1
|
Jia Z, Xu K, Li R, Yang S, Chen L, Zhang Q, Li S, Sun X. The critical role of Sirt1 in ischemic stroke. Front Pharmacol 2025; 16:1425560. [PMID: 40160465 PMCID: PMC11949987 DOI: 10.3389/fphar.2025.1425560] [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: 04/30/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Ischemic stroke, the most prevalent form of stroke, is responsible for the highest disability rates globally and ranks as the primary cause of mortality worldwide. Sirt1, extensively investigated in neurodegenerative disorders, is the most well-known and earliest member of the sirtuins family. However, its mechanism of action during ischemic stroke remains ambiguous. The literature examination revealed the intricate involvement of Sirt1 in regulating both physiological and pathological mechanisms during ischemic stroke. Sirt1 demonstrates deacetylation effects on PGC-1α, HMGB1, FOXOs, and p53. It hinders the activation of NLRP3 inflammasome and NF-κB while also engaging with AMPK. It regulates inflammatory response, oxidative stress, mitochondrial dysfunction, autophagy, pro-death, and necrotic apoptosis. Therefore, the potential of Sirt1 as a therapeutic target for the management of ischemic stroke is promising.
Collapse
Affiliation(s)
- Ziyi Jia
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ke Xu
- The Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ruobing Li
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Siyu Yang
- The Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Long Chen
- The Fourth Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qianwen Zhang
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shulin Li
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaowei Sun
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
2
|
Wei X, Guo H, Huang G, Luo H, Gong L, Meng P, Liu J, Zhang W, Mei Z. SIRT1 Alleviates Mitochondrial Fission and Necroptosis in Cerebral Ischemia/Reperfusion Injury via SIRT1-RIP1 Signaling Pathway. MedComm (Beijing) 2025; 6:e70118. [PMID: 40008377 PMCID: PMC11850763 DOI: 10.1002/mco2.70118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 12/20/2024] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Programmed cell death, including necroptosis, plays a critical role in the pathogenesis of cerebral ischemia/reperfusion injury (CIRI). Silent information regulator 1 (SIRT1) has been identified as a potential therapeutic target for CIRI, yet its precise role in regulating necroptosis remains controversial. Furthermore, the potential interaction between SIRT1 and receptor-interacting protein kinase 1 (RIP1) in this context is not fully understood. Sanpian Decoction (SPD), a classical traditional herbal formula, was previously shown to enhance SIRT1 expression in our studies. Our findings demonstrated that, both in vivo and in vitro, CIRI was associated with a decrease in SIRT1 levels and phosphorylated dynamin-related protein 1 (p-DRP1) at Ser637, alongside an increase in RIP1 and other necroptosis-related proteins. Co-immunoprecipitation and immunofluorescence analyses revealed a weakened interaction between SIRT1 and RIP1. Furthermore, abnormal mitochondrial fission and dysfunction were mediated through the phosphoglycerate mutase 5-DRP1 pathway. Notably, SPD treatment improved neurological outcomes and reversed these pathological changes by enhancing the SIRT1-RIP1 interaction. In conclusion, this study suggests that SIRT1 is a promising therapeutic target for CIRI, capable of inhibiting necroptosis and mitigating mitochondrial fission via the SIRT1-RIP1 pathway. SPD exhibits therapeutic potential by activating SIRT1, thereby attenuating necroptosis and mitochondrial fission during CIRI.
Collapse
Affiliation(s)
- Xuan Wei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Hanjing Guo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Guangshan Huang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Haoyue Luo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Lipeng Gong
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Pan Meng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Jiyong Liu
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine DiagnosticsHunan University of Chinese MedicineChangshaHunanChina
| | - Wenli Zhang
- School of PharmacyHunan University of Chinese MedicineChangshaHunanChina
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
- Third‐Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese MedicineCollege of Medicine and Health SciencesChina Three Gorges UniversityYichangHubeiChina
| |
Collapse
|
3
|
Pei J, Wei Y, Lv L, Tao H, Zhang H, Ma Y, Han L. Preliminary evidence for the presence of programmed cell death in pressure injuries. J Tissue Viability 2024; 33:720-725. [PMID: 39095251 DOI: 10.1016/j.jtv.2024.07.012] [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: 01/03/2024] [Revised: 05/23/2024] [Accepted: 07/21/2024] [Indexed: 08/04/2024]
Abstract
Pressure injuries (PIs) are a common healthcare problem worldwide and are considered to be the most expensive chronic wounds after arterial ulcers. Although the gross factors including ischemia-reperfusion (I/R) have been identified in the etiology of PIs, the precise cellular and molecular mechanisms contributing to PIs development remain unclear. Various forms of programmed cell death including apoptosis, autophagy, pyroptosis, necroptosis and ferroptosis have been identified in PIs. In this paper, we present a detailed overview on various forms of cell death; discuss the recent advances in the roles of cell death in the occurrence and development of PIs and found much of the evidence is novel and based on animal experiments. Herein, we also state critical evaluation of the existing data and future perspective in the field. A better understanding of the programmed cell death mechanism in PIs may have important implications in driving the development of new preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Juhong Pei
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yuting Wei
- School of Nursing, Lanzhou University, Lanzhou, Gansu, China
| | - Lin Lv
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Hongxia Tao
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - HongYan Zhang
- Department of Nursing, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - YuXia Ma
- School of Nursing, Lanzhou University, Lanzhou, Gansu, China
| | - Lin Han
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China; School of Nursing, Lanzhou University, Lanzhou, Gansu, China; Department of Nursing, Gansu Provincial Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
4
|
Mahmoud SA, Elkhoely A, El-Sayed EK, Ahmed AAE. Enhanced upregulation of SIRT1 via pioglitazone and ligustrazine confers protection against ethanol-induced gastric ulcer in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6177-6195. [PMID: 38441571 PMCID: PMC11329587 DOI: 10.1007/s00210-024-03026-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/23/2024] [Indexed: 08/18/2024]
Abstract
Gastric ulcer is a disturbing disease that impacts many people worldwide. Pioglitazone (Piog), a thiazolidinedione, and ligustrazine (Ligu), a natural component of Ligusticum chuanxiong possess gastroprotective properties. However, the underlying mechanism is not well elucidated. The present study aimed to investigate the gastroprotective effects of Piog (15 mg/kg, p.o.), Ligu (15 mg/kg, p.o.), and their combination against ethanol-induced gastric ulcer in rats. Omeprazole (10 mg/kg) was used as a standard. Pre-treatment for 7 days with Piog, Ligu, and (Piog+Ligu) effectively alleviated ethanol-predisposed oxidative stress and inflammation through restoring HO-1, GSH, and SOD tissue levels and decreasing elevated MDA, TNF-α, ICAM, I-NOS, and IL-1β contents. Moreover, Piog, Ligu, and (Piog+Ligu) markedly inhibited the ethanol-induced increase of gastric NF-KB and BAX. In contrast, this pre-treatment regimen significantly accelerated protein expression of SIRT1, Nrf2, and Bcl-2, along with autophagic proteins, ATG5 and Beclin. Interestingly, macroscopic, histopathological examination and mucin content were in harmony with previous results, where pre-treatment with Piog, Ligu, and (Piog+Ligu) showed a declined mucosal injury as evidenced by the remarkable decrease of the ulcer area percentage by 62.3%, 38.7%, and 91.2%, respectively, compared to the ethanol-ulcerated group. In conclusion, Piog and Ligu exhibited remarkable gastroprotective properties. Our study was the first to show that Piog, Ligu, and (Piog+Ligu) ameliorated oxidative stress, inflammation, and apoptosis and accelerated the autophagic process via the upregulation of the upstream SIRT1 protein. It is worth mentioning that future studies are needed to pave the way for the clinical use of Piog and Ligu as gastro-protective agents.
Collapse
Affiliation(s)
- Sara A Mahmoud
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, Egypt
| | - Abeer Elkhoely
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, Egypt.
| | - Elsayed K El-Sayed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, Egypt
| | - Amany A E Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, Egypt
| |
Collapse
|
5
|
Gong Y, Qiu B, Zheng H, Li X, Wang Y, Wu M, Yan M, Gong Y. Unacylated ghrelin attenuates acute liver injury and hyperlipidemia via its anti-inflammatory and anti-oxidative activities. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:49-56. [PMID: 38164484 PMCID: PMC10722475 DOI: 10.22038/ijbms.2023.70831.15388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 08/16/2023] [Indexed: 01/03/2024]
Abstract
Objectives Liver injury and hyperlipidemia are major issues that have drawn more and more attention in recent years. The present study aimed to investigate the effects of unacylated ghrelin (UAG) on acute liver injury and hyperlipidemia in mice. Materials and Methods UAG was injected intraperitoneally once a day for three days. Three hours after the last administration, acute liver injury was induced by intraperitoneal injection of carbon tetrachloride (CCl4), and acute hyperlipidemia was induced by intraperitoneal injection of poloxamer 407, respectively. Twenty-four hours later, samples were collected for serum biochemistry analysis, histopathological examination, and Western blotting. Results In acute liver injury mice, UAG significantly decreased liver index, serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), reduced malondialdehyde (MDA) concentration and increased superoxide dismutase(SOD) in liver tissue. NF-kappa B (NF-κB) protein expression in the liver was down-regulated. In acute hyperlipidemia mice, UAG significantly decreased serum total cholesterol (TC), triglyceride (TG), ALT, and AST, as well as hepatic TG levels. Meanwhile, hepatic MDA decreased and SOD increased significantly. Moreover, UAG improved the pathological damage in the liver induced by CCl4 and poloxamer 407, respectively. Conclusion Intraperitoneal injection of UAG exhibited hepatoprotective and lipid-lowering effects on acute liver injury and hyperlipidemia, which is attributed to its anti-inflammatory and anti-oxidant activities.
Collapse
Affiliation(s)
- Yating Gong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Beibei Qiu
- Department of Pathology, Feicheng Hospital Affiliated to Shandong First Medical University, Qingdao, China
| | - Haotian Zheng
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Xiangbo Li
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Yifan Wang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Mengran Wu
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Meixing Yan
- Department of Pharmacy, Qingdao Women and Children’s Hospital, Qingdao, China
| | - Yanling Gong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| |
Collapse
|
6
|
Zhang X, Zeng Z, Liu Y, Liu D. Emerging Relevance of Ghrelin in Programmed Cell Death and Its Application in Diseases. Int J Mol Sci 2023; 24:17254. [PMID: 38139082 PMCID: PMC10743592 DOI: 10.3390/ijms242417254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Ghrelin, comprising 28 amino acids, was initially discovered as a hormone that promotes growth hormones. The original focus was on the effects of ghrelin on controlling hunger and satiation. As the research further develops, the research scope of ghrelin has expanded to a wide range of systems and diseases. Nevertheless, the specific mechanisms remain incompletely understood. In recent years, substantial studies have demonstrated that ghrelin has anti-inflammatory, antioxidant, antiapoptotic, and other effects, which could affect the signaling pathways of various kinds of programmed cell death (PCD) in treating diseases. However, the regulatory mechanisms underlying the function of ghrelin in different kinds of PCD have not been thoroughly illuminated. This review describes the relationship between ghrelin and four kinds of PCD (apoptosis, necroptosis, autophagy, and pyroptosis) and then introduces the clinical applications based on the different features of ghrelin.
Collapse
Affiliation(s)
- Xue Zhang
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.Z.); (Z.Z.); (Y.L.)
| | - Zihan Zeng
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.Z.); (Z.Z.); (Y.L.)
| | - Yaning Liu
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.Z.); (Z.Z.); (Y.L.)
| | - Dan Liu
- School of Pharmacy, Nanchang University, Nanchang 330006, China
| |
Collapse
|
7
|
Tuero C, Becerril S, Ezquerro S, Neira G, Frühbeck G, Rodríguez A. Molecular and cellular mechanisms underlying the hepatoprotective role of ghrelin against NAFLD progression. J Physiol Biochem 2023; 79:833-849. [PMID: 36417140 DOI: 10.1007/s13105-022-00933-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/12/2022] [Indexed: 11/24/2022]
Abstract
The underlying mechanisms for the development and progression of nonalcoholic fatty liver disease (NAFLD) are complex and multifactorial. Within the last years, experimental and clinical evidences support the role of ghrelin in the development of NAFLD. Ghrelin is a gut hormone that plays a major role in the short-term regulation of appetite and long-term regulation of adiposity. The liver constitutes a target for ghrelin, where this gut-derived peptide triggers intracellular pathways regulating lipid metabolism, inflammation, and fibrosis. Interestingly, circulating ghrelin levels are altered in patients with metabolic diseases, such as obesity, type 2 diabetes, and metabolic syndrome, which, in turn, are well-known risk factors for the pathogenesis of NAFLD. This review summarizes the molecular and cellular mechanisms involved in the hepatoprotective action of ghrelin, including the reduction of hepatocyte lipotoxicity via autophagy and fatty acid β-oxidation, mitochondrial dysfunction, endoplasmic reticulum stress and programmed cell death, the reversibility of the proinflammatory phenotype in Kupffer cells, and the inactivation of hepatic stellate cells. Together, the metabolic and inflammatory pathways regulated by ghrelin in the liver support its potential as a therapeutic target to prevent NAFLD in patients with metabolic disorders.
Collapse
Affiliation(s)
- Carlota Tuero
- Department of General Surgery, Clínica Universidad de Navarra, School of Medicine, University of Navarra, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Silvia Ezquerro
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
| | - Gabriela Neira
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain.
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
8
|
Han L, Pei J, Tao H, Guo X, Wei Y, Yang Z, Zhang H. The potential role of ferroptosis in the physiopathology of deep tissue injuries. Int Wound J 2023; 21:e14466. [PMID: 37905685 PMCID: PMC10828531 DOI: 10.1111/iwj.14466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/15/2023] [Indexed: 11/02/2023] Open
Abstract
Deep tissue injuries (DTIs) are a serious type of pressure injuries that mainly occur at the bony prominences and can develop rapidly, making prevention and treatment more difficult. Although consistent research efforts have been made over the years, the cellular and molecular mechanisms contributing to the development of DTIs remain unclear. More recently, ferroptosis, a novel regulatory cell death (RCD) type, has been identified that is morphological, biochemical and genetic criteria distinct from apoptosis, autophagy and other known cell death pathways. Ferroptosis is characterized by iron overload, iron-dependent lipid peroxidation and shrunken mitochondria. We also note that some of the pathological features of DTI are known to be key features of the ferroptosis pathway. Numerous studies have confirmed that ferroptosis may be involved in chronic wounds, including DTIs. Here, we elaborate on the basic pathological features of ferroptosis. We also present the evidence that ferroptosis is involved in the pathology of DTIs and highlight a future perspective on this emerging field, desiring to provide more possibilities for the prevention and treatment of DTIs.
Collapse
Affiliation(s)
- Lin Han
- Department of NursingGansu Provincial HospitalLanzhouChina
- School of NursingLanzhou UniversityLanzhouChina
| | - Juhong Pei
- The First Clinical Medical CollegeLanzhou UniversityLanzhouChina
| | - Hongxia Tao
- The First Clinical Medical CollegeLanzhou UniversityLanzhouChina
| | | | - Yuting Wei
- School of NursingLanzhou UniversityLanzhouChina
| | - Zhuang Yang
- School of NursingLanzhou UniversityLanzhouChina
| | - Hongyan Zhang
- Department of NursingGansu Provincial HospitalLanzhouChina
| |
Collapse
|
9
|
Wei W, Li H, Deng Y, Zheng X, Zhou Y, Xue X. The combination of Alisma and Atractylodes ameliorates cerebral ischaemia/reperfusion injury by negatively regulating astrocyte-derived exosomal miR-200a-3p/141-3p by targeting SIRT1. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116597. [PMID: 37146842 DOI: 10.1016/j.jep.2023.116597] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/22/2023] [Accepted: 05/03/2023] [Indexed: 05/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The combination of Alisma and Atractylodes (AA), a classical traditional Chinese herbal decoction, may protect against cerebral ischaemia/reperfusion injury (CIRI). However, the underlying mechanism has not been characterized. Intriguingly, exosomal microRNAs (miRNAs) have been recognized as vital factors in the pharmacology of Chinese herbal decoctions. AIM OF THE STUDY The aim of the present study was to assess whether the neuroprotective effect of AA was dependent on the efficient transfer of miRNAs via exosomes in the brain. MATERIALS AND METHODS Bilateral common carotid artery ligation (BCAL) was used to induce transient global cerebral ischaemia/reperfusion (GCI/R) in C57BL/6 mice treated with/without AA. Neurological deficits were assessed with the modified neurological severity score (mNSS) and Morris water maze (MWM) test. Western blot (WB) analysis was used to detect the expression of sirtuin 1 (SIRT1) in the cerebral cortex. The inflammatory state was quantitatively evaluated by measuring the expression of phospho-Nuclear factor kappa B (p-NF-κB), Interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) using WB analysis and glial fibrillary acidic protein (GFAP) immunohistochemical staining. The protein expression of zonula occluden-1 (ZO-1), occludin, caudin-5 and CD31 was examined by immunohistochemical staining to determine blood‒brain barrier (BBB) permeability. Exosomes were extracted from the brain interstitial space by ultracentrifugation and identified by transmission electron microscopy (TEM), WB analysis and nanoparticle tracking analysis (NTA). The origin of exosomes was clarified by measuring the specific mRNAs within exosomes via Real Time Quantitative PCR (RT‒qPCR). Differential miRNAs in exosomes were identified using microarray screening and were validated by RT‒qPCR. Exosomes were labelled with fluorescent dye (PKH26) and incubated with bEnd.3 cells, the supernatant was collected, IL-1β/TNF-α expression was measured using enzyme-linked immunosorbent assay (ELISA), total RNA was extracted, and miR-200a-3p/141-3p expression was examined by RT‒qPCR. In addition, the levels of miR-200a-3p/141-3p in oxygen glucose deprivation/reoxygenation (OGD/R)-induced bEnd.3 cells were quantified. The direct interaction between miR-200a-3p/141-3p and the SIRT1 3' untranslated region (3'UTR) was measured by determining SIRT1 expression in bEnd.3 cells transfected with the miR-200a-3p/141-3p mimic/inhibitor. RESULTS Severe neurological deficits and memory loss caused by GCI/R in mice was markedly ameliorated by AA treatment, particularly in the AA medium-dose group. Moreover, AA-treated GCI/R-induced mice showed significant increases in SIRT1, ZO-1, occludin, caudin-5, and CD31 expression levels and decreases in p-NF-κB, IL-1β, TNF-α, and GFAP expression levels compared with those in untreated GCI/R-induced mice. Furthermore, we found that miR-200a-3p/141-3p was enriched in astrocyte-derived exosomes from GCI/R-induced mice and could be inhibited by treatment with a medium dose of AA. The exosomes mediated the transfer of miR-200a-3p/141-3p into bEnd.3 cells, promoted IL-1β and TNF-α release and downregulated the expression of SIRT1. No significant changes in the levels of miR-200a-3p/141-3p were observed in OGD/R-induced bEnd.3 cells. The miR-200a-3p/141-3p mimic/inhibitor decreased/increased SIRT1 expression in bEnd.3 cells, respectively. CONCLUSION Our findings demonstrated that AA attenuated inflammation-mediated CIRI by inhibiting astrocyte-derived exosomal miR-200a-3p/141-3p by targeting the SIRT1 gene, which provided further evidence and identified a novel regulatory mechanism for the neuroprotective effects of AA.
Collapse
Affiliation(s)
- Wei Wei
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Huihong Li
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - YunFei Deng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - XiaoQing Zheng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yangjie Zhou
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.
| | - Xiehua Xue
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China; Fujian Key Laboratory of Rehabilitation Technology, Fujian Key Laboratory of Cognitive Rehabilitation, Fuzhou, China.
| |
Collapse
|
10
|
The Effect of Ghrelin on Apoptosis, Necroptosis and Autophagy Programmed Cell Death Pathways in the Hippocampal Neurons of Amyloid-β 1–42-Induced Rat Model of Alzheimer’s Disease. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10457-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
11
|
Chen Y, Sun Y, Luo Z, Lin J, Qi B, Kang X, Ying C, Guo C, Yao M, Chen X, Wang Y, Wang Q, Chen J, Chen S. Potential Mechanism Underlying Exercise Upregulated Circulating Blood Exosome miR-215-5p to Prevent Necroptosis of Neuronal Cells and a Model for Early Diagnosis of Alzheimer's Disease. Front Aging Neurosci 2022; 14:860364. [PMID: 35615585 PMCID: PMC9126031 DOI: 10.3389/fnagi.2022.860364] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/21/2022] [Indexed: 02/05/2023] Open
Abstract
Exercise is crucial for preventing Alzheimer's disease (AD), although the exact underlying mechanism remains unclear. The construction of an accurate AD risk prediction model is beneficial as it can provide a theoretical basis for preventive exercise prescription. In recent years, necroptosis has been confirmed as an important manifestation of AD, and exercise is known to inhibit necroptosis of neuronal cells. In this study, we extracted 67 necroptosis-related genes and 32 necroptosis-related lncRNAs and screened for key predictive AD risk genes through a random forest analysis. Based on the neural network Prediction model, we constructed a new logistic regression-based AD risk prediction model in order to provide a visual basis for the formulation of exercise prescription. The prediction model had an area under the curve (AUC) value of 0.979, indicative of strong predictive power and a robust clinical application prospect. In the exercise group, the expression of exosomal miR-215-5p was found to be upregulated; miR-215-5p could potentially inhibit the expressions of IDH1, BCL2L11, and SIRT1. The single-cell SCENIC assay was used to identify key transcriptional regulators in skeletal muscle. Among them, CEBPB and GATA6 were identified as putative transcriptional regulators of miR-215. After "skeletal muscle removal of load," the expressions of CEBPB and GATA6 increased substantially, which in turn led to the elevation of miR-215 expression, thereby suggesting a putative mechanism for negative feedback regulation of exosomal homeostasis.
Collapse
Affiliation(s)
- Yisheng Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinrong Lin
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Beijie Qi
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xueran Kang
- Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chenting Ying
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenyang Guo
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mengxuan Yao
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
| | | | - Yi Wang
- Huashan Hospital, Fudan University, Shanghai, China
| | - Qian Wang
- Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an, China
| | - Jiwu Chen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Xin Y, Zhang Y, Deng S, Hu X. Vagus Nerve Stimulation Attenuates Acute Skeletal Muscle Injury Induced by Hepatic Ischemia/Reperfusion Injury in Rats. Front Pharmacol 2022; 12:756997. [PMID: 35046803 PMCID: PMC8762262 DOI: 10.3389/fphar.2021.756997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Vagus nerve stimulation (VNS) has a protective effect on distal organ injury after ischemia/reperfusion (I/R) injury. We aimed to investigate the protective efficacy of VNS on hepatic I/R injury-induced acute skeletal muscle injury and explore its underlying mechanisms. To test this hypothesis, male Sprague-Dawley rats were randomly divided into three groups: sham group (sham operation, n = 6); I/R group (hepatic I/R with sham VNS, n = 6); and VNS group (hepatic I/R with VNS, n = 6). A hepatic I/R injury model was prepared by inducing hepatic ischemia for 1 h (70%) followed by hepatic reperfusion for 6 h. VNS was performed during the entire hepatic I/R process. Tissue and blood samples were collected at the end of the experiment for biochemical assays, molecular biological preparations, and histological examination. Our results showed that throughout the hepatic I/R process, VNS significantly reduced inflammation, oxidative stress, and apoptosis, while significantly increasing the protein levels of silent information regulator 1 (SIRT1) and decreasing the levels of acetylated forkhead box O1 and Ac-p53, in the skeletal muscle. These data suggest that VNS can alleviate hepatic I/R injury-induced acute skeletal muscle injury by suppressing inflammation, oxidative stress, and apoptosis, potentially via the SIRT1 pathway.
Collapse
Affiliation(s)
- Ying Xin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yifeng Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Simin Deng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xinqun Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
13
|
Zhang W, Miao R, Tang J, Su Q, Li P, Pi H. Low temperature exerts protective effects by inhibiting mitochondria-mediated apoptosis pathway following pressure injury to rat muscle. Rev Esc Enferm USP 2021; 55:e20200319. [PMID: 34528993 DOI: 10.1590/1980-220x-reeusp-2020-0319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 01/08/2021] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE We aimed to determine the effect of different low-temperature range interventions at different time-points in a rat model of pressure injury (PI) produced by Ischemia/Reperfusion (I/R) injury. METHODS Sprague-Dawley rats were randomly assigned to blank control, injury control, and temperature intervention groups. Rats in the injury control and temperature intervention groups (involving exposure to different temperature range at different time-points) were subjected to three cycles of I/R injury with 2-h ischemia and 0.5-h reperfusion to induce PI. RESULTS The muscle tissues exhibited degenerative changes after compression. Low temperature intervention of 16-18°C in the ischemia period resulted in the lowest degree of tissue damage and significantly decreased levels of Bcl-2-associated X protein (Bax), caspase-9, and caspase-3. Moreover, it resulted in the highest expression level of B-cell lymphoma 2 (Bcl-2) and lowest expression levels of Bax, caspase-9, and caspase-3 in muscle tissues among all intervention groups. CONCLUSION Low-temperature intervention at 16-18°C during the ischemia period showed optimal effects on the expressions of apoptotic factors during the development of PI with I/R-induced tissue damage.
Collapse
Affiliation(s)
- Wenyu Zhang
- Capital Medical University School of Nursing, Beijing, China.,Medical School of Chinese PLA, Beijing, China.,College of Social Administration, Department of Senior Citizens Welfare, Beijing, China
| | - Ran Miao
- Capital Medical University, Beijing Chao-Yang Hospital, Medical Research Center, Beijing, 100853, China
| | | | - Qingqing Su
- Medical School of Chinese PLA, Beijing, China
| | - Peifeng Li
- Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Hongying Pi
- Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
14
|
Zenitani M, Hosoda H, Nose S, Kangawa K, Kawahara H, Oue T. Importance of plasma ghrelin levels with special reference to nutritional metabolism and energy expenditure in pediatric patients with severe motor and intellectual disabilities. Clin Nutr ESPEN 2021; 42:180-187. [PMID: 33745575 DOI: 10.1016/j.clnesp.2021.01.043] [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: 12/18/2020] [Revised: 01/23/2021] [Accepted: 01/29/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND & AIMS Nutritional metabolism is complex in pediatric patients with severe motor and intellectual disability (SMID), and therefore, appropriate estimation of the energy requirements is difficult. Focusing on ghrelin's role in energy metabolism regulation, we investigated plasma ghrelin levels in pediatric SMID patients and analyzed its nutritional significance as a regulatory marker of energy reserve. METHODS Fasting plasma total, acyl, and des-acyl ghrelin levels in 40 patients with SMID, including cerebral palsy (CP) (n = 20) and muscular disease (MD) (n = 8), and healthy controls (n = 13) were investigated. The correlations of plasma ghrelin levels with anthropometry, blood nutritional markers, energy intake, and resting energy expenditure (REE) measured with indirect calorimetry were analyzed. A p value < 0.05 was considered significant. RESULTS SMID patients had significantly higher acyl ghrelin, and lower body mass index (BMI), z-scores of body weight (BW), body height and BMI, and albumin than controls. CP patients had significantly higher total and acyl ghrelin, z-score of the mid-upper arm circumference (MUAC), retinol-binding protein, transthyretin, creatinine, and glucose than MD patients. Total and acyl ghrelin in CP patients and des-acyl ghrelin in MD patients had significant negative correlations with MUAC and upper arm fat area. In CP patients, total and acyl ghrelin had significant positive correlations with REE/BW (kcal/kg), and total ghrelin was predictive of REE/BW (r2 = 0.625, p < 0.0001). CONCLUSIONS An increase in acyl ghrelin observed in SMID patients possibly indicates energy reserve deficiency. In CP patients, total and acyl ghrelin inversely reflected total body fat mass, resulting in strongly positive correlations with REE/BW. The measurement of plasma ghrelin may be useful to assess nutritional metabolism and energy reserve in pediatric SMID patients, such as CP and MD patients.
Collapse
Affiliation(s)
- Masahiro Zenitani
- Department of Pediatric Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan.
| | - Hiroshi Hosoda
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.
| | - Satoko Nose
- Department of Pediatric Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan.
| | - Kenji Kangawa
- National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.
| | - Hisayoshi Kawahara
- Department of Pediatric Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan.
| | - Takaharu Oue
- Department of Pediatric Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan.
| |
Collapse
|
15
|
Wang H, Dou S, Zhu J, Shao Z, Wang C, Cheng B. Regulatory effects of ghrelin on endoplasmic reticulum stress, oxidative stress, and autophagy: Therapeutic potential. Neuropeptides 2021; 85:102112. [PMID: 33333485 DOI: 10.1016/j.npep.2020.102112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/29/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Ghrelin is a regulatory peptide that is the endogenous ligand of the growth hormone secretagogue 1a (GHS-R1a) which belongs to the G protein-coupled receptor family. Ghrelin and GHS-R1a are widely expressed in the central and peripheral tissues and play therapeutic potential roles in the cytoprotection of many internal organs. Endoplasmic reticulum stress (ERS), oxidative stress, and autophagy dysfunction, which are involved in various diseases. In recent years, accumulating evidence has suggested that ghrelin exerts protective effects by regulating ERS, oxidative stress, and autophagy in diverse diseases. This review article summarizes information about the roles of the ghrelin system on ERS, oxidative stress, and autophagy in multiple diseases. It is suggested that ghrelin positively affects the treatment of diseases and may be considered as a therapeutic drug in many illnesses.
Collapse
Affiliation(s)
- Huiqing Wang
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Shanshan Dou
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Junge Zhu
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Ziqi Shao
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Chunmei Wang
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Baohua Cheng
- Neurobiology Institute, Jining Medical University, 272067 Jining, China.
| |
Collapse
|
16
|
Broussy S, Laaroussi H, Vidal M. Biochemical mechanism and biological effects of the inhibition of silent information regulator 1 (SIRT1) by EX-527 (SEN0014196 or selisistat). J Enzyme Inhib Med Chem 2021; 35:1124-1136. [PMID: 32366137 PMCID: PMC7241506 DOI: 10.1080/14756366.2020.1758691] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The human sirtuin silent information regulator 1 (SIRT1) is a NAD+-dependent deacetylase enzyme. It deacetylates many protein substrates, including histones and transcription factors, thereby controlling many physiological and pathological processes. Several synthetic inhibitors and activators of SIRT1 have been developed, and some therapeutic applications have been explored. The indole EX-527 and its derivatives are among the most potent and selective SIRT1 inhibitors. EX-527 has been often used as a pharmacological tool to explore the effect of SIRT1 inhibition in various cell types. Its therapeutic potential has, therefore, been evaluated in animal models for several pathologies, including cancer. It has also been tested in phase II clinical trial for the treatment of Huntington’s disease (HD). In this review, we will provide an overview of the literature on EX-527, including its mechanism of inhibition and biological studies.
Collapse
Affiliation(s)
- Sylvain Broussy
- Université de Paris, Faculté de Pharmacie de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, Paris, France
| | - Hanna Laaroussi
- Université de Paris, Faculté de Pharmacie de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, Paris, France
| | - Michel Vidal
- Université de Paris, Faculté de Pharmacie de Paris, CiTCoM, 8038 CNRS, U 1268 INSERM, Paris, France.,Service biologie du médicament, toxicologie, AP-HP, Hôpital Cochin, Paris, France
| |
Collapse
|
17
|
Nikseresht S, Khodagholi F, Ahmadiani A. Protective effects of ex-527 on cerebral ischemia-reperfusion injury through necroptosis signaling pathway attenuation. J Cell Physiol 2019; 234:1816-1826. [PMID: 30067864 DOI: 10.1002/jcp.27055] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/25/2018] [Indexed: 12/16/2022]
Abstract
Necroptosis, a novel type of programmed cell death, is involved in ischemia-reperfusion-induced brain injury. Sirtuin 1 (Sirt1), as a well-known member of histone deacetylase class III, plays pivotal roles in inflammation, metabolism, and neuron loss in cerebral ischemia. We explored the relationship between Sirt1 and the necroptosis signaling pathway and its downstream events by administration of ex-527, as a selective and potent inhibitor of Sirt1, and necrostatin-1 (nec-1), as a necroptosis inhibitor, in an animal model of focal cerebral ischemia. Our data showed different patterns of sirt1 and necroptosis critical regulators, including receptor-interacting protein kinase 3 and mixed lineage kinase domain-like protein gene expressions in the prefrontal cortex and the hippocampus after ischemia-reperfusion. We found that ex-527 microinjection reduces the infarction volume of ischemic brains and improves the survival rate, but not stroke-associated neurological deficits. Additionally, treatment with ex-527 effectively abolished the elevation of the critical regulators of necroptosis, whereas necroptosis inhibition through nec-1 microinjection did not influence Sirt1 expression levels. Our data also demonstrated that the ex-527 relieves ischemia-induced perturbation of necroptosis-associated metabolic enzymes activity in downstream. This study provides a new approach to the possible neuroprotective potential of ex-527 orchestrated by necroptosis pathway inhibition to alleviate ischemia-reperfusion brain injury.
Collapse
Affiliation(s)
- Sara Nikseresht
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|