
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
8
|
Tian K, Yang Y, Zhou K, Deng N, Tian Z, Wu Z, Liu X, Zhang F, Jiang Z. The role of ROS-induced pyroptosis in CVD. Front Cardiovasc Med 2023; 10:1116509. [PMID: 36873396 PMCID: PMC9978107 DOI: 10.3389/fcvm.2023.1116509] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Cardiovascular disease (CVD) is the number one cause of death in the world and seriously threatens human health. Pyroptosis is a new type of cell death discovered in recent years. Several studies have revealed that ROS-induced pyroptosis plays a key role in CVD. However, the signaling pathway ROS-induced pyroptosis has yet to be fully understood. This article reviews the specific mechanism of ROS-mediated pyroptosis in vascular endothelial cells, macrophages, and cardiomyocytes. Current evidence shows that ROS-mediated pyroptosis is a new target for the prevention and treatment of cardiovascular diseases such as atherosclerosis (AS), myocardial ischemia-reperfusion injury (MIRI), and heart failure (HF).
Collapse
Affiliation(s)
- Kaijiang Tian
- The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Yu Yang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Kun Zhou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Nianhua Deng
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Zhen Tian
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Zefan Wu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Xiyan Liu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| | - Fan Zhang
- The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Zhisheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China
| |
Collapse
|
9
|
Li Y, Feng L, Bai L, Jiang H. Study of Therapeutic Mechanisms of Puerarin against Sepsis-Induced Myocardial Injury by Integrating Network Pharmacology, Bioinformatics Analysis, and Experimental Validation. Crit Rev Immunol 2023; 43:25-42. [PMID: 37824375 DOI: 10.1615/critrevimmunol.2023050050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Myocardial injury is the most prevalent and serious complication of sepsis. The potential of puerarin (Pue) to treat sepsis-induced myocardial injury (SIMI) has been recently reported. Nevertheless, the specific anti-SIMI mechanisms of Pue remain largely unclear. Integrating network pharmacology, bioinformatics analysis, and experimental validation, we aimed to clarify the anti-SIMI mechanisms of Pue, thereby furnishing novel therapeutic targets. Pue-associated targets were collected from HIT, GeneCards, SwissTargetPrediction, SuperPred, and CTD databases. SIMI-associated targets were acquired from GeneCards and DisGeNET. Differentially expressed genes (DEGs) were identified from GEO database. Potential anti-SIMI targets of Pue were determined using VennDiagram. ClusterProfiler was employed for GO and KEGG analyses. STRING database and Cytoscape were used for protein-protein interaction (PPI) network construction, and cytoHubba was used for hub target screening. PyMOL and AutoDock were utilized for molecular docking. An in vitro SIMI model was built to further verify the therapeutic mechanisms of Pue. Seventy-three Pue-SIMI-DEG intersecting target genes were obtained. GO and KEGG analyses revealed that the targets were principally concentrated in cellular response to chemical stress, response to oxidative stress (OS), and insulin and neurotrophin signaling pathways. Through PPI analysis and molecular docking, AKT1, CASP3, TP53, and MAPK3 were identified as the pivotal targets. In vivo experiments indicated that Pue promoted cell proliferation, downregulated AKT1, CASP3, TP53, and MAPK3, and inhibited inflammation, myocardial injury, OS, and apoptosis in the cell model. Pue might inhibit inflammation, myocardial injury, OS, and apoptosis to treat SIMI by reducing AKT1, CASP3, TP53, and MAPK3.
Collapse
Affiliation(s)
- Yin Li
- Department of Emergency, Huadong Hospital Fudan University, Shanghai 200040, China
| | - Lei Feng
- Department of Emergency, Huadong Hospital Fudan University, Shanghai 200040, China
| | - Lin Bai
- Department of Emergency, Huadong Hospital Fudan University, Shanghai 200040, China
| | - Hao Jiang
- Department of Emergency, Huadong Hospital Fudan University, Shanghai 200040, China
| |
Collapse
|
10
|
Ye D, Xu Y, Shi Y, Fan M, Lu P, Bai X, Feng Y, Hu C, Cui K, Tang X, Liao J, Huang W, Xu F, Liang X, Huang J. Anti-PANoptosis is involved in neuroprotective effects of melatonin in acute ocular hypertension model. J Pineal Res 2022; 73:e12828. [PMID: 36031799 DOI: 10.1111/jpi.12828] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/12/2022] [Accepted: 08/24/2022] [Indexed: 11/29/2022]
Abstract
Acute ocular hypertension (AOH) is the most important characteristic of acute glaucoma, which can lead to retinal ganglion cell (RGC) death and permanent vision loss. So far, approved effective therapy is still lacking in acute glaucoma. PANoptosis (pyroptosis, apoptosis, and necroptosis), which consists of three key modes of programmed cell death-apoptosis, necroptosis, and pyroptosis-may contribute to AOH-induced RGC death. Previous studies have demonstrated that melatonin (N-acetyl-5-methoxytryptamine) exerts a neuroprotective effect in many retinal degenerative diseases. However, whether melatonin is anti-PANoptotic and neuroprotective in the progression of acute glaucoma remains unclear. Thus, this study aimed to explore the role of melatonin in AOH retinas and its underlying mechanisms. The results showed that melatonin treatment attenuated the loss of ganglion cell complex thickness, retinal nerve fiber layer thickness, and RGC after AOH injury, and improved the amplitudes of a-wave, b-wave, and oscillatory potentials in the electroretinogram. Additionally, the number of terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive cells was decreased, and the upregulation of cleaved caspase-8, cleaved caspase-3, Bax, and Bad and downregulation of Bcl-2 and p-Bad were inhibited after melatonin administration. Meanwhile, both the expression and activation of MLKL, RIP1, and RIP3, along with the number of PI-positive cells, were reduced in melatonin-treated mice, and p-RIP3 was in both RGC and microglia/macrophage after AOH injury. Furthermore, melatonin reduced the expression of NLRP3, ASC, cleaved caspase-1, gasdermin D (GSDMD), and cleaved GSDMD, and decreased the number of Iba1/interleukin-1β-positive cells. In conclusion, melatonin ameliorated retinal structure, prevented retinal dysfunction after AOH, and exerted a neuroprotective effect via inhibition of PANoptosis in AOH retinas.
Collapse
Affiliation(s)
- Dan Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuxun Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Matthew Fan
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Peng Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xue Bai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yanlin Feng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Chenyang Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jing Liao
- Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Nanning, China
| | - Wei Huang
- Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Nanning, China
| | - Fan Xu
- Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Nanning, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jingjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
11
|
Liu R, Luo X, Li J, Lei Y, Zeng F, Huang X, Lan Y, Yang F. Melatonin: A window into the organ-protective effects of sepsis. Biomed Pharmacother 2022; 154:113556. [PMID: 35994818 DOI: 10.1016/j.biopha.2022.113556] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/22/2022] [Accepted: 08/14/2022] [Indexed: 11/02/2022] Open
Abstract
Sepsis is an uncontrolled host response to infection. In some cases, it progresses to multi-organ insufficiency, leading to septic shock and increased risk of mortality. Various organ support strategies are currently applied clinically, but they are still inadequate in terms of reducing mortality. Melatonin is a hormone that regulates sleep and wakefulness, and it is associated with a reduced risk of death in patients with sepsis. Evidence suggests that melatonin may help protect organ function from sepsis-related damage. Here, we review information related to the role of melatonin in protecting organ function during sepsis and explore its potential clinical applications, with the aim of providing an effective therapeutic strategy for treating sepsis-induced organ insufficiency.
Collapse
Affiliation(s)
- Rongan Liu
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoxiu Luo
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiajia Li
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Lei
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fan Zeng
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaobo Huang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunping Lan
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Fuxun Yang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
12
|
Ye J, Li M, Li Q, Jia Z, Hu X, Zhao G, Zhi S, Hong G, Lu Z. Activation of STIM1/Orai1‑mediated SOCE in sepsis‑induced myocardial depression. Mol Med Rep 2022; 26:259. [PMID: 35713214 DOI: 10.3892/mmr.2022.12775] [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: 02/04/2021] [Accepted: 02/21/2022] [Indexed: 11/05/2022] Open
Abstract
Unbalanced Ca2+ homeostasis serves an essential role in the occurrence and development of septic myocardial injury. However, the mechanism of Ca2+ homeostasis in septic myocardial depression is poorly understood due to the complexity of Ca2+ transporters in excitable cells. It was therefore hypothesized that cardiac dysfunction, myocardial injury and cardiac apoptosis in septic myocardial depression are associated with elevated intracellular Ca2+ concentrations caused by stromal interaction molecule 1 (STIM1)/Orai calcium release‑activated calcium modulator 1 (Orai1)‑mediated store‑operated Ca2+ entry (SOCE). A septic myocardial depression model was established using the cecal ligation and puncture operation (CLP) in mice and was simulated in H9C2 cells via lipopolysaccharide (LPS) stimulation. Cardiac function, myocardial injury, cardiac apoptosis and the expression levels of Bax, Bcl‑2, STIM1 and Orai1 were quantified in vivo at 6, 12 and 24 h. Changes in the intracellular Ca2+ concentration, SOCE and the distribution of STIM1 were assessed in vitro within 6 h. The morphological changes of heart tissue were observed by hematoxylin‑eosin staining. Myocardial cellular apoptosis was determined by TUNEL method. The expression of Bax, Bcl‑2, STIM1 and Orai1 were visualized by western blot. Cytosolic calcium concentration and SOCE were evaluated by confocal microscopy. The results demonstrated that cardiac contractile function was significantly reduced at 6 h and morphological changes in cardiac tissues, as well as the myocardial apoptosis rate, were markedly increased at 6, 12 and 24 h following CLP. mRNA and protein expression levels of Bax/Bcl‑2 were significantly enhanced at 6 and 12 h and glycosylation of Orai1 in the myocardium of septic mice was significantly increased at 6 h following CLP. The intracellular Ca2+ concentration, SOCE, was significantly increased at 1‑2 h and the clustering and distribution of STIM1 were markedly changed in H9C2 cells at 1 and 2 h. These findings suggested that myocardial dysfunction, cardiac injury and myocardial depression may be related to increased intracellular Ca2+ concentration resulting from STIM1/Orai1‑mediated SOCE, which may provide a potential method to alleviate septic myocardial depression.
Collapse
Affiliation(s)
- Jingjing Ye
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Mengfang Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Qiao Li
- Ultrasound Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhijun Jia
- Ultrasound Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiyi Hu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Guangju Zhao
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Shaoce Zhi
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Guangliang Hong
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhongqiu Lu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
13
|
Theofilis P, Vordoni A, Kalaitzidis RG. The Role of Melatonin in Chronic Kidney Disease and Its Associated Risk Factors: A New Tool in Our Arsenal? Am J Nephrol 2022; 53:565-574. [PMID: 35767942 DOI: 10.1159/000525441] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/25/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND The increasing incidence of chronic kidney disease (CKD), as a consequence of the high prevalence of arterial hypertension and type 2 diabetes mellitus (T2DM), warrants the need for developing effective treatment approaches. In this regard, the pineal gland-derived hormone melatonin may represent an appealing treatment approach of CKD and its associated risk factors. SUMMARY Targeting the adverse pathophysiology surrounding CKD and its associated risk factors has been the concept of pharmacologic treatment developed for its management. This review article aimed to present the role of melatonin in this direction, by providing an overview of melatonin's physiology followed by its effect as a therapeutic agent in arterial hypertension and T2DM. KEY MESSAGES Melatonin, the primary darkness hormone, possesses pleiotropic mechanisms of action which may have important implications in various pathologic states since its receptors are situated across various organ systems. As a treatment tool in arterial hypertension, melatonin may be efficacious in reducing both daytime and nocturnal blood pressure by influencing endothelial function, oxidative stress, the autonomic nervous system, and the renin-angiotensin system. Melatonin may also increase insulin sensitivity and β-cell function. However, late meal intake may be detrimental in glucose regulation, as consumption close to melatonin peak concentrations may induce hyperglycemia and insulin resistance. This finding may explain the inconsistent glycose regulation achieved with melatonin in clinical trials and meta-analyses. Additionally, the presence of genetic variants to melatonin receptor 2 may predispose to T2DM development. Finally, we present the available preclinical evidence supporting melatonin's efficacy in ameliorating CKD's pathophysiology since melatonin supplementation has not been adequately explored in patients with CKD. The combined use of stem cells with melatonin is an appealing therapeutic approach which ought to be assessed further.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- General Hospital of Nikaia-Piraeus Agios Panteleimon, Center for Nephrology "G. Papadakis", Piraeus, Greece
| | - Aikaterini Vordoni
- General Hospital of Nikaia-Piraeus Agios Panteleimon, Center for Nephrology "G. Papadakis", Piraeus, Greece
| | - Rigas G Kalaitzidis
- General Hospital of Nikaia-Piraeus Agios Panteleimon, Center for Nephrology "G. Papadakis", Piraeus, Greece
| |
Collapse
|
14
|
Chen R, Yang M. Melatonin Inhibits OGD/R-Induced H9c2 Cardiomyocyte Pyroptosis via Regulation of MT2/miR-155/FOXO3a/ARC Axis. Int Heart J 2022; 63:327-337. [PMID: 35354753 DOI: 10.1536/ihj.21-571] [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] [Indexed: 11/18/2022]
Abstract
Emerging literature suggests that pyroptosis plays a critical role in ischemia/hypoxia (I/R) -induced myocardial injury. Melatonin has been implicated in attenuating I/R-induced injury of cardiomyocytes. Nevertheless, whether melatonin inhibits I/R-induced pyroptosis of cardiomyocytes and the underlying molecular mechanisms remain unexploited.H9c2 cardiomyocytes were cultured under oxygen-glucose deprivation/reperfusion (OGD/R) condition to establish a myocardial pyroptosis model in vitro. OGD/R-induced pyroptosis was evaluated by CCK-8 assay, IL-1β and IL-18 release, and western blotting. Luciferase reporter assay was utilized to validate the association between miR-155 and Forkhead box O3a (FOXO3a).Melatonin could inhibit OGD/R-induced pyroptosis of H9c2 cells and upregulation of FOXO3a contributed to the antipyroptotic effect of melatonin. Melatonin reduced miR-155 expression, which led to FOXO3a upregulation and inhibition of pyroptosis in OGD/R-exposed H9c2 cells. miR-155 inhibitor enhanced the antipyroptotic effect of melatonin in OGD/R-exposed H9c2 cells. Melatonin-induced downregulation of miR-155 and upregulation of FOXO3a were reversed by melatonin receptor 2 (MT2) siRNA. Melatonin treatment also led to an increased level of apoptosis repressor with caspase recruitment domain (ARC), which was inhibited by FOXO3a siRNA. Moreover, silencing ARC by siRNA significantly blocked the antipyroptotic actions of melatonin, whereas ARC overexpression enhanced the antipyroptotic actions of melatonin in OGD/R-exposed H9c2 cells.Our findings demonstrated that melatonin prevented OGD/R-induced pyroptosis via regulating the MT2/miR-155/FOXO3a/ARC axis in cardiomyocytes.
Collapse
Affiliation(s)
- Rui Chen
- Department of Cardiology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine
| | - Min Yang
- Prevention Treatment Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine
| |
Collapse
|
15
|
Zhou T, Li Z, Chen H. Melatonin alleviates lipopolysaccharide (LPS) / adenosine triphosphate (ATP)-induced pyroptosis in rat alveolar Type II cells (RLE-6TN) through nuclear factor erythroid 2-related factor 2 (Nrf2)-driven reactive oxygen species (ROS) downregulation. Bioengineered 2022; 13:1880-1892. [PMID: 35109747 PMCID: PMC8973817 DOI: 10.1080/21655979.2021.2018981] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pyroptosis has pivotal parts within disease development, rendering this attractive mechanism for novel therapeutics. This investigation aimed at analyzing melatonin roles within pyroptosis together with related mechanistics. RLE-6TN cultures were exposed to varying LPS doses for 4.5 h followed by concomitant culturing in the presence of ATP (5 mM) for 0.5 h to induce injury, and the roles of melatonin, N-Acety-L-cysteine (NAC - a ROS scavenger), ML385 (specific Nrf2 inhibitor) were examined. Apoptosis analysis was performed through lactate dehydrogenase (LDH) activity assays, together with propidium iodide (PI) stain-assay. Intracellular ROS were quantified through 2, 7-dichlorodihydrofluorescein diacetate (DCFH-DA). Pyrolysis-associated proteins, such as nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3), apoptosis-associated speck-like protein containing a CARD (ASC), cysteine aspartate-specific protease-1 P20 (Caspase-1 P20), gasdermin D-N (GSDMD-N), and mature interleukin-1β (IL-1β), were identified through Western blotting. Dataset outcomes demonstrated LPS/ATP induce RLE-6TN cell pyroptosis, while melatonin alleviated this phenomenon, visualized through increased cell survival rate, reduction of LDH discharge and PI+ cellular count. Moreover, melatonin effectively reduced NLRP3 inflammasome triggering in RLE-6TN cells. Meanwhile, this study demonstrated melatonin thwarting over NLRP3 inflammasome triggering was depending on ROS. In addition, this study found that melatonin activated Nrf2/Heme Oxygenase-1 (HO-1) pathway, with pyroptotic-inhibiting function of melatonin was reverted through a bespoke Nrf2-inhibitor and siNrf2. In summary, this study concluded that melatonin prevents RLE-6TN cellular pyroptosis through Nrf2-triggered ROS downregulation.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Pulmonary and Critical care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhaodong Li
- Basic Medicine College, Chongqing Medical University,1# Medical College Road, Yuzhong District, Chongqing, China
| | - Hong Chen
- Department of Pulmonary and Critical care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Zhang X, Zhao X, Tie S, Li J, Su W, Tan M. A smart cauliflower-like carrier for astaxanthin delivery to relieve colon inflammation. J Control Release 2022; 342:372-387. [PMID: 35038495 DOI: 10.1016/j.jconrel.2022.01.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/24/2021] [Accepted: 01/07/2022] [Indexed: 12/12/2022]
Abstract
As a fat-soluble carotenoid, astaxanthin has excellent antioxidant and anti-inflammation biological activities, but its poor biocompatibility and low stability limit application of astaxanthin in the food industry. In this study, cauliflower-like carriers (CCs) were constructed based on caseinate, chitosan-triphenylphosphonium (TPP) and sodium alginate through an electrostatic self-assembly method to improve the biocompatibility, stability and targeting transport properties of astaxanthin. The smart CCs showed pH-response release and mitochondrial targeted characteristics. In vitro studies demonstrated that the CCs could improve the internalization of astaxanthin, and significantly inhibited the excessive production of reactive oxygen species and the depolarization of mitochondrial membrane potential caused by oxidative stress. In vivo studies revealed that the astaxanthin-loaded CCs could effectively relieve the colitis induced by dextran sodium sulfate and protect the integrity of the colon tissue structure. The astaxanthin-loaded CCs could significantly inhibit the expression of inflammation factors such as interleukin-1β, interleukin-6, tumor necrosis factor alpha, cyclooxygenase-2, myeloperoxidase, inducible nitric oxide synthase, and nitric oxide. Moreover, the astaxanthin-loaded CCs could maintain the expression of zonula occludens-1, increase the abundance of Firmicutes and Lactobacillaceae in the intestine. In a word, the constructed astaxanthin delivery system provided a potential application for the oral uptake hydrophobic bio-activator in intervention of ulcerative colitis.
Collapse
Affiliation(s)
- Xuedi Zhang
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Xue Zhao
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Shanshan Tie
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Jiaxuan Li
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Wentao Su
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Mingqian Tan
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China.
| |
Collapse
|
17
|
Wei A, Liu J, Li D, Lu Y, Yang L, Zhuo Y, Tian W, Cong H. Syringaresinol attenuates sepsis-induced cardiac dysfunction by inhibiting inflammation and pyroptosis in mice. Eur J Pharmacol 2021; 913:174644. [PMID: 34801532 DOI: 10.1016/j.ejphar.2021.174644] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 12/17/2022]
Abstract
The mortality of sepsis-induced cardiac dysfunction (SICD) is very high due to the complex pathophysiological mechanism. Syringaresinol (SYR) is a natural abstract which possesses anti-inflammatory property. The present study aims was to identify the protective impact of SYR on sepsis-induced cardiac dysfunction and investigate the specific mechanisms. We found that SYR improved the cardiac function and alleviated myocardial injury in mice that subjected to cecal ligation and puncture, in addition, SIRT1 expression was significantly elevated after SYR treatment compared to sepsis group both in vivo and in vitro, along with suppression of NLRP3 activation and proinflammatory cytokines release. However, SIRT1 inhibitor EX427 abolished the impact of SYR on LPS-induced pyroptosis in cardiomyocytes. Furthermore, molecular docking analysis predicted that there is high affinity between SYR and estrogen receptor (ER), ER inhibitor ICI182780, the specific ERβ inhibitor PHTP and the specific ERαinhibitor AZD9496 were used to examine the role of ER in the protective effect of SYR against SICD, and the results suggested that ER activation was essential for the cardioprotective function of SYR. In conclusion, SYR ameliorates SICD via the ER/SIRT1/NLRP3/GSDMD pathway.
Collapse
Affiliation(s)
- Ao Wei
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, 300222, China
| | - Jingjing Liu
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, 300222, China
| | - Dihua Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Yanmin Lu
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Yuzhen Zhuo
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China.
| | - Wencong Tian
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Hongliang Cong
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, 300222, China.
| |
Collapse
|
18
|
Feng R, Adeniran SO, Huang F, Li Y, Ma M, Zheng P, Zhang G. The ameliorative effect of melatonin on LPS-induced Sertoli cells inflammatory and tight junctions damage via suppression of the TLR4/MyD88/NF-κB signaling pathway in newborn calf. Theriogenology 2021; 179:103-116. [PMID: 34871925 DOI: 10.1016/j.theriogenology.2021.11.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 11/20/2021] [Accepted: 11/25/2021] [Indexed: 12/23/2022]
Abstract
The blood-testicular barrier (BTB) is involved in spermatogenesis, protects sperm development, and plays a crucial role in the reproductive process. Tight junctions (TJs) between Sertoli cells (SCs) are the key structure of (BTB), and if its structure is damaged, BTB function is affected. The cellular inflammation caused by Gram-negative bacteria affects the structural integrity of TJs. Melatonin (MT) has anti-inflammatory effects; however, the effect of MT in newborn calf SCs is unknown. Therefore, this experiment studied the protective effect of MT. The results showed that LPS upregulated TLR4, MyD88, and NF-κB expressions, in turn, activated the TLR4/MyD88/NF-κB signaling pathway, produced a large amount of IL-6 and IL-1β, downregulated the expression of ZO-1 and Occludin, and reduced the viability of SCs, which resulted in the inflammatory response of SCs and damage of TJs. The addition of MT decreased TLR4, MyD88, and NF-κB expressions, it then inhibited the activation of TLR4/MyD88/NF-κB signaling pathway, downregulated the expression of IL-6 and IL-1β, upregulated the expression of ZO-1 and Occludin, and increased the cell viability, thereby alleviating the inflammatory response of SCs, and restored the TJs structure. Overall, our results reveal that MT can alleviate LPS-induced in newborn calf SCs Inflammation and TJs injury through TLR4/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Rui Feng
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Samson O Adeniran
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Fushuo Huang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Yulong Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Mingjun Ma
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Peng Zheng
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China
| | - Guixue Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, China.
| |
Collapse
|
19
|
Lu J, Xu L, Zeng Z, Xue C, Li J, Chen X, Zhou P, Lin S, Liao Y, Du X, Yang R, Zheng S. Normothermic ex vivo Heart Perfusion Combined With Melatonin Enhances Myocardial Protection in Rat Donation After Circulatory Death Hearts via Inhibiting NLRP3 Inflammasome-Mediated Pyroptosis. Front Cell Dev Biol 2021; 9:733183. [PMID: 34532321 PMCID: PMC8438322 DOI: 10.3389/fcell.2021.733183] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/11/2021] [Indexed: 01/06/2023] Open
Abstract
Objective The adoption of hearts from donation after circulatory death (DCD) is a promising approach for the shortage of suitable organs in heart transplantation. However, DCD hearts suffer from serious ischemia/reperfusion injury (IRI). Recent studies demonstrate that nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome-mediated pyroptosis is a novel target to ameliorate myocardial IRI. Melatonin is shown to inhibit NLRP3 inflammasome-mediated pyroptosis. Therefore, this study is designed to verify the hypothesis that melatonin can protect the heart graft preserved with ex vivo heart perfusion (EVHP) against myocardial IRI via inhibiting NLRP3 inflammasome-mediated pyroptosis in a rat model of DCD. Methods Donor-heart rats were randomly divided into three groups: (1) Control group: non-DCD hearts were harvested from heart-beating rats and immediately preserved with allogenic blood-based perfusate at constant flow for 105 min in the normothermic EVHP system; (2) DCD-vehicle group; and (3) DCD-melatonin group: rats were subjected to the DCD procedure with 25 min of warm ischemia injury and preserved by the normothermic EVHP system for 105 min. Melatonin (200 μmol/L) or vehicle was perfused in the cardioplegia and throughout the whole EVHP period. Cardiac functional assessment was performed every 30 min during EVHP. The level of oxidative stress, inflammatory response, apoptosis, and NLRP3 inflammasome-mediated pyroptosis of heart grafts submitted to EVHP were evaluated. Results Twenty five-minute warm ischemia injury resulted in a significant decrease in the developed pressure (DP), dP/dt max , and dP/dt min of left ventricular of the DCD hearts, while the treatment with melatonin significantly increased the DP, dP/dt max of the left ventricular of DCD hearts compared with DCD-vehicle group. Furthermore, warm ischemia injury led to a significant increase in the level of oxidative stress, inflammatory response, apoptosis, and NLRP3 inflammasome-mediated pyroptosis in the hearts preserved with EVHP. However, melatonin added in the cardioplegia and throughout the EVHP period significantly attenuated the level of oxidative stress, inflammatory response, apoptosis, and NLRP3 inflammasome-mediated pyroptosis compared with DCD-vehicle group. Conclusion EVHP combined with melatonin post-conditioning attenuates myocardial IRI in DCD hearts by inhibiting NLRP3 inflammasome-mediated pyroptosis, which might expand the donor pool by the adoption of transplantable DCD hearts.
Collapse
Affiliation(s)
- Jun Lu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liwei Xu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zifeng Zeng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chuqing Xue
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiale Li
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiong Chen
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pengyu Zhou
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoyan Lin
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuhui Liao
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Xianjin Du
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ronghua Yang
- Department of Burn Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|