1
|
Lyu Z, Ji X, Chen G, An B. Atractylodin ameliorates lipopolysaccharide and d-galactosamine-induced acute liver failure via the suppression of inflammation and oxidative stress. Int Immunopharmacol 2019; 72:348-357. [DOI: 10.1016/j.intimp.2019.04.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 03/28/2019] [Accepted: 04/02/2019] [Indexed: 12/21/2022]
|
2
|
Bao LH, Zhang YN, Zhang JN, Gu L, Yang HM, Huang YY, Xia N, Zhang H. Urate inhibits microglia activation to protect neurons in an LPS-induced model of Parkinson's disease. J Neuroinflammation 2018; 15:131. [PMID: 29720230 PMCID: PMC5932803 DOI: 10.1186/s12974-018-1175-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/23/2018] [Indexed: 01/30/2023] Open
Abstract
Background Multiple risk factors contribute to the progression of Parkinson’s disease, including oxidative stress and neuroinflammation. Epidemiological studies have revealed a link between higher urate level and a lower risk of developing PD. However, the mechanistic basis for this association remains unclear. Urate protects dopaminergic neurons from cell death induced by oxidative stress. Here, we investigated a novel role of urate in microglia activation in a lipopolysaccharide (LPS)-induced PD model. Methods We utilized Griess, ELISA, real-time PCR, Western blot, immunohistochemistry, and immunofluorescence to detect the neuroinflammation. For Griess, ELISA, Western blot, and immunofluorescence assay, cells were seeded in 6-well plates pre-coated with poly-l-lysine (PLL) and incubated for 24 h with the indicated drugs. For real-time PCR assay, cells were seeded in 6-well plates pre-coated with PLL and incubated for 6 h with the indicated drugs. For animal experiments, rats were injected with urate or its vehicle twice daily for five consecutive days before and after stereotaxic surgery. Rats were killed and brain tissues were harvested after 4 weeks of LPS injection. Results In cultured BV2 cells and rat primary microglia, urate suppressed proinflammatory cytokine production and inducible cyclooxygenase 2 and nitric oxide synthase expression to protect dopaminergic neurons from the toxic effects of activated microglia. The neuroprotective effects of urate may also be associated with the stimulation of anti-inflammatory factors interleukin 10 and transforming growth factor β1. Intracellular urate level was increased in a dose-dependent manner upon co-treatment with urate and LPS as compared with LPS alone, an effect that was abrogated by pretreatment with probenecid (PBN), an inhibitor of both glucose transporter 9 and urate transporter 1 (URAT1). PBN also abolished the anti-inflammatory effect of urate. Consistent with these in vitro observations, the number of tyrosine hydroxylase-positive neurons was decreased and the loss of motor coordination was reversed by urate administration in an LPS-induced rat model of PD. Additionally, increased plasma urate level abolished the reduction of URAT1 expression, the increase in the expression of interleukin-1β, and the number of ionized calcium-binding adaptor molecule 1-positive microglia along with changes in their morphology. Conclusions Urate protects neurons against cytotoxicity induced by microglia activation via modulating urate transporter-mediated intracellular urate level. Electronic supplementary material The online version of this article (10.1186/s12974-018-1175-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Li-Hui Bao
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Ya-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Jian-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Li Gu
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Hui-Min Yang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Yi-Ying Huang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Ning Xia
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders and Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
3
|
Zhang H, Jia R, Wang F, Qiu G, Qiao P, Xu X, Wu D. Catalpol protects mice against Lipopolysaccharide/D-galactosamine-induced acute liver injury through inhibiting inflammatory and oxidative response. Oncotarget 2017; 9:3887-3894. [PMID: 29423091 PMCID: PMC5790508 DOI: 10.18632/oncotarget.23242] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/01/2017] [Indexed: 01/10/2023] Open
Abstract
The purpose of this study was to investigate the protective effect of catalpol on Lipopolysaccharide (LPS)/D-galactosamine (D-gal)-induced acute liver injury in mice. The mouse model was established by injection of LPS and D-gal. Catalpol (2.5, 5, 10 mg/kg) were pretreated intraperitoneally 1 h before LPS and D-gal. The survival rate, AST, ALT, MDA, MPO activity, hepatic tissue histology, TNF-α level, and NF-κB activation were assayed. The results revealed that catalpol dose-dependently elevated the survival rate. Furthermore, catalpol reduced the activities of AST, ALT, MDA, and MPO. The production of TNF-α was also inhibited by treatment of catalpol. In addition, catalpol inhibited LPS/D-gal-induced NF-κB activation. The expression of Nrf2 and HO-1 were up-regulated by treatment of catalpol. These results indicated that pretreatment with catalpol could attenuate LPS/D-gal-induced acute liver injury in mice and the underlying mechanism may due to the inhibition of NF-κB signaling pathway and the activation of Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Haogang Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Ruichun Jia
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Fujing Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Gongcai Qiu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Pengfei Qiao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Xunzheng Xu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Dequan Wu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| |
Collapse
|
4
|
Silachev DN, Usatikova EA, Pevzner IB, Zorova LD, Babenko VA, Gulyaev MV, Pirogov YA, Plotnikov EY, Zorov DB. Effect of Anesthetics on Efficiency of Remote Ischemic Preconditioning. BIOCHEMISTRY (MOSCOW) 2017; 82:1006-1016. [PMID: 28988529 DOI: 10.1134/s0006297917090036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Remote ischemic preconditioning of hind limbs (RIPC) is an effective method for preventing brain injury resulting from ischemia. However, in numerous studies RIPC has been used on the background of administered anesthetics, which also could exhibit neuroprotective properties. Therefore, investigation of the signaling pathways triggered by RIPC and the effect of anesthetics is important. In this study, we explored the effect of anesthetics (chloral hydrate and Zoletil) on the ability of RIPC to protect the brain from injury caused by ischemia and reperfusion. We found that RIPC without anesthesia resulted in statistically significant decrease in neurological deficit 24 h after ischemia, but did not affect the volume of brain injury. Administration of chloral hydrate or Zoletil one day prior to brain ischemia produced a preconditioning effect by their own, decreasing the degree of neurological deficit and lowering the volume of infarct with the use of Zoletil. The protective effects observed after RIPC with chloral hydrate or Zoletil were similar to those observed when only the respective anesthetic was used. RIPC was accompanied by significant increase in the level of brain proteins associated with the induction of ischemic tolerance such as pGSK-3β, BDNF, and HSP70. However, Zoletil did not affect the level of these proteins 24 h after injection, and chloral hydrate caused increase of only pGSK-3β. We conclude that RIPC, chloral hydrate, and Zoletil produce a significant neuroprotective effect, but the simultaneous use of anesthetics with RIPC does not enhance the degree of neuroprotection.
Collapse
Affiliation(s)
- D N Silachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Cai J, Peng Y, Chen T, Liao H, Zhang L, Chen Q, He Y, Wu P, Xie T, Pan Q. Chloral Hydrate Treatment Induced Apoptosis of Macrophages via Fas Signaling Pathway. Med Sci Monit 2016; 22:4836-4843. [PMID: 27941708 PMCID: PMC5167102 DOI: 10.12659/msm.901772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND There are recent reports on several anesthetics that have anti-inflammatory and anti-infective effects apart from their uses for pain relief and muscle relaxation. Chloral hydrate is a clinical anesthetic drug and sedative that has also been reported to attenuate inflammatory response, but the mechanisms are not clearly understood. MATERIAL AND METHODS This study investigated the effect of chloral hydrate treatment on the apoptosis of macrophages and explored the underlying mechanisms. RAW264.7 macrophages were treated with various concentrations of chloral hydrate for various lengths of time. Morphological changes were observed under a light microscope and apoptosis was detected with annexin-V-FITC/PI double-staining assay, Hochest 33258 and DNA ladder assay, the expression of Fas/FasL was detected with a flow cytometer, and the Fas signaling pathway was assessed by Western blotting. RESULTS The results showed that chloral hydrate treatment induced the morphology of RAW264.7 macrophages to change shape from typical fusiform to round in a concentration- and time-dependent manner, and was finally suspended in the supernatant. For the induction of apoptosis, chloral hydrate treatment induced the apoptosis of RAW264.7 macrophages from early-to-late stage apoptosis in a concentration- and time-dependent manner. For the mechanism, chloral hydrate treatment induced higher expression of Fas on RAW264.7 macrophages, and was also associated with changes in the expression of proteins involved in Fas signaling pathways. CONCLUSIONS Chloral hydrate treatment can induce the apoptosis of RAW264.7 macrophages through the Fas signaling pathway, which may provide new options for adjunctive treatment of acute inflammation.
Collapse
Affiliation(s)
- Jun Cai
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Yanxia Peng
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Ting Chen
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Huanjin Liao
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Lifang Zhang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Qiuhua Chen
- Department of Immunology and Rheumatology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Yiming He
- Department of Immunology and Rheumatology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Ping Wu
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Tong Xie
- Department of Immunology and Rheumatology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Qingjun Pan
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| |
Collapse
|
6
|
Liu JH, Feng D, Zhang YF, Shang Y, Wu Y, Li XF, Pei L. Chloral Hydrate Preconditioning Protects Against Ischemic Stroke via Upregulating Annexin A1. CNS Neurosci Ther 2015. [PMID: 26205642 DOI: 10.1111/cns.12435] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIMS Preconditioning is promising for treating cerebral ischemic stroke. Annexin A1 (ANXA1) is a homeostatic antiinflammatory mediator that participates in countering against ischemic injuries. We investigated whether chloral hydrate preconditioning (CH) exerts neuroprotection via regulation of ANXA1 in stroke. METHODS Adult male C57BL/6J mice or ANXA1 knockout (ANXA1(-/-) ) mice were randomly allocated to control (NCH) and CH groups [2%, 6%, and 10% chloral hydrate (i.p.) 1 h before the middle cerebral artery occlusion (MCAO)]. Neurological performances were evaluated by modified 7-point neurological scales and rotarod test. Cerebral infarction was analyzed by triphenyltetrazolium chloride (TTC) staining and MRI. The expression of ANXA1, pro-inflammatory (TNF-α, IL-1β, IL-6), and antiinflammatory (IL-4, IL-10, TGF-β) cytokines was investigated by RT-PCR, western blot, and immunofluorescence. RESULTS Chloral hydrate preconditioning significantly improved the neurological outcomes and reduced the infarction and brain edema after ischemia. In addition, CH increased the expression of ANXA1 in the microglia, decreased the levels of TNF-α, IL-1β, and IL-6, while elevated the levels of IL-4, IL-10, and TGF-β in the MCAO mice. Furthermore, both ANXA1 blocker Boc1 (5 mg/kg, i.c.v.) or ANXA1 gene deficiency restrained the protective effects of CH against stroke. CONCLUSIONS Chloral hydrate preconditioning protects against ischemic injuries through upregulating the expression of ANXA1, and the followed antiinflammatory mechanisms.
Collapse
Affiliation(s)
- Jiu-Hong Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Feng
- Department of Pain Clinic, Institute of Anesthesia and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Fen Zhang
- Department of Neurobiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - You Shang
- Department of Pain Clinic, Institute of Anesthesia and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Feng Li
- Department of Neurology, The People's Hospital of Guangxi Zhuang-Autonomous Region, Guilin, China
| | - Lei Pei
- Department of Neurobiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Pan Q, Liu Y, Zhu X, Liu H. Chloral hydrate-dependent reduction in the peptidoglycan-induced inflammatory macrophage response is associated with lower expression levels of toll-like receptor 2. Exp Ther Med 2014; 7:1305-1310. [PMID: 24940429 PMCID: PMC3991540 DOI: 10.3892/etm.2014.1587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 02/21/2014] [Indexed: 12/24/2022] Open
Abstract
The aim of this study was to investigate the effect and mechanism of action of chloral hydrate on the peptidoglycan (PGN)-induced inflammatory macrophage response. The effect of chloral hydrate on the production of tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6) by murine peritoneal macrophages with PGN-stimulation was investigated. In addition, RAW264.7 cells transfected with a nuclear factor-κB (NF-κB) luciferase reporter plasmid stimulated by PGN were used to study the effect of chloral hydrate on the levels NF-κB activity. Flow cytometry and western blotting were performed to investigate the expression levels of toll-like receptor 2 (TLR2) in the treated RAW264.7 cells. It was identified that chloral hydrate reduced the levels of IL-6 and TNF-α produced by the peritoneal macrophages stimulated with PGN. The levels of NF-κB activity of the RAW264.7 cells stimulated by PGN decreased following treatment with chloral hydrate, which was associated with a reduction in the expression levels of TLR2 and reduced levels of TLR2 signal transduction. These data demonstrate that chloral hydrate reduced the magnitude of the PGN-induced inflammatory macrophage response associated with lower expression levels of TLR2.
Collapse
Affiliation(s)
- Qingjun Pan
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical College, Zhangjiang, Guangdong 524001, P.R. China
| | - Yuan Liu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, SAR 999077, P.R. China
| | - Xuezhi Zhu
- Guangdong Yuehai Feed Group Co. Ltd., Zhangjiang, Guangdong 524001, P.R. China
| | - Huafeng Liu
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical College, Zhangjiang, Guangdong 524001, P.R. China
| |
Collapse
|
8
|
Lee JK, Jung JS, Park SH, Sim YB, Suh HW. Deficiency of alpha-calcitonin gene-related peptide induces inflammatory responses and lethality in sepsis. Cytokine 2013; 64:548-54. [PMID: 24021706 DOI: 10.1016/j.cyto.2013.07.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 07/08/2013] [Accepted: 07/30/2013] [Indexed: 01/15/2023]
Abstract
In the present study, we examined the role of alpha-calcitonin gene-related peptide (αCGRP) on expression of neuropeptides in the brain, inflammatory responses, and survival rate in septic shock condition. We examined expression of neuropeptides such as αCGRP, proopiomelanocortin (POMC), corticotrophin releasing hormone (CRH), and proenkephalin (ProENK) in the hippocampus and hypothalamus in C57BL/6 (WT) or αCGRP-/- (KO) mice subjected to sepsis. Cecal ligation and puncture (CLP) or lipopolysaccharide/D-galactosamine (LPS/D-GalN) treatment showed significant increases of hippocampal and hypothalamic αCGRP, POMC, CRH, and ProENK mRNA levels in WT mice, but not ProENK mRNA in the hypothalamus at 6h after on-set of sepsis. However, enhanced mRNA levels of POMC, CRH, and ProENK genes were not increased in the hippocampus and hypothalamus of CLP-subjected KO mice at 6h following sepsis. KO mice treated with LPS/D-GalN displayed a significant enhancement of plasma corticosterone, aspartate aminotransferase, and alanine aminotransferase levels compared to LPS/D-GalN treated WT mice at 12h after induction of sepsis. In addition, plasma levels of pro-inflammatory cytokines, such as IL-1β and TNF-α, were also further increased in KO mice compared to WT mice at 24h after CLP or LPS/D-GalN treatment. Interestingly, mRNA expressions of IL-6 and IL-10, anti-inflammatory cytokines, were synergistically enhanced in liver and lymph node of KO mice compared to WT mice at 6h after CLP. However, plasma level of IL-10 but not IL-6 was significantly decreased in KO mice compared to WT mice at 24h after CLP or LPS/D-GalN challenge. The survival rate of KO mice was significantly reduced compared to WT mice following mild (1 punch) and moderate (2 punch) CLP and LPS/D-GalN administration. Taken together, our findings suggest that the activation of αCGRP may induce other neuropeptides associated with immunomodulation at CNS level and modulate immune responses as enhancing anti-inflammatory cytokines and reducing pro-inflammatory cytokines during the sepsis.
Collapse
Affiliation(s)
- Jin-Koo Lee
- Department of Pharmacology, College of Medicine, Institute of Bio-Science Technology, Dankook University, Cheonan 330-714, Republic of Korea; Translational Research Center, Institute of Bio-Science Technology, Dankook University, Cheonan 330-714, Republic of Korea
| | | | | | | | | |
Collapse
|
9
|
Li S, Bao H, Han L, Liu L. Effects of propofol on early and late cytokines in lipopolysaccharide-induced septic shock in rats. J Biomed Res 2013; 24:389-94. [PMID: 23554654 PMCID: PMC3596685 DOI: 10.1016/s1674-8301(10)60052-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Indexed: 12/16/2022] Open
Abstract
Objective It has been reported that the intravenous anesthetic propofol (PPF) has anti-inflammatory effects in vitro and in patients. The purpose of this study was to investigate whether PPF has anti-inflammatory effects in lipopolysaccharide (LPS)-induced septic shock by inhibiting the induction of pro-inflammatory cytokines [interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α)] and high mobility group box 1 (HMGB1) in rats. Methods Thirty six male Wistar rats were randomly assigned to one of three groups (control group, PPF + LPS group and LPS group; n = 12 per group). Control group rats received a 0.9% NaCl solution (NS) by the tail vein. The PPF + LPS group rats received PPF (10 mg/kg bolus, followed by infusion at 10 mg/(kg·h) through a femoral vein catheter) 1 h before LPS (7.5 mg/kg) administration via the tail vein. The LPS group rats received injection of LPS (7.5 mg/kg) via the tail vein. Hemodynamic effects were recorded as well as mortality rates, and plasma cytokine con-centrations (TNF-α, IL-6, HMGB1) were measured for the 24-h observation period. Results The mean arterial pressure and heart rate of the PPF + LPS group were more stable than those of the LPS group. The mortality at 24 h after the administration of the LPS injection was much higher in the LPS group (58.3%) compared to the PPF + LPS group (25.0%). Plasma concentrations of cytokines (IL-6 and TNF-α) and HMGB1 were significantly reduced in the PPF + LPS group compared to the LPS group (P < 0.05). Conclusion Pretreatment with PPF reduced the mortality rate of rats and attenuated the pro-inflammatory cytokine responses in an endotoxin shock model through an anti-inflammatory action inhibiting induction of HMGB1.
Collapse
Affiliation(s)
- Sha Li
- Department of Anesthesiology, Nanjing First Hospital Affiliated to Nanjing Medical University, Nanjing 210006, Jiangsu Province, China
| | | | | | | |
Collapse
|
10
|
Protective effects of probiotic Lactobacillus casei Zhang against endotoxin- and d-galactosamine-induced liver injury in rats via anti-oxidative and anti-inflammatory capacities. Int Immunopharmacol 2013; 15:30-7. [DOI: 10.1016/j.intimp.2012.10.026] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 10/23/2012] [Accepted: 10/26/2012] [Indexed: 12/18/2022]
|
11
|
Chen EQ, Gong DY, Leng XH, Bai L, Liu C, Wang LC, Tang H. Inhibiting the expression of hepatocyte nuclear factor 4 alpha attenuates lipopolysaccharide/D-galactosamine-induced fulminant hepatic failure in mice. Hepatobiliary Pancreat Dis Int 2012; 11:624-9. [PMID: 23232634 DOI: 10.1016/s1499-3872(12)60235-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Hepatocyte nuclear factor 4 alpha (HNF4alpha) plays an important role in regulating cytokine-induced inflammatory responses. This study aimed to investigate the role of HNF4alpha in the development of fulminant hepatic failure (FHF) induced by lipopolysaccharide/D-galactosamine (LPS/D-GalN). METHODS The FHF model was induced by simultaneous intraperitoneal injection of LPS/D-GalN in mice. Three days prior to LPS/D-GalN administration, HNF4alpha short-hairpin interfering RNA expression plasmid or physiological saline was injected via the tail vein with a hydrodynamics-based procedure. The degree of hepatic damage and cumulative survival rate were subsequently assessed. RESULTS The expression of HNF4alpha was increased in the early stage after LPS/D-GalN administration. Inhibiting the expression of HNF4alpha reduced serum levels of alanine aminotransferase and aspartate aminotransferase, alleviated histological injury, and improved the survival of mice with FHF. In addition, both serum and hepatic tumor necrosis factor alpha expression were suppressed when HNF4alpha expression was inhibited in mice with FHF. CONCLUSION Inhibiting HNF4alpha expression protects mice from FHF induced by LPS/D-GalN, but the exact mechanism behind this needs further investigation.
Collapse
Affiliation(s)
- En-Qiang Chen
- Center for Infectious Diseases, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | | | |
Collapse
|