1
|
Yang XR, Wen R, Yang N, Zhang TN. Role of sirtuins in sepsis and sepsis-induced organ dysfunction: A review. Int J Biol Macromol 2024; 278:134853. [PMID: 39163955 DOI: 10.1016/j.ijbiomac.2024.134853] [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: 04/29/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 08/22/2024]
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis causes a high mortality rate and current treatment focuses on supportive therapies but lacks specific therapeutic targets. Notably, sirtuins (SIRTs) shows potential clinical application in the treatment of sepsis. It has been demonstrated that SIRTs, the nicotinamide adenine dinucleotide+(NAD+)-dependent deacetylases that regulate key signaling pathways in eukaryotes and prokaryotes, are involved in a variety of biological processes. To date, seven mammalian yeast Sir2 homologs have been identified. SIRTs can regulate inflammation, oxidative stress, apoptosis, autophagy, and other pathways that play important roles in sepsis-induced organ dysfunction. However, the existing studies on SIRTs in sepsis are too scattered, and there is no relevant literature to integrate them. This review innovatively summarizes the different mechanisms of SIRTs in sepsis organ dysfunction according to the different systems, and focuses on SIRT agonists, inhibitors, and targeted drugs that have been proved to be effective in the treatment of sepsis, so as to integrate the clinical research and basic research closely. We searched PubMed for all literature related to SIRTs and sepsis since its inception using the following medical subject headings: sirtuins, SIRTs, and sepsis. Data on the mechanisms of SIRTs in sepsis-induced organ damage and their potential as targets for disease treatment were extracted.
Collapse
Affiliation(s)
- Xin-Ru Yang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ri Wen
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ni Yang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Tie-Ning Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
2
|
Zhang YM, Wei RM, Zhang JY, Liu S, Zhang KX, Kong XY, Ge YJ, Li XY, Chen GH. Resveratrol prevents cognitive deficits induced by sleep deprivation via modulating sirtuin 1 associated pathways in the hippocampus. J Biochem Mol Toxicol 2024; 38:e23698. [PMID: 38501767 DOI: 10.1002/jbt.23698] [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: 04/24/2023] [Revised: 02/28/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Accumulating evidence confirms that sleep insufficiency is a high risk factor for cognitive impairment, which involves inflammation and synaptic dysfunction. Resveratrol, an agonist of the Sirt1, has demonstrated anti-inflammation and neuroprotective effects in models of Alzheimer's disease, Parkinson's disease, and schizophrenia. However, the beneficial effects of resveratrol on sleep deprivation-induced cognitive deficits and its underlying molecular mechanisms are unclear. In the present study, thirty-two male C57BL/6 J mice were randomly divided into a Control+DMSO group, Control+Resveratrol group, SD+DMSO group, and SD+Resveratrol group. The mice in the SD+Resveratrol group underwent 5 days of sleep deprivation after pretreatment with resveratrol (50 mg/kg) for 2 weeks, while the mice in the SD+DMSO group only underwent sleep deprivation. After sleep deprivation, we evaluated spatial learning and memory function using the Morris water maze test. We used general molecular biology techniques to detect changes in levels of pro-inflammatory cytokines and Sirt1/miR-134 pathway-related synaptic plasticity proteins. We found that resveratrol significantly reversed sleep deprivation-induced learning and memory impairment, elevated interleukin-1β, interleukin-6, and tumor necrosis factor-α levels, and decreased brain-derived neurotrophic factor, tyrosine kinase receptor B, postsynaptic density protein-95, and synaptophysin levels by activating the Sirt1/miR-134 pathway. In conclusion, resveratrol is a promising agent for preventing sleep deprivation-induced cognitive dysfunction by reducing pro-inflammatory cytokines and improving synaptic function via the Sirt1/miR-134 pathway.
Collapse
Affiliation(s)
- Yue-Ming Zhang
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, PR China
| | - Ru-Meng Wei
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, PR China
| | - Jing-Ya Zhang
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, PR China
| | - Shuang Liu
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, PR China
| | - Kai-Xuan Zhang
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, PR China
| | - Xiao-Yi Kong
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, PR China
| | - Yi-Jun Ge
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, PR China
| | - Xue-Yan Li
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, PR China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, PR China
| |
Collapse
|
3
|
Shehata AH, Anter AF, Ahmed ASF. Role of SIRT1 in sepsis-induced encephalopathy: Molecular targets for future therapies. Eur J Neurosci 2023; 58:4211-4235. [PMID: 37840012 DOI: 10.1111/ejn.16167] [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: 08/12/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/17/2023]
Abstract
Sepsis induces neuroinflammation, BBB disruption, cerebral hypoxia, neuronal mitochondrial dysfunction, and cell death causing sepsis-associated encephalopathy (SAE). These pathological consequences lead to short- and long-term neurobehavioural deficits. Till now there is no specific treatment that directly improves SAE and its associated behavioural impairments. In this review, we discuss the underlying mechanisms of sepsis-induced brain injury with a focus on the latest progress regarding neuroprotective effects of SIRT1 (silent mating type information regulation-2 homologue-1). SIRT1 is an NAD+ -dependent class III protein deacetylase. It is able to modulate multiple downstream signals (including NF-κB, HMGB, AMPK, PGC1α and FoxO), which are involved in the development of SAE by its deacetylation activity. There are multiple recent studies showing the neuroprotective effects of SIRT1 in neuroinflammation related diseases. The proposed neuroprotective action of SIRT1 is meant to bring a promising therapeutic strategy for managing SAE and ameliorating its related behavioural deficits.
Collapse
Affiliation(s)
- Alaa H Shehata
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Aliaa F Anter
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Al-Shaimaa F Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
4
|
Liu Y, Yang H, Luo N, Fu Y, Qiu F, Pan Z, Li X, Jian W, Yang X, Xue Q, Luo Y, Yu B, Liu Z. An Fgr kinase inhibitor attenuates sepsis-associated encephalopathy by ameliorating mitochondrial dysfunction, oxidative stress, and neuroinflammation via the SIRT1/PGC-1α signaling pathway. J Transl Med 2023; 21:486. [PMID: 37475042 PMCID: PMC10360347 DOI: 10.1186/s12967-023-04345-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/11/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) is characterized by diffuse brain dysfunction, long-term cognitive impairment, and increased morbidity and mortality. The current treatment for SAE is mainly symptomatic; the lack of specific treatment options and a poor understanding of the underlying mechanism of disease are responsible for poor patient outcomes. Fgr is a member of the Src family of tyrosine kinases and is involved in the innate immune response, hematologic cancer, diet-induced obesity, and hemorrhage-induced thalamic pain. This study investigated the protection provided by an Fgr kinase inhibitor in SAE and the underlying mechanism(s) of action. METHODS A cecal ligation and puncture (CLP)-induced mouse sepsis model was established. Mice were treated with or without an Fgr inhibitor and a PGC-1α inhibitor/activator. An open field test, a novel object recognition test, and an elevated plus maze were used to assess neurobehavioral changes in the mice. Western blotting and immunofluorescence were used to measure protein expression, and mRNA levels were measured using quantitative PCR (qPCR). An enzyme-linked immunosorbent assay was performed to quantify inflammatory cytokines. Mitochondrial membrane potential and morphology were measured by JC-1, electron microscopy, and the MitoTracker Deep Red probe. Oxidative stress and mitochondrial dysfunction were analyzed. In addition, the regulatory effect of Fgr on sirtuin 1 (SIRT1) was assessed. RESULTS CLP-induced sepsis increased the expression of Fgr in the hippocampal neurons. Pharmacological inhibition of Fgr attenuated CLP-induced neuroinflammation, the survival rate, cognitive and emotional dysfunction, oxidative stress, and mitochondrial dysfunction. Moreover, Fgr interacted with SIRT1 and reduced its activity and expression. In addition, activation of SIRT1/PGC-1α promoted the protective effects of the Fgr inhibitor on CLP-induced brain dysfunction, while inactivation of SIRT1/PGC-1α counteracted the benefits of the Fgr inhibitor. CONCLUSIONS To our knowledge, this is the first report of Fgr kinase inhibition markedly ameliorating SAE through activation of the SIRT1/PGC-1α pathway, and this may be a promising therapeutic target for SAE.
Collapse
Affiliation(s)
- Yuqiang Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China.
| | - Han Yang
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Nanbo Luo
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yifei Fu
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Fang Qiu
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Zhenglong Pan
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiongjuan Li
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Wenling Jian
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xinping Yang
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qingsheng Xue
- Department of Anesthesiology, Ruijin Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Buwei Yu
- Department of Anesthesiology, Ruijin Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Zhiheng Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China.
| |
Collapse
|
5
|
Cao E, Xu J, Gong Y, Yuan J, Chen A, Liu J, Fan Y, Fan X, Kuang X. Effect of the Lipoxin Receptor Agonist BML-111 on Cigarette Smoke Extract-Induced Macrophage Polarization and Inflammation in RAW264.7 Cells. Int J Chron Obstruct Pulmon Dis 2023; 18:919-932. [PMID: 37229441 PMCID: PMC10204758 DOI: 10.2147/copd.s395569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 04/29/2023] [Indexed: 05/27/2023] Open
Abstract
Background Macrophages are known to play a crucial role in the chronic inflammation associated with Chronic Obstructive Pulmonary Disease (COPD). BML-111, acting as a lipoxin A4 (LXA4) receptor agonist, has shown to be effective in protecting against COPD. However, the precise mechanism by which BML-111 exerts its protective effect remains unclear. Methods In order to establish a cell model of inflammation, cigarette smoke extract (CSE) was used on the RAW264.7 cell line. Afterwards, an Enzyme-linked immunosorbent assay (ELISA) kit was employed to measure concentrations of tumor necrosis factor-α (TNF-α), interleukin-1beta (IL-1β), interleukin-18 (IL-18), and interleukin-10 (IL-10) in the cell supernatants of the RAW264.7 cells.In this study, we examined the markers of macrophage polarization using two methods: quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis. Additionally, we detected the expression of Notch-1 and Hes-1 through Western blotting. Results BML-111 effectively suppressed the expression of pro-inflammatory cytokines TNF-α, IL-1β, and IL-18, as well as inflammasome factors NLRP3 and Caspase-1, while simultaneously up-regulating the expression of the anti-inflammatory cytokine IL-10 induced by CSE. Moreover, BML-111 reduced the expression of iNOS, which is associated with M1 macrophage polarization, and increased the expression of Arg-1, which is associated with M2 phenotype. Additionally, BML-111 downregulated the expression of Hes-1 and the ratio of activated Notch-1 to Notch-1 induced by CSE. The effect of BML-111 on inflammation and macrophage polarization was reversed upon administration of the Notch-1 signaling pathway agonist Jagged1. Conclusion BML-111 has the potential to suppress inflammation and modulate M1/M2 macrophage polarization in RAW264.7 cells. The underlying mechanism may involve the Notch-1 signaling pathway.
Collapse
Affiliation(s)
- En Cao
- Department of Pathology, Basic Medical College of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Jun Xu
- Department of Pathology, Basic Medical College of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Yuanqi Gong
- Department of Critical Care Medicine/ICU (Intensive Care Unit), Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Jingjing Yuan
- Department of Physiology, School of Basic Medicine, Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Anbang Chen
- Department of Pathology, Basic Medical College of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Jiayi Liu
- The Basic Medical School of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Yunfei Fan
- The Basic Medical School of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Xiangyang Fan
- The Basic Medical School of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Xiaodong Kuang
- Department of Pathology, Basic Medical College of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| |
Collapse
|
6
|
Zhao YS, Li JY, Li ZC, Wang LL, Gan CL, Chen J, Jiang SY, Aschner M, Ou SY, Jiang YM. Sodium Para-aminosalicylic Acid Inhibits Lead-Induced Neuroinflammation in Brain Cortex of Rats by Modulating SIRT1/HMGB1/NF-κB Pathway. Neurochem Res 2023; 48:238-249. [PMID: 36063295 PMCID: PMC9825627 DOI: 10.1007/s11064-022-03739-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 01/11/2023]
Abstract
Lead (Pb) is considered to be a major environmental pollutant and occupational health hazard worldwide which may lead to neuroinflammation. However, an effective treatment for Pb-induced neuroinflammation remains elusive. The aim of this study was to investigate the mechanisms of Pb-induced neuroinflammation, and the therapeutic effect of sodium para-aminosalicylic acid (PAS-Na, a non-steroidal anti-inflammatory drug) in rat cerebral cortex. The results indicated that Pb exposure induced pathological damage in cerebral cortex, accompanied by increased levels of inflammatory factors tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β). Moreover, Pb decreased the expression of silencing information regulator 2 related enzyme 1 (SIRT1) and brain-derived neurotrophic factor (BDNF), and increased the levels of high mobile group box 1 (HMGB1) expression and p65 nuclear factor-κB (NF-κB) phosphorylation. PAS-Na treatment ameliorated Pb-induced histopathological changes in rat cerebral cortex. Moreover, PAS-Na reduced the Pb-induced increase of TNF-α and IL-1β levels concomitant with a significant increase in SIRT1 and BDNF levels, and a decrease in HMGB1 and the phosphorylation of p65 NF-κB expression. Thus, PAS-Na may exert anti-inflammatory effects by mediating the SIRT1/HMGB1/NF-κB pathway and BDNF expression. In conclusion, in this novel study PAS-Na was shown to possess an anti-inflammatory effect on cortical neuroinflammation, establishing its efficacy as a potential treatment for Pb exposures.
Collapse
Affiliation(s)
- Yue-Song Zhao
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Jun-Yan Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Hengyang Center for Disease Control and Prevention, Hengyang, China
| | - Zhao-Cong Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Lei-Lei Wang
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Cui-Liu Gan
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Jing Chen
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Si-Yang Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Shi-Yan Ou
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, No. 22, Shuang-yong Rd., Nanning, 530021, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China.
| |
Collapse
|
7
|
Xu QQ, Su ZR, Hu Z, Yang W, Xian YF, Lin ZX. Patchouli alcohol ameliorates the learning and memory impairments in an animal model of Alzheimer's disease via modulating SIRT1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154441. [PMID: 36108371 DOI: 10.1016/j.phymed.2022.154441] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/14/2022] [Accepted: 09/04/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases. Patchouli alcohol (PA), a major active ingredient isolated from Pogostemonis Herba, exhibits extensive bioactivity in the central nervous system (CNS) and exerts neuroprotective effects. PURPOSE This study aimed to investigate the anti-AD effects of PA in an animal model of AD and to elucidate the underlying molecular mechanisms. METHODS The gas chromatography (GC) was used to determine the ability of PA to pass the blood-brain barrier (BBB) in rats after oral administration. The sporadic AD rat model was established by intracerebroventricularly (ICV) injection with streptozotocin (STZ). PA (25 and 50 mg/kg) was given to rat orally once daily for 42 consecutive days. Morris water maze (MWM) test was performed to determine the learning and memory functions of the STZ-induced AD rats. EX527, a silent information regulator 1 (SIRT1) selective inhibitor, was used to investigate the involvement of SIRT1 in the anti-AD effects of PA in rats. RESULTS PA could penetrate the BBB. MWM test results showed that PA could significantly ameliorate the learning and memory deficits induced by STZ in rats. Meanwhile, PA enhanced the expression of SIRT1, and markedly alleviated the tau pathology by inhibiting the hyperacetylation (at the site of Lys174) and hyperphosphorylation (at the sites of Thr181, Thr205, Ser396 and Ser404) of tau protein. PA also efficiently suppressed the activation of microglia and astrocytes, and the beta-amyloid (Aβ) expression and the deacetylation of nuclear factor-kappa B (NF-κB) at Lys 310 (K310) in the STZ-treated AD rats. EX527, a SIRT1 selective inhibitor, could partially abolish the cognitive deficits improving effect of PA and inhibit the down-regulation of acetylated tau and acetylated NF-κB p65, suggesting that PA exhibited neuroprotective effects against AD via upregulating SIRT1. CONCLUSION This study reported for the first time that PA could penetrate the BBB to exert its protective effects on the brain after a single-dose oral administration. The current experimental findings also amply demonstrated that PA could improve the cognitive and memory impairments in the STZ-induced AD rat model. The underlying mechanisms involve the alleviations of neuroinflammation, tau pathology and Aβ deposition via modulating of SIRT1 and NF-κB pathways. All these findings strongly suggest that PA is a promising naturally occurring compound worthy of further development into an anti-AD pharmaceutical.
Collapse
Affiliation(s)
- Qing-Qing Xu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Zi-Ren Su
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhen Hu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Wen Yang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China; Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China; Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
| |
Collapse
|
8
|
Yan J, Tang X, Zhou ZQ, Zhang J, Zhao Y, Li S, Luo A. Sirtuins functions in central nervous system cells under neurological disorders. Front Physiol 2022; 13:886087. [PMID: 36111151 PMCID: PMC9468898 DOI: 10.3389/fphys.2022.886087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 07/25/2022] [Indexed: 11/14/2022] Open
Abstract
The sirtuins (SIRTs), a class of NAD+ -dependent deacylases, contain seven SIRT family members in mammals, from SIRT1 to SIRT7. Extensive studies have revealed that SIRT proteins regulate virous cell functions. Central nervous system (CNS) decline resulted in progressive cognitive impairment, social and physical abilities dysfunction. Therefore, it is of vital importance to have a better understanding of potential target to promote homeostasis of CNS. SIRTs have merged as the underlying regulating factors of the process of neurological disorders. In this review, we profile multiple functions of SIRT proteins in different cells during brain function and under CNS injury.
Collapse
Affiliation(s)
- Jing Yan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaole Tang
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhi-qiang Zhou
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yilin Zhao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiyong Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Shiyong Li, ; Ailin Luo,
| | - Ailin Luo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Shiyong Li, ; Ailin Luo,
| |
Collapse
|
9
|
Sepsis-Induced Brain Dysfunction: Pathogenesis, Diagnosis, and Treatment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1328729. [PMID: 36062193 PMCID: PMC9433216 DOI: 10.1155/2022/1328729] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/30/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022]
Abstract
Dysregulated host response to infection, which cause life-threatening organ dysfunction, was defined as sepsis. Sepsis can cause acute and long-term brain dysfunction, namely, sepsis-associated encephalopathy (SAE) and cognitive impairment. SAE refers to changes in consciousness without direct evidence of central nervous system infection. It is highly prevalent and may cause poor outcomes in sepsis patients. Cognitive impairment seriously affects the life quality of sepsis patients and increases the medical burden. The pathogenesis of sepsis-induced brain dysfunction is mainly characterized by the interaction of systemic inflammation, blood-brain barrier (BBB) dysfunction, neuroinflammation, microcirculation dysfunction, and brain dysfunction. Currently, the diagnosis of sepsis-induced brain dysfunction is based on clinical manifestation of altered consciousness along with neuropathological examination, and the treatment is mainly involves controlling sepsis. Although treatments for sepsis-induced brain dysfunction have been tested in animals, clinical treat sepsis-induced brain dysfunction is still difficult. Therefore, we review the underlying mechanisms of sepsis-induced brain injury, which mainly focus on the influence of systemic inflammation on BBB, neuroinflammation, brain microcirculation, and the brain function, which want to bring new mechanism-based directions for future basic and clinical research aimed at preventing or ameliorating brain dysfunction.
Collapse
|
10
|
Li Y, Ji M, Yang J. Current Understanding of Long-Term Cognitive Impairment After Sepsis. Front Immunol 2022; 13:855006. [PMID: 35603184 PMCID: PMC9120941 DOI: 10.3389/fimmu.2022.855006] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is recognized as a life-threatening multi-organ dysfunction resulting from a dysregulated host response to infection. Although the incidence and mortality of sepsis decrease significantly due to timely implementation of anti-infective and support therapies, accumulating evidence suggests that a great proportion of survivors suffer from long-term cognitive impairment after hospital discharge, leading to decreased life quality and substantial caregiving burdens for family members. Several mechanisms have been proposed for long-term cognitive impairment after sepsis, which are not mutually exclusive, including blood-brain barrier disruption, neuroinflammation, neurotransmitter dysfunction, and neuronal loss. Targeting these critical processes might be effective in preventing and treating long-term cognitive impairment. However, future in-depth studies are required to facilitate preventive and/or treatment strategies for long-term cognitive impairment after sepsis.
Collapse
Affiliation(s)
- Ying Li
- Department of Anesthesiology, Jiangyin Hospital, Affiliated to Southeast University Medical School, Jiangyin, China
| | - Muhuo Ji
- Department of Anesthesiology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Pre-treatment and continuous administration of simvastatin during sepsis improve metabolic parameters and prevent CNS injuries in survivor rats. Mol Cell Biochem 2022; 477:2657-2667. [DOI: 10.1007/s11010-022-04463-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
|
12
|
Xin YY, Wang JX, Xu AJ. Electroacupuncture ameliorates neuroinflammation in animal models. Acupunct Med 2022; 40:474-483. [PMID: 35229660 DOI: 10.1177/09645284221076515] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Neuroinflammation refers to a wide range of immune responses occurring in the brain or spinal cord. It is closely related to a variety of neurodegenerative diseases, for which it potentially represents a new direction for treatment. Electroacupuncture (EA) is one method of acupuncture treatment, which can be used as an adjuvant therapy for many diseases. This review focuses on molecular mechanisms of EA in the reduction of neuroinflammation, summarizes relevant basic research and outlines future directions for investigation. Findings: A growing body of basic research has shown that EA can ameliorate neuroinflammation centrally (in animal models of ischemic stroke, Alzheimer’s disease, traumatic brain injury, spinal cord injury, Parkinson’s disease and vascular dementia) and peripherally (e.g. after a surgical insult or injection of lipopolysaccharide) and that its effects involve different molecular mechanisms, including activation of the α7 nicotinic acetylcholine receptor signaling pathway and P2 type purinergic receptors, inhibition of nuclear factor κB, and mitigation of damage secondary to oxidative stress and NOD-like receptor protein 3 inflammasome activation. Conclusions: EA is capable of regulating multiple cell signal transduction pathways to alleviate neuroinflammation in animal models. Although the findings of animal studies are encouraging, further prospective clinical trials are needed to verify the efficacy of EA for the treatment of neuroinflammation.
Collapse
Affiliation(s)
- Yue-yang Xin
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-xu Wang
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ai-jun Xu
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
miR-197 Participates in Lipopolysaccharide-Induced Cardiomyocyte Injury by Modulating SIRT1. Cardiol Res Pract 2022; 2022:7687154. [PMID: 35223094 PMCID: PMC8872679 DOI: 10.1155/2022/7687154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/11/2022] [Indexed: 11/25/2022] Open
Abstract
Sepsis is a systemic inflammation and is capable of inducing myocarditis, which is a major leading cause of death in patients. Studies have found that miR-197 is correlated with the prognosis of patients with inflammatory heart disease, but its effect on sepsis-induced cardiomyocyte injury remains unclear. We treated H9c2 cells with lipopolysaccharide (LPS), then detected the cell viability via the cell counting kit-8 (CCK-8) assay and quantified miR-197 expression via quantitative real-time polymerase chain reaction (qRT-PCR). Then, we investigated the role of miR-197 in LPS-induced H9c2 cells by CCK-8 assay, flow cytometry, lactate dehydrogenase (LDH) measurement, enzyme-linked immunosorbent assay (ELISA), qRT-PCR, and western blot. Subsequently, silent information regulator 1 (SIRT1) was downregulated in H9c2 cells to explore its interaction with miR-197 under LPS induction. LPS induced miR-197 overexpression in H9c2 cells. LPS restrained viability, the expressions of B-cell lymphoma-2 (Bcl-2) and SIRT1, but promoted apoptosis, LDH release, and levels of interleukin-6 (IL-6), interleukin-1β (IL-1β), acetyl (AC)-p53, BCL2-associated X (Bax), and cleaved caspase-3 in H9c2 cells. miR-197 inhibition reversed the effects of LPS on H9c2 cells. The protective role of miR-197 downregulation in LPS-induced H9c2 cells was reversed by SIRT1 silencing. miR-197 contributed to LPS-induced cardiomyocyte injury by modulating SIRT1, which might be used as a molecular marker in the management of sepsis.
Collapse
|
14
|
Chuang Y, Van I, Zhao Y, Xu Y. Icariin ameliorate Alzheimer's disease by influencing SIRT1 and inhibiting Aβ cascade pathogenesis. J Chem Neuroanat 2021; 117:102014. [PMID: 34407393 DOI: 10.1016/j.jchemneu.2021.102014] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/23/2022]
Abstract
Of all types of dementia, Alzheimer's disease is the type that has the highest proportion of cases and is the cause of substantial medical and economic burden. The mechanism of Alzheimer's disease is closely associated with the aggregation of amyloid-β protein and causes neurotoxicity and extracellular accumulation in the brain and to intracellular neurofibrillary tangles caused by tau protein hyperphosphorylation in the brain tissue. Previous studies have demonstrated that sirtuin1 downregulation is involved in the pathological mechanism of Alzheimer's disease. The decrease of sirtuin1 level would cause Alzheimer's disease by means of promoting the amyloidogenic pathway to generate amyloid-β species and thereby triggering amyloid-β cascade reaction, such as tau protein hyperphosphorylation, neuron autophagy, neuroinflammation, oxidative stress, and neuron apoptosis. Currently, there is no effective treatment for Alzheimer's disease, it is necessary to develop new treatment strategies. According to the theory of traditional Chinese medicine and based on the mechanism of the disease, tonifying the kidneys is one of the principles for the treatment of Alzheimer's disease and Epimedium is a well-known Chinese medicine for tonifying kidney. Therefore, investigating the influence of the components of Epimedium on the pathological characteristics of Alzheimer's disease may provide a reference for the treatment of Alzheimer's disease in the future. In this article, we summarise the effects and mechanism of icariin, the main ingredient extracted from Epimedium, in ameliorating Alzheimer's disease by regulating sirtuin1 to inhibit amyloid-β protein and improve other amyloid-β cascade pathogenesis.
Collapse
Affiliation(s)
- Yaochen Chuang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China; Kiang Wu Nursing College of Macau, Macao, 999078, China
| | - Iatkio Van
- Kiang Wu Nursing College of Macau, Macao, 999078, China.
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China.
| |
Collapse
|
15
|
Li Z, Liu Z, Lu H, Dai W, Chen J, He L. RvD1 Attenuated Susceptibility to Ischemic AKI in Diabetes by Downregulating Nuclear Factor-κ B Signal and Inhibiting Apoptosis. Front Physiol 2021; 12:651645. [PMID: 34326777 PMCID: PMC8315138 DOI: 10.3389/fphys.2021.651645] [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: 01/10/2021] [Accepted: 04/28/2021] [Indexed: 11/19/2022] Open
Abstract
Background Acute kidney injury (AKI), when occurring in diabetic kidney disease (DKD), is known to be more severe and difficult to recover from. Inflammation and apoptosis may contribute to the heightened sensitivity of, and non-recovery from, AKI in patients with DKD. Resolvin D1 (RvD1) is a potent lipid mediator which can inhibit the inflammatory response and apoptosis in many diseases. However, it has been reported that the RvD1 levels were decreased in diabetes, which may explain why DKD is more susceptible to AKI. Methods For animal experiments, diabetic nephropathy (DN) mice were induced by streptozotocin (STZ) injection intraperitoneally. Renal ischemia–reperfusion was used to induce AKI. Blood urea nitrogen (BUN) and serum creatinine were determined using commercial kits to indicate renal function. Renal apoptosis was examined by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay. Real-time polymerase chain reaction (PCR) was used to detect the marker of inflammatory response. Western blot was used to detect the expression of nuclear factor-κB (NF-κB)-related proteins. For clinical study, 12 cases diagnosed with DKD were enrolled in this study, and an equal number of non-diabetic renal disease patients (NDKD) were recruited as a control group. The serum RvD1 in DKD or NDKD patients were detected through an ELISA kit. Results In clinical study, we found that the serum RvD1 levels were decreased in DKD patients compared to those in NDKD patients. Decreased serum RvD1 levels were responsible for the susceptibility to ischemic AKI in DKD patients. In animal experiments, both the serum RvD1 and renal ALX levels were downregulated. RvD1 treatment could ameliorate renal function and histological damage after ischemic injury in DN mice. RvD1 treatment also could inhibit the inflammatory response. Di-tert-butyl dicarbonate (BOC-2) treatment could deteriorate renal function and histological damage after ischemic injury in non-diabetic mice. RvD1 could inhibit the NF-κB activation and suppress inflammatory response mainly by inhibiting NF-κB signaling. Conclusion RvD1 attenuated susceptibility to ischemic AKI in diabetes by downregulating NF-κB signaling and inhibiting apoptosis. Downregulated serum RvD1 levels could be the crucial factor for susceptibility to ischemic AKI in diabetes.
Collapse
Affiliation(s)
- Zheng Li
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiwen Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hengcheng Lu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wenni Dai
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Junxiang Chen
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Liyu He
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
16
|
Sepsis Induces Physical and Mental Impairments in a Mouse Model of Post-Intensive Care Syndrome. J Clin Med 2021; 10:jcm10081593. [PMID: 33918862 PMCID: PMC8068824 DOI: 10.3390/jcm10081593] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 12/31/2022] Open
Abstract
Post-intensive care syndrome (PICS) is a physical, cognitive, and mental impairment observed in intensive care unit (ICU) survivors. Although this is an emerging problem in the ICU, how sepsis induces the characteristic symptoms of PICS remains unclear. To develop a model of PICS, we induced sepsis in male C57/B6 mice via sublethal cecum slurry injection and subsequently treated them using ICU-like interventions. At 1–2 weeks post-sepsis induction, we simultaneously evaluated the abilities of the surviving mice using the following behavioral tests: (1) a grip strength test (GST) and a treadmill test for physical assessment, (2) a novel object recognition test (NORT) for cognitive assessment, and (3) an open field test (OFT) and a marble burying test (MBT) for mental assessment. The surviving mice showed a range of deficits, including muscle weakness with significantly decreased grip strength in the GST; decreased total mileage during the treadmill test; anxiety and decreased activity, with significantly decreased time in the central area, and increased duration of immobility in the OFT; and an increased number of buried marbles in the MBT. Given these physical and mental impairments in the surviving mice, our model has the potential to elucidate mechanistic insights and to discover therapeutic targets and new interventions for PICS.
Collapse
|
17
|
Granja MG, Alves LP, Leardini-Tristão M, Saul ME, Bortoni LC, de Moraes FM, Ferreira EC, de Moraes BPT, da Silva VZ, Dos Santos AFR, Silva AR, Gonçalves-de-Albuquerque CF, Bambini-Junior V, Weyrich AS, Rondina MT, Zimmerman GA, de Castro-Faria-Neto HC. Inflammatory, synaptic, motor, and behavioral alterations induced by gestational sepsis on the offspring at different stages of life. J Neuroinflammation 2021; 18:60. [PMID: 33632243 PMCID: PMC7905683 DOI: 10.1186/s12974-021-02106-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 02/09/2021] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The term sepsis is used to designate a systemic condition of infection and inflammation associated with hemodynamic changes that result in organic dysfunction. Gestational sepsis can impair the development of the central nervous system and may promote permanent behavior alterations in the offspring. The aim of our work was to evaluate the effects of maternal sepsis on inflammatory cytokine levels and synaptic proteins in the hippocampus, neocortex, frontal cortex, and cerebellum of neonatal, young, and adult mice. Additionally, we analyzed the motor development, behavioral features, and cognitive impairments in neonatal, young and adult offspring. METHODS Pregnant mice at the 14th embryonic day (E14) were intratracheally instilled with saline 0.9% solution (control group) or Klebsiella spp. (3 × 108 CFU) (sepsis group) and started on meropenem after 5 h. The offspring was sacrificed at postnatal day (P) 2, P8, P30, and P60 and samples of liver, lung, and brain were collected for TNF-α, IL-1β, and IL-6 measurements by ELISA. Synaptophysin, PSD95, and β-tubulin levels were analyzed by Western blot. Motor tests were performed at all analyzed ages and behavioral assessments were performed in offspring at P30 and P60. RESULTS Gestational sepsis induces a systemic pro-inflammatory response in neonates at P2 and P8 characterized by an increase in cytokine levels. Maternal sepsis induced systemic downregulation of pro-inflammatory cytokines, while in the hippocampus, neocortex, frontal cortex, and cerebellum an inflammatory response was detected. These changes in the brain immunity were accompanied by a reduction of synaptophysin and PSD95 levels in the hippocampus, neocortex, frontal cortex, and cerebellum, in all ages. Behavioral tests demonstrated motor impairment in neonates, and depressive-like behavior, fear-conditioned memory, and learning impairments in animals at P30 and P60, while spatial memory abilities were affected only at P60, indicating that gestational sepsis not only induces an inflammatory response in neonatal mouse brains, but also affects neurodevelopment, and leads to a plethora of behavioral alterations and cognitive impairments in the offspring. CONCLUSION These data suggest that maternal sepsis may be causatively related to the development of depression, learning, and memory impairments in the litter.
Collapse
Affiliation(s)
- Marcelo Gomes Granja
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz - Fiocruz, Rio de Janeiro, Brazil
- Programa de Pós-graduação em Biologia Molecular e Celular, Universidade Federal do Estado do Rio de Janeiro - UNIRIO, Rio de Janeiro, Brazil
| | - Letícia Pires Alves
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz - Fiocruz, Rio de Janeiro, Brazil
- Programa de Pós-graduação em Biologia Molecular e Celular, Universidade Federal do Estado do Rio de Janeiro - UNIRIO, Rio de Janeiro, Brazil
| | - Marina Leardini-Tristão
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz - Fiocruz, Rio de Janeiro, Brazil
| | - Michelle Edelman Saul
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz - Fiocruz, Rio de Janeiro, Brazil
- Faculdade de Medicina, Universidade Estácio de Sá - UNESA, Rio de Janeiro, Brazil
| | - Letícia Coelho Bortoni
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz - Fiocruz, Rio de Janeiro, Brazil
- Faculdade de Medicina, Universidade Estácio de Sá - UNESA, Rio de Janeiro, Brazil
| | - Flávia Maciel de Moraes
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz - Fiocruz, Rio de Janeiro, Brazil
| | - Erica Camila Ferreira
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz - Fiocruz, Rio de Janeiro, Brazil
| | - Bianca Portugal Tavares de Moraes
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz - Fiocruz, Rio de Janeiro, Brazil
- Programa de Pós-graduação em Neurociências, Universidade Federal Fluminense - UFF, Niterói, Rio de Janeiro, Brazil
| | - Victória Zerboni da Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz - Fiocruz, Rio de Janeiro, Brazil
| | | | - Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz - Fiocruz, Rio de Janeiro, Brazil
| | | | - Victorio Bambini-Junior
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, PR1 2HE, Lancashire, Preston, England, UK
| | - Andrew S Weyrich
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, PR1 2HE, Lancashire, Preston, England, UK
| | - Matthew T Rondina
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine and Pathology, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine and GRECC, George E. Wahlen VAMC, Salt Lake City, UT, USA
| | - Guy A Zimmerman
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
| | | |
Collapse
|
18
|
Kalinkovich A, Pouyrovsky M, Nasyrova R, Livshits G. Biased activation of inflammation pro-resolving receptors as an evolving supportive strategy in schizophrenia treatment. Schizophr Res 2021; 228:295-297. [PMID: 33497903 DOI: 10.1016/j.schres.2021.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/03/2021] [Accepted: 01/11/2021] [Indexed: 10/22/2022]
Affiliation(s)
- Alexander Kalinkovich
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | - Michael Pouyrovsky
- Maale HaCarmel Mental Health Center, Affiliated to Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Regina Nasyrova
- V. M. Bekhterev National Research Medical Center for Psychiatry and Neurology, Russian Federation Ministry of Health, Bekhterev Street, St. Petersburg, Russia
| | - Gregory Livshits
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Israel; Adelson School of Medicine, Ariel University, Ariel, Israel.
| |
Collapse
|
19
|
Baicalin Ameliorates Cognitive Impairment and Protects Microglia from LPS-Induced Neuroinflammation via the SIRT1/HMGB1 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4751349. [PMID: 33029280 PMCID: PMC7527898 DOI: 10.1155/2020/4751349] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/20/2020] [Accepted: 09/04/2020] [Indexed: 12/16/2022]
Abstract
Systemic inflammation often induces neuroinflammation and disrupts neural functions, ultimately causing cognitive impairment. Furthermore, neuronal inflammation is the key cause of many neurological conditions. It is particularly important to develop effective neuroprotectants to prevent and control inflammatory brain diseases. Baicalin (BAI) has a wide variety of potent neuroprotective and cognitive enhancement properties in various models of neuronal injury through antioxidation, anti-inflammation, anti-apoptosis, and stimulating neurogenesis. Nevertheless, it remains unclear whether BAI can resolve neuroinflammation and cognitive decline triggered by systemic or distant inflammatory processes. In the present study, intraperitoneal lipopolysaccharide (LPS) administration was used to establish neuroinflammation to evaluate the potential neuroprotective and anti-inflammatory effects of BAI. Here, we report that BAI activated silent information regulator 1 (SIRT1) to deacetylate high-mobility group box 1 (HMGB1) protein in response to acute LPS-induced neuroinflammation and cognitive deficits. Furthermore, we demonstrated the anti-inflammatory and cognitive enhancement effects and the underlying molecular mechanisms of BAI in modulating microglial activation and systemic cytokine production, including tumor necrosis factor- (TNF-) α and interleukin- (IL-) 1β, after LPS exposure in mice and in the microglial cell line, BV2. In the hippocampus, BAI not only reduced reactive microglia and inflammatory cytokine production but also modulated SIRT1/HMGB1 signaling in microglia. Interestingly, pretreatment with SIRT1 inhibitor EX-527 abolished the beneficial effects of BAI against LPS exposure. Specifically, BAI treatment inhibited HMGB1 release via the SIRT1/HMGB1 pathway and reduced the nuclear translocation of HMGB1 in LPS-induced BV2 cells. These effects were reversed in BV2 cells by silencing endogenous SIRT1. Taken together, these findings indicated that BAI reduced microglia-associated neuroinflammation and improved acute neurocognitive deficits in LPS-induced mice via SIRT1-dependent downregulation of HMGB1, suggesting a possible novel protection against acute neurobehavioral deficits, such as delayed neurocognitive recovery after anesthesia and surgery challenges.
Collapse
|
20
|
S100A9 Upregulation Contributes to Learning and Memory Impairments by Promoting Microglia M1 Polarization in Sepsis Survivor Mice. Inflammation 2020; 44:307-320. [PMID: 32918665 DOI: 10.1007/s10753-020-01334-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/16/2020] [Accepted: 08/25/2020] [Indexed: 12/17/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is a clinical syndrome of brain dysfunction secondary to sepsis, which is characterized by long-term neurocognitive deficits such as memory, attention, and executive dysfunction. However, the mechanisms underlying SAE remain unclear. By using transcriptome sequencing approach, we showed that hippocampal S100A9 was significantly increased in sepsis induced by cecal ligation and puncture (CLP) or lipopolysaccharide (LPS) challenge. Thus, we used S100A9 inhibitor Paquinimod to study the role of S100A9 in cognitive impairments in CLP-induced and LPS-induced mice models of SAE. Sepsis survivor mice underwent behavioral tests or the hippocampal tissues subjected to Western blotting, real-time quantitative PCR, and immunohistochemistry. Our results showed that CLP-induced and LPS-induced memory impairments were accompanied with increased expressions of hippocampal microglia Iba1 and CD86 (M1 markers), but reduced expression of Arg1 (M2 marker). Notably, S100A9 inhibition significantly improved the survival rate and learning and memory impairments in sepsis survivors, with a shift from M1 to M2 phenotype. Taken together, our study suggests that S100A9 upregulation might contribute to learning and memory impairments by promoting microglia M1 polarization in sepsis survivors, whereas S100A9 inhibition might provide a potential therapeutic target for SAE.
Collapse
|
21
|
Protective function of exosomes from adipose tissue-derived mesenchymal stem cells in acute kidney injury through SIRT1 pathway. Life Sci 2020; 255:117719. [DOI: 10.1016/j.lfs.2020.117719] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 12/19/2022]
|
22
|
Simvastatin Prevents Long-Term Cognitive Deficits in Sepsis Survivor Rats by Reducing Neuroinflammation and Neurodegeneration. Neurotox Res 2020; 38:871-886. [PMID: 32524380 DOI: 10.1007/s12640-020-00222-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/09/2020] [Accepted: 05/01/2020] [Indexed: 02/07/2023]
Abstract
Sepsis-associated encephalopathy causes brain dysfunction that can result in cognitive impairments in sepsis survivor patients. In previous work, we showed that simvastatin attenuated oxidative stress in brain structures related to memory in septic rats. However, there is still a need to evaluate the long-term impact of simvastatin administration on brain neurodegenerative processes and cognitive damage in sepsis survivors. Here, we investigated the possible neuroprotective role of simvastatin in neuroinflammation, and neurodegeneration conditions of brain structures related to memory in rats at 10 days after sepsis survival. Male Wistar rats (250-300 g) were submitted to cecal ligation and puncture (CLP, n = 42) or remained as non-manipulated (naïve, n = 30). Both groups were treated (before and after the surgery) by gavage with simvastatin (20 mg/kg) or an equivalent volume of saline and observed for 10 days. Simvastatin-treated rats that survived to sepsis showed a reduction in the levels of nitrate, IL1-β, and IL-6 and an increase in Bcl-2 protein expression in the prefrontal cortex and hippocampus, and synaptophysin only in the hippocampus. Immunofluorescence revealed a reduction of glial activation, neurodegeneration, apoptosis, and amyloid aggregates confirmed by quantification of GFAP, Iba-1, phospho Ser396-tau, total tau, cleaved caspase-3, and thioflavin-S in the prefrontal cortex and hippocampus. In addition, treated animals presented better performance in tasks involving habituation memory, discriminative, and aversive memory. These results suggest that statins exert a neuroprotective role by upregulation of the Bcl-2 and gliosis reduction, which may prevent the cognitive deficit observed in sepsis survivor animals.
Collapse
|
23
|
CXCR5 Knockdown Attenuates Hippocampal Neurogenesis Deficits and Cognitive Impairment in a Mouse Model of Sepsis-associated Encephalopathy. Neuroscience 2020; 433:212-220. [DOI: 10.1016/j.neuroscience.2020.03.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/28/2019] [Accepted: 03/08/2020] [Indexed: 01/21/2023]
|
24
|
Heimfarth L, Carvalho AMS, Quintans JDSS, Pereira EWM, Lima NT, Bezerra Carvalho MT, Barreto RDSS, Moreira JCF, da Silva-Júnior EF, Schmitt M, Bourguignon JJ, de Aquino TM, Araújo-Júnior JXD, Quintans-Júnior LJ. Indole-3-guanylhydrazone hydrochloride mitigates long-term cognitive impairment in a neonatal sepsis model with involvement of MAPK and NFκB pathways. Neurochem Int 2020; 134:104647. [DOI: 10.1016/j.neuint.2019.104647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/09/2019] [Accepted: 12/18/2019] [Indexed: 01/20/2023]
|
25
|
Liu J, Peng L, Li J. The Lipoxin A4 Receptor Agonist BML-111 Alleviates Inflammatory Injury and Oxidative Stress in Spinal Cord Injury. Med Sci Monit 2020; 26:e919883. [PMID: 31971927 PMCID: PMC6996263 DOI: 10.12659/msm.919883] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Spinal cord injury (SCI) has a high incidence and causes serious harm. Lipoxin A4 (LXA4) receptor agonist BML-111 was reported to regulate inflammation and oxidative stress. The goal of this study was to assess whether BML-111 could protect against SCI by suppressing inflammation and oxidative stress. Material/Methods We developed a rat SCI model, then BML-111 was intraperitoneally injected into SCI rats to observe the BML-111 function. The pathological changes of SCI were observed with hematoxylin and eosin (HE) staining. Motor function of rats were assessed by the modified Tarlov’s scale. ELISA was used to assess the changes in levels of TNF-α, IL-1β, and IL-6. Western blot analysis was performed to assess the expressions of TNF-α, IL-1β, IL-6, Bcl2, Bax, and cleaved caspase3 in spinal cord tissue. TOS and TAS in rat serum were detected by xylenol orange method and ABTS method, respectively. The apoptotic cells in spinal cord tissue were observed with TUNEL assay. Results The results indicated that BML-111 effectively improved the SCI and motor function of rats. BML-111 treatment decreased the levels of TNF-α, IL-1β, and IL-6 in serum and spinal cord tissue, as well as decreasing the levels of TOS and TAS and cell apoptosis. Conclusions BML-111 alleviated inflammation and oxidative stress in SCI rats.
Collapse
Affiliation(s)
- Jian Liu
- Department of Orthopedics, Chongqing Emergency Medical Center, Chongqing, China (mainland)
| | - Lei Peng
- Department of Orthopedics, Chongqing Emergency Medical Center, Chongqing, China (mainland)
| | - Jie Li
- Department of Orthopedics, Chongqing Emergency Medical Center, Chongqing, China (mainland)
| |
Collapse
|
26
|
Mei L, He M, Zhang C, Miao J, Wen Q, Liu X, Xu Q, Ye S, Ye P, Huang H, Lin J, Zhou X, Zhao K, Chen D, Zhou J, Li C, Li H. Paeonol attenuates inflammation by targeting HMGB1 through upregulating miR-339-5p. Sci Rep 2019; 9:19370. [PMID: 31852965 PMCID: PMC6920373 DOI: 10.1038/s41598-019-55980-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/02/2019] [Indexed: 12/26/2022] Open
Abstract
Sepsis is a life-threatening disease caused by infection. Inflammation is a key pathogenic process in sepsis. Paeonol, an active ingredient in moutan cortex (a Chinese herb), has many pharmacological activities, such as anti-inflammatory and antitumour actions. Previous studies have indicated that paeonol inhibits the expression of HMGB1 and the transcriptional activity of NF-κB. However, its underlying mechanism is still unknown. In this study, microarray assay and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) results confirmed that paeonol could significantly up-regulate the expression of miR-339-5p in RAW264.7 cells stimulated by LPS. Dual-luciferase assays indicated that miR-339-5p interacted with the 3′ untranslated region (3′-UTR) of HMGB1. Western blot, immunofluorescence and enzyme-linked immunosorbent assay (ELISA) analyses indicated that miR-339-5p mimic and siHMGB1 both negatively regulated the expression and secretion of inflammatory cytokines (e.g., HMGB1, IL-1β and TNF-α) in LPS-induced RAW264.7 cells. Studies have confirmed that IKK-β is targeted by miR-339-5p, and we further found that paeonol could inhibit IKK-β expression. Positive mutual feedback between HMGB1 and IKK-β was observed when we silenced HMGB1 or IKK-β. These results indicated that paeonol could attenuate the inflammation mediated by HMGB1 and IKK-β by upregulating miR-339-5p expression. In addition, we constructed CLP model mice by cecal ligation and puncture. Paeonol was used to intervene to investigate its anti-inflammatory effect in vivo. The results showed that paeonol could improve the survival rate of sepsis mice and protect the kidney of sepsis mice.
Collapse
Affiliation(s)
- Liyan Mei
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Meihong He
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Chaoying Zhang
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Jifei Miao
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Quan Wen
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xia Liu
- School of Basic Medical Sciences, Guiyang University of Chinese Medicine, Guiyang, Guizhou Province, 550025, China
| | - Qin Xu
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Sen Ye
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Peng Ye
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Huina Huang
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Junli Lin
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Xiaojing Zhou
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Kai Zhao
- School of Nursing Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Dongfeng Chen
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Jianhong Zhou
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Chun Li
- School of Nursing Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Hui Li
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China.
| |
Collapse
|
27
|
Park JY, Han S, Ka HI, Joo HJ, Soh SJ, Yoo KH, Yang Y. Silent mating-type information regulation 2 homolog 1 overexpression is an important strategy for the survival of adapted suspension tumor cells. Cancer Sci 2019; 110:2773-2782. [PMID: 31348594 PMCID: PMC6726698 DOI: 10.1111/cas.14147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 07/02/2019] [Accepted: 07/10/2019] [Indexed: 12/16/2022] Open
Abstract
Characterization of circulating tumor cells (CTC) is important to prevent death caused by the metastatic spread of cancer cells because CTC are associated with distal metastasis and poor prognosis of breast cancer. We have previously developed suspension cells (SC) using breast cancer cell lines and demonstrated their high metastatic potential. As survival of CTC is highly variable from a few hours to decades, herein we cultured SC for an extended time and named them adapted suspension cells (ASC). Silent mating‐type information regulation 2 homolog 1 (SIRT1) expression increased in ASC, which protected the cells from apoptosis. High SIRT1 expression was responsible for the suppression of nuclear factor kappa B (NF‐κB) activity and downregulation of reactive oxygen species (ROS) in ASC. As the inhibition of NF‐κB and ROS production in SIRT1‐depleted ASC contributed to the development of resistance to apoptotic cell death, maintenance of a low ROS level and NF‐κB activity in ASC is a crucial function of SIRT1. Thus, SIRT1 overexpression may play an important role in growth adaptation of SC because SIRT1 expression is increased in long‐term rather than in short‐term cultures.
Collapse
Affiliation(s)
- Ji Young Park
- Department of Biological Sciences, Research Center for Cellular Heterogeneity, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| | - Sora Han
- Department of Biological Sciences, Research Center for Cellular Heterogeneity, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| | - Hye In Ka
- Department of Biological Sciences, Research Center for Cellular Heterogeneity, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| | - Hyun Jeong Joo
- Department of Biological Sciences, Research Center for Cellular Heterogeneity, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| | - Su Jung Soh
- Department of Biological Sciences, Research Center for Cellular Heterogeneity, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| | - Kyung Hyun Yoo
- Department of Biological Sciences, Research Center for Cellular Heterogeneity, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| | - Young Yang
- Department of Biological Sciences, Research Center for Cellular Heterogeneity, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| |
Collapse
|
28
|
Tian M, Qingzhen L, Zhiyang Y, Chunlong C, Jiao D, Zhang L, Li W. Attractylone attenuates sepsis-associated encephalopathy and cognitive dysfunction by inhibiting microglial activation and neuroinflammation. J Cell Biochem 2019; 120:7101-7108. [PMID: 30672013 DOI: 10.1002/jcb.27983] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 10/08/2018] [Indexed: 01/24/2023]
Abstract
Multiple studies demonstrated that sepsis is a life-threatening state of organ dysfunction caused by infection and can induce neuroinflammation and cognitive impairment. The aim of this study was to evaluate the protective effects of attractylone (Atr) on sepsis-associated encephalopathy (SAE) and cognitive dysfunction. Moreover, we studied the underlying molecular mechanisms. We used an LPS-induced sepsis mouse model and evaluated the cognitive function with the Morris water maze and open field test. Neuronal damage in the hippocampus was assessed by immunohistochemical analysis. BV2 cells were used to identify the protective mechanism of Atr. The result showed that Atr attenuated LPS-induced cognitive impairment, neural apoptosis, inflammatory factors, and microglial activation. The in vitro experiment showed that Atr promoted silent information regulator 1 (SIRT1) expression and suppressed NFκB expression. Downregulation of SIRT1 reversed the protective effect of Atr in the LPS condition. Moreover, Atr-induced SIRT1 expression promoted BV2 from LPS-induced M1 to M2 phenotype. Taken together, these results indicated that Atr was a potential therapeutic agent for SAE and cognitive dysfunction.
Collapse
Affiliation(s)
- Mi Tian
- Department of Anesthesiology, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liu Qingzhen
- Department of Anesthesiology, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Zhiyang
- Department of Anesthesiology, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chen Chunlong
- Department of Anesthesiology, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Duan Jiao
- Department of Anesthesiology, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lidong Zhang
- Department of Anesthesiology, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiyan Li
- Department of Anesthesiology, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|