51
|
Wang J, Yan X, Wang T, Fang L. Potential effect of astragaloside IV on the lipopolysaccharide induced inflammation via the inactivation of NF-κB signaling pathway. Pharmacogn Mag 2021. [DOI: 10.4103/pm.pm_267_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
52
|
Protective Role of Coenzyme Q10 in Acute Sepsis-Induced Liver Injury in BALB/c Mice. BIOMED RESEARCH INTERNATIONAL 2021; 2020:7598375. [PMID: 33381582 PMCID: PMC7762638 DOI: 10.1155/2020/7598375] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 11/18/2022]
Abstract
Sepsis increases the risk of the liver injury development. According to the research works, coenzyme Q10 exhibits hepatoprotective properties in vivo as well as in vitro. Current work aimed at investigating the protective impacts of coenzyme Q10 against liver injury in septic BALB/c mice. The male BALB/c mice were randomly segregated into 4 groups: the control group, the coenzyme Q10 treatment group, the puncture and cecal ligation group, and the coenzyme Q10+cecal ligation and puncture group. Cecal ligation and puncture was conducted after gavagaging the mice with coenzyme Q10 during two weeks. Following 48 h postcecal ligation and puncture, we estimated hepatic biochemical parameters and histopathological changes in hepatic tissue. We evaluated the expression of factors associated with autophagy, pyroptosis, and inflammation. Findings indicated that coenzyme Q10 decreased the plasma levels in alkaline phosphatase, alanine aminotransferase, and aspartate aminotransferase in the cecal ligation and puncture group. Coenzyme Q10 significantly inhibited the elevation of sequestosome-1, interleukin-1β, oligomerization domain-like receptor 3 and nucleotide-binding, interleukin-6, and tumor necrosis factor-α expression levels; coenzyme Q10 also increased beclin 1 levels. Coenzyme Q10 might be a significant agent in the treatment of liver injury induced by sepsis.
Collapse
|
53
|
Zuo W, Tian R, Chen Q, Wang L, Gu Q, Zhao H, Huang C, Liu Y, Li J, Yang X, Xu L, Zhang B, Liu Z. miR-330-5p inhibits NLRP3 inflammasome-mediated myocardial ischaemia-reperfusion injury by targeting TIM3. Cardiovasc Drugs Ther 2020; 35:691-705. [PMID: 33137205 DOI: 10.1007/s10557-020-07104-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND/AIMS The Nod-like receptor protein-3 (NLRP3) inflammasome signalling pathway is involved in the inflammatory reaction of myocardial ischaemia-reperfusion (I/R) injury. Our previous study showed that miR-330-5p was differentially expressed in both cerebral and myocardial I/R injury, and thus might be a biomarker for I/R injury-related diseases. Another study also indicated that miR-330-5p could promote NLRP3 inflammasome activation in renal IRI. However, the role of miR-330-5p in myocardial I/R injury-induced inflammatory responses is unknown. This study aimed to investigate the role of miR-330-5p in NLRP3 inflammasome-mediated myocardial I/R injury. METHODS Myocardial I/R injury was induced in mice by occlusion of the left anterior descending coronary artery for 45 min followed by reperfusion. For NLRP3 inflammasome stimulation in vitro, cardiomyocytes were treated with 2 h of oxygen and glucose deprivation (OGD) or LPS (100 ng/ml). Myocardial miR-330-5p expression was examined by PCR at different treatment times. A miR-330-5p antagomir and an agomir were used to regulate miR-330-5p expression. To evaluate the role of miR-330-5p in myocardial I/R injury, 2,3,5-triphenyltetrazolium chloride (TTC) staining, echocardiography, and immunoblotting were used to assess infarct volume, cardiac function, and NLRP3 inflammasome activation respectively. A luciferase binding assay was used to examine whether miR-330-5p could directly bind to the T cell immunoglobulin domain and mucin domain-containing molecule-3 (TIM3). Finally, the role of the miR-330-5p/TIM3 axis in regulating apoptosis and NLRP3 inflammasome formation was evaluated with flow cytometry assays and immunofluorescence staining. RESULTS Compared to that in the model group, the inhibition of miR-330-5p significantly aggravated myocardial I/R injury, resulting in increased infarct volume and more severe cardiac dysfunction. Moreover, inhibition of miR-330-5p significantly increased the levels of NLRP3 inflammasome-related proteins, including caspase-1, IL-1β, IL-18 and TNF-α, in both in-vivo and in-vitro models. Furthermore, TIM3 was confirmed as a potential target of miR-330-5p. As predicted, suppression of TIM3 by siRNA ameliorated the anti-miR-330-5p-mediated activation of the NLRP3 inflammasome induced by OGD and LPS, thus decreasing cardiomyocyte apoptosis. CONCLUSIONS Our study indicated that the miR-330-5p/TIM3 axis was involved in the regulatory mechanism of NLRP3 inflammasome-mediated myocardial inflammation.
Collapse
Affiliation(s)
- Wei Zuo
- Department of Pharmacy, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Ran Tian
- Department of Cardiology, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Qian Chen
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Lun Wang
- Department of Cardiology, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Qing Gu
- Department of Cardiology, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Hongmei Zhao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Chunmei Huang
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Yingxian Liu
- Department of Cardiology, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Jingyi Li
- Department of Cardiology, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Xinglin Yang
- Department of Cardiology, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Lihong Xu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Beijing, China
| | - Bo Zhang
- Department of Pharmacy, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China.
| | - Zhenyu Liu
- Department of Cardiology, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
54
|
Batra N, De Souza C, Batra J, Raetz AG, Yu AM. The HMOX1 Pathway as a Promising Target for the Treatment and Prevention of SARS-CoV-2 of 2019 (COVID-19). Int J Mol Sci 2020; 21:E6412. [PMID: 32899231 PMCID: PMC7503392 DOI: 10.3390/ijms21176412] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/26/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
The coronavirus disease of 2019 (COVID-19) or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is a global pandemic with increasing incidence and mortality rates. Recent evidence based on the cytokine profiles of severe COVID-19 cases suggests an overstimulation of macrophages and monocytes associated with reduced T-cell abundance (lymphopenia) in patients infected with SARS-CoV-2. The SARS-CoV-2 open reading frame 3 a (ORF3a) protein was found to bind to the human HMOX1 protein at a high confidence through high-throughput screening experiments. The HMOX1 pathway can inhibit platelet aggregation, and can have anti-thrombotic and anti-inflammatory properties, amongst others, all of which are critical medical conditions observed in COVID-19 patients. Here, we review the potential of modulating the HMOX1-ORF3a nexus to regulate the innate immune response for therapeutic benefits in COVID-19 patients. We also review other potential treatment strategies and suggest novel synthetic and natural compounds that may have the potential for future development in clinic.
Collapse
Affiliation(s)
- Neelu Batra
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (N.B.); (C.D.S.); (A.G.R.)
| | - Cristabelle De Souza
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (N.B.); (C.D.S.); (A.G.R.)
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Jyoti Batra
- Gladstone Institute, San Francisco, CA 94158, USA;
| | - Alan G. Raetz
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (N.B.); (C.D.S.); (A.G.R.)
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (N.B.); (C.D.S.); (A.G.R.)
| |
Collapse
|
55
|
The expressions of NLRP1, NLRP3, and AIM2 inflammasome complexes in the contusive spinal cord injury rat model and their responses to hormonal therapy. Cell Tissue Res 2020; 381:397-410. [DOI: 10.1007/s00441-020-03250-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
|
56
|
Baek HS, Min HJ, Hong VS, Kwon TK, Park JW, Lee J, Kim S. Anti-Inflammatory Effects of the Novel PIM Kinase Inhibitor KMU-470 in RAW 264.7 Cells through the TLR4-NF-κB-NLRP3 Pathway. Int J Mol Sci 2020; 21:5138. [PMID: 32698512 PMCID: PMC7403980 DOI: 10.3390/ijms21145138] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
PIM kinases, a small family of serine/threonine kinases, are important intermediates in the cytokine signaling pathway of inflammatory disease. In this study, we investigated whether the novel PIM kinase inhibitor KMU-470, a derivative of indolin-2-one, inhibits lipopolysaccharide (LPS)-induced inflammatory responses in RAW 264.7 cells. We demonstrated that KMU-470 suppressed the production of nitric oxide and inducible nitric oxide synthases that are induced by LPS in RAW 264.7 cells. Furthermore, KMU-470 inhibited LPS-induced up-regulation of TLR4 and MyD88, as well as the phosphorylation of IκB kinase and NF-κB in RAW 264.7 cells. Additionally, KMU-470 suppressed LPS-induced up-regulation at the transcriptional level of various pro-inflammatory cytokines such as IL-1β, TNF-α, and IL-6. Notably, KMU-470 inhibited LPS-induced up-regulation of a major component of the inflammasome complex, NLRP3, in RAW 264.7 cells. Importantly, PIM-1 siRNA transfection attenuated up-regulation of NLRP3 and pro-IL-1β in LPS-treated RAW 264.7 cells. Taken together, these findings indicate that PIM-1 plays a key role in inflammatory signaling and that KMU-470 is a potential anti-inflammatory agent.
Collapse
Affiliation(s)
- Hye Suk Baek
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Korea; (H.S.B.); (H.J.M.); (T.K.K.); (J.W.P.)
| | - Hyeon Ji Min
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Korea; (H.S.B.); (H.J.M.); (T.K.K.); (J.W.P.)
| | | | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Korea; (H.S.B.); (H.J.M.); (T.K.K.); (J.W.P.)
| | - Jong Wook Park
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Korea; (H.S.B.); (H.J.M.); (T.K.K.); (J.W.P.)
| | - Jinho Lee
- Department of Chemistry, Keimyung University, Daegu 42601, Korea;
| | - Shin Kim
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Korea; (H.S.B.); (H.J.M.); (T.K.K.); (J.W.P.)
- Institute of Medical Science, Keimyung University, Daegu 42601, Korea
| |
Collapse
|
57
|
Sun Y, Xia Y, Liu X, Liu J, He W, Ye H, Yuan X. Dexmedetomidine alleviates LPS‑induced acute lung injury via regulation of the p38/HO‑1 pathway. Mol Med Rep 2020; 22:2442-2450. [PMID: 32705282 PMCID: PMC7411448 DOI: 10.3892/mmr.2020.11330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/27/2020] [Indexed: 12/11/2022] Open
Abstract
Acute lung injury (ALI) is a common critical illness in clinical anesthesia and the intensive care unit that can cause acute hypoxic respiratory insufficiency. Despite various therapeutic regimes having been investigated, there is currently no effective pharmacotherapy available to treat ALI. Previous studies have reported that the NOD-like receptor protein 3 (NLRP3) signaling pathway plays an important role in the inflammatory response and is involved in the pathogenesis of ALI. Moreover, dexmedetomidine (Dex), an α2-adrenergic receptor activating agent, has been routinely used as an adjuvant therapy in treating inflammatory diseases, including ALI. However, the precise pathological mechanisms of Dex in ALI remain to be elucidated. Thus, the present study aimed to investigate the effects of the p38/heme oxygenase 1 (HO-1) signaling pathways in the pathological mechanisms of Dex in ALI. Newborn male Sprague-Dawley rats (n=48) were randomly divided into four groups (n=12 each), and an intravenous injection of lipopolysaccharide (LPS) was used to successfully induce the ALI model, with increased pulmonary damage, cell apoptosis, interleukin-1β (IL-1β) secretion and edema fluid in lungs. Moreover, the mRNA and protein expression levels of NLRP3 were significantly upregulated, while that of HO-1 were downregulated by LPS treatment. Furthermore, the levels of phosphorylated p38 were also upregulated in ALI rats. It was demonstrated that Dex administration significantly alleviated LPS-induced ALI, downregulated the secretion of IL-1β, decreased the expression of NLRP3, inhibited the phospho-activation of p38 and increased HO-1 expression. In addition, pharmacological inhibition of p38 using the inhibitor SB20380 further enhanced the effect of Dex. Collectively, these preliminarily results identified the effects of Dex intervention on the pathogenesis of ALI via the regulation of p38/HO-1 signaling pathways, which impacted the inflammatory effects, thus providing a theoretical basis and novel evidence for the development of new targets for clinical treatment of ALI.
Collapse
Affiliation(s)
- Yingying Sun
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Yin Xia
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Xinghui Liu
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Junxia Liu
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Weitian He
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Hongwu Ye
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Xianren Yuan
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| |
Collapse
|
58
|
Cornelius DC, Travis OK, Tramel RW, Borges-Rodriguez M, Baik CH, Greer M, Giachelli CA, Tardo GA, Williams JM. NLRP3 inflammasome inhibition attenuates sepsis-induced platelet activation and prevents multi-organ injury in cecal-ligation puncture. PLoS One 2020; 15:e0234039. [PMID: 32555710 PMCID: PMC7299389 DOI: 10.1371/journal.pone.0234039] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/06/2020] [Indexed: 12/27/2022] Open
Abstract
Sepsis is characterized by organ dysfunction due to a dysregulated immune response to infection. Currently, no effective treatment for sepsis exists. Platelets are recognized as mediators of the immune response and may be a potential therapeutic target for the treatment of sepsis. We previously demonstrated that NLRP3 inflammasome activation in sepsis-induced activated platelets was associated with multi-organ injury in the cecal-ligation puncture (CLP) rat model of sepsis. In this study, we tested the hypothesis that inhibition of NLRP3 would inhibit platelet activation and attenuate multi-organ injury in the CLP rat. CLP (n = 10) or Sham (n = 10) surgery were performed in male and female Sprague-Dawley rats. A subset of CLP rats were treated with MCC950 (50mg/kg/d), a specific NLRP3 inhibitor (CLP+MCC950, n = 10). At 72 hrs. post-CLP, blood and organs were harvested for analysis of platelet activation, NLRP3 activation, inflammation and end organ damage. Platelet activation increased from 8±0.8% in Sham to 16±1% in CLP, and was reduced to 9±1% in CLP+M rats (p<0.05). NLRP3 activation was also increased in platelets of CLP vs Sham. NLRP3 expression was unchanged in kidney and lung after CLP, but Caspase 1 expression and IL-1β were increased. MCC950 treatment attenuated NLRP3 activation in platelets. Plasma, kidney, and lung levels of NLRP3 inflammasome associated cytokines, IL-1ß and IL-18, were significantly increased in CLP compared to Sham rats. Inhibition of NLRP3 normalized cytokine levels. Glomerular injury, pulmonary edema, and endothelial dysfunction markers were increased in CLP rats vs Sham. MCC950 treatment significantly decreased renal and pulmonary injury and endothelial dysfunction in CLP+M. Our results demonstrate a role for NLRP3 in contributing to platelet activation and multi-organ injury in sepsis.
Collapse
Affiliation(s)
- Denise C. Cornelius
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- * E-mail:
| | - Olivia K. Travis
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Robert W. Tramel
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Marivee Borges-Rodriguez
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Cedar H. Baik
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Mallory Greer
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Chelsea A. Giachelli
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Geilda A. Tardo
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Jan M. Williams
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| |
Collapse
|
59
|
Khodir AE, Samra YA, Said E. A novel role of nifuroxazide in attenuation of sepsis-associated acute lung and myocardial injuries; role of TLR4/NLPR3/IL-1β signaling interruption. Life Sci 2020; 256:117907. [PMID: 32504751 DOI: 10.1016/j.lfs.2020.117907] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/23/2020] [Accepted: 06/01/2020] [Indexed: 12/20/2022]
Abstract
Acute lung injury (ALI) and the subsequent multi-system organ failure is a serious health problem with devastating impacts on the health care systems. Indeed, the world has been facing an un-preceded situation in the past couple of months following COVID-19 infestation and the associated high-mortality rates mainly attributed to sepsis and the associated multiple organ failures of particular concern; acute respiratory distress syndrome post lung injury. The current study provides evidence on the ameliorative impact of nifuroxazide, and FDA approved antidiarrheal drug in attenuation of lipopolysaccharide (LPS)-induced ALI and myocarditis when administrated either in prophylactic or curative regimens. Nifuroxazide administration was associated with a significant improvement in lung and heart histopathological characteristics and architecture with retraction of LPS-induced inflammatory-infiltration. This was associated with retraction in serum biomarkers of cellular injury of which; LDH, CK-MB, and ALP. Nifuroxazide administration was associated with a significant improvement in both lung and heart oxidative status. Such positive outcomes were underlined by a significant inhibitory effect of nifuroxazide on lung and heart contents of toll-like receptor (4) (TLR4)/the inflammasome NALPR3/interleukin- 1β (IL-1β). In conclusion: Nifuroxazide attenuates LPS-induced ALI and myocardial injury via interruption of TLR4/NALPR3/IL-1β signaling. Thus it can offer a potential approach for attenuation of sepsis in critically ill patients.
Collapse
Affiliation(s)
- Ahmed E Khodir
- Dep. of Pharmacology and Biochemistry, Delta University for Science and Technology, International Coastal Road, Gamasa City, Mansoura, Dakahliya, Egypt
| | - Yara A Samra
- Dep. of Biochemistry, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt
| | - Eman Said
- Dep. of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt.
| |
Collapse
|
60
|
Human Umbilical Cord Mesenchymal Stromal Cells Attenuate Systemic Sepsis in Part by Enhancing Peritoneal Macrophage Bacterial Killing via Heme Oxygenase-1 Induction in Rats. Anesthesiology 2020; 132:140-154. [PMID: 31764154 DOI: 10.1097/aln.0000000000003018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Mesenchymal stromal cells have therapeutic potential in sepsis, but the mechanism of action is unclear. We tested the effects, dose-response, and mechanisms of action of cryopreserved, xenogeneic-free human umbilical cord mesenchymal stromal cells in a rat model of fecal peritonitis, and examined the role of heme oxygenase-1 in protection. METHODS Separate in vivo experiments evaluated mesenchymal stromal cells in fecal sepsis, established dose response (2, 5, and 10 million cells/kg), and the role of heme oxygenase-1 in mediating human umbilical cord-derived mesenchymal stromal/stem cell effects. Ex vivo studies utilized pharmacologic blockers and small inhibitory RNAs to evaluate mechanisms of mesenchymal stromal cell enhanced function in (rodent, healthy and septic human) macrophages. RESULTS Human umbilical cord mesenchymal stromal cells reduced injury and increased survival (from 48%, 12 of 25 to 88%, 14 of 16, P = 0.0033) in fecal sepsis, with dose response studies demonstrating that 10 million cells/kg was the most effective dose. Mesenchymal stromal cells reduced bacterial load and peritoneal leukocyte infiltration (from 9.9 ± 3.1 × 10/ml to 6.2 ± 1.8 × 10/ml, N = 8 to 10 per group, P < 0.0001), and increased heme oxygenase-1 expression in peritoneal macrophages, liver, and spleen. Heme oxygenase-1 blockade abolished the effects of mesenchymal stromal cells (N = 7 or 8 per group). Mesenchymal stromal cells also increased heme oxygenase-1 expression in macrophages from healthy donors and septic patients. Direct ex vivo upregulation of macrophage heme oxygenase-1 enhanced macrophage function (phagocytosis, reactive oxygen species production, bacterial killing). Blockade of lipoxin A4 production in mesenchymal stromal cells, and of prostaglandin E2 synthesis in mesenchymal stromal cell/macrophage cocultures, prevented upregulation of heme oxygenase-1 in macrophages (from 9.6 ± 5.5-fold to 2.3 ± 1.3 and 2.4 ± 2.3 respectively, P = 0.004). Knockdown of heme oxygenase-1 production in macrophages ablated mesenchymal stromal cell enhancement of macrophage phagocytosis. CONCLUSIONS Human umbilical cord mesenchymal stromal cells attenuate systemic sepsis by enhancing peritoneal macrophage bacterial killing, mediated partly via upregulation of peritoneal macrophage heme oxygenase-1. Lipoxin A4 and prostaglandin E2 play key roles in the mesenchymal stromal cell and macrophage interaction.
Collapse
|
61
|
Luo M, Meng J, Yan J, Shang F, Zhang T, Lv D, Li C, Yang X, Luo S. Role of the Nucleotide-Binding Domain-Like Receptor Protein 3 Inflammasome in the Endothelial Dysfunction of Early Sepsis. Inflammation 2020; 43:1561-1571. [DOI: 10.1007/s10753-020-01232-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
62
|
Abstract
There is abundant evidence that infectious sepsis both in humans and mice with polymicrobial sepsis results in robust activation of complement. Major complement activation products involved in sepsis include C5a anaphylatoxin and its receptors (C5aR1 and C5aR2) and, perhaps, the terminal complement activation product, C5b-9. These products (and others) also cause dysfunction of the innate immune system, with exaggerated early proinflammatory responses, followed by decline of the innate immune system, leading to immunosuppression and multiorgan dysfunction. Generation of C5a during sepsis also leads to activation of neutrophils and macrophages and ultimate appearance of extracellular histones, which have powerful proinflammatory and prothrombotic activities. The distal complement activation product, C5b-9, triggers intracellular Ca fluxes in epithelial and endothelial cells. Histones activate the NLRP3 inflammasome, products of which can damage cells. C5a also activates MAPKs and Akt signaling pathways in cardiomyocytes, causing buildup of [Ca]i, defective action potentials and substantial cell dysfunction, resulting in cardiac and other organ dysfunction. Cardiac dysfunction can be quantitated by ECHO-Doppler parameters. In vivo interventions that block these complement-dependent products responsible for organ dysfunction in sepsis reduce the intensity of sepsis. The obvious targets in sepsis are C5a and its receptors, histones, and perhaps the MAPK pathways. Blockade of C5 has been considered in sepsis, but the FDA-approved antibody (eculizumab) is known to compromise defenses against neisseria and pneumonococcal bacteria, and requires immunization before the mAb to C5 can be used clinically. Small molecular blocking agents for C5aRs are currently in development and may be therapeutically effective for treatment of sepsis.
Collapse
|
63
|
Zhang Y, Dong A, Xie K, Yu Y. Protective Effects of Hydrogen on Myocardial Mitochondrial Functions in Septic Mice. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1568209. [PMID: 32083123 PMCID: PMC7011313 DOI: 10.1155/2020/1568209] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/13/2020] [Indexed: 11/17/2022]
Abstract
Enhancement of mitochondrial physiological function prevents sepsis-induced dysfunction. The present study aimed to elucidate the mechanism by which hydrogen (H2) affects mitochondrial function in a wild-type (WT) and homozygous nuclear factor erythroid 2-related factor 2 (Nrf2) knockout (KO, Nrf2-/-) murine model of sepsis. In myocardial tissues with severe sepsis, H2 gas treatment reduced mitochondrial dysfunction, whereas zinc protoporphyrin (ZnPPIX) negated these beneficial effects. H2 treatment upregulated the protein expression of mitofusin-2 (Mfn2), peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), and protein heme oxygenase-1 (HO-1) in WT mice with severe sepsis but not in their Nrf2-/- counterparts, and this upregulation was inhibited in the presence of ZnPPIX. In conclusion, the mechanism by which H2 limits organ damage in mice with severe sepsis involves HO-1, whereas the mechanism that limits severe sepsis-related mitochondrial dysfunction involves both HO-1 and Nrf2.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Aili Dong
- Department of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Keliang Xie
- Department of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Yonghao Yu
- Department of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
64
|
Danielski LG, Giustina AD, Bonfante S, Barichello T, Petronilho F. The NLRP3 Inflammasome and Its Role in Sepsis Development. Inflammation 2019; 43:24-31. [DOI: 10.1007/s10753-019-01124-9] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
65
|
Hemolysis Derived Products Toxicity and Endothelium: Model of the Second Hit. Toxins (Basel) 2019; 11:toxins11110660. [PMID: 31766155 PMCID: PMC6891750 DOI: 10.3390/toxins11110660] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/02/2019] [Accepted: 11/06/2019] [Indexed: 12/16/2022] Open
Abstract
Vascular diseases are multifactorial, often requiring multiple challenges, or ‘hits’, for their initiation. Intra-vascular hemolysis illustrates well the multiple-hit theory where a first event lyses red blood cells, releasing hemolysis-derived products, in particular cell-free heme which is highly toxic for the endothelium. Physiologically, hemolysis derived-products are rapidly neutralized by numerous defense systems, including haptoglobin and hemopexin which scavenge hemoglobin and heme, respectively. Likewise, cellular defense mechanisms are involved, including heme-oxygenase 1 upregulation which metabolizes heme. However, in cases of intra-vascular hemolysis, those systems are overwhelmed. Heme exerts toxic effects by acting as a damage-associated molecular pattern and promoting, together with hemoglobin, nitric oxide scavenging and ROS production. In addition, it activates the complement and the coagulation systems. Together, these processes lead to endothelial cell injury which triggers pro-thrombotic and pro-inflammatory phenotypes. Moreover, among endothelial cells, glomerular ones display a particular susceptibility explained by a weaker capacity to counteract hemolysis injury. In this review, we illustrate the ‘multiple-hit’ theory through the example of intra-vascular hemolysis, with a particular focus on cell-free heme, and we advance hypotheses explaining the glomerular susceptibility observed in hemolytic diseases. Finally, we describe therapeutic options for reducing endothelial injury in hemolytic diseases.
Collapse
|
66
|
Heme oxygenase-1/carbon monoxide as modulators of autophagy and inflammation. Arch Biochem Biophys 2019; 678:108186. [PMID: 31704095 DOI: 10.1016/j.abb.2019.108186] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 10/10/2019] [Accepted: 11/04/2019] [Indexed: 12/29/2022]
Abstract
Heme oxygenase-1 (HO-1) catalyzes heme degradation to generate biliverdin-IXα, carbon monoxide (CO), and iron. The HO-1/CO system confers cytoprotection in animal models of organ injury and disease, via modulation of inflammation and apoptosis. Recent studies have uncovered novel anti-inflammatory targets of HO-1/CO including regulation of the autophagy and inflammasome pathways. Autophagy is a lysosome-dependent program for the turnover of cellular organelles such as mitochondria, proteins, and pathogens; which may downregulate inflammatory processes. Therapeutic modulation of autophagy by CO has been demonstrated in models of sepsis. The nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome regulates the maturation of pro-inflammatory cytokines. CO can regulate NLRP3 inflammasome activation and associated pro-inflammatory cytokines production and promote the resolution of inflammation by upregulating the synthesis of specialized pro-resolving mediators (SPMs). Mitochondria may represent a proximal target of HO-1/CO action. HO-1 may localize to mitochondria in response to stress, while CO can moderate mitochondrial dysfunction and regulate mitochondrial autophagy (mitophagy) and biogenesis. The interplay between mitochondrial autophagy, mitochondrial dysfunction, and the regulation and resolution of inflammation may make important contributions to the protection afforded by HO-1/CO in cellular and organ injury models. Recent studies have continued to explore the potential of CO for clinical applications.
Collapse
|
67
|
Vitali SH, Fernandez-Gonzalez A, Nadkarni J, Kwong A, Rose C, Mitsialis SA, Kourembanas S. Heme oxygenase-1 dampens the macrophage sterile inflammasome response and regulates its components in the hypoxic lung. Am J Physiol Lung Cell Mol Physiol 2019; 318:L125-L134. [PMID: 31664855 DOI: 10.1152/ajplung.00074.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Exposure to hypoxia causes an inflammatory reaction in the mouse lung, and this response can be modulated by overexpressing the hypoxia-inducible stress-response enzyme, heme oxygenase-1 (HO-1). We hypothesized that the inflammasome activity may be a central pathway by which HO-1 controls pulmonary inflammation following alveolar hypoxia. Therefore, we investigated whether HO-1 controls inflammasome activation by altering its expression in macrophages primed with classic NOD-like receptor containing a pyrin domain 3 (NLRP3) inducers, and in murine lungs lacking HO-1 and exposed to acute hypoxia. We found that lack of HO-1 activated lipopolysaccharide (LPS) and ATP-treated bone marrow-derived macrophages, causing an increase in secreted levels of cleaved interleukin (IL)-1B, IL-18, and caspase-1, markers of increased inflammasome activity, whereas HO-1 overexpression suppressed IL-1B, NLRP3, and IL-18. The production of cleaved IL-1B and the activation of caspase-1 in LPS- and ATP-primed macrophages were inhibited by hemin, an HO-1 inducer, and two HO-1 enzymatic products [bilirubin and carbon monoxide (CO)]. Exposure of mice to hypoxia induced the expression of several inflammasome mRNA components (IL-1B, Nlrp3, and caspase-1), and this was further augmented by HO-1 deficiency. This pronounced inflammasome activation was detected as increased protein levels of apoptosis-associated speck-like protein containing a COOH-terminal caspase recruitment domain, IL-18, procaspase-1, and cleaved caspase-1 in the lungs of hypoxic mice. Systemically, Hmox1-deficient mice showed increased basal levels of IL-18 that were further increased after 48 h of hypoxic exposure. Taken together, these finding point to a pivotal role for HO-1 in the control of baseline and hypoxic inflammasome signaling, perhaps through the antioxidant properties of bilirubin and CO's pleiotropic effects.
Collapse
Affiliation(s)
- Sally H Vitali
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts.,Division of Critical Care Medicine, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, Massachusetts.,Department of Anesthesia, Harvard Medical School, Boston, Massachusetts
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Janhavi Nadkarni
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts.,Division of Critical Care Medicine, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, Massachusetts
| | - April Kwong
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts.,Division of Critical Care Medicine, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, Massachusetts
| | - Chase Rose
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts.,Division of Critical Care Medicine, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, Massachusetts
| | - S Alex Mitsialis
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Stella Kourembanas
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
68
|
Lian M, Xue Z, Qiao X, Liu C, Zhang S, Li X, Huang C, Song Q, Yang W, Chen X, Wang T. Movable Hollow Nanoparticles as Reactive Oxygen Scavengers. Chem 2019. [DOI: 10.1016/j.chempr.2019.05.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
69
|
|
70
|
Karimi A, Ghodsi R, Kooshki F, Karimi M, Asghariazar V, Tarighat-Esfanjani A. Therapeutic effects of curcumin on sepsis and mechanisms of action: A systematic review of preclinical studies. Phytother Res 2019; 33:2798-2820. [PMID: 31429161 DOI: 10.1002/ptr.6467] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/28/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022]
Abstract
Sepsis is a complex disease that begins with an infectious disorder and causes excessive immune responses. Curcumin is considered as an active component of turmeric that can improve the condition in sepsis due to its anti-inflammatory and antioxidant properties. PubMed, Embase, Google Scholar, Web of Science, and Scopus databases were searched. Searching was not limited to a specific publication period. Only English-language original articles, which had examined the effect of curcumin on sepsis, were included. At first, 1,098 articles were totally found, and 209 articles were selected after excluding duplicated data; 46 articles were remained due to the curcumin effects on sepsis. These included 23 in vitro studies and 23 animal studies. Our results showed that curcumin and various analogs of curcumin can have an inhibitory effect on sepsis-induced complications. Curcumin has the ability to inhibit the inflammatory, oxidative coagulation factors, and regulation of immune responses in sepsis. Despite the promising evidence of the therapeutic effects of curcumin on the sepsis complication, further studies seem necessary to investigate its effect and possible mechanisms of action in human studies.
Collapse
Affiliation(s)
- Arash Karimi
- Student Research Committee, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Ghodsi
- Student Research Committee, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fateme Kooshki
- Student Research Committee, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mozhdeh Karimi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Asghariazar
- Student Research Committee, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Tarighat-Esfanjani
- Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
71
|
Galangin Suppresses Renal Inflammation via the Inhibition of NF- κB, PI3K/AKT and NLRP3 in Uric Acid Treated NRK-52E Tubular Epithelial Cells. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3018357. [PMID: 31240210 PMCID: PMC6556363 DOI: 10.1155/2019/3018357] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/22/2019] [Accepted: 04/16/2019] [Indexed: 01/10/2023]
Abstract
Renal inflammation can result in renal injury. Uric acid (UA) is the final product of purine metabolism in humans and because of the lack of urate oxidase, UA may accumulate in tissues, including kidney, causing inflammation. Galangin was isolated from a traditional Chinese medicine plant and possesses several beneficial effects, working as an anti-oxidant, anti-mutagenic, anti-tumor, anti-inflammatory, anti-microbial, and anti-viral agent. Therefore, this study aimed at investigating the molecular mechanism of galangin in the attenuation of UA induced renal inflammation in normal rat kidney epithelial cells NRK-52E. Our findings suggested that galangin treatment efficiently protected NRK-52E cells against UA induced renal inflammation by decreasing tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-18, prostaglandin E2 (PGE2), and nitric oxide (NO) release, and it inhibited nitric oxide synthase (iNOS), prostaglandin endoperoxide synthase 2 (PTGS2), TNF-α, IL-1β, and IL-18 mRNA expression. In addition, galangin was not exerting any cytotoxicity at the concentrations that were effective against inflammation as assessed by CCK8 assay. Moreover, western blotting showed that galangin treatment effectively inhibited nuclear factor-kappa B (NF-κB), phosphatidylinositol 3 kinase (PI3K)/protein kinase B (AKT) and nucleotide-binding domain- (NOD-) like receptor protein 3 (NLRP3) signaling pathway activation. Taken together, these findings suggested that galangin plays a pivotal role in renal inflammation by suppressing inflammatory responses, which might be closely associated with the inhibition of NLRP3 inflammasome, NF-κB and PI3K/AKT signaling pathway activation.
Collapse
|
72
|
Chen H, Ding Y, Chen W, Feng Y, Shi G. Glibenclamide alleviates inflammation in oleic acid model of acute lung injury through NLRP3 inflammasome signaling pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1545-1554. [PMID: 31123394 PMCID: PMC6511253 DOI: 10.2147/dddt.s196040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/02/2019] [Indexed: 12/31/2022]
Abstract
Background: Pulmonary fat embolism (PFE) is one of the important causes of acute lung injury (ALI), but its pathogenesis is unclear. In recent years, it has been found that the NLRP3 inflammasome is closely related to inflammatory response. However, there are no reports about the involvement of NLRP3 in PFE- associated ALI. Glibenclamide is a kind of hypoglycaemic drug with anti-inflammatory effect. It has been reported to have the anti-inflammatory effect related to inhibiting NLRP3. Objective: To determine whether NLRP3 inflammasome was involved in ALI induced by PFE or whether glibenclamide had therapeutic effects on such lung injury, we designed this experiment. Materials and methods: The rat model of intravenous injection of oleic acid (OA) was used to simulate PFE. Rats were divided into three groups: control, OA and glibenclamide treatment group. Blood free fatty acid (FFA) concentration was determined by ACS-ACOD. Histopathological examinations were taken to assess the severity of lung injury. The expression of NLRP3 pathway and its downstream products were analyzed by IHC, WB, qPCR and ELISA. Results: Four hours after intravenous OA injection, the typical pathological manifestations of ALI accompanied by elevated levels of plasma FFAs were found. The activity of NLRP3 inflammasomes increased in OA group, too. Pretreatment with glibenclamide partly inhibited the increase in NLRP3, caspase-1 and IL-1β expression induced by OA, simultaneously attenuated the lung injury. But it has little effect on the expression of Toll-like receptor 4 (TLR4) expression in this experiment. Conclusion: NLRP3 inflammasome, one of the main components of innate immune response, involved in ALI induced by OA. Glibenclamide can alleviate this kind of ALI by inhibiting rather the NLRP3/caspase-1/IL-1β signaling pathway than the levels of FFAs or TLR4 pathway.
Collapse
Affiliation(s)
- Hong Chen
- Department of Respiration and Critical Care Disease, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China
| | - Yongjie Ding
- Department of Respiration and Critical Care Disease, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China
| | - Wei Chen
- Department of Respiration and Critical Care Disease, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China
| | - Yun Feng
- Department of Respiration and Critical Care Disease, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China
| | - Guochao Shi
- Department of Respiration and Critical Care Disease, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China
| |
Collapse
|
73
|
Cornelius DC, Baik CH, Travis OK, White DL, Young CM, Austin Pierce W, Shields CA, Poudel B, Williams JM. NLRP3 inflammasome activation in platelets in response to sepsis. Physiol Rep 2019; 7:e14073. [PMID: 31054188 PMCID: PMC6499866 DOI: 10.14814/phy2.14073] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 01/20/2023] Open
Abstract
Sepsis is a complex syndrome characterized by organ dysfunction and a dysregulated immune host response to infection. There is currently no effective treatment for sepsis, but platelets have been proposed as a potential therapeutic target for the treatment of sepsis. We hypothesized that the NLRP3 inflammasome is activated in platelets during sepsis and may be associated with multiorgan injury in response to polymicrobial sepsis. Polymicrobial sepsis was induced by cecal ligation and puncture (CLP) in 12- to 13-week-old male Sprague-Dawley rats. The necrotic cecum was removed at 24 h post-CLP. At 72 h post-CLP, activated platelets were significantly increased in CLP versus Sham rats. Colocalization of NLRP3 inflammasome components was observed in platelets from CLP rats at 72 h post-CLP. Plasma, pulmonary, and renal levels of IL-1β and IL-18 were significantly higher in CLP rats compared to Sham controls. Soluble markers of endothelial permeability were increased in CLP versus Sham. Renal and pulmonary histopathology were markedly elevated in CLP rats compared to Sham controls. NLRP3 is activated in platelets in response to CLP and is associated with inflammation, endothelial permeability and multiorgan injury. Our results indicate that activated platelets may play a role to cause multiorgan injury in sepsis and may have therapeutic potential for the treatment of sepsis multiorgan injury.
Collapse
Affiliation(s)
- Denise C. Cornelius
- Department of Emergency MedicineUniversity of Mississippi Medical CenterJacksonMississippi
- Department of PharmacologyUniversity of Mississippi Medical CenterJacksonMississippi
- Cardiovascular Renal‐Research CenterUniversity of Mississippi Medical CenterJacksonMississippi
| | - Cedar H. Baik
- Department of Emergency MedicineUniversity of Mississippi Medical CenterJacksonMississippi
| | - Olivia K. Travis
- Department of PharmacologyUniversity of Mississippi Medical CenterJacksonMississippi
| | - Dakota L. White
- Department of Emergency MedicineUniversity of Mississippi Medical CenterJacksonMississippi
| | - Cassandra M. Young
- Department of PharmacologyUniversity of Mississippi Medical CenterJacksonMississippi
| | - W. Austin Pierce
- Department of Emergency MedicineUniversity of Mississippi Medical CenterJacksonMississippi
| | - Corbin A. Shields
- Department of PharmacologyUniversity of Mississippi Medical CenterJacksonMississippi
| | - Bibek Poudel
- Department of PharmacologyUniversity of Mississippi Medical CenterJacksonMississippi
| | - Jan M. Williams
- Department of PharmacologyUniversity of Mississippi Medical CenterJacksonMississippi
- Cardiovascular Renal‐Research CenterUniversity of Mississippi Medical CenterJacksonMississippi
| |
Collapse
|
74
|
He DK, Chen JF, Shao YR, Zhou FQ, Shen J. Adenovirus-delivered angiopoietin-1 ameliorates phosgene-induced acute lung injury via inhibition of NLRP3 inflammasome activation. Inhal Toxicol 2019; 30:187-194. [PMID: 30257148 DOI: 10.1080/08958378.2018.1492648] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Angiopoietin-1 (Ang1) is reported to have the ability to attenuate endothelial permeability and inflammation during the stress condition and is considered to play a critical role in vascular stabilization. The aim of this study was to investigate the mechanisms involved in the protective effects of adenovirus-delivered Ang1 in phosgene-induced acute lung injury (ALI). METHODS ALI was induced in rats by phosgene exposure at 8.33 g/m3 for 5 min, followed by an intravenous injection of adenovirus-Ang1 (Ad/Ang1). The histologic changes of the lung were evaluated with H&E staining. The levels of cytokines in the serum and bronchoalveolar lavage fluid (BALF) were determined by ELISA. NLRP3 inflammasome activation was assessed with immunohistochemistry, RT-PCR, Western blotting and TUNEL staining. RESULTS Histologic analyses suggested that reduced severity in phosgene-induced ALI with Ad/Ang1 treatment. Reduced levels of IL-1β, IL-18 and IL-33 were found in both serum and BALF samples from Ad/Ang1-treated ALI rats induced by phosgene. Moreover, immunohistochemistry analysis revealed that Ad/Ang1 treatment inhibited the NLRP3 inflammasome activation. Decreased mRNA and protein levels of NLRP3 and caspase-1 were found in phosgene-exposed rats treated with Ad/Ang1. In addition, TUNEL staining indicated a decrease in pyroptosis in phosgene-exposed rats treated with Ad/Ang1. CONCLUSIONS Ang1 exerts beneficial effects on phosgene-induced lung injury via inhibition of NLRP3 inflammasome activation. Disruption of NLRP3 inflammasome activation might be served as therapeutic modality for the treatment of phosgene-induced ALI.
Collapse
Affiliation(s)
- Dai-Kun He
- a Center of Emergency & Intensive Care Unit, Jinshan Hospital , Fudan University , Shanghai , China.,b Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital , Fudan University , Shanghai , China.,c Medical Research Centre for Chemical Injury, Emergency and Critical Care, Jinshan Hospital , Fudan University , Shanghai , China
| | - Jun-Feng Chen
- a Center of Emergency & Intensive Care Unit, Jinshan Hospital , Fudan University , Shanghai , China.,b Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital , Fudan University , Shanghai , China.,c Medical Research Centre for Chemical Injury, Emergency and Critical Care, Jinshan Hospital , Fudan University , Shanghai , China
| | - Yi-Ru Shao
- a Center of Emergency & Intensive Care Unit, Jinshan Hospital , Fudan University , Shanghai , China.,b Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital , Fudan University , Shanghai , China.,c Medical Research Centre for Chemical Injury, Emergency and Critical Care, Jinshan Hospital , Fudan University , Shanghai , China
| | - Fang-Qing Zhou
- a Center of Emergency & Intensive Care Unit, Jinshan Hospital , Fudan University , Shanghai , China.,b Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital , Fudan University , Shanghai , China.,c Medical Research Centre for Chemical Injury, Emergency and Critical Care, Jinshan Hospital , Fudan University , Shanghai , China
| | - Jie Shen
- a Center of Emergency & Intensive Care Unit, Jinshan Hospital , Fudan University , Shanghai , China.,b Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital , Fudan University , Shanghai , China.,c Medical Research Centre for Chemical Injury, Emergency and Critical Care, Jinshan Hospital , Fudan University , Shanghai , China
| |
Collapse
|
75
|
Protective effect of oxytocin on LPS-induced acute lung injury in mice. Sci Rep 2019; 9:2836. [PMID: 30808956 PMCID: PMC6391417 DOI: 10.1038/s41598-019-39349-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 01/21/2019] [Indexed: 12/16/2022] Open
Abstract
Oxytocin (OT), a neurohypophyseal hormone synthesized in the paraventricular and supraoptic nuclei of the hypothalamus, has been reported to have an anti- inflammatory effect. However, its role in acute lung injury (ALI) has never been investigated. The aim of this study was to explore the therapeutic effects and potential mechanism action of OT on lipopolysaccharide (LPS)-induced ALI. Mice were treated with OT 30 min before the intraperitoneal injection of LPS. After 2 h, the effects of OT on lung histopathological changes, lung wet/dry (W/D) ratio, myeloperoxidase (MPO) activity, levels of inflammatory cytokines in the bronchoalveolar lavage fluid (BALF), and expression of inflammation proteins were detected. The results showed that OT significantly reduced LPS-induced pathological injury, W/D ratio, MPO activity, and the levels of interleukin (IL)-1β, IL-18 and IL-6. Further, OT also inhibited LPS-induced Toll-like receptor 4 expression and NLR family pyrin domain containing 3 inflammasome activation. OT receptor antagonist (L-368,899) was given 90 min before injecting OT to further demonstrate the role of OT in LPS-induced ALI. The results showed OT could not alleviate the aforementioned inflammatory reactions after administering L-368,899. In conclusion, the present results indicated that OT could reduce inflammatory responses of LPS-induced ALI.
Collapse
|
76
|
Yang Y, Yang F, Yu X, Wang B, Yang Y, Zhou X, Cheng R, Xia S, Zhou X. miR-16 inhibits NLRP3 inflammasome activation by directly targeting TLR4 in acute lung injury. Biomed Pharmacother 2019; 112:108664. [PMID: 30784935 DOI: 10.1016/j.biopha.2019.108664] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/03/2019] [Accepted: 02/04/2019] [Indexed: 12/12/2022] Open
Abstract
Acute lung injury (ALI) is the leading cause of human death, and it is widely accepted that the runaway inflammation is an important risk for the development of ALI. In the present study, we aimed to investigate the effect of miR-16 on lipopolysaccharide-induced acute lung injury in mice, especially focusing on Toll-like receptor 4 (TLR4) and NF-kB signaling pathway as well as NOD-like receptor protein 3 (NLRP3) inflammasome activation. We established in vivo and in vitro model of ALI using LPS and demonstrated that miR-16 expression was down-regulated in lung tissue as well as A549 cells after 8 h of LPS treatment. Furthermore, when miR-16 levels in lung tissues were up-regulated by miR-16 agomir, it was confirmed that the mRNA and protein levels of NF-κB, NLRP3 inflammasome, and inflammatory factors were decreased by the miR-16 by directly targeting TLR4. We also treated A549 cells with miR-16 mimics and anti-miR-16 to confirm the results. Overall, our experiments showed that miR-16 protects against acute lung injury in mice by regulating the TLR4/ NF-κB pathway and attenuating inflammatory response. This work suggests a potential novel therapeutic approach to combat ALI.
Collapse
Affiliation(s)
- Yuan Yang
- Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Feng Yang
- Department of Pediatrics, The Central Hospital of Enshi Autonomous Prefecture, Enshi City 445000, Hubei Province, China
| | - Xinqiao Yu
- Department of Pediatrics, The Central Hospital of Enshi Autonomous Prefecture, Enshi City 445000, Hubei Province, China
| | - Beibei Wang
- Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Yang Yang
- Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Xiaoyu Zhou
- Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Rui Cheng
- Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Shiwen Xia
- Department of Neonatology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, Hubei Province, China.
| | - Xiaoguang Zhou
- Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China.
| |
Collapse
|
77
|
The protective effect of epigallocatechin 3-gallate on mouse pancreatic islets via the Nrf2 pathway. Surg Today 2019; 49:536-545. [PMID: 30730004 DOI: 10.1007/s00595-019-1761-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/20/2018] [Indexed: 12/30/2022]
Abstract
PURPOSE Epigallocatechin 3-gallate (EGCG), a green tea polyphenol, has been shown to have anti-oxidant and anti-inflammatory effects in vitro and in vivo. The aim of this study was to investigate the effects and mechanism of EGCG on isolated pancreatic islets as pre-conditioning for pancreatic islet transplantation. METHODS The pancreatic islets were divided into two groups: an islet culture medium group (control) and an islet culture medium with EGCG (100 µM) group. We investigated the islet viability, Nrf2 expression, reactive oxygen species (ROS) production, and heme oxygenase-1 (HO-1) mRNA. Five hundred islet equivalents after 12 h of culture for the EGCG 100 µM and control group were transplanted under the kidney capsule of streptozotocin-induced diabetic ICR mice. RESULTS The cell viability and insulin secretion ability in the EGCG group were preserved, and the nuclear translocation of Nrf2 was increased in the EGCG group (p < 0.01). While the HO-1 mRNA levels were also higher in the EGCG group than in the control group (p < 0.05), the ROS production was lower (p < 0.01). An in vivo functional assessment showed that the blood glucose level had decreased in the EGCG group after transplantation (p < 0.01). CONCLUSION EGCG protects the viability and function of islets by suppressing ROS production via the Nrf2 pathway.
Collapse
|
78
|
Wang J, Gong S, Wang F, Niu M, Wei G, He Z, Gu T, Jiang Y, Liu A, Chen P. Granisetron protects polymicrobial sepsis-induced acute lung injury in mice. Biochem Biophys Res Commun 2019; 508:1004-1010. [DOI: 10.1016/j.bbrc.2018.12.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 12/05/2018] [Indexed: 01/08/2023]
|
79
|
Jiang L, Jiang Q, Yang S, Huang S, Han X, Duan J, Pan S, Zhao M, Guo S. GYY4137 attenuates LPS-induced acute lung injury via heme oxygenase-1 modulation. Pulm Pharmacol Ther 2018; 54:77-86. [PMID: 30605726 DOI: 10.1016/j.pupt.2018.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 12/16/2018] [Accepted: 12/30/2018] [Indexed: 12/16/2022]
Abstract
GYY4137, a slow-releasing hydrogen sulfide (H2S) donor, has been reported to exert anti-inflammatory activity and protect against sepsis. Heme oxygenase-1 (HO-1) is an important anti-inflammatory heat shock protein and plays a similar effect on sepsis. This study investigated the role of GYY4137 in acute lung injury (ALI) via HO-1 regulation. Lung injury was assessed in mice challenged with intratracheal lipopolysaccharide (LPS) and the mechanism of anti-inflammatory effects of GYY4137 was investigated in mice and RAW264.7 cells. GYY4137 reduced the LPS-mediated pulmonary injury and neutrophil infiltration, and inhibited the LPS-induced production of proinflammatory cytokines, inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expression. Moreover, GYY4137 suppressed the LPS-evoked NF-κB activation in RAW264.7 cells. GYY4137, not time-expired GYY4137 significantly induced HO-1 expression compared with the LPS group. The beneficial effects of GYY4137 above were reversed by the HO-1 inhibitor tin protoporphyrin (SnPP). These results suggest an anti-inflammatory effect and a therapeutic role of GYY4137 in LPS-induced ALI via HO-1 regulation.
Collapse
Affiliation(s)
- Lei Jiang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Jiang
- School of Statistics, Beijing Normal University, Beijing, China
| | - Songlin Yang
- Department of ICU, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shicong Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoli Han
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Duan
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shangha Pan
- The Key Hepatosplenic Surgery Laboratory, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingyan Zhao
- Department of ICU, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Shuliang Guo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
80
|
Sun X, Wang W, Dai J, Huang J, Shi M, Chu X, Wang F, Guo C, Wang C, Pang L, Wang Y. Donor heart preservation with a novel long-term and slow-releasing hydrogen sulfide system. Nitric Oxide 2018; 81:1-10. [DOI: 10.1016/j.niox.2018.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/08/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022]
|
81
|
Xu G, Shi D, Zhi Z, Ao R, Yu B. Melatonin ameliorates spinal cord injury by suppressing the activation of inflammasomes in rats. J Cell Biochem 2018; 120:5183-5192. [PMID: 30257055 DOI: 10.1002/jcb.27794] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 09/10/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Guanghui Xu
- Orthopedics Department Shanghai Pudong Hospital, Pudong Medical Center, Fudan University Pudong Shanghai China
| | - Dong Shi
- Radiology Department The 251st Hospital of Chinese PLA Zhangjiakou China
| | - Zhongzheng Zhi
- Orthopedics Department Shanghai Pudong Hospital, Pudong Medical Center, Fudan University Pudong Shanghai China
| | - Rongguang Ao
- Orthopedics Department Shanghai Pudong Hospital, Pudong Medical Center, Fudan University Pudong Shanghai China
| | - Baoqing Yu
- Orthopedics Department Shanghai Pudong Hospital, Pudong Medical Center, Fudan University Pudong Shanghai China
| |
Collapse
|
82
|
Abstract
The purpose of this study was to investigate the protective effects of Saikosaponin a (SSa), a triterpene saponin derived from Radix bupleuri, on lipopolysaccharide (LPS)-induced acute lung injury (ALI) using a murine model. The mice were given SSa 1 h after intranasal instillation of LPS. Then, lung histopathological examination, the wet/dry (W/D) ratio, myeloperoxidase (MPO), and inflammatory cytokines in bronchoalveolar lavage fluid (BALF) were detected in this study. The results showed that SSa reduced lung pathological injury induced by LPS. Furthermore, LPS-induced lung W/D ratio, MPO activity, and inflammatory cytokines TNF-α and IL-1β in BALF were significantly inhibited by SSa. In addition, SSa suppressed LPS-induced NF-κB activation and NLRP3 inflammasome expression. In conclusion, we found that SSa played a critical anti-inflammatory effect through inhibition of NF-κB and NLRP3 signaling pathways and protected against LPS-induced ALI.
Collapse
Affiliation(s)
- Zhi-An Du
- Department of Intensive Care Units, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Mei-Na Sun
- Department of Intensive Care Units, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Zhan-Sheng Hu
- Department of Intensive Care Units, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China.
| |
Collapse
|
83
|
Abstract
Initially described as an interferon (IFN)γ‐inducing factor, interleukin (IL)‐18 is indeed involved in Th1 and NK cell activation, but also in Th2, IL‐17‐producing γδ T cells and macrophage activation. IL‐18, a member of the IL‐1 family, is similar to IL‐1β for being processed by caspase 1 to an 18 kDa‐biologically active mature form. IL‐18 binds to its specific receptor (IL‐18Rα, also known as IL‐1R7) forming a low affinity ligand chain. This is followed by recruitment of the IL‐18Rβ chain. IL‐18 then uses the same signaling pathway as IL‐1 to activate NF‐kB and induce inflammatory mediators such as adhesion molecules, chemokines and Fas ligand. IL‐18 also binds to the circulating high affinity IL‐18 binding protein (BP), such as only unbound free IL‐18 is active. IL‐18Rα may also bind IL‐37, another member of the IL‐1 family, but in association with the negative signaling chain termed IL‐1R8, which transduces an anti‐inflammatory signal. IL‐18BP also binds IL‐37 and this acts as a sink for the anti‐inflammatory properties of IL‐37. There is now ample evidence for a role of IL‐18 in various infectious, metabolic or inflammatory diseases such as influenza virus infection, atheroma, myocardial infarction, chronic obstructive pulmonary disease, or Crohn's disease. However, IL‐18 plays a very specific role in the pathogenesis of hemophagocytic syndromes (HS) also termed Macrophage Activation Syndrome. In children affected by NLRC4 gain‐of‐function mutations, IL‐18 circulates in the range of tens of nanograms/mL. HS is treated with the IL‐1 Receptor antagonist (anakinra) but also specifically with IL‐18BP. Systemic juvenile idiopathic arthritis or adult‐onset Still's disease are also characterized by high serum IL‐18 concentrations and are treated by IL‐18BP.
Collapse
Affiliation(s)
- Gilles Kaplanski
- Assistance Publique-Hôpitaux de Marseille, Centre Hospitalier Universitaire Conception, Service de Médecine Interne et Immunologie Clinique, Aix-Marseille Université, Marseille, France.,Vascular Research Center Marseille, Faculté de Pharmacie, Aix-Marseille Université, INSERM UMR_S1076, Marseille, France
| |
Collapse
|
84
|
Yanling Q, Xiaoning C, Fei B, Liyun F, Huizhong H, Daqing S. Inhibition of NLRP9b attenuates acute lung injury through suppressing inflammation, apoptosis and oxidative stress in murine and cell models. Biochem Biophys Res Commun 2018; 503:436-443. [PMID: 29655793 DOI: 10.1016/j.bbrc.2018.04.079] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 04/09/2018] [Indexed: 02/07/2023]
Abstract
Acute lung injury (ALI), known a severe disease along with high morbidity and mortality, is lacking of specific therapies. Inflammation, apoptosis and oxidative stress are critical pathologies that contribute to ALI. Recently, there is study indicated that NLRP9b, a NOD-like receptor (NLR) member, is critical in modulation of inflammatory response. However, the effects of NLRP9b on sepsis-associated ALI, and the underlying molecular mechanism have not been understood. In the present study, the wild type (WT) and NLRP9b-knockout (NLRP9b-/-) mice with C57B/L6 background were subjected to a cecal ligation and puncture (CLP) for ALI murine model establishment. The findings indicated that NLRP9b-/- improved the survival rate of CLP-induced ALI mice, and inhibited pulmonary histopathological alterations, inflammation, and apoptosis. NLRP9b-/- reduced the activation of inhibitor of κBα/nuclear factor kappa B (IκBα/NF-κB), apoptosis-associated speck-like protein containing a Caspase-recruitment domain (ASC)/Casapse-1 and Caspase-3/poly (ADP-ribose) polymerase (PARP) signaling pathways in CLP-challenged mice with ALI. In vitro, mouse epithelial cells (MLE-12) were incubated with lipopolysaccharide (LPS) or recombinant NLRP9b caused a significant increased of pro-inflammatory cytokines or chemokine, and reactive oxygen species (ROS) generation; however, these changes were markedly alleviated by NLRP9-knockdown using its specific siRNA sequence. Pre-treatment of MLE-12 cells with ROS scavenger of N-acetylcysteine (NAC) remarkably decreased lipopolysaccharide (LPS)- and rMuNLRP9-induced production of ROS, and the secretion of inflammatory cytokines or chemokine, as well as the activity of IκBα/NF-κB, ASC/Casapse-1 and Caspase-3/PARP signaling pathways. Together, the findings here suggested that NLRP9b played an essential role in lung inflammation, apoptosis and oxidative stress of sepsis-induced ALI animal model or in LPS-induced MLE-12 cells, providing that NLRP9b inhibition might be a potential therapeutic option for ALI.
Collapse
Affiliation(s)
- Qiu Yanling
- Department of Pediatric, Baoji Maternal and Child Health Hospital, Baoji City 721000, Shaanxi, China
| | - Cheng Xiaoning
- Department of Pediatric, The Second Affiliated Hospital, Shaanxi University of Traditional Chinese Medicine, Shanxi 712000, China
| | - Bai Fei
- Department of Child Healthcare, Northwest Women and Children Hospital, Xi'an, Shaanxi 710061, China
| | - Fang Liyun
- Department of Pediatric, Xi'an NO.3 Hospital, Xi'an, Shaanxi 710018, China
| | - Hu Huizhong
- Department of Pediatric, Xi'an NO.3 Hospital, Xi'an, Shaanxi 710018, China
| | - Sun Daqing
- Department of Pediatric, Xi'an NO.3 Hospital, Xi'an, Shaanxi 710018, China.
| |
Collapse
|
85
|
Heme Oxygenase-1 Reduces Sepsis-Induced Endoplasmic Reticulum Stress and Acute Lung Injury. Mediators Inflamm 2018; 2018:9413876. [PMID: 30013453 PMCID: PMC6022325 DOI: 10.1155/2018/9413876] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/04/2018] [Indexed: 12/29/2022] Open
Abstract
Background Sepsis leads to severe acute lung injury/acute respiratory distress syndrome (ALI/ARDS) that is associated with enhanced endoplasmic reticulum (ER) stress. Heme oxygenase-1 (HO-1), an ER-anchored protein, exerts antioxidant and protective functions under ALI. However, the role of HO-1 activation in the development of endoplasmic reticulum (ER) stress during sepsis remains unknown. Methods Cecal ligation and puncture (CLP) model was created to induce septic ALI. Lung tissue ER stress was measured 18 hours after CLP. The effects of HO-1 on ER stress during septic ALI were investigated in vivo using HO-1 agonist hemin and antagonist ZnPP. Results Compared with the sham group, ER stress in septic lung increased significantly 18 hours after CLP, which was significantly reduced by pretreatment with the ER inhibitor 4-phenylbutyrate (4-PBA). The lung injury score and the lung wet to dry (W/D) ratio in lungs were significantly reduced in septic rats after ER stress inhibition. Similarly, lung ER stress-related genes' (PERK, eIF2-α, ATF4, and CHOP) levels were attenuated after ER stress inhibition. Furthermore, HO-1 activation by hemin reduced p-PERK, p-eIF2-α, ATF4, and CHOP protein expression and oxidative stress and lung cell apoptosis. Additionally, HO-1 antagonist could aggregate the ER stress-related ALI. Conclusions ER stress was activated during CLP-induced ALI, which may represent a mechanism by which CLP induces ALI. HO-1 activation could inhibit CLP-induced lung ER stress and attenuate CLP-induced ALI.
Collapse
|
86
|
Pereira MLM, Marinho CRF, Epiphanio S. Could Heme Oxygenase-1 Be a New Target for Therapeutic Intervention in Malaria-Associated Acute Lung Injury/Acute Respiratory Distress Syndrome? Front Cell Infect Microbiol 2018; 8:161. [PMID: 29868517 PMCID: PMC5964746 DOI: 10.3389/fcimb.2018.00161] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 04/26/2018] [Indexed: 01/17/2023] Open
Abstract
Malaria is a serious disease and was responsible for 429,000 deaths in 2015. Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is one of the main clinical complications of severe malaria; it is characterized by a high mortality rate and can even occur after antimalarial treatment when parasitemia is not detected. Rodent models of ALI/ARDS show similar clinical signs as in humans when the rodents are infected with murine Plasmodium. In these models, it was shown that the induction of the enzyme heme oxygenase 1 (HO-1) is protective against severe malaria complications, including cerebral malaria and ALI/ARDS. Increased lung endothelial permeability and upregulation of VEGF and other pro-inflammatory cytokines were found to be associated with malaria-associated ALI/ARDS (MA-ALI/ARDS), and both were reduced after HO-1 induction. Additionally, mice were protected against MA-ALI/ARDS after treatment with carbon monoxide- releasing molecules or with carbon monoxide, which is also released by the HO-1 activity. However, high HO-1 levels in inflammatory cells were associated with the respiratory burst of neutrophils and with an intensification of inflammation during episodes of severe malaria in humans. Here, we review the main aspects of HO-1 in malaria and ALI/ARDS, presenting the dual role of HO-1 and possibilities for therapeutic intervention by modulating this important enzyme.
Collapse
Affiliation(s)
- Marcelo L M Pereira
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Claudio R F Marinho
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Sabrina Epiphanio
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
87
|
Singh N, Ahmad Z, Baid N, Kumar A. Host heme oxygenase-1: Friend or foe in tackling pathogens? IUBMB Life 2018; 70:869-880. [PMID: 29761622 DOI: 10.1002/iub.1868] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/14/2018] [Indexed: 12/26/2022]
Abstract
Infectious diseases are a major challenge in management of human health worldwide. Recent literature suggests that host immune system could be modulated to ameliorate the pathogenesis of infectious disease. Heme oxygenase (HMOX1) is a key regulator of cellular signaling and it could be modulated using pharmacological reagents. HMOX1 is a cytoprotective enzyme that degrades heme to generate carbon monoxide (CO), biliverdin, and molecular iron. CO and biliverdin (or bilirubin derived from it) can restrict the growth of a few pathogens. Both of these also induce antioxidant pathways and anti-inflammatory pathways. On the other hand, molecular iron can induce proinflammatory pathway besides making the cellular environment oxidative in nature. Since microbial infections often induce oxidative stress in host cells/tissues, role of HMOX1 has been analyzed in the pathogenesis of number of infections. In this review, we have described the role of HMOX1 in pathogenesis of bacterial infections caused by Mycobacterium species, Salmonella and in microbial sepsis. We have also provided a succinct overview of the role of HMOX1 in parasitic infections such as malaria and leishmaniasis. In the end, we have also elaborated the role of HMOX1 in viral infections such as AIDS, hepatitis, dengue, and influenza. © 2018 IUBMB Life, 70(9):869-880, 2018.
Collapse
Affiliation(s)
- Nisha Singh
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, Punjab, India
| | - Zeeshan Ahmad
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, Punjab, India
| | - Navin Baid
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, Punjab, India
| | - Ashwani Kumar
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh, Punjab, India
| |
Collapse
|
88
|
Liu Z, Qu M, Yu L, Song P, Chang Y. Artesunate Inhibits Renal Ischemia-Reperfusion-Mediated Remote Lung Inflammation Through Attenuating ROS-Induced Activation of NLRP3 Inflammasome. Inflammation 2018; 41:1546-1556. [DOI: 10.1007/s10753-018-0801-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
89
|
Transduced PEP-1-Heme Oxygenase-1 Fusion Protein Attenuates Lung Injury in Septic Shock Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6403861. [PMID: 29682161 PMCID: PMC5848134 DOI: 10.1155/2018/6403861] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/20/2017] [Accepted: 09/26/2017] [Indexed: 11/21/2022]
Abstract
Oxidative stress and inflammation have been identified to play a vital role in the pathogenesis of lung injury induced by septic shock. Heme oxygenase-1 (HO-1), an effective antioxidant and anti-inflammatory and antiapoptotic substance, has been used for the treatment of heart, lung, and liver diseases. Thus, we postulated that administration of exogenous HO-1 protein transduced by cell-penetrating peptide PEP-1 has a protective role against septic shock-induced lung injury. Septic shock produced by cecal ligation and puncture caused severe lung damage, manifested in the increase in the lung wet/dry ratio, oxidative stress, inflammation, and apoptosis. However, these changes were reversed by treatment with the PEP-1-HO-1 fusion protein, whereas lung injury in septic shock rats was alleviated. Furthermore, the septic shock upregulated the expression of Toll-like receptor 4 (TLR4) and transcription factor NF-κB, accompanied by the increase of lung injury. Administration of PEP-1-HO-1 fusion protein reversed septic shock-induced lung injury by downregulating the expression of TLR4 and NF-κB. Our study indicates that treatment with HO-1 protein transduced by PEP-1 confers protection against septic shock-induced lung injury by its antioxidant, anti-inflammatory, and antiapoptotic effects.
Collapse
|
90
|
Wu D, Shi L, Li P, Ni X, Zhang J, Zhu Q, Qi Y, Wang B. Intermedin 1-53 Protects Cardiac Fibroblasts by Inhibiting NLRP3 Inflammasome Activation During Sepsis. Inflammation 2018; 41:505-514. [PMID: 29192367 DOI: 10.1007/s10753-017-0706-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Sepsis is a disease that occurs as a result of systemic inflammatory response syndrome (SIRS) in response to an infection, contributing to multiple organ dysfunction and a high mortality rate. Interleukin-lβ (IL-1β) is a cytokine that plays critical roles in inflammation and cardiac dysfunction during severe sepsis. Intermedin1-53 (IMD1-53) has been recently discovered to possess potential endogenous anti-inflammatory and strong cardiovascular protective effects. To investigate whether IMD1-53 can inhibit the NLRP3/caspase-1/IL-1β pathway to alleviate cardiac injury and rescue heart function, sepsis was induced in vivo by caecal ligation and puncture (CLP) surgery, and lipopolysaccharides were used as septic stressors for cardiac fibroblasts (CFs) in vitro. The expressions of IMD1-53 receptors in sepsis rat heart were increased. After IMD1-53 treatment, inflammation caused by sepsis in vivo was greatly reduced, as shown by the downregulation of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), nucleotide-binding domain and leucine-rich repeat containing family, pyrin containing 3 (NLRP3), pro-IL-1β, caspase 1, and nuclear translocation of nuclear factor-κB (NF-kB) protein levels. In addition, cardiac function was significantly improved and mean arterial blood pressure (MABP) increased by 34.8% (P < 0.05) which almost back to normal. Surprisingly, IMD1-53 inhibited cell apoptosis, as caspase 3 activity and Bax expression was significantly reduced in the heart upon IMD1-53 treatment. IMD1-53 abolished the upregulation of ASC, NLRP3, and caspase 1 protein levels in CFs induced by lipopolysaccharide (LPS). IMD1-53 increased cell survival rates and inhibited IL-1β production in the cell culture medium. IMD1-53 can protect against inflammation and heart injury during sepsis via attenuating the NLRP3/caspase-1/IL-1β pathway.
Collapse
Affiliation(s)
- Di Wu
- The Peking University Aerospace School of Clinical Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Lin Shi
- The Peking University People's Hospital, Beijing, 100191, China
| | - Pengyang Li
- Texas Heart Institute, Houston, TX, 77030, USA
| | - Xianqiang Ni
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Jinsheng Zhang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Qing Zhu
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Yongfen Qi
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
| | - Bin Wang
- The Peking University Aerospace School of Clinical Medicine, Peking University Health Science Center, Beijing, 100191, China.
- The First Affiliated Hospital of Shantou University Medical College, Guangdong, 515041, China.
| |
Collapse
|
91
|
Liu Q, Ci X, Wen Z, Peng L. Diosmetin Alleviates Lipopolysaccharide-Induced Acute Lung Injury through Activating the Nrf2 Pathway and Inhibiting the NLRP3 Inflammasome. Biomol Ther (Seoul) 2018; 26:157-166. [PMID: 28365974 PMCID: PMC5839494 DOI: 10.4062/biomolther.2016.234] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 01/03/2023] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a common clinical syndrome of diffuse lung inflammation with high mortality rates and limited therapeutic methods. Diosmetin, an active component from Chinese herbs, has long been noticed because of its antioxidant and anti-inflammatory activities. The aim of this study was to evaluate the effects of diosmetin on LPS-induced ALI model and unveil the possible mechanisms. Our results revealed that pretreatment with diosmetin effectively alleviated lung histopathological changes, which were further evaluated by lung injury scores. Diosmetin also decreased lung wet/dry ratios, as well as total protein levels, inflammatory cell infiltration and proinflammatory cytokine (eg. TNF-α, IL-1β and IL-6) overproduction in bronchoalveolar lavage fluid (BALF). Additionally, increased MPO, MDA and ROS levels induced by LPS were also markly suppressed by diosmetin. Furthermore, diosmetin significantly increased the expression of Nrf2 along with its target gene HO-1 and blocked the activation of NLRP3 inflammasome in the lung tissues, which might be central to the protective effects of diosmetin. Further supporting these results, in vitro experiments also showed that diosmetin activated Nrf2 and HO-1, as well as inhibited the NLRP3 inflammasome in both RAW264.7 and A549 cells. The present study highlights the protective effects of diosmetin on LPS-induced ALI via activation of Nrf2 and inhibition of NLRP3 inflammasome, bringing up the hope of its application as a therapeutic drug towards LPS-induced ALI.
Collapse
Affiliation(s)
- Qinmei Liu
- Department of Respiration, The First Hospital, Jilin University, Changchun 130021, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun 130061, China
| | - Zhongmei Wen
- Department of Respiration, The First Hospital, Jilin University, Changchun 130021, China
| | - Liping Peng
- Department of Respiration, The First Hospital, Jilin University, Changchun 130021, China
| |
Collapse
|
92
|
Hydrogen Sulfide Attenuates LPS-Induced Acute Kidney Injury by Inhibiting Inflammation and Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6717212. [PMID: 29636853 PMCID: PMC5831990 DOI: 10.1155/2018/6717212] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 11/21/2017] [Accepted: 12/04/2017] [Indexed: 12/29/2022]
Abstract
In order to investigate the protective mechanism of hydrogen sulfide (H2S) in sepsis-associated acute kidney injury (SA-AKI), ten AKI patients and ten healthy controls were enrolled. In AKI patients, levels of creatinine (Cre), urea nitrogen (BUN), tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), and myeloperoxidase (MPO) activity as well as concentrations of malondialdehyde (MDA) and hydrogen peroxide (H2O2) were significantly increased compared with those of controls. However, plasma level of H2S decreased and was linearly correlated with levels of Cre and BUN. After that, an AKI mouse model by intraperitoneal lipopolysaccharide (LPS) injection was constructed for in vivo study. In AKI mice, H2S levels decreased with the decline of 3-MST activity and expression; similar changes were observed in other indicators mentioned above. However, the protein expressions of TLR4, NLRP3, and caspase-1 in mice kidney tissues were significantly increased 6 h after LPS injection. NaHS could improve renal function and kidney histopathological changes, attenuate LPS-induced inflammation and oxidative stress, and inhibit expressions of TLR4, NLRP3, and caspase-1. Our study demonstrated that endogenous H2S is involved in the pathogenesis of SA-AKI, and exogenous H2S exerts protective effects against LPS-induced AKI by inhibiting inflammation and oxidative stress via the TLR4/NLRP3 signaling pathway.
Collapse
|
93
|
Liu S, Tian L, Chai G, Wen B, Wang B. Targeting heme oxygenase-1 by quercetin ameliorates alcohol-induced acute liver injury via inhibiting NLRP3 inflammasome activation. Food Funct 2018; 9:4184-4193. [PMID: 29993075 DOI: 10.1039/c8fo00650d] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Quercetin can ameliorate alcohol-induced acute liver injury via inducing heme oxygenase-1 and inhibiting NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Shu Liu
- Department of Geriatrics
- The First Affiliated Hospital of China Medical University
- China
| | - Lei Tian
- Department of Gastroenterology
- The First Affiliated Hospital of Jinzhou Medical University
- China
| | - Guangrui Chai
- Department of Ophthalmology
- Shengjing Hospital of China Medical University
- China
| | - Bo Wen
- Department of Geriatrics
- The First Affiliated Hospital of China Medical University
- China
| | - Bingyuan Wang
- Department of Geriatrics
- The First Affiliated Hospital of China Medical University
- China
| |
Collapse
|
94
|
Fu J, Sun H, Zhang Y, Xu W, Wang C, Fang Y, Zhao J. Neuroprotective Effects of Luteolin Against Spinal Cord Ischemia–Reperfusion Injury by Attenuation of Oxidative Stress, Inflammation, and Apoptosis. J Med Food 2018; 21:13-20. [PMID: 28976796 DOI: 10.1089/jmf.2017.4021] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Jian Fu
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Emergency Medicine of Hebei Province, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haibin Sun
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yongzhe Zhang
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wei Xu
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chuan Wang
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yanwei Fang
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jianhui Zhao
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
95
|
Bai C, Li T, Sun Q, Xin Q, Xu T, Yu J, Wang Y, Wei L. Protective effect of baicalin against severe burn‑induced remote acute lung injury in rats. Mol Med Rep 2017; 17:2689-2694. [PMID: 29207058 DOI: 10.3892/mmr.2017.8120] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 10/03/2017] [Indexed: 11/05/2022] Open
Abstract
Baicalin exhibits antibacterial, anti‑viral, anti‑oxidative, antipyretic, analgesic, anti‑inflammatory and anti‑tumor properties. The chemical scavenges oxygen free radicals, protects the cardiovascular system and neurons, protects the liver, and has been used for the prevention and treatment of diabetes‑associated complications. The present study investigated the effect of baicalin on severe burn‑induced remote acute lung injury (ALI). The present study demonstrated that baicalin significantly decreased the lung wet‑to‑dry weight ratio, improved pulmonary histological alterations and reduced the expression of high mobility group protein B1 in the rat model of ALI. In addition, treatment with baicalin decreased tumor necrosis factor‑α, interleukin (IL)‑8, IL‑1β and IL‑18 concentrations in the serum, reduced myeloperoxidase activity and malondialdehyde content, and increased the level of superoxide dismutase in the serum in treated model rats with ALI. As a result, baicalin significantly suppressed nucleotide‑binding oligomerization, NACHT, LRR and PYD domains‑containing protein 3 (NLRP3), caspase‑1, nuclear factor‑κB and matrix metalloproteinase‑9 protein expression in the rat model of ALI. The results of the present study suggested that baicalin may serve a protective role against ALI in rats through the NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Chongfeng Bai
- Postdoctoral Research Station of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Tao Li
- Department of Cardiothoracic Surgery, 401 Hospital of People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Qing Sun
- Department of Hyperbaric Medicine, 401 Hospital of People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Qun Xin
- Department of General Surgery, 401 Hospital of People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Tongyi Xu
- Department of Cardiothoracic Surgery, 401 Hospital of People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Jinjian Yu
- Department of Cardiothoracic Surgery, 401 Hospital of People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Yun Wang
- Department of Cardiothoracic Surgery, 401 Hospital of People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Li Wei
- Postdoctoral Research Station of Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
96
|
Xu F, Wang F, Wen T, Sang W, He X, Li L, Zeng N. Protective effect of cinnamic acid in endotoxin-poisoned mice. Phytother Res 2017; 31:1946-1953. [PMID: 29024091 DOI: 10.1002/ptr.5944] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 08/25/2017] [Accepted: 09/11/2017] [Indexed: 01/16/2023]
Abstract
In this work, we aimed to evaluate the protective effect of cinnamic acid (CD) on lipopolysaccharide (LPS; Escherichia coli 055:B5)-induced endotoxin-poisoned mice and clarify the underlying mechanisms. The mice were administrated CD 5 d before 15 mg/kg LPS challenge. 12 hr later, thymus was separated for determination of thymus indexes. Lung and spleen tissues were collected for histologic examination and the wet/dry weight ratio of lung was calculated, and serum was acquired for tumor necrosis factor-α (TNF-α), interleukin (IL)-18, and IL-1β measurement. Moreover, the expression of NOD-like receptor (NLR) family, pyrin domain-containing 3 (NLRP3) inflammasome was determined in lung. CD increased the thymus indexes and decreased lung wet/dry weight ratio. In addition, CD improved the lung and spleen histopathological changes induced by LPS and decreased the number of neutrophils in lung tissues. CD also inhibited the pro-inflammatory cytokines (TNF-α, IL-18, and IL-1β) production in serum. Furthermore, CD suppressed the LPS-induced NLRP3, Caspase-1, and IL-1β mRNA expression in lung, as well as the expression of NLRP3 and Caspase-1 (p20) protein. CD may have protective effects in endotoxin-poisoned mice via inhibiting the activation of NLRP3 inflammasome, and can be considered as a potential therapeutic candidate for diseases involved in endotoxin poisoning such as sepsis.
Collapse
Affiliation(s)
- Feng Xu
- Department of Pharmacology, College of Pharmacy, Chengdu University of TCM, Chengdu, Sichuan Province, China
| | - Feng Wang
- Department of Pharmacology, College of Pharmacy, Chengdu University of TCM, Chengdu, Sichuan Province, China
| | - Taoqun Wen
- Department of Pharmacology, College of Pharmacy, Chengdu University of TCM, Chengdu, Sichuan Province, China
| | - Wentao Sang
- Department of Pharmacology, College of Pharmacy, Chengdu University of TCM, Chengdu, Sichuan Province, China
| | - Xinyu He
- Department of Pharmacology, College of Pharmacy, Chengdu University of TCM, Chengdu, Sichuan Province, China
| | - Ling Li
- Department of Pharmacology, College of Pharmacy, Chengdu University of TCM, Chengdu, Sichuan Province, China
| | - Nan Zeng
- Department of Pharmacology, College of Pharmacy, Chengdu University of TCM, Chengdu, Sichuan Province, China
| |
Collapse
|
97
|
Luo M, Hu L, Li D, Wang Y, He Y, Zhu L, Ren W. MD-2 regulates LPS-induced NLRP3 inflammasome activation and IL-1beta secretion by a MyD88/NF-κB-dependent pathway in alveolar macrophages cell line. Mol Immunol 2017; 90:1-10. [DOI: 10.1016/j.molimm.2017.06.035] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/18/2017] [Accepted: 06/19/2017] [Indexed: 11/30/2022]
|
98
|
Nrf2/HO-1 mediates the neuroprotective effect of mangiferin on early brain injury after subarachnoid hemorrhage by attenuating mitochondria-related apoptosis and neuroinflammation. Sci Rep 2017; 7:11883. [PMID: 28928429 PMCID: PMC5605716 DOI: 10.1038/s41598-017-12160-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/04/2017] [Indexed: 02/07/2023] Open
Abstract
Early brain injury (EBI) is involved in the process of cerebral tissue damage caused by subarachnoid hemorrhage (SAH), and multiple mechanisms, such as apoptosis and inflammation, participate in its development. Mangiferin (MF), a natural C-glucoside xanthone, has been reported to exert beneficial effects against several types of organ injury by influencing various biological progresses. The current study aimed to investigate the potential of MF to protect against EBI following SAH via histological and biological assessments. A rat perforation model of SAH was established, and MF was subsequently administered via intraperitoneal injection at a low and a high dose. High-dose MF significantly lowered the mortality of SAH animals and ameliorated their neurological deficits and brain edema. MF also dose-relatedly attenuated SAH-induced oxidative stress and decreased cortical cell apoptosis by influencing mitochondria-apoptotic proteins. In addition, MF downregulated the activation of the NLRP3 inflammasome and NF-κB as well as the production of inflammatory cytokines, and the expression of Nrf2 and HO-1 was upregulated by MF. The abovementioned findings indicate that MF is neuroprotective against EBI after SAH and Nrf2/HO-1 cascade may play a key role in mediating its effect through regulation of the mitochondrial apoptosis pathway and activation of the NLRP3 inflammasome and NF-κB.
Collapse
|
99
|
Rahim I, Djerdjouri B, Sayed RK, Fernández-Ortiz M, Fernández-Gil B, Hidalgo-Gutiérrez A, López LC, Escames G, Reiter RJ, Acuña-Castroviejo D. Melatonin administration to wild-type mice and nontreated NLRP3 mutant mice share similar inhibition of the inflammatory response during sepsis. J Pineal Res 2017; 63. [PMID: 28370493 DOI: 10.1111/jpi.12410] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 03/24/2017] [Indexed: 12/18/2022]
Abstract
The NLRP3 inflammasome is involved in the innate immune response during inflammation. Moreover, melatonin blunts the NF-κB/NLRP3 connection during sepsis. Thus, we compared the roles of the NLRP3 inflammasome and/or melatonin treatment in the septic response of wild-type and NLRP3-/- mice. Mouse myocardial tissue was used for this purpose. The nuclear turnover of NF-κB was enhanced during sepsis, with an increase in TNFα, iNOS, and pro-IL-1β. The lack of inflammasome in NLRP3-/- mice significantly reduced that response and blunted IL-1β maturation due to the lack of caspase-1. Clock and Bmal1 did not change in both mouse strains, enhancing Chrono expression in mutants. RORα, which positively regulates Bmal1, was enhanced at a similar extend in both mouse strains, whereas the expression of the Bmal1 repressor, Rev-Erbα, increased in WT but was depressed in NLRP3-/- mice. Nampt, transcriptionally controlled by Bmal1, increased in WT mice together with Sirt1, whereas they remained unchanged in NLRP3-/- mice. Melatonin treatment reduced the septic response in a comparable manner as did the lack of NLRP3, but unlike the latter, it normalized the clock genes turnover through the induction of RORα and repression of Rev-Erbα and Per2, leading to enhanced Nampt and Sirt1. The lack of NLRP3 inflammasome converts sepsis to a moderate inflammatory disease and identifies NLRP3 as a main target for the treatment of sepsis. The efficacy of melatonin in counteracting the NLRP3 inflammasome activation further confirms the indoleamine as a useful therapeutic drug against this serious condition.
Collapse
Affiliation(s)
- Ibtissem Rahim
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Département de Biologie et Physiologie Cellulaire, Faculté des Sciences de la Nature et de la Vie, Université Blida 1, Blida, Algeria
- Faculté des Sciences Biologiques, Laboratoire de Biologie Cellulaire et Moléculaire, Université des Sciences et de la Technologie Houari Boumediene, Bab-Ezzouar, Algiers, Algeria
| | - Bahia Djerdjouri
- Faculté des Sciences Biologiques, Laboratoire de Biologie Cellulaire et Moléculaire, Université des Sciences et de la Technologie Houari Boumediene, Bab-Ezzouar, Algiers, Algeria
| | - Ramy K Sayed
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | - Marisol Fernández-Ortiz
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Beatriz Fernández-Gil
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Agustín Hidalgo-Gutiérrez
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Luis C López
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- CIBERfes, Ibs.Granada, and UGC de Laboratorios Clínicos, Complejo Hospitalario de Granada, Granada, Spain
| | - Germaine Escames
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- CIBERfes, Ibs.Granada, and UGC de Laboratorios Clínicos, Complejo Hospitalario de Granada, Granada, Spain
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX, USA
| | - Darío Acuña-Castroviejo
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- CIBERfes, Ibs.Granada, and UGC de Laboratorios Clínicos, Complejo Hospitalario de Granada, Granada, Spain
| |
Collapse
|
100
|
Ohbuchi A, Kono M, Kitagawa K, Takenokuchi M, Imoto S, Saigo K. Quantitative analysis of hemin-induced neutrophil extracellular trap formation and effects of hydrogen peroxide on this phenomenon. Biochem Biophys Rep 2017; 11:147-153. [PMID: 28955779 PMCID: PMC5614717 DOI: 10.1016/j.bbrep.2017.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/10/2017] [Accepted: 07/20/2017] [Indexed: 12/20/2022] Open
Abstract
Formation of neutrophil extracellular traps (NETs) can perpetuate sterile inflammation; thus, it is important to clarify their pathophysiological characteristics. Free heme, derived via hemolysis, is a major contributor to organ damage, and reportedly induces neutrophil activation as well as reactive oxygen species (ROS) production and NET formation. For this study, we examined hemin (Fe3+ -protoporphyrin IX)-induced NET formation quantitatively in vitro as well as the effects of oxidative stress. NETs formed in vitro from cultured neutrophils were quantitatively detected by using nuclease treatment and Sytox Green, a nucleic acid stain. Hemin-induced NET production was found to be in a dose-dependent manner, NADPH oxidase-dependent and toll-like receptor (TLR)-4 independent. Additionally, the iron molecule in the porphyrin ring was considered essential for the formation of NETs. In the presence of low concentrations of hydrogen peroxide, low concentrations of hemin-induced NETs were enhanced, unlike those of phorbol myristate acetate (PMA)-induced NETs. Quantitative analysis of NET formation may prove to be a useful tool for investigating NET physiology, and hemin could function as a possible therapeutic target for hemolysis-related events.
Collapse
Key Words
- DPI, diphenyleneiodonium
- ELISA, Enzyme-Linked Immuno-Sorbent Assay
- Extracellular trap
- HO-1, heme oxygenase-1
- Hemin
- Hydrogen peroxide
- LPS, lipopolysaccharide
- MPO, myeloperoxidase
- NADPH oxidase, nicotinamide adenine dinucleotide phosphate oxidase
- NET, neutrophil extracellular traps
- Neutrophil
- PAD4, peptidylarginine deiminases 4
- PMA, phorbol myristate acetate
- Quantitative detection
- ROS, reactive oxygen species
- TAK-242 (PubChem CID: 11703255)
- TLR, toll-like receptor
- diphenylene iodonium (PubChem CID: 3101)
- hemin (PubChem CID: 121225420)
- hydrogen peroxide (PubChem CID: 784)
- phorbol myristate acetate (PubChem CID: 22833501)
- polymyxin B (PubChem CID: 4868)
- protoporphyrin IX (PubChem CID: 4971)
- sytox green (PubChem CID: 46863923)
Collapse
Affiliation(s)
- Ayako Ohbuchi
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, 7-2-1 Kamiono, Himeji, Hyogo 670-8524, Japan
| | - Mari Kono
- Scientific Research Division, Scientific Affairs, Sysmex Corporation, 1-3-2 Murotani, Nishi-ku, Kobe, Hyogo 651-2241, Japan
| | - Kaihei Kitagawa
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, 7-2-1 Kamiono, Himeji, Hyogo 670-8524, Japan
| | - Mariko Takenokuchi
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, 7-2-1 Kamiono, Himeji, Hyogo 670-8524, Japan
| | - Shion Imoto
- Department of Health Science, Kobe Tokiwa University, 2-6-2 Otani-cho, Nagata-ku, Kobe, Hyogo 653-0838, Japan
| | - Katsuyasu Saigo
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, 7-2-1 Kamiono, Himeji, Hyogo 670-8524, Japan
- Corresponding author.
| |
Collapse
|