51
|
LTB4 Promotes Acute Lung Injury via Upregulating the PLCε-1/TLR4/NF-κB Pathway in One-Lung Ventilation. DISEASE MARKERS 2022; 2022:1839341. [PMID: 35059042 PMCID: PMC8766192 DOI: 10.1155/2022/1839341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/23/2021] [Indexed: 11/21/2022]
Abstract
Background Mechanical ventilation (MV) can provoke acute lung injury (ALI) by increasing inflammation activation and disrupting the barrier in lung tissues even causing death. However, the inflammation-related molecules and pathways in MV-induced ALI remain largely unknown. Hence, the purposes of this study are to examine the role and mechanism of a novel inflammation-related molecule, leukotriene B4 (LTB4), in ALI. Methods The functions of LTB4 in one-lung ventilation (OLV) model were detected by the loss-of-function experiments. H&E staining was used to examine the pathologic changes of lung tissues. Functionally, PLCε-1 knockdown and Toll-like receptor 4 (TLR4)/NF-κB pathway inhibitor were used to detect the regulatory effects of LTB4 on the phospholipase Cε (PLCε-1)/TLR4/nuclear factor-kappa B (NF-κB) pathway. The levels of genes and proteins were determined by RT-qPCR and western blotting assay. The levels of inflammation cytokines and chemokines were measured by ELISA. Results Here, we found LTA4H, leukotriene B (4) receptor 1 (BLT1), LTB4, and PLCε-1 upregulated in OLV rats and associated with inflammatory activation and lung permeability changes of lung tissues. Inhibition of LTB4 alleviated the OLV-induced ALI by inhibiting inflammatory activation and lung permeability changes of lung tissues. For mechanism analyses, LTB4 promoted OLV-induced ALI by activating the PLCε-1/TLR4/NF-κB pathway. Conclusion LTB4 induced ALI in OLV rats by activating the PLCε-1/TLR4/NF-κB pathway. Our findings might supply a new potential therapeutic for OLV-induced ALI.
Collapse
|
52
|
Guan S, Jin T, Han S, Fan W, Chu H, Liang Y. Dihydroartemisinin alleviates morphine-induced neuroinflammation in BV-2 cells. Bioengineered 2021; 12:9401-9410. [PMID: 34854364 PMCID: PMC8810002 DOI: 10.1080/21655979.2021.1982311] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Morphine tolerance poses a great challenge for clinicians, whose pathogenesis has a close connection with microglial activation and neuroinflammation. Dihydroartemisinin (DHA) that derives from artemisinin, may serve as a potential anti-inflammatory drug. In this study, the effects as well as the underlying mechanism of DHA on suppressing microglial activation and neuroinflammation were explored. The microglial cell line BV-2 cells were induced by morphine and treated with DHA or minocycline. With the application of CCK-8, the cell viability was detected. Western blot was employed to assess the expressions of Ki67, IBa-1, and TLR4 and quantitative real-time PCR (qRT-PCR) was adopted to evaluate miRNA-16 (miR-16) expression. With the adoption of ELISA kits and qRT-PCR, the release of inflammatory cytokines was evaluated. Besides, luciferase reporter assay was applied to testify the binding relationship between miR-16 and TLR4. NF-κB expression was measured by immunofluorescence. DHA reduced cell viability and decreased protein expression of Ki67 and IBa-1 in morphine-induced BV-2 cells. Additionally, DHA contributed to the declined release of pro-inflammatory cytokines. miR-16 was down-regulated by morphine but was up-regulated by DHA concentration-dependently in BV-2 cells. The inhibition of miR-16 partly abolished the inhibitory effects of DHA on morphine-induced microglial activation and neuroinflammation. Moreover, TLR4 was found to be bound to miR-16, and the inhibitory effect of DHA on TLR4/NF-κB was partly reversed by miR-16 inhibition. In conclusion, DHA remarkably suppressed microglial activation and neuroinflammation through regulating miR-16-mediated TLR4/NF-κB signaling. This study may provide a new solution to improve clinical analgesic efficacy of morphine.
Collapse
Affiliation(s)
- Sen Guan
- Department of Anesthesiology, Women's and Children's Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
| | - Tingting Jin
- Department of Anesthesiology, Women's and Children's Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
| | - Shuai Han
- Department of Anesthesiology, Women's and Children's Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
| | - Wenjie Fan
- Department of Anesthesiology, Women's and Children's Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
| | - Haichen Chu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yongxin Liang
- Department of Anesthesiology, Women's and Children's Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
53
|
Mabrey FL, Morrell ED, Wurfel MM. TLRs in COVID-19: How they drive immunopathology and the rationale for modulation. Innate Immun 2021; 27:503-513. [PMID: 34806446 PMCID: PMC8762091 DOI: 10.1177/17534259211051364] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is both a viral illness and a disease of immunopathology. Proximal events within the innate immune system drive the balance between deleterious inflammation and viral clearance. We hypothesize that a divergence between the generation of excessive inflammation through over activation of the TLR associated myeloid differentiation primary response (MyD88) pathway relative to the TIR-domain-containing adaptor-inducing IFN-β (TRIF) pathway plays a key role in COVID-19 severity. Both viral elements and damage associated host molecules act as TLR ligands in this process. In this review, we detail the mechanism for this imbalance in COVID-19 based on available evidence, and we discuss how modulation of critical elements may be important in reducing severity of disease.
Collapse
Affiliation(s)
- F Linzee Mabrey
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, 7284University of Washington, USA
| | - Eric D Morrell
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, 7284University of Washington, USA
| | - Mark M Wurfel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, 7284University of Washington, USA
| |
Collapse
|
54
|
Hu M, Yang J, Xu Y. Isoorientin suppresses sepsis-induced acute lung injury in mice by activating an EPCR-dependent JAK2/STAT3 pathway. J Mol Histol 2021; 53:97-109. [PMID: 34787735 DOI: 10.1007/s10735-021-10039-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/29/2021] [Indexed: 10/19/2022]
Abstract
Sepsis is a systemic inflammatory syndrome, and acute lung injury (ALI) is one of the most common fatal complications of sepsis. Isoorientin (ISO) exerts a momentous role in the regulation of inflammation. However, whether ISO has a protective effect on sepsis-induced ALI remains unknown. This research aimed to elucidate the function of ISO on sepsis-induced ALI and its mechanism. In this study, the sepsis-induced ALI was established in the male C57BL/6 J mice. Functionally, ISO reduced the total protein concentration in BALF, lung wet/dry ratio and the numbers of neutrophils and macrophages in BALF as well as ameliorated lung injury. Besides, ISO treatment decreased the cytokine expressions and oxidative stress, and repressed the adhesion and migration of inflammatory cells induced by CLP. Mechanistically, ISO reduced the shedding of EPCR in the endothelial cell membrane; ISO treatment activated the JAK2/STAT3 signaling pathway through EPCR and the JAK2/STAT3 pathway inhibitors repressed the anti-inflammatory and antioxidant effects of ISO. In general, ISO suppressed sepsis-induced ALI in mice by activating an EPCR-dependent JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Mu Hu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiaotong, University School of Medicine, No. 999 Xiwang Road, Jiading District, Shanghai, 201801, China.
| | - Jielai Yang
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiaotong, University School of Medicine, No. 999 Xiwang Road, Jiading District, Shanghai, 201801, China
| | - Yang Xu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiaotong, University School of Medicine, No. 999 Xiwang Road, Jiading District, Shanghai, 201801, China
| |
Collapse
|
55
|
Kong L, Deng J, Zhou X, Cai B, Zhang B, Chen X, Chen Z, Wang W. Sitagliptin activates the p62-Keap1-Nrf2 signalling pathway to alleviate oxidative stress and excessive autophagy in severe acute pancreatitis-related acute lung injury. Cell Death Dis 2021; 12:928. [PMID: 34635643 PMCID: PMC8505515 DOI: 10.1038/s41419-021-04227-0] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/15/2021] [Accepted: 09/29/2021] [Indexed: 12/29/2022]
Abstract
Acute lung injury (ALI) is a complication of severe acute pancreatitis (SAP). Sitagliptin (SIT) is a DPP4 inhibitor that exerts anti-inflammatory and antioxidant effects; however, its mechanism of action in SAP-ALI remains unclear. In this study, we investigated the effects of SIT on SAP-ALI and the specific pathways involved in SAP-induced lung inflammation, including oxidative stress, autophagy, and p62-Kelch-like ECH-associated protein 1 (Keap1)-NF-E2-related factor 2 (Nrf2) signalling pathways. Nrf2 knockout (Nrf2-/-) and wild-type (WT) mice were pre-treated with SIT (100 mg/kg), followed by caerulein and lipopolysaccharide (LPS) administration to induce pancreatic and lung injury. BEAS-2B cells were transfected with siRNA-Nrf2 and treated with LPS, and the changes in inflammation, reactive oxygen species (ROS) levels, and autophagy were measured. SIT reduced histological damage, oedema, and myeloperoxidase activity in the lung, decreased the expression of pro-inflammatory cytokines, and inhibited excessive autophagy and ROS production via the activation of the p62-Keap1-Nrf2 signalling pathway and promotion of the nuclear translocation of Nrf2. In Nrf2-knockout mice, the anti-inflammatory effect of SIT was reduced, resulting in ROS accumulation and excessive autophagy. In BEAS-2B cells, LPS induced ROS production and activated autophagy, further enhanced by Nrf2 knockdown. This study demonstrates that SIT reduces SAP-ALI-associated oxidative stress and excessive autophagy through the p62-Keap1-Nrf2 signalling pathway and nuclear translocation of Nrf2, suggesting its therapeutic potential in SAP-ALI.
Collapse
Affiliation(s)
- Lingming Kong
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Deng
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiang Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Binbin Cai
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Baofu Zhang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaohu Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zongjing Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Weiming Wang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
56
|
Wang W, Yang N, Yang YH, Wen R, Liu CF, Zhang TN. Non-Coding RNAs: Master Regulators of Inflammasomes in Inflammatory Diseases. J Inflamm Res 2021; 14:5023-5050. [PMID: 34616171 PMCID: PMC8490125 DOI: 10.2147/jir.s332840] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/21/2021] [Indexed: 12/11/2022] Open
Abstract
Emerging data indicates that non-coding RNAs (ncRNAs) represent more than just “junk sequences” of the genome and have been found to be involved in multiple diseases by regulating various biological process, including the activation of inflammasomes. As an important aspect of innate immunity, inflammasomes are large immune multiprotein complexes that tightly regulate the production of pro-inflammatory cytokines and mediate pyroptosis; the activation of the inflammasomes is a vital biological process in inflammatory diseases. Recent studies have emphasized the function of ncRNAs in the fine control of inflammasomes activation either by directly targeting components of the inflammasomes or by controlling the activity of various factors that control the activation of inflammasomes; consequently, ncRNAs may represent potential therapeutic targets for inflammatory diseases. Understanding the precise role of ncRNAs in controlling the activation of inflammasomes will help us to design targeted therapies for multiple inflammatory diseases. In this review, we summarize the regulatory role and therapeutic potential of ncRNAs in the activation of inflammasomes by focusing on a range of inflammatory diseases, including microbial infection, sterile inflammatory diseases, and fibrosis-related diseases. Our goal is to provide new ideas and perspectives for future research.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Ni Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yu-Hang Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Ri Wen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Chun-Feng Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Tie-Ning Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
57
|
Gao W, Zhang Y. Depression of lncRNA MINCR antagonizes LPS-evoked acute injury and inflammatory response via miR-146b-5p and the TRAF6-NFkB signaling. Mol Med 2021; 27:124. [PMID: 34602057 PMCID: PMC8489090 DOI: 10.1186/s10020-021-00367-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 08/31/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Inflammation plays an important role in the development of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). The long non-coding RNA (lncRNA) MINCR is closely related to inflammation injury. This study was performed to explore the protective effects and mechanisms of MINCR in lipopolysaccharide (LPS)-induced lung injury and inflammation. METHODS The expression levels of MINCR and miR-146b-5p in lung tissue status were detected by using quantitative real-time polymerase chain reaction (qRT-PCR), hematoxylin and eosin staining, immunohistochemical staining, and terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Enzyme-linked immunosorbent assay and Western blotting analysis were used to detect the expression of inflammatory factors such as tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-10 in lung tissue. The relationship between MINCR, miR-146b-5p, and TRAF6 was explored using bioinformatics analysis and luciferase assay. RESULTS The expression levels of MINCR were increased in a mouse model of LPS-induced ALI and small airway epithelial cells (SAECs). shMINCR resulted in increased cell viability and decreased apoptosis, which protected against LPS-induced cell damage. shMINCR can inhibit the formation of neutrophil extracellular traps, neutrophil numbers, myeloperoxidase activity, and the production of inflammatory cytokines IL-6, IL-1β, and TNF-α induced by LPS. The silencing of miR-146b-5p reversed the effects of MINCR on LPS-induced lung damage. Sh-MINCR decreased the expression levels of TRAF6 and p-P65 in LPS-induced SAECs and lung tissues. Co-transfection of sh-MINCR with miR-146b-5p inhibitor reversed the effect of sh-MINCR on the expression of TRAF6 and p-P65. CONCLUSIONS MINCR may induce alveolar epithelial cell injury and inflammation and aggravate the progression of ALI/ARDS through miR-146b-5p and TRAF6/NF-κB pathways, which would provide a promising target for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Wei Gao
- Department of Critical Care Medicine, The Second Hospital of Shandong University, Jinan, 250033, Shandong, People's Republic of China
| | - Ying Zhang
- Department of Respiratory, The Second Hospital of Shandong University, No.247 Beiyuan Avenue, Jinan, 250033, Shandong, People's Republic of China.
| |
Collapse
|
58
|
Chicken bone marrow mesenchymal stem cells improve lung and distal organ injury. Sci Rep 2021; 11:17937. [PMID: 34508136 PMCID: PMC8433226 DOI: 10.1038/s41598-021-97383-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are associated with pulmonary protection and longevity. We separated chicken bone marrow-derived mesenchymal stem cells (BM-MSCs); investigated whether BM-MSCs can improve lipopolysaccharide (LPS)-induced lung and distal organ injury; and explored the underlying mechanisms. Ninety-six male ICR (6 weeks old) mice were randomly divided into three groups: Sham, LPS, and LPS + MSC groups. The mice were intratracheally injected with 5 mg/kg LPS to induce acute lung injury (ALI). The histopathological severity of injury to the lung, liver, kidney, heart, and aortic tissues was detected. Wet/dry ratio, protein concentrations in bronchoalveolar lavage fluid (BALF), BALF cell counts, inflammatory cytokine levels in serum, inflammatory cytokine gene expression, and oxidative stress-related indicators were detected. In addition, a survival analysis was performed in sixty male ICR mice (6 weeks old, 18–20 g). This study used chicken BM-MSCs, which are easier to obtain and more convenient than other animal or human MSCs, and have MSC-associated properties, such as a colony forming ability, multilineage differentiation potential, and certain phenotypes. BM-MSCs administration significantly improved the survival rate, systemic inflammation, and the histopathological severity of lung, liver, kidney, and aortic injury during ALI. BM-MSCs administration reduced the levels of inflammatory factors in BALF, the infiltration of neutrophils, and oxidative stress injury in lung tissue. In addition, BM-MSCs administration reduced TRL4 and Mdy88 mRNA expression during ALI. Chicken BM-MSCs serve as a potential alternative resource for stem cell therapy and exert a prominent effect on LPS-induced ALI and extrapulmonary injury, in part through TRL4/Mdy88 signaling and inhibition of neutrophil inflammation and oxidative stress injury.
Collapse
|
59
|
Jiang Z, Shen J, Ding J, Yuan Y, Gao L, Yang Z, Zhao X. USP18 mitigates lipopolysaccharide-induced oxidative stress and inflammation in human pulmonary microvascular endothelial cells through the TLR4/NF-κB/ROS signaling. Toxicol In Vitro 2021; 75:105181. [PMID: 33930521 DOI: 10.1016/j.tiv.2021.105181] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/19/2021] [Accepted: 04/25/2021] [Indexed: 12/16/2022]
Abstract
As a type I interferon response gene, ubiquitin-specific protease 18 (USP18) has been shown to be widely involved in oxidative stress and immune regulation processes. However, the relationship between USP18 and acute lung injury (ALI) is unclear. This study aimed to analyze the role of USP18 in the pathogenesis of ALI. Lipopolysaccharide (LPS) treatment up-regulated the expression of USP18 mRNA and protein in human pulmonary microvascular endothelial cells (hPMVECs). USP18 overexpression increased the viability of LPS-induced hPMVECs, and reduced LPS-induced cell damage. Additionally, USP overexpression increased the activity of SOD and CAT, and reduced the production of NO and MDA in LPS-induced hPMVECs. Moreover, overexpression of USP18 inhibited the secretion of IL-1β, IL-6, TNF-α, and IL-18 in LPS-induced hPMVECs. USP18 overexpression restrained LPS-induced upregulation of TLR4 and the excessive phosphorylation of p65 and IκBα, as well as the production of reactive oxygen species (ROS). TLR4 agonist MPLA attenuated the inhibitory effect of USP18 overexpression on LPS-induced oxidative stress and inflammation in hPMVECs. In addition, USP18 ameliorated LPS induced ALI in vivo. In conclusion, USP18 may resist LPS-induced oxidative stress and inflammatory response in hPMVECs by inhibiting the TLR4/NF-κB/ROS signaling pathway, which may provide new and complementary strategies for ALI treatment.
Collapse
Affiliation(s)
- Zeyu Jiang
- Department of Anesthesiology, The First People's Hospital of Changzhou, PR China
| | - Jiang Shen
- Department of Anesthesiology, The First People's Hospital of Changzhou, PR China
| | - Jie Ding
- Department of Anesthesiology, The First People's Hospital of Changzhou, PR China.
| | - Yan Yuan
- Department of Anesthesiology, The First People's Hospital of Changzhou, PR China
| | - Lulu Gao
- Department of Anesthesiology, The First People's Hospital of Changzhou, PR China
| | - Zhuocheng Yang
- Department of Anesthesiology, The First People's Hospital of Changzhou, PR China
| | - Xin Zhao
- Department of Anesthesiology, The First People's Hospital of Changzhou, PR China
| |
Collapse
|
60
|
Ma X, Tian D, Lv W, Gao B, Ma Z, Zheng X. Anti-inflammatory effects of microRNA-223 on sepsis-induced lung injury in rats by targeting the Toll-like receptor signaling pathway. Exp Ther Med 2021; 22:964. [PMID: 34335906 PMCID: PMC8290467 DOI: 10.3892/etm.2021.10396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 02/24/2021] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to investigate the mediation of micro RNA (miR)-223 on the anti-inflammatory effect of the Toll-like receptor (TLR) signaling pathway on sepsis-induced lung injury in rats via negatively regulating the expression of interleukin (IL)-6. Sprague-Dawley rats were used in the present study. It was determined whether miR-223 is differentially expressed in the lung using reverse transcription-quantitative PCR techniques and the content of cytokines in bronchoalveolar lavage (BAL) fluid was detected. The protein expression levels of TLR4 and nuclear factor (NF)-κB p65 were examined by western blotting and the pathological changes in the lung tissues of the sepsis group were observed. Hematoxylin and eosin was used to stain the lung tissues. The alveoli in the sham group exhibited a normal structure and morphology. In the sepsis group, the alveoli of the lung tissues were surrounded by numerous neutrophils, the mesenchyme was swollen, regions of the alveolar wall exhibited fibrosis and the alveolar wall was thickened. Furthermore, in the sepsis group, miR-223 expression was increased in the lung tissues when compared with that in the sham group. The content of cytokines, IL-6 and IL-1β in the BAL fluid was significantly increased when compared with that of the sham group and TLR4 and NF-κB were also highly expressed. In addition, when compared with RAW264.7 cells that were overexpressing miR-223, the content of IL-6 and IL-1β in the supernatant and protein expression of TLR and NF-κB in cells were markedly decreased. Thus, it was demonstrated that miR-223 negatively regulated the expression of IL-6, mediating the TLR4/NF-κB signaling pathway and exerting an anti-inflammatory effect in sepsis-induced lung injury.
Collapse
Affiliation(s)
- Xuena Ma
- Department of Critical Medicine, The Fourth Central Hospital of Baoding City, Baoding, Hebei 072350, P.R. China
| | - Dan Tian
- Department of Critical Medicine, The Fourth Central Hospital of Baoding City, Baoding, Hebei 072350, P.R. China
| | - Weina Lv
- Department of Critical Medicine, The Fourth Central Hospital of Baoding City, Baoding, Hebei 072350, P.R. China
| | - Baoyu Gao
- Department of Otorhinolaryngology, The Fourth Central Hospital of Baoding City, Baoding, Hebei 072350, P.R. China
| | - Zhiyong Ma
- Department of Critical Medicine, The Fourth Central Hospital of Baoding City, Baoding, Hebei 072350, P.R. China
| | - Xiaotuo Zheng
- Department of Clinical Laboratory, The Fourth Central Hospital of Baoding City, Baoding, Hebei 072350, P.R. China
| |
Collapse
|
61
|
Sun X, Wu B, Geng L, Zhang J, Qin G. Xiaokang Liuwei Dihuang decoction ameliorates the immune infertility of male rats induced by lipopolysaccharide through regulating the levels of sex hormones, reactive oxygen species, pro-apoptotic and immune factors. Biomed Pharmacother 2021; 139:111514. [DOI: 10.1016/j.biopha.2021.111514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/11/2021] [Accepted: 03/14/2021] [Indexed: 12/16/2022] Open
|
62
|
Dong R, Zhang X, Zhao Z. Ulinastatin as an Adjuvant Therapy to Restricting Volumes of Resuscitation Fluid Strategy for Patients with Septic Shock after Initial Management. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:4231454. [PMID: 34221069 PMCID: PMC8221865 DOI: 10.1155/2021/4231454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/09/2021] [Indexed: 12/29/2022]
Abstract
Septic shock is the most serious complication of sepsis, leading to unacceptably high morbidity and mortality worldwide. Fluid resuscitation using crystalloids has become the mainstay of early and aggressive treatment of severe sepsis and septic shock, while increased daily fluid balances from day 2 until day 7 have been related with increased mortality. Recently, pharmacological management has been recommended to combine with appropriate fluid resuscitation for the treatment of septic shock. In this study, we compared the clinical efficacy of restricting volumes of resuscitation fluid strategy with or without intravenous infusion of ulinastatin (UTI) in treating patients with septic shock and additionally examined the patient's changes of the extravascular lung water index (EVLWI), pulmonary vascular permeability index (PVPI), systemic vascular resistance index (SVRI), cardiac function, lactic acid (LA) level, coagulation function, and renal function. The study included 182 patients with septic shock, among which 89 patients had undergone restricting volumes of resuscitation fluid strategy with intravenous infusion of UTI and 93 patients had undergone restricting volumes of resuscitation fluid strategy alone. It was found that patients with septic shock after restricting volumes of resuscitation fluid strategy with intravenous infusion of UTI showed an increased SVRI concomitant with declined PVPI and EVLWI, increased mean artery pressure (MAP), cardiac output (CO), left ventricular ejection fraction (LVEF), stroke volume (SV), and heart rate (HR), declined levels of cardiac troponin I (cTnI), N-terminal pro-B-type natriuretic peptide (NT-proBNP), and C-reactive protein (CRP), reduced LA level along with shortened prothrombin time (PT) and partially activated thrombin time (PATT), and decreased levels of blood urea nitrogen (BUN), creatinine (Cr), and uric acid (UA) when comparable to those after restricting volumes of resuscitation fluid strategy alone (P < 0.05). We also observed fewer scores of the Acute Physiology and Chronic Health Evaluation (APACHE II) and the sequential organ failure assessment (SOFA) in patients undergoing restricting volumes of resuscitation fluid strategy with intravenous infusion of UTI than those undergoing restricting volumes of resuscitation fluid strategy alone (P < 0.05). According to the above data, it is concluded that UTI as an adjuvant therapy for restricting volumes of resuscitation fluid strategy in treating septic shock may decrease the LA level, attenuate the inflammatory response, reduce vascular permeability, prevent pulmonary edema, and restore cardiac and renal functions.
Collapse
Affiliation(s)
- Rensong Dong
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xi Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhi Zhao
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
63
|
MiR-124-3p helps to protect against acute respiratory distress syndrome by targeting p65. Biosci Rep 2021; 40:224100. [PMID: 32391561 PMCID: PMC7253404 DOI: 10.1042/bsr20192132] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/23/2022] Open
Abstract
Background: Acute respiratory distress syndrome (ARDS) is a severe form of acute lung injury that has a high mortality rate and leads to substantial healthcare costs. MicroRNA-124-3p (miR-124-3p) helps to suppress inflammation during a pulmonary injury. However, its mechanism of action is largely unknown, and its role in ARDS remains to be determined. Methods: Mice and NR8383 cells were exposed to lipopolysaccharides (LPS) to induce ARDS, and their miR-124-3p levels were determined. After a miRNA agomir was administrated to the mice, their pulmonary injuries were evaluated by H&E staining and assays for peripheral inflammatory cytokine levels. The direct interaction between miR-124-3p and p65 was predicted, and then confirmed by a luciferase activity assay. The role played by miRNA-124-3p in regulating p65 expression was further examined by transfection with its agomir, and its role in cell apoptosis was investigated by observing the effects of miRNA overexpression in vitro and in vivo. Results: After exposure to LPS, there was a consistent decrease in miR-124-3p expression in the lungs of mice and in NR8383 cells. After treatment with the miR-124-3p agomir, the degrees of pulmonary injury (e.g. alveolar hemorrhage and interstitial edema), and the increases in IL-1β, IL-6, and TNF-α levels induced by LPS were significantly attenuated. Overexpression of miR-124-3p in NC8383 cells and lung tissues significantly suppressed LPS-induced p65 expression and cell apoptosis. Conclusions: These results suggest that miR-124-3p directly targeted p65, and thereby decreased the levels of inflammation and pulmonary injury in a mouse model of ARDS.
Collapse
|
64
|
Akpinar E, Kutlu Z, Kose D, Aydin P, Tavaci T, Bayraktutan Z, Yuksel TN, Yildirim S, Eser G, Dincer B. Protective Effects of Idebenone against Sepsis Induced Acute Lung Damage. J INVEST SURG 2021; 35:560-568. [PMID: 33722148 DOI: 10.1080/08941939.2021.1898063] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND/AIMS Sepsis is an uncontrolled systemic infection, withcomplex pathophysiology that may result in acute lung organ damage and cause multiple organ failure. Although much research has been conducted to illuminate sepsis's complex pathophysiology, sepsis treatment protocols are limited, and sepsis remains an important cause of mortality andmorbidity in intensive care units.Various studies have shown that idebenone (IDE) possesses strong antioxidant properties, which inhibit lipid peroxidation and protect cells from oxidative damage. The present study aimed to evaluate the protective effects of IDE against lung injury in a cecal ligation and puncture (CLP)-induced sepsis rat model. METHODS Male albino Wistar rats were used. The animals were divided into a healthy control (no treatment), CLP, IDE control (200 mg/kg), and CLP + IDE subgroups (50 mg/kg, 100 mg/kg, and 200 mg/kg), with nine rats in each group.IDE was administered 1 h after CLP induction.To evaluate the protective effects of IDE, lung tissues were collected 16 h after sepsis for biochemical, immunohistochemical staining, and histopathological examination. RESULTS IDE significantly ameliorated sepsis-induced disturbances in oxidative stress-related factors, with its effects increasing in accordance with the dose.IDE also abolished histopathological changes in lung tissues associated with CLP.Furthermore, interleukin 1 beta (IL-1β)and tumor necrosis factor-alpha (TNF-α) immunopositivity markedly decreased in the septic rats following IDE treatment. CONCLUSIONS IDE largely mitigated the inflammatory response in sepsis-induced lung injury by decreasing free radicals and preventing lipid peroxidation. The results suggest that IDE may represent a potential novel therapeutic drug for sepsis treatment.
Collapse
Affiliation(s)
- Erol Akpinar
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Zerrin Kutlu
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey
| | - Duygu Kose
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey.,Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| | - Pelin Aydin
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey.,Department of Anesthesiology and Reanimation, Educational and Research Hospital, Erzurum, Turkey
| | - Taha Tavaci
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Zafer Bayraktutan
- Department of Biochemistry, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Tugba Nurcan Yuksel
- Department of Pharmacology, Faculty of Medicine, Namik Kemal University, Tekirdag, Turkey
| | - Serkan Yildirim
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Gizem Eser
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Busra Dincer
- Department of Pharmacology, Faculty of Pharmacy, Erzincan Binali Yildirim University, Erzincan, Turkey
| |
Collapse
|
65
|
Tu Q, Zhu Y, Yuan Y, Guo L, Liu L, Yao L, Zou Y, Li J, Chen F. Gypenosides Inhibit Inflammatory Response and Apoptosis of Endothelial and Epithelial Cells in LPS-Induced ALI: A Study Based on Bioinformatic Analysis and in vivo/vitro Experiments. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:289-303. [PMID: 33531796 PMCID: PMC7846875 DOI: 10.2147/dddt.s286297] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022]
Abstract
Introduction Severe inflammatory response leads to poor prognosis of acute lung injury (ALI), the role of gypenosides (GPs) on ALI is not fully clear. The study aimed at investigating the effects of GPs on ALI. Methods We firstly established LPS-induced ALI mice model. Then, we tested whether GPs contributed to alleviate inflammatory response and lung injury of ALI in vivo. In order to identify specific mechanisms of the phenomenon, we conducted a bioinformatic analysis of LPS-induced ALI mice based on GEO database to identify hub differentially expressed genes (DEGs). PPI network of the DEGs was used to find hub-genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were conducted based on the DAVID database to identify which pathways the genes enriched. Then, we tested whether GPs inhibited lung injury and inflammatory response via the enriched pathways. We also tested whether GPs inhibited the apoptosis of endothelial and epithelial cells secondary to severe inflammation. Results We found GPs significantly alleviated lung injury and improved the survival rate of LPS-induced ALI mice in vivo. Bioinformatic analysis identified 20 hub-genes from DEGs, they were mainly enriched in NF-κB and TNF-α pathways. GPs could reduce the lung injury and inflammatory response via inhibiting NF-κB and TNF-α pathways in vivo. Our results indicated that GPs also inhibited inflammatory response of epithelial and endothelial cells via NF-κB and TNF-α pathways in vitro. Severe inflammatory response could also lead to apoptosis of endothelial and epithelial cells. Our results indicated that GPs effectively inhibited the apoptosis of endothelial and epithelial cells. Conclusion Our study suggested GPs contributed to alleviated lung injury in vivo and inhibited inflammation and apoptosis of endothelial and epithelial cells in vitro, providing novel strategies for the prevention and therapy for ALI.
Collapse
Affiliation(s)
- Qing Tu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Yabing Zhu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Yuan Yuan
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Long Guo
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Lu Liu
- School of Anesthesiology, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Liangfang Yao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Yun Zou
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Jinbao Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Feng Chen
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| |
Collapse
|
66
|
Wang R, Song W, Xie C, Zhong W, Xu H, Zhou Q, Deng Y, Hong Y, Li X, Fang M. Urinary Trypsin Inhibitor Protects Tight Junctions of Septic Pulmonary Capillary Endothelial Cells by Regulating the Functions of Macrophages. J Inflamm Res 2021; 14:1973-1989. [PMID: 34045879 PMCID: PMC8149216 DOI: 10.2147/jir.s303577] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/19/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Our previous study found that urinary trypsin inhibitor (ulinastatin, UTI) protected tight junctions (TJs) of lung endothelia via TNF-α inhibition, thereby alleviating pulmonary capillary permeability in septic rats. As the activated macrophage is the main source of TNF-α in sepsis, we speculate that UTI may exert the above effects by regulating the functions of macrophages. METHODS Bone-marrow derived macrophages (BMDM) were divided into control, lipopolysaccharide (LPS), UTI+LPS and UTI groups. TNF-α, TGF-β, IL-10, CD86, CD206 and MCP-1 expression were assessed by Western blot. The phagocytosis and migration of BMDM were detected. Pulmonary microvascular endothelial cells (PMVECs) were cultured with the conditioned medium (CM) from each group of BMDM above. Sprague-Dawley rats were divided into sham, cecal ligation and puncture (CLP), and UTI+CLP groups. Western blot and immunofluorescence were used to detected zonula occludens-1 (ZO-1), occludin and claudin-5 expression in PMVECs, as well as TNF-α, TGF-β, iNOS, CD86 and CD206 expression in lungs. Pulmonary capillary permeability was assessed by extravasated Evans blue, lung injury score (LIS), wet-to-dry weight ratio and electron microscope. RESULTS TNF-α and CD86 expression were increased in LPS-treated BMDM, but were reversed by UTI pretreatment. TGF-β, IL-10 and CD206 expression were the opposite. UTI markedly decreased phagocytosis and migration of LPS-treated BMDM. ZO-1, occludin and claudin-5 expression were markedly decreased in PMVECs of the CM-LPS group, but significantly increased in the CM-UTI+LPS group. TNF-α, iNOS and CD86 expression were increased in the lungs of CLP-rats but decreased with UTI pretreatment, while TGF-β and CD206 expression were the opposite. UTI markedly ameliorated the lung EB leakage, improved LIS, reduced the wet-to-dry ratio and revised the damaged TJs of PMVECs in CLP-rats. CONCLUSION UTI effectively inhibits the conversion of M1 macrophage but increases M2, reduces the phagocytosis and migration, which helps to protect endothelia TJs and reduce pulmonary capillary permeability during sepsis.
Collapse
Affiliation(s)
- Ruijie Wang
- Department of Intensive Care Unit, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
| | - Wenliang Song
- Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, People’s Republic of China
| | - Chengyuan Xie
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Wenhong Zhong
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
| | - Hui Xu
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
- Shantou University Medical College, Shantou, People’s Republic of China
| | - Qiuping Zhou
- Department of Intensive Care Unit, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
| | - Yiyu Deng
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
| | - Yimei Hong
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
| | - Xin Li
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
| | - Ming Fang
- Department of Intensive Care Unit, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
- Correspondence: Ming Fang Department of Intensive Care Unit, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, 510080, People’s Republic of ChinaTel +8613527774075 Email
| |
Collapse
|
67
|
Meng Y, Qiu S, Sun L, Zuo J. Knockdown of exosome‑mediated lnc‑PVT1 alleviates lipopolysaccharide‑induced osteoarthritis progression by mediating the HMGB1/TLR4/NF‑κB pathway via miR‑93‑5p. Mol Med Rep 2020; 22:5313-5325. [PMID: 33174011 PMCID: PMC7646997 DOI: 10.3892/mmr.2020.11594] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis is a chronic degenerative joint disease. Long non‑coding RNA plasmacytoma variant translocation 1 (PVT1) is involved in the progression of osteoarthritis and exosomes serve a central role in intercellular communication. However, whether PVT1 can be mediated by exosomes in osteoarthritis has not been reported. Whole blood was drawn from osteoarthritis patients and healthy volunteers. Lipopolysaccharide (LPS) was used to stimulate human normal chondrocytes (C28/I2) to construct a cell damage model in vitro. Protein levels were examined via western blot analysis. eThe expression of PVT1, microRNA (miR)‑93‑5p and high mobility groupprotein B1 (HMGB1) was evaluated through reverse transcription‑quantitative PCR. Cell viability and apoptosis were determined through CCK‑8 or flow cytometric assay. Inflammatory cytokines were measured via ELISA. The relationship between PVT1 or HMGB1 and miR‑93‑5p was confirmed by dual‑luciferase reporter assay. PVT1, HMGB1 and exosomal PVT1 were upregulated while miR‑93‑5p was downregulated in osteoarthritis patient serum and LPS‑induced C28/I2 cells. Exosomes from osteoarthritis patient serum and LPS‑treated C28/I2 cells increased PVT1 expression in C28/I2 cells. PVT1 depletion reversed the decrease of viability and the increase of apoptosis, inflammation responses and collagen degradation of C28/I2 cells induced by LPS. PVT1 regulated HMGB1 expression via sponging miR‑93‑5p. miR‑93‑5p inhibition abolished PVT1 silencing‑mediated viability, apoptosis, inflammation responses and collagen degradation of LPS‑stimulated C28/I2 cells. HMGB1 increase overturned miR‑93‑5p upregulation‑mediated viability, apoptosis, inflammation responses and collagen degradation of LPS‑stimulated C28/I2 cells. Furthermore, PVT1 modulated the Toll‑like receptor 4/NF‑κB pathway through an miR‑93‑5p/HMGB1 axis. In summary, exosome‑mediated PVT1 regulated LPS‑induced osteoarthritis progression by modulating the HMGB1/TLR4/NF‑κB pathway via miR‑93‑5p, providing a new route for possible osteoarthritis treatment.
Collapse
Affiliation(s)
- Yong Meng
- Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Siqiang Qiu
- Department of Spine Surgery, The Fourth People's Hospital of Jinan, Jinan, Shandong 250031, P.R. China
| | - Long Sun
- Department of Orthopedics, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Jinliang Zuo
- Department of Spine Surgery, The Fourth People's Hospital of Jinan, Jinan, Shandong 250031, P.R. China
| |
Collapse
|
68
|
Yu Y, Sun H, Zhu L, Ji L, Liu H. Downregulating lncRNA PRNCR1 ameliorates LPS-induced pulmonary vascular endothelial cell injury by modulating miR-330-5p/TLR4 axis. J Biochem Mol Toxicol 2020; 35:e22644. [PMID: 33049095 DOI: 10.1002/jbt.22644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/08/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022]
Abstract
Pulmonary vascular endothelial cell (PVEC) injury following acute lung injury or acute respiratory distress syndrome seriously affects disease development. Recently, accumulating evidence has suggested that long noncoding RNA (lncRNA) exerts significant effects in vascular endothelial cell injury. However, PRNCR1, a novel lncRNA, remains scarcely understood in terms of its functions in PVEC injury. Both in vivo and in vitro models of PVEC injury were constructed by lipopolysaccharide (LPS) administration. The relative expressions of PRNCR1, miR-330-5p, and TLR4 were detected by quantitative reverse transcription-polymerase chain reaction, Western blot, and immunohistochemistry. Besides, gain and loss assays of PRNCR1/miR-330-5p were conducted to verify their effects on LPS-induced PVEC injury. Cell Counting Kit-8 assay used to measure cell viability and flow cytometry was used to detect apoptosis. Besides, the protein levels of caspase 3, nuclear factor-κB (NF-κB), and inflammatory cytokines (including tumor necrosis factor-α, interleukin-1β [IL-1β], and IL-6) were evaluated via Western blot and enzyme-linked immunosorbent assay. Moreover, a dual-luciferase activity experiment and RNA immunoprecipitation were applied to confirm the targeting relationship between PRNCR1 and miR-330-5p, miR-330-5p, and TLR4. PRNCR1 and TLR4 levels were significantly upregulated in LPS-treated PVEC, both in vivo and in vitro, while miR-330-5p were downregulated. Inhibiting PRNCR1 or overexpressing miR-330-5p markedly attenuated LPS-induced PVEC injury, expressions of TLR4, NF-κB, and inflammatory cytokines. Mechanistically, PRNCR1 functioned as a competitive endogenous RNA by sponging miR-330-5p and then promoting TLR4 expression. PRNCR1 was upregulated in LPS-induced PVEC and aggravated its injury via modulating the miR-330-5p/TLR4 axis.
Collapse
Affiliation(s)
- Yingqing Yu
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongzhi Sun
- Department of Intensive Medicine, Second Hospital of Jilin University, Changchun, Jilin, China
| | - Lei Zhu
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Lianfeng Ji
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Haibo Liu
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
69
|
Brandão SCS, Ramos JDOX, Dompieri LT, Godoi ETAM, Figueiredo JL, Sarinho ESC, Chelvanambi S, Aikawa M. Is Toll-like receptor 4 involved in the severity of COVID-19 pathology in patients with cardiometabolic comorbidities? Cytokine Growth Factor Rev 2020; 58:102-110. [PMID: 32988728 PMCID: PMC7505161 DOI: 10.1016/j.cytogfr.2020.09.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/09/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
The severe form of COVID-19 is marked by an abnormal and exacerbated immunological host response favoring to a poor outcome in a significant number of patients, especially those with obesity, diabetes, hypertension, and atherosclerosis. The chronic inflammatory process found in these cardiometabolic comorbidities is marked by the overexpression of pro-inflammatory cytokines such as interleukin-6 (IL-6) and tumoral necrosis factor-alpha (TNF-α), which are products of the Toll-Like receptors 4 (TLR4) pathway. The SARS-CoV-2 initially infects cells in the upper respiratory tract and, in some patients, spread very quickly, needing respiratory support and systemically, causing collateral damage in tissues. We hypothesize that this happens because the SARS-CoV-2 spike protein interacts strongly with TLR4, causing an intensely exacerbated immune response in the host's lungs, culminating with the cytokine storm, accumulating secretions and hindering blood oxygenation, along with the immune system attacks the body, leading to multiple organ failure.
Collapse
Affiliation(s)
- Simone Cristina Soares Brandão
- Department of Medicine, Cardiology and Nuclear Imaging Division, Clinical Hospital, Federal University of Pernambuco, Recife, Pernambuco, Brazil.
| | | | | | | | - José Luiz Figueiredo
- Department of Surgery, Experimental Surgery Unit, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Emanuel Sávio Cavalcanti Sarinho
- Department of Medicine, Allergy and Clinical Immunology Division, Clinical Hospital, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Sarvesh Chelvanambi
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Masanori Aikawa
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
70
|
Banchini F, Vallisa D, Maniscalco P, Capelli P. Iron overload and Hepcidin overexpression could play a key role in COVID infection, and may explain vulnerability in elderly, diabetics, and obese patients. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:e2020013. [PMID: 32921750 PMCID: PMC7716981 DOI: 10.23750/abm.v91i3.9826] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/04/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND The COVID epidemic hit like a tsunami worldwide. At the time of its arrival in Italy, available literary data were meager, and most of them concerned its epidemiology. World Health Organization proposed guidelines in march 2020, a strategy of treatment has been developed, and a significant number of subsequent articles have been published to understand, prevent, and cure COVID patients. METHODS From the observation of two patients, we performed a careful analysis of scientific literature to unearth the relation between COVID infection, clinical manifestations as pneumonia and thrombosis, and to find out why it frequently affects obese, diabetics, and elderly patients. RESULTS The analysis shows that hepcidin could represent one of such correlating factors. Hepcidin is most elevated in older age, in non-insulin diabetics patients and in obese people. It is the final target therapy of many medicaments frequently used. Viral disease, and in particular SARS-CoV19, could induce activation of the hepcidin pathway, which in turn is responsible for an increase in the iron load. Excess of iron can lead to cell death by ferroptosis and release into the bloodstream, such as free iron, which in turn has toxic and pro-coagulative effects. CONCLUSIONS Overexpression of hepcidin and iron overload might play a crucial role in COVID infection, becoming potential targets for treatment. Hepcidin could also be considered as a biomarker to measure the effectiveness of our treatments and the restoration of iron homeostasis the final intent. (www.actabiomedica.it).
Collapse
Affiliation(s)
- Filippo Banchini
- Department of General Surgery, Guglielmo da Saliceto Hospital, Piacenza, Italy.
| | - Daniele Vallisa
- Department of Hematology , Guglielmo da Saliceto Hospital, Piacenza, Italy.
| | - Pietro Maniscalco
- Orthopedics and Traumatology Department, Guglielmo da Saliceto Hospital, Piacenza, Italy.
| | - Patrizio Capelli
- Department of General Surgery, Guglielmo da Saliceto Hospital, Piacenza, Italy.
| |
Collapse
|
71
|
Xia H, Ge Y, Wang F, Ming Y, Wu Z, Wang J, Sun S, Huang S, Chen M, Xiao W, Yao S. Protectin DX ameliorates inflammation in sepsis-induced acute lung injury through mediating PPARγ/NF-κB pathway. Immunol Res 2020; 68:280-288. [PMID: 32845434 DOI: 10.1007/s12026-020-09151-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 08/19/2020] [Indexed: 01/08/2023]
Abstract
Previous reports have demonstrated that the newly identified lipid mediator protectin DX (PDX) could effectively attenuate multiple organ injuries in sepsis. The aim of our study was to clarify whether PDX could improve acute lung injury (ALI) induced by sepsis and elucidate the relevant potential mechanism. After inducing sepsis by the cecal ligation and puncture approach, mice were treated with a high or low dose of PDX. Pathological changes in the pulmonary tissue were analyzed by hematoxylin-eosin staining, and lung injury score was evaluated. Lung permeability and edema were assessed by lung wet/dry ratio, and protein and cellular load of the bronchoalveolar lavage fluid (BALF). Inflammatory cytokine levels in BALF were measured by ELISA and the expression of PPARγ in the lung tissue was analyzed by immunoblotting. The results suggested that PDX could diminish the inflammatory response in lung tissue after sepsis by upregulating PPARγ and inhibiting the phosphorylation and activation of NF-κB p65. PDX treatment lowered the levels of pro-inflammation cytokines IL-1β, IL-6, TNF-α, and MCP-1, and the levels of anti-inflammatory cytokine IL-10 was increased in the BALF. It also improved lung permeability and reduced lung injury. Furthermore, the protective effect of PDX on lung tissue could be reversed by GW9662, a specific PPAR-γ antagonist. Taken together, our study indicated that PDX could ameliorate the inflammatory response in ALI by activating the PPARγ/NF-κB pathway in a mouse model of sepsis.
Collapse
Affiliation(s)
- Haifa Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Anesthesia and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
| | - Yangyang Ge
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Anesthesia and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
| | - Fuquan Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Anesthesia and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
| | - Yu Ming
- College of Health Science and Nursing, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Zhouyang Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Anesthesia and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
| | - Jingxu Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Anesthesia and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
| | - Shujun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Anesthesia and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
| | - Shiqian Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Anesthesia and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
| | - Ming Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Institute of Anesthesia and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
| | - Weimin Xiao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Institute of Anesthesia and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Institute of Anesthesia and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
72
|
Zhang G, Du Y, Sun N, Sun Y, Zhang L, Li X, Li X. Ulinastatin enhances autophagy against radiation-induced lung injury in mice. Transl Cancer Res 2020; 9:4162-4172. [PMID: 35117785 PMCID: PMC8798660 DOI: 10.21037/tcr-19-3018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 06/12/2020] [Indexed: 01/09/2023]
Abstract
Background To investigate the enhancement of autophagy by ulinastatin for protecting against radiation-induced lung injury (RILI) in mice. Methods Forty C57BL/6 mice were equally divided into (I) control (C), (II) irradiation (R), (III) ulinastatin (U), (IV) 3-methyladenine (3-MA) (M), and (V) ulinastatin plus 3-MA (U+M) groups. Three mice in each group were infected with adeno-associated virus (AAV) carrying green fluorescent protein (GFP)-1A/1B-light chain 3 (GFP-LC3) in the lung for the marker of autophagy. All mice in R, U, M and U+M groups were given chest irradiation (1 Gy/min, 12 min), following injection with normal saline in C and U groups, ulinastatin (500,000 IU/kg·d, i.p., 7 d) in U group, 3-MA (10 mg/kg·d, i.p., 7 d) in M group, and ulinastatin plus 3-MA in U+M group. The effects of ulinastatin on lung injury and autophagy were evaluated by electron microscope (EM), immunohistochemistry, mRNA expression levels of collagen alpha-1 (COL1A1), collagen alpha-2 (COL1A2), α-smooth muscle actin (α-SMA) and transforming growth factor β1 (TGF-β1), and protein levels of LC3, α-SMA, COL1A2, TGF-β1, matrix metalloproteinase-2 (MMP-2) and MMP-9. Results EM observation revealed that the radiation caused the injury of type I and II alveolar epithelial cells, which was improved by ulinastatin treatment associated with increased the numbers of autophagosomes. GFP-LC3 signals was significantly enhanced by ulinastatin detected by immune histochemical tests. At transcriptional and/or translational levels, ulinastatin significantly enhanced the expression levels of TGF-β1 and LC3 but reduced COL1A1, COL1A2, α-SMA, MMP-2 and MMP-9 after radiation-induced RILI. Conclusions Ulinastatin reduces RILI by enhancing autophagy, which might be a potential therapeutic drug in the protection against RILI.
Collapse
Affiliation(s)
- Guoxing Zhang
- Department of Intensive Care Unit, Jilin Cancer Hospital, Changchun, China
| | - Yujun Du
- Department of Kidney, The First Hospital of Jilin University, Changchun, China
| | - Ni Sun
- Department of Intensive Care Unit, Jilin Cancer Hospital, Changchun, China
| | - Yu Sun
- Department of Intensive Care Unit, Jilin Cancer Hospital, Changchun, China
| | - Liying Zhang
- Department of Intensive Care Unit, Jilin Cancer Hospital, Changchun, China
| | - Xiaohua Li
- Department of Infectious Diseases, The First Hospital of Jilin University, Changchun, China
| | - Xiujiang Li
- Department of Intensive Care Unit, Jilin Cancer Hospital, Changchun, China
| |
Collapse
|
73
|
Zheng D, Liu D, Kuang Y, Xu J, Xu G, Tai Q. Toll-like receptor 7 deficiency mitigates hyperoxia-induced acute lung injury in mice. Biomed Pharmacother 2020; 129:110345. [PMID: 32535385 DOI: 10.1016/j.biopha.2020.110345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/10/2020] [Accepted: 05/30/2020] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Toll-like receptor (TLR) 7 is an important mediator in inflammation. However, its role in hyperoxia-induced acute lung injury (HALI) remains to be elucidated. METHODS C57BL/6 wild-type and C57BL/6 background TLR 7 deficiency mice were exposed to hyperoxia to stimulate HALI in airtight cages. Animals were sacrificed at 72 h post hyperoxia or room air exposure. Lung injury indicators were measured. Moreover, soluble epoxide hydrolase (sEH) activity was detected by a 14, 15-EET/DHET ELISA kit. Activation of activator protein (AP)-1 and nuclear factor kappa-B (NF-κB) was detected with enzyme linked immunosorbent assay kits. RESULTS Our data revealed that pulmonary histological assay and wet to dry weight ratio, myeloperoxidase and malondialdehyde activity were reduced in TLR 7 deficiency mice compared with wild-type mice. Moreover, hyperoxia-caused elevation of sEH activity was reduced in TLR 7 deficiency mice. Transcription factors AP-1 activation was significantly inhibited in TLR 7 deficiency mice compared with wild-type mice. Similarly, the activation of NF-κB was reduced in TLR 7 deficiency mice. Tumor necrosis factor-α and interleukin-1β, potent proinflammatory cytokines, were reduced in TLR 7 deficiency mice. CONCLUSION TLR 7 deficiency is associated with inhibition of inflammation in HALI in mice.
Collapse
Affiliation(s)
- Donghua Zheng
- Department Of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, GuangDong, 510000, China
| | - Dawei Liu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, GuangDong, 510000, China
| | - Yukun Kuang
- The Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, GuangDong, 510000, China
| | - Jinghong Xu
- Department Of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, GuangDong, 510000, China
| | - Guixing Xu
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, GuangDong, 510000, China.
| | - Qiang Tai
- Department Of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, GuangDong, 510000, China.
| |
Collapse
|
74
|
Sulzbacher MM, Sulzbacher LM, Passos FR, Bilibio BLE, Althaus WF, Weizenmann L, de Oliveira K, Frizzo MN, Ludwig MS, Heck TG. A single dose of eHSP72 attenuates sepsis severity in mice. Sci Rep 2020; 10:9198. [PMID: 32513986 PMCID: PMC7280184 DOI: 10.1038/s41598-020-66011-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
High levels of extracellular 72 kDa heat shock protein (eHSP72) can be detected in the serum of septic patients and are associated with increased oxidative profiles and elevated rates of mortality among these patients. However, a possible immunomodulatory role for this protein, resulting in tissue protection during sepsis, has never been assessed. In this study, we investigated whether eHSP72 administration could attenuate the severity of sepsis in a mouse peritonitis model. Animals (90-day-old male C57BL/6J mice) were divided into Sepsis (n = 8) and Sepsis + eHSP72 (n = 9) groups, which both received injections of 20% fecal solution [1 mg/g body weight (wt), intraperitoneal (i.p.)], to trigger peritonitis induced-sepsis, whereas a Control group (n = 7) received a saline injection. eHSP72 was administered (1.33 ng/g body wt) to the Sepsis+eHSP72 group, 12 h after sepsis induction. All animals were evaluated for murine sepsis score (MSS), hemogram, core temperature, and glycemia (before and 4, 12, and 24 h after sepsis induction). Treatment with eHSP72 promoted reduced sepsis severity 24 h after sepsis induction, based on MSS scores (Control = 1.14 ± 1.02; Sepsis = 11.07 ± 7.24, and Sepsis + eHSP72 = 5.62 ± 1.72, P < 0.001) and core temperatures (°C; Control = 37.48 ± 0.58; Sepsis = 35.17 ± 2.88, and Sepsis + eHSP72 = 36.94 ± 2.02; P = 0.006). eHSP72 treatment also limited the oxidative profile and respiratory dysfunction in mice with sepsis. Although sepsis modified glycemic levels and white and red blood cell counts, these variables were not influenced by eHSP72 treatment (P > 0.05). Finally, eHSP72 improved the survival rate after sepsis (P = 0.0371). Together, our results indicated that eHSP72 may ameliorate sepsis severity and possibly improve some sepsis indices in mice.
Collapse
Affiliation(s)
- Maicon Machado Sulzbacher
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil. .,Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil.
| | - Lucas Machado Sulzbacher
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil
| | - Felipe Rafael Passos
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil
| | - Bruna Letícia Endl Bilibio
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil.,Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil
| | - Wellington Felipe Althaus
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil
| | - Luana Weizenmann
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil.,Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil
| | - Kauana de Oliveira
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil
| | - Matias Nunes Frizzo
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil.,Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil
| | - Mirna Stela Ludwig
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil.,Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil
| | - Thiago Gomes Heck
- Research Group in Physiology, Department of Life Sciences, Regional University of Northwestern Rio Grande do Sul State (UNIJUÍ), Ijuí, RS, Brazil. .,Postgraduate Program in Integral Attention to Health (PPGAIS-UNIJUÍ/UNICRUZ), Ijuí, RS, Brazil.
| |
Collapse
|
75
|
Liu P, Feng Y, Li H, Chen X, Wang G, Xu S, Li Y, Zhao L. Ferrostatin-1 alleviates lipopolysaccharide-induced acute lung injury via inhibiting ferroptosis. Cell Mol Biol Lett 2020; 25:10. [PMID: 32161620 PMCID: PMC7045739 DOI: 10.1186/s11658-020-00205-0] [Citation(s) in RCA: 400] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/17/2020] [Indexed: 02/07/2023] Open
Abstract
Background Ferroptosis is a newly recognized type of cell death, which is different from traditional necrosis, apoptosis or autophagic cell death. However, the position of ferroptosis in lipopolysaccharide (LPS)-induced acute lung injury (ALI) has not been explored intensively so far. In this study, we mainly analyzed the relationship between ferroptosis and LPS-induced ALI. Methods In this study, a human bronchial epithelial cell line, BEAS-2B, was treated with LPS and ferrostatin-1 (Fer-1, ferroptosis inhibitor). The cell viability was measured using CCK-8. Additionally, the levels of malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), and iron, as well as the protein level of SLC7A11 and GPX4, were measured in different groups. To further confirm the in vitro results, an ALI model was induced by LPS in mice, and the therapeutic action of Fer-1 and ferroptosis level in lung tissues were evaluated. Results The cell viability of BEAS-2B was down-regulated by LPS treatment, together with the ferroptosis markers SLC7A11 and GPX4, while the levels of MDA, 4-HNE and total iron were increased by LPS treatment in a dose-dependent manner, which could be rescued by Fer-1. The results of the in vivo experiment also indicated that Fer-1 exerted therapeutic action against LPS-induced ALI, and down-regulated the ferroptosis level in lung tissues. Conclusions Our study indicated that ferroptosis has an important role in the progression of LPS-induced ALI, and ferroptosis may become a novel target in the treatment of ALI patients.
Collapse
Affiliation(s)
- Pengfei Liu
- 1Department of Anesthesiology, The 2nd Clinical Medical College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, 518020 China.,2Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632 China
| | - Yetong Feng
- 3Health Science Center, School of Basic Medical Sciences, Shenzhen University, Shenzhen, 518037 China
| | - Hanwei Li
- 1Department of Anesthesiology, The 2nd Clinical Medical College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, 518020 China.,4Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, 510280 China
| | - Xin Chen
- 5Department of Laboratory Medicine, The 2nd Clinical Medicine College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, 518020 China
| | - Guangsuo Wang
- 6Department of Thoracic Surgery, The 2nd Clinical Medicine College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, 518020 China
| | - Shiyuan Xu
- 4Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, 510280 China
| | - Yalan Li
- 7Department of Anesthesiology, First Affiliated Hospital of Jinan University, Guangzhou, 510632 China.,2Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632 China
| | - Lei Zhao
- 1Department of Anesthesiology, The 2nd Clinical Medical College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, 518020 China.,2Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632 China
| |
Collapse
|
76
|
Attenuation of Sepsis-Induced Cardiomyopathy by Regulation of MicroRNA-23b Is Mediated Through Targeting of MyD88-Mediated NF-κB Activation. Inflammation 2019; 42:973-986. [PMID: 30734878 DOI: 10.1007/s10753-019-00958-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Myocardial cell injury or cardiomyopathy is associated with excessive inflammatory response and apoptosis of cardiac myocytes during sepsis. MicroRNA-23b (miR-23b) is a multifunctional miRNA that is considered to regulate immunosuppression in sepsis. The aim of this study was to examine the effect of miR-23b on cardiomyopathy induced by sepsis and to explore the potential mechanism involved. Sprague-Dawley rats were subjected to cecal ligation and puncture (CLP), and the level of miR-23b at different time points was measured by quantitative real-time polymerase chain reaction (qPCR). Then, we overexpressed miR-23b in vivo and in vitro. The rats were subjected to CLP 7 days after transfection. Cardiac function, inflammatory response, and heart tissues were examined 3 days thereafter. In an in vitro experiment, H9C2 cardiomyoblasts were stimulated with lipopolysaccharide (LPS) after transfection of miR-23b, following which apoptosis and the level of NF-κB were analyzed. The expression of miR-23b was upregulated during polymicrobial sepsis, and transfection of miR-23b lentivirus improved the outcome of sepsis-induced cardiomyopathy by attenuating inflammatory responses and protecting against histopathological damage. In in vitro experiments, elevated miR-23b inhibited excessive apoptosis of cardiomyocytes, which may be because activation of the NF-κB signaling pathway was inhibited by the decreased levels of TRAF6 and IKKβ. Therefore, miR-23b improved sepsis-induced cardiomyopathy by attenuating the inflammatory response, suppressing apoptosis, and preventing NF-κB activation via targeted inhibition of TRAF6 and IκκB. These results indicated that miR-23b may represent a novel therapeutic approach for clinical treatment of sepsis-induced cardiomyopathy.
Collapse
|
77
|
Chen Y, Xu Z, Song Q, Wang Z, Ji Z, Qiu Z, Cheng F, Jiang H. [Mechanism of ulinastatin in reducing lung inflammatory injury in rats with hemorrhagic shock]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:1232-1238. [PMID: 31801723 DOI: 10.12122/j.issn.1673-4254.2019.10.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the effect of ulinastatin on the inflammatory mediators and their signaling pathways miR-146a/TLR4/NF-κB in rats with hemorrhagic shock. METHODS Seventy-two SD rats were randomly assigned into shock without resuscitation group (SR group, n=24), acetated Ringer's solution resuscitation group (AR group, n=24) and ulinastatin treatment group (n=24). In all the 3 groups hemorrhagic shock models were established by femoral artery bleeding (with the mean arterial pressure maintained at 30-40 mmHg) without resuscitation (in SR group) or with resuscitation (in AR and ulinastatin groups) using acetated Ringer's solution for 30 min at 60 min after the onset of shock. At 1, 4, and 6 h after the shock onset or immediately after shock if the rats died, the lung tissues were taken for measurement of mRNA expressions of miR-146a, tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1), IL-4, IL-6 and IL-10 using real-time quantitative PCR and the protein expressions of TLR4, MyD88, IκB-α, p-IκB-α, NF-κB p65, IRAK4, p-IRAK4 (Thr345, Ser346), p-IRAK4 (Thr342) and TRAF6 using Western blotting. The lung histopathology of the rats was examined under optical microscope with HE staining. RESULTS Compared with the SR group, the rats in the AR group showed slightly alleviated inflammatory infiltration in the lung tissues with significantly increased mRNA levels of miR-146a, IL-4 and IL-10 (P < 0.05) and protein expressions of IκB-α, p-IRAK4 (Thr342) and p-IRAK4 (Thr345, ser346) (P < 0.05), and decreased mRNA levels of TNF-α, IL-1 and IL-6 (P < 0.05) and protein expressions of TLR4, MyD88, NF-κB p65, p-IκB-α, IRAK-4 and TRAF6 (P < 0.05). Compared with those in AR group, the rats in ulinastatin group showed further alleviation of inflammatory lung tissue injury, with increased mRNA levels of miR-146a, IL-4 and IL-10 (P < 0.01) and protein expressions of IκB-α, p-IRAK4 and p-IRAK4 (P < 0.01) and decreased mRNA levels of TNF-α, IL-1 and IL-6 (P < 0.01) and protein expressions of TLR4, MyD88, NF-κB p65, p-IκB-α, IRAK-4 and TRAF6 (P < 0.01). CONCLUSIONS Ulinastatin combined with acetated Ringer's solution resuscitation alleviates lung inflammations in rats with hemorrhagic shock possibly by enhancing miR-146a expression to regulate TLR4/NF-κB signaling pathway through a negative feedback mechanism and thus modulate the balance of pro-inflammatory and anti-inflammatory factors.
Collapse
Affiliation(s)
- Ying Chen
- Department of Emergency Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Zhipeng Xu
- Department of Emergency Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Qi Song
- Department of Emergency Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Zhenjie Wang
- Department of Emergency Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Zhong Ji
- Department of Emergency Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Zhaolei Qiu
- Department of Emergency Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Feng Cheng
- Department of Emergency Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Hai Jiang
- Department of Emergency Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| |
Collapse
|
78
|
Ji S, Wang L. μ‐Opioid receptor signalling via PI3K/Akt pathway ameliorates lipopolysaccharide‐induced acute respiratory distress syndrome. Exp Physiol 2019; 104:1555-1561. [PMID: 31272134 DOI: 10.1113/ep087783] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/02/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Suzhen Ji
- Department of EmergencyCangzhou Central Hospital Xinhua West Road Cangzhou 061000 Hebei China
| | - Leilei Wang
- Department of NeurosurgeryCangzhou Central Hospital Xinhua West Road Cangzhou 061000 Hebei China
| |
Collapse
|
79
|
Wang B, Wang J, Lu D, Qi N, Liu Q. The Defensive Action of LYRM03 on LPS-Induced Acute Lung Injury by NF-κB/TLR4/NLRP3 Signals. J INVEST SURG 2019; 34:284-296. [PMID: 31274341 DOI: 10.1080/08941939.2019.1634165] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The aim of the current investigation was to study the role of 3-amino-2-hydroxy-4-phenyl-valyl-isoleucine (LYRM03) in lipopolysaccharide (LPS)-induced acute lung injury (ALI) and investigate its potential pathogenesis. An LPS-induced ALI model was produced with LPS (5 mg/kg) followed by 24 h of injury. Rats were randomly assigned to 6 groups for in vivo experiments: (1) Sham, (2) LYRM03 (20 mg/kg), (3) LPS, (4) LPS plus LYRM03 (5 mg/kg), (5) LPS plus LYRM03 (10 mg/kg), and (6) LPS plus LYRM03 (20 mg/kg). The rat alveolar macrophage cell line (NR8383) cells were divided into 6 groups for in vitro experiments: (1) Sham, (2) LYRM03 (200 μmol/L), (3) LPS (100 ng/mL), (4) LPS plus LYRM03 (50 μmol/L), (5) LPS plus LYRM03 (100 μmol/L), and (6) LPS plus LYRM03 (200 μmol/L). Further study about siRNA targeting NF-κB p65, TLR4, and NLRP3 to explore the potential mechanism of LYRM03 in the LPS-induced ALI models have been done. Therefore, LYRM03 decreased LPS-induced ALI and NR8383 activation as demonstrated through hematoxylin-eosin staining and western blot analysis in vivo and in vitro. LYRM03 ameliorated the content of protein in bronchoalveolar lavage fluid, myeloperoxidase in the lung and malondialdehyde (MDA) in serum. In addition, LYRM03 ameliorated the levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-18 (IL-18) in the serum of rats and the supernatant of NR8383 cells. Moreover, LYRM03 significantly inhibited the activities of nuclear factor kappa B (NF-κB), myeloid differentiation factor 88 (MyD88), and toll-like receptor 4 (TLR4). LYRM03 also reduced the increase in the inflammasome, including apoptosis-related speck-like protein containing CARD (ASC), and NOD-like receptor 3 (NLRP3), in LPS-stimulated rats and NR8383 cells. The extent of injury and lung injury scores in the LYRM03 (20 mg/kg) + siRNA targeting NF-κB p65, TLR4, or NLRP3 + LPS-treated rats were higher than that in the LYRM03 (20 mg/kg) + LPS-treated rats. In summary, LYRM03 conferred an intensely lung defensive action on LPS-induced ALI in vivo and in vitro, which could be associated with the abatement of TLR4-induced NLRP3/NF-κB.
Collapse
Affiliation(s)
- Bin Wang
- Department of Critical Care Medicine, Rizhao People Hospital, Rizhao, People's Republic of China
| | - Jiaoyue Wang
- Department of Critical Care Medicine, Rizhao People Hospital, Rizhao, People's Republic of China
| | - Daopeng Lu
- Department of Emergency, Jinan Medical Emergency Center, Jinan, People's Republic of China
| | - Na Qi
- Department of Respiratory Medicine, Hengshui People Hospital, Hengshui, People's Republic of China
| | - Qin Liu
- Department of Emergency, Jinan Medical Emergency Center, Jinan, People's Republic of China
| |
Collapse
|
80
|
Kulikov OA, Ageev VP, Marochkina EE, Dolgacheva IS, Minayeva OV, Inchina VI. Efficacy of liposomal dosage forms and hyperosmolar salines in experimental pharmacotherapy of acute lung injury. RESEARCH RESULTS IN PHARMACOLOGY 2019. [DOI: 10.3897/rrpharmacology.5.35529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Introduction: Hypertonic sodium chloride solutions and liposomal drugs with pulmotropic effect are of great interest for the treatment of acute lung injury (ALI). The results of the studies on the efficacy of hypertonic solutions and liposomes in ALI treatment are currently controversial.Materials and methods: For the experiment, liposomes with dexamethasone, N-acetylcysteine (NAC), aprotinin and dye Cyanine-7 (Cy-7) were obtained. A liposome analysis was performed by means of spectrophotometry. ALI was modeled in rats by the administration of the damaging agents into the trachea. The experimental agents were injected once intravenously after the modeling of ALI. For experimental therapy used liposomal agents, 7.5% hypertonic saline (HS) and HyperHAES solutions in the respective groups. The efficacy of the therapy was assessed by the survival of animals, functional indicators of the cardiovascular and respiratory systems, and by the lung-body ratio. The biodistribution of liposomes after intravenous administration was investigated in mice through using a fluorescent dye Cy-7. The biodistribution of liposomes with Cy-7 was assessed using bioimaging according to the fluorescence intensity of internal organs (lungs, liver, and kidneys) and blood, expressed as dye concentration according to the calibration dependence of dye concentrarion on fluorescence intensity.Results and discussion: All the studied liposomal drugs were effective for the pharmacological correction of ALI. Hypertonic solutions, unlike liposomal drugs, were less likely to prevent the development of pulmonary edema. All the studied therapeutic agents increased the survival rate of the laboratory animals with ALI. The most effective experimental agent was liposomal dexamethasone. The use of drugs in form of simple liposomes with average diameter of 350 nm provided for a higher concentration of the drug in the lungs within the first 40 minutes after intravenous administration.Conclusion: Intravenous administration of liposomal forms is promising for the pharmacotherapy of acute lung injury.
Collapse
|
81
|
Xu Q, Yan Q, Chen S. Use of ulinastatin was associated with reduced mortality in critically ill patients with sepsis. J Thorac Dis 2019; 11:1911-1918. [PMID: 31285884 DOI: 10.21037/jtd.2019.05.03] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Ulinastatin has anti-inflammatory properties and could potentially benefit critically ill septic patients. Nevertheless, clinical studies have yielded conflicting results. The present study examined the efficacy of ulinastatin in intensive care unit (ICU) patients with sepsis and/or septic shock. Methods All septic patients admitted to the ICU of Wuhu No. 2 People's Hospital between 2014 and 2017 were screened for potential eligibility for this retrospective study. The primary outcome was 28-day mortality, and its correlation with ulinastatin was assessed using multiple logistic regression models. Results The study included 263 patients, with an overall 28-day mortality of 38%. Patients receiving ulinastatin showed significantly lower mortality than the control patients (31% vs. 55%; P<0.001). Ulinastatin use was associated with significantly reduced risk of death (OR: 0.317, 95% CI: 0.158-0.621; P=0.001) after adjustment for age, Sequential Organ Failure Assessment score, vasopressor use, and patient type as determined with a multivariable regression model. Conclusions Treatment with ulinastatin was associated with a decrease in 28-day mortality in critically ill septic patients.
Collapse
Affiliation(s)
- Qiancheng Xu
- Department of Critical Care Medicine, Wuhu No. 2 People's Hospital, Wannan Medical College, Wuhu 241000, China
| | - Qian Yan
- Department of Critical Care Medicine, Wuhu No. 2 People's Hospital, Wannan Medical College, Wuhu 241000, China
| | - Shanghua Chen
- Department of Critical Care Medicine, Wuhu No. 2 People's Hospital, Wannan Medical College, Wuhu 241000, China
| |
Collapse
|
82
|
Ding Y, Wang L, Zhao Q, Wu Z, Kong L. MicroRNA‑93 inhibits chondrocyte apoptosis and inflammation in osteoarthritis by targeting the TLR4/NF‑κB signaling pathway. Int J Mol Med 2018; 43:779-790. [PMID: 30569118 PMCID: PMC6317687 DOI: 10.3892/ijmm.2018.4033] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 12/07/2018] [Indexed: 01/16/2023] Open
Abstract
Osteoarthritis (OA) is a serious disease of the articular cartilage, and inflammation has been implicated in its pathogenesis. Previously, microRNAs (miRNAs) have been proposed as novel regulators of inflammation, however, the functional role of microRNAs in regulating inflammation in OA remains to be fully elucidated. The aim of the present study was to investigate the roles of miRNAs in OA inflammation and the underlying molecular mechanism. Firstly, the miRNA expression patterns were analyzed in the articular cartilage tissues from experimental OA mice using an miRNA microarray. miRNA (miR)-93 was identified with particular interest due to its reported effects on apoptosis and inflammation suppression. Subsequently, the expression of miR-93 was further validated in the articular cartilage tissues of OA mice and lipopolysaccharide (LPS)-stimulated primary chondrocytes. Using this LPS-induced chondrocyte injury model, the overexpression of miR-93 enhanced cell viability, improved cell apoptosis and attenuated the inflammatory response, as reflected by reductions in pro-inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6. In addition, Toll-like receptor 4 (TLR4), an important regulator of the nuclear factor-κB (NF-κB) signaling pathway, was identified as a direct target of miR-93 in chondrocytes. Furthermore, the restoration of TLR4 markedly abrogated the inhibitory effects of miR-93 on the chondrocyte apoptosis and inflammation induced by LPS. In addition, the overexpression of miR-93 by agomir-miR-93 significantly inhibited the levels of pro-inflammatory cytokines and cell apoptosis, whereas antagomir-93 exacerbated apoptosis and inflammation in vivo. Taken together, the results of the study suggested that miR-93 may be a promising therapeutic target for the treatment of human OA.
Collapse
Affiliation(s)
- Yanjie Ding
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Laifang Wang
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Qing Zhao
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Zhenzhen Wu
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Lingli Kong
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| |
Collapse
|
83
|
What's New in Shock, November 2018? Shock 2018; 50:501-503. [PMID: 30320673 DOI: 10.1097/shk.0000000000001223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
84
|
Ulinastatin is effective in reducing mortality for critically ill patients with sepsis: a causal mediation analysis. Sci Rep 2018; 8:14360. [PMID: 30254204 PMCID: PMC6156583 DOI: 10.1038/s41598-018-32533-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/10/2018] [Indexed: 12/29/2022] Open
Abstract
Ulinastatin has been found to have anti-inflammatory effect for patients with sepsis. However, its clinical effects were conflicting. The study aimed to investigate the cost-effectiveness of ulinastatin and to perform mediation analysis to explore the proportion of the total effects that can be explained by inflammatory responses. This is a retrospective study involving critically ill patients with sepsis from January 2014 to July 2017. A total of 263 patients were included in the study, involving 179 patients in the ulinastatin group and 84 in the control group. Ulinastatin group showed significantly lower 28-day mortality rate than that in the control group (31% vs. 55%; p < 0.001). Both total (46330 [26000,83500] vs. 19870 [8747,41140] RMB; p < 0.01) and drug cost (18210 [9492,31920] vs. 7230 [2675,19270] RMB; p < 0.01) were significantly higher in the ulinastatin group than the control group. In multivariable model, the adjusted odds ratio for ulinastatin was 0.304 (95% CI: 0.152 to 0.592; p = 0.001). The mediation analysis showed that the use of ulinastatin was able to reduce the probability of death by 23.5%. The average causal mediation effect of delta C-reactive protein (CRP) was 8%, accounting for 35% of the total effect.
Collapse
|