1
|
Zhang J, Zhao H, Wang F, Zhou J, Li M, Li H, Ren M, Wang L, Ren Q, Zhong X, Jiang X, Zhang Z. Asiaticoside alleviates lipopolysaccharide-induced acute lung injury by blocking Sema4D/CD72 and inhibiting mitochondrial dysfunction in RAW264.7 cell and mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7573-7587. [PMID: 38664244 PMCID: PMC11450039 DOI: 10.1007/s00210-024-03091-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/08/2024] [Indexed: 10/04/2024]
Abstract
Acute lung injury (ALI) is a common disease with complex pathogenesis. However, the treatment is mainly symptomatic with limited clinical options. Asiaticoside (AS), a Chinese herbal extract, has protective effects against LPS-induced ALI in mice and inhibits nitric oxide and prostaglandin E2 synthesis; however, the specific mechanism of AS in the prevention and treatment of LPS-induced ALI needs further study. Sema4D/CD72 pathway, mitochondrial dysfunction, and miRNA-21 are closely associated with inflammation. Therefore, the present study aimed to explore whether AS exerts its therapeutic effect on ALI by influencing Sema4D/CD72 pathway and mitochondrial dysfunction, restoring the balance of inflammatory factors, and influencing miRNA-21 expression. Cell and animal experiments were performed to investigate the effect of AS on ALI. Lipopolysaccharide (LPS) was used to establish the ALI model. CCK8 and flow cytometry were used to detect the cell viability and apoptosis rate. HE staining and wet-to-dry weight ratio (W/D) of lung tissue were determined. The expressions of Sema4D, CD72, NF-κB p65, Bax, Bcl2, and caspase 3 in RAW264.7 cells and lung tissues were detected by western blot, and the levels of IL-10 and IL-1β induced by LPS in supernatant of RAW264.7 cells and BALF were measured by ELISA. And the expression of miRNA-21 in cells and lung tissues was detected by fluorescence quantitative PCR. The result shows that AS treatment suppressed LPS-induced cell damage and lung injury in mice. AS treatment could alleviate the pathological changes such as inflammatory infiltration and histopathological changes in the lungs caused by LPS, and reduce the ratio of W/D. AS significantly alleviated the decrease of mitochondrial membrane potential induced by LPS, inhibited the increase of ROS production, and reduced the expression of mitochondrial fission proteins Drp1 and Fis1. The high-dose AS group significantly downregulated the expression of Sema4D, CD72, phosphorylated NF-κB p65, and apoptosis-related proteins, decreased the pro-inflammatory factor IL-1β, and enhanced the level of anti-inflammatory factor IL-10. In addition, AS promoted miRNA-21 expression. These effects inhibited apoptosis and restored the balance between anti- and pro-inflammatory factors. This represents the inaugural report elucidating the mechanism by which AS inhibits the Sema4D/CD72 signaling pathway. These findings offer novel insights into the potential application of AS in both preventing and treating ALI.
Collapse
Affiliation(s)
- Jianhua Zhang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Anesthesiology, Luzhou People's Hospital, Luzhou, China
| | - Hao Zhao
- Department of Pharmacy, The Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, China
| | - Fang Wang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jie Zhou
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Mao Li
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hua Li
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Meiping Ren
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Lulu Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qingyi Ren
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaolin Zhong
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xian Jiang
- Department of Anesthesiology, Luzhou People's Hospital, Luzhou, China.
| | - Zhuo Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.
| |
Collapse
|
2
|
Fan J, Zheng S, Wang M, Yuan X. The critical roles of caveolin-1 in lung diseases. Front Pharmacol 2024; 15:1417834. [PMID: 39380904 PMCID: PMC11458383 DOI: 10.3389/fphar.2024.1417834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
Caveolin-1 (Cav-1), a structural and functional component in the caveolae, plays a critical role in transcytosis, endocytosis, and signal transduction. Cav-1 has been implicated in the mediation of cellular processes by interacting with a variety of signaling molecules. Cav-1 is widely expressed in the endothelial cells, smooth muscle cells, and fibroblasts in the various organs, including the lungs. The Cav-1-mediated internalization and regulation of signaling molecules participate in the physiological and pathological processes. Particularly, the MAPK, NF-κB, TGFβ/Smad, and eNOS/NO signaling pathways have been involved in the regulatory effects of Cav-1 in lung diseases. The important effects of Cav-1 on the lungs indicate that Cav-1 can be a potential target for the treatment of lung diseases. A Cav-1 scaffolding domain peptide CSP7 targeting Cav-1 has been developed. In this article, we mainly discuss the structure of Cav-1 and its critical roles in lung diseases, such as pneumonia, acute lung injury (ALI), asthma, chronic obstructive pulmonary disease (COPD), pulmonary hypertension, pulmonary fibrosis, and lung cancer.
Collapse
Affiliation(s)
| | | | | | - Xiaoliang Yuan
- Department of Respiratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
3
|
Yin Y, Zeng Z, Wei S, Shen Z, Cong Z, Zhu X. Using the sympathetic system, beta blockers and alpha-2 agonists, to address acute respiratory distress syndrome. Int Immunopharmacol 2024; 139:112670. [PMID: 39018694 DOI: 10.1016/j.intimp.2024.112670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
Acute Respiratory Distress Syndrome (ARDS) manifests as an acute inflammatory lung injury characterized by persistent hypoxemia, featuring a swift onset, high mortality, and predominantly supportive care as the current therapeutic approach, while effective treatments remain an area of active investigation. Adrenergic receptors (AR) play a pivotal role as stress hormone receptors, extensively participating in various inflammatory processes by initiating downstream signaling pathways. Advancements in molecular biology and pharmacology continually unveil the physiological significance of distinct AR subtypes. Interventions targeting these subtypes have the potential to induce specific alterations in cellular and organismal functions, presenting a promising avenue as a therapeutic target for managing ARDS. This article elucidates the pathogenesis of ARDS and the basic structure and function of AR. It also explores the relationship between AR and ARDS from the perspective of different AR subtypes, aiming to provide new insights for the improvement of ARDS.
Collapse
Affiliation(s)
- Yiyuan Yin
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Zhaojin Zeng
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Senhao Wei
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Ziyuan Shen
- Department of Anaesthesiology, Peking University Third Hospital, Beijing, China
| | - Zhukai Cong
- Department of Anaesthesiology, Peking University Third Hospital, Beijing, China.
| | - Xi Zhu
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
4
|
Lu X, Li G, Liu Y, Luo G, Ding S, Zhang T, Li N, Geng Q. The role of fatty acid metabolism in acute lung injury: a special focus on immunometabolism. Cell Mol Life Sci 2024; 81:120. [PMID: 38456906 PMCID: PMC10923746 DOI: 10.1007/s00018-024-05131-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/06/2024] [Accepted: 01/17/2024] [Indexed: 03/09/2024]
Abstract
Reputable evidence from multiple studies suggests that excessive and uncontrolled inflammation plays an indispensable role in mediating, amplifying, and protracting acute lung injury (ALI). Traditionally, immunity and energy metabolism are regarded as separate functions regulated by distinct mechanisms, but recently, more and more evidence show that immunity and energy metabolism exhibit a strong interaction which has given rise to an emerging field of immunometabolism. Mammalian lungs are organs with active fatty acid metabolism, however, during ALI, inflammation and oxidative stress lead to a series metabolic reprogramming such as impaired fatty acid oxidation, increased expression of proteins involved in fatty acid uptake and transport, enhanced synthesis of fatty acids, and accumulation of lipid droplets. In addition, obesity represents a significant risk factor for ALI/ARDS. Thus, we have further elucidated the mechanisms of obesity exacerbating ALI from the perspective of fatty acid metabolism. To sum up, this paper presents a systematical review of the relationship between extensive fatty acid metabolic pathways and acute lung injury and summarizes recent advances in understanding the involvement of fatty acid metabolism-related pathways in ALI. We hold an optimistic believe that targeting fatty acid metabolism pathway is a promising lung protection strategy, but the specific regulatory mechanisms are way too complex, necessitating further extensive and in-depth investigations in future studies.
Collapse
Affiliation(s)
- Xiao Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Guorui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Guoqing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Tianyu Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| |
Collapse
|
5
|
Zhang B, Shen J. Dexmedetomidine activates the PKA/CREB pathway and inhibits proinflammatory factor expression through β2 adrenergic receptors. Immun Inflamm Dis 2024; 12:e1176. [PMID: 38411331 PMCID: PMC10898205 DOI: 10.1002/iid3.1176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 01/02/2024] [Accepted: 01/18/2024] [Indexed: 02/28/2024] Open
Abstract
INTRODUCTION Dexmedetomidine (DEX) is primarily utilized for sedation in the context of general anesthesia or intensive care. However, the exact regulatory mechanism by which DEX affects cytokines remains unclear. This study aims to investigate the underlying mechanism by which DEX inhibits proinflammatory factors through activation of the β2 adrenergic receptor (β2 AR). METHODS The inflammatory cell model of human mononuclear macrophage (THP-1) cells induced by lipopolysaccharide (LPS) was established to study the effect of DEX on the expression of cell-related inflammatory factors. ADRA2A gene knockout THP-1 cells (THP-1KO ) were constructed by CRISPR technology, and the effect of DEX on the expression of inflammatory factors of THP-1KO cells was detected. The target sites of DEX on β2 AR were screened by molecular docking. Reversion experiments were performed using ADRB2-siRNA. Western blot was used to detect the activation of β2 AR/PKA/CREB pathway and NF-κB, and ELISA was used to detect the release level of inflammatory factors. RESULTS The results demonstrated a significant reduction in the expression levels of MCP-1, interleukin-06, and IL-8 in both THP-1 and THP-1KO cells when induced by LPS following treatment with DEX. Additionally, DEX treatment led to an increase in IL-10 expression. Immunofluorescence analysis revealed an upregulation of β2 AR expression after DEX treatment. Western blot results indicated that DEX notably enhanced the activation of the β2 AR and PKA/CREB pathways, while concurrently suppressing the activation of NF-κB. Notably, the use of ADRB2 siRNA reversed the effects of DEX treatment and promoted the release of inflammatory cytokines. CONCLUSION DEX initiates the activation of the PKA/CREB pathway through the activation of β2 AR. Simultaneously, it exerts an inhibitory effect on the activation of NF-κB, consequently reducing the transcription of proinflammatory factors while increasing the transcription of anti-inflammatory factors.
Collapse
Affiliation(s)
- Baocheng Zhang
- Department of Emergency and Critical Care CenterJinshan Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Jie Shen
- Department of Emergency and Critical Care CenterJinshan Hospital Affiliated to Fudan UniversityShanghaiChina
| |
Collapse
|
6
|
Şenol N, Şahin M, Şahin U. The protective role of 5-hydroxy-1,4-naphthoquinone against the harmful effects of 50 Hz electric field in rat lung tissue. Electromagn Biol Med 2023; 42:133-143. [PMID: 37811636 DOI: 10.1080/15368378.2023.2265935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 09/16/2023] [Indexed: 10/10/2023]
Abstract
There is strong scientific evidence that the electric field is harmful to life. Exposure to an electric field (EF) can cause lung toxicity and respiratory disorders. In addition, the electric field has been shown to cause tissue damage through inflammation and apoptosis. Juglone (JUG) is one of the powerful antioxidants with anti-apoptotic and anti-inflammatory, various pharmacological properties in the biological system. In this study, we evaluated the efficacy of JUG against the potential adverse effects of electric field on the lung. Twenty-four Wistar albino rats were randomly divided into three groups; control group (Cont), EF group, and EF exposure+JUG-treated group (EJUG). After routine histological procedures, sections stained with hematoxylin-eosin (H&E) showed significant changes in lung tissues in the EF group compared to the Cont group. Significant protective effects were observed in the building volumes and histopathology in the EJUG group. Our immunohistochemical and gene expression results increased the expression of caspase-3 and tumor necrosis factor alpha (TNF-α) in the EF group (p < 0.05). Juglon increased cytokine signal suppressor (SOCS) expression (p < 0.001). These findings were consistent with the antioxidant effect of JUG treatment. We reasoned that exposure to EF damaged rat lung tissues and administration of JUG alleviated the complications caused by 50 Hz EF.
Collapse
Affiliation(s)
- Nurgül Şenol
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Süleyman Demirel University, Isparta, Türkiye
| | - Melda Şahin
- Department of Bioengineering, Institute of Science, Süleyman Demirel University, Isparta, Türkiye
| | - Uğur Şahin
- Department of Chemistry, Faculty of Art and Science, University of Süleyman Demirel, Isparta, Türkiye
- Genetic Research Unit, Innovative Technologies Application and Research Center, Süleyman Demirel University, Isparta, Türkiye
| |
Collapse
|
7
|
Shu Y, Jin S. Caveolin-1 in endothelial cells: A potential therapeutic target for atherosclerosis. Heliyon 2023; 9:e18653. [PMID: 37554846 PMCID: PMC10405014 DOI: 10.1016/j.heliyon.2023.e18653] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/10/2023] Open
Abstract
Atherosclerosis (AS) is a chronic vascular disease characterized by lipid accumulation and the activation of the inflammatory response; it remains the leading nation-wide cause of death. Early in the progression of AS, stimulation by pro-inflammatory agonists (TNF-α, LPS, and others), oxidized lipoproteins (ox-LDL), and biomechanical stimuli (low shear stress) lead to endothelial cell activation and dysfunction. Consequently, it is crucial to investigate how endothelial cells respond to different stressors and ways to alter endothelial cell activation in AS development, as they are the earliest cells to respond. Caveolin-1 (Cav1) is a 21-24-kDa membrane protein located in caveolae and highly expressed in endothelial cells, which plays a vital role in regulating lipid transport, inflammatory responses, and various cellular signaling pathways and has atherogenic effects. This review summarizes recent studies on the structure and physiological functions of Cav1 and outlines the potential mechanisms it mediates in AS development. Included are the roles of Cav1 in the regulation of endothelial cell autophagy, response to shear stress, modulation of the eNOS/NO axis, and transduction of inflammatory signaling pathways. This review provides a rationale for proposing Cav1 as a novel target for the prevention of AS, as well as new ideas for therapeutic strategies for early AS.
Collapse
Affiliation(s)
- Yan Shu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, 430077, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, 430077, China
| |
Collapse
|
8
|
Wu C, Guo L, Muhataer X, Li Q, Lian Z, Li Y, Wang W, Ding W, Zhou Y, Yang X, Chen M. Interaction between the PI3K/AKT pathway and mitochondrial autophagy in macrophages and the leukocyte count in rats with LPS-induced pulmonary infection. Open Life Sci 2023; 18:20220588. [PMID: 37077346 PMCID: PMC10106970 DOI: 10.1515/biol-2022-0588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 02/26/2023] [Accepted: 03/02/2023] [Indexed: 04/21/2023] Open
Abstract
This study examined the effects of the PI3K/AKT pathway and mitochondrial autophagy in macrophages and the leukocyte count after pulmonary infection. Sprague‒Dawley rats were subjected to tracheal injection of lipopolysaccharide (LPS) to establish animal models of pulmonary infection. By inhibiting the PI3K/AKT pathway or inhibiting/inducing mitochondrial autophagy in macrophages, the severity of the pulmonary infection and the leukocyte count were altered. The PI3K/AKT inhibition group did not show a significant difference in leukocyte counts compared with the infection model group. Mitochondrial autophagy induction alleviated the pulmonary inflammatory response. The infection model group had significantly higher levels of LC3B, Beclin1, and p-mTOR than the control group. The AKT2 inhibitor group exhibited significantly increased levels of LC3B and Beclin1 compared with the control group (P < 0.05), and the Beclin1 level was significantly higher than that in the infection model group (P < 0.05). Compared with the infection model group, the mitochondrial autophagy inhibitor group exhibited significantly decreased levels of p-AKT2 and p-mTOR, whereas the levels of these proteins were significantly increased in the mitochondrial autophagy inducer group (P < 0.05). PI3K/AKT inhibition promoted mitochondrial autophagy in macrophages. Mitochondrial autophagy induction activated the downstream gene mTOR of the PI3K/AKT pathway, alleviated pulmonary inflammatory reactions, and decreased leukocyte counts.
Collapse
Affiliation(s)
- Chao Wu
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Tianshan District, 830001Urumqi, China
| | - Lianghua Guo
- Department of Respiratory Medicine, Mindong Hospital Affiliated to Fujian Medical University, 355000Fu’an City, China
| | - Xirennayi Muhataer
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Tianshan District, 830001Urumqi, China
| | - Qifeng Li
- Xinjiang Institute of Pediatrics, Children’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830054, China
| | - Zhichuang Lian
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Tianshan District, 830001Urumqi, China
| | - Yafang Li
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Tianshan District, 830001Urumqi, China
| | - Wenyi Wang
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Tianshan District, 830001Urumqi, China
| | - Wei Ding
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Tianshan District, 830001Urumqi, China
| | - Yuan Zhou
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Tianshan District, 830001Urumqi, China
| | - Xiaohong Yang
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Tianshan District, 830001Urumqi, China
| | - Muzhi Chen
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No. 318, Chaowang Road, Gongshu District, 310005Hangzhou, China
| |
Collapse
|
9
|
Cupping alleviates lung injury through the adenosine/A 2BAR pathway. Heliyon 2022; 8:e12141. [PMID: 36544817 PMCID: PMC9761715 DOI: 10.1016/j.heliyon.2022.e12141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/09/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Background Acute lung injury (ALI) is a serious condition. Inflammation plays a crucial role in the pathogenesis of ALI. Cupping, as a part of traditional Chinese medicine, is still a popular complementary and alternative therapy for a variety of ailments including respiratory diseases. However, reliable scientific data about cupping therapy are scarce. Adenosine, a purine nucleoside produced under metabolic stress by the action of extracellular ectonucleotidases (i.e. CD39 and CD73), can attenuate ALI through the A2BAR receptor. The aim of this study was to investigate the protective effect of cupping in a rat model of ALI and the role of adenosine in it. Methods Male adult rats were subjected to ALI by intratracheal LPS instillation (0.3 mg/kg). Immediately after intratracheal LPS instillation, vacuum pressure was applied to a sanitized plastic bell cup on the back of the rat by suction for 10 min. Pulmonary injury and inflammation were assessed at 4 h after LPS challenge. The role of adenosine and A2BAR in cupping's protection after LPS instillation were evaluated. Results Cupping alleviated LPS-induced lung injury, reduced inflammation and inhibited NF-kB activation in rats. Cupping upregulated CD39 and CD73 mRNA expression of the skin tissue at the cupping site and increased circulating levels of adenosine. Administration of PSB1115, a specific adenosine A2BAR receptor antagonist, abolished cupping's beneficial effects in LPS-induced ALI. Conclusions Cupping attenuates lung inflammation and injury through the adenosine/A2BAR pathway. The current study provides evidence-based information about cupping therapy in ALI.
Collapse
|
10
|
CHEN Z, RUAN B, LONG G, LIN W. Adipose tissue-derived mesenchymal stem cells attenuate lung inflammation and fibrosis in the bleomycin-induced pulmonary fibrosis rat model via caveolin-1/NF-kB signaling axis. Physiol Res 2022; 71:657-666. [PMID: 36047729 PMCID: PMC9841806 DOI: 10.33549/physiolres.934892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Stem cells have emerged as promising therapeutic options for several human diseases, including pulmonary fibrosis (PF). In this study, we investigated the therapeutic effects of adipose tissue-derived mesenchymal stem cells (ADMSCs) in the bleomycin-induced PF model rats and the underlying mechanisms. The PF model rats were generated by intratracheal injections of 5 mg/kg bleomycin sulfate. The ADMSC group rats were generated by injecting 2×10(6) ADMSCs via the tail vein at 0, 12, and 24 h after bleomycin injection. The control, PF, and ADMSC group rats were sacrificed on day 21 after bleomycin injections and the changes in lung histology and the levels of pro-inflammatory cytokines, collagen I, and caveolin-1 (Cav-1), and the activity of the NF-kappaB signaling pathway in the lung tissues was assessed by hematoxylin-eosin staining, ELISA, and western blotting assays. The lung tissues of the PF model rats showed significant infiltration of neutrophils, tissue destruction, and collagen deposition, but these effects were abrogated by the ADMSCs. The levels of pro-inflammatory cytokines such as IL-6, IL-1beta, and TGF-beta1 were elevated in the lung tissues and the bronchoalveolar lavage fluid (BALF) of the bleomycin-induced PF model rats, but these effects were reversed by the ADMSCs. The lung tissues of the PF model rats showed significant downregulation of Cav-1 and significantly higher activation of the pro-inflammatory NF-kappaB pathway. However, administration of the ADMSCs restored the expression levels of Cav-1 and suppressed the NF-kappaB signaling pathway in the lungs of the bleomycin-induced PF model rats. In conclusion, this study demonstrated that the ADMSCs protected against bleomycin-induced PF in the rat model by modulating the Cav-1/NF-kappaB axis.
Collapse
Affiliation(s)
- Zhe CHEN
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Wenling, Zhejiang, China
| | - Bingqing RUAN
- Department of Internal Medicine, Wenling Women’s and Children’s Hospital, Zhejiang, China
| | - Guangyan LONG
- Department of Infectious Diseases, The First People’s Hospital of Wenling, Zhejiang, China
| | - Wei LIN
- Department of Respirology, The First People’s Hospital of Wenling, Zhejiang, China
| |
Collapse
|
11
|
Zhi J, Duan Q, Wang QY, Du X, Yang D. Dexmedetomidine reduces IL-4 and IgE expression through downregulation of theTLR4/NF-κB signaling pathway to alleviate airway hyperresponsiveness in OVA mice. Pulm Pharmacol Ther 2022; 75:102147. [PMID: 35863724 DOI: 10.1016/j.pupt.2022.102147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/25/2022] [Accepted: 07/14/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Airway hyperresponsiveness (AHR) is a clinical manifestation of airflow limitation due to abnormal tracheal and bronchial sensitivity and is the main basis for the diagnosis of asthma. Patients with AHR are at high risk of perioperative tracheal and bronchospasm, which can lead to hypoxaemia and haemodynamic instability and, in severe cases, to a life-threatening 'silent lung'. It is therefore important to reduce the incidence or intensity of AHR episodes in the perioperative period. The inflammatory response is key to the development and progression of AHR. HYPOTHESIS/PURPOSE Based on the modulatory role of dexmedetomidine (DEX) in the inflammatory response, we hypothesised that dexmedetomidine (DEX) attenuates inflammatory properties by inhibiting the toll-like receptor 4 (TLR4)/nuclear factor (NF-κB) signalling pathway and can reduce the respiratory parameters of mechanical ventilation in ovalbumin-induced allergic airway hyperresponsiveness. STUDY DESIGN BABL/C mice were divided into control and OVA groups (ovalbumin-induced allergy. Ten mice in all OVA models were randomly selected for in vivo invasive lung function monitoring to analyse airway resistance parameters and demonstrate successful model establishment. The remaining OVA mice were treated with dexmedetomidine 25 μg/kg for 5 days (OVA + DEX group) or dexmedetomidine 25 μg/kg + yohimbine 1 mg/kg for 5 days (OVA + DEX + yohimbine). After treatment, bronchoalveolar lavage fluid (BAL) and peripheral blood (ELISA) and lung tissue (H&E and PAS) were collected for analysis of inflammatory factors, and lung tissue was verified by PCR for genes and proteins that do correlate with inflammatory mediators. RESULTS All airway resistance parameters were increased in OVA mice by invasive lung function monitoring. Proximal airway resistance (parameter Rn) and total respiratory resistance (parameter Rrs) were attenuated after dexmedetomidine intervention treatment. Dexmedetomidine reduced total inflammatory cell count and inflammatory infiltration of lung tissue in BALF and down-regulated IL-4 and IgE levels in BALF and peripheral blood, as shown by Giemsa, H&E, PAS staining and ELISA; this mechanism of action was found to be related to the TLR4/NFκB pathway, but not to TLR4/NFκB, as measured by PCR. CONCLUSION Dexmedetomidine reduces hyperresponsiveness and airway inflammatory responses. This mechanism of action may be related to the TLR4/NFκB signalling pathway. Overall conclusions are presented in.
Collapse
Affiliation(s)
- Juan Zhi
- Department of Anesthesia, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing, 100144, China.
| | - Qirui Duan
- Department of Anesthesia, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing, 100144, China.
| | - Qian Yu Wang
- Department of Anesthesia, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing, 100144, China
| | - Xiyu Du
- Huadong Hospital Affillated to Fudan University, 211 Yan'an West Road, Jing'an District, Shanghai, 200041, China.
| | - Dong Yang
- Department of Anesthesia, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing, 100144, China.
| |
Collapse
|
12
|
Han J, Liu X, Wang L. Dexmedetomidine protects against acute lung injury in mice via the DUSP1/MAPK/NF-κB axis by inhibiting miR-152-3p. Pulm Pharmacol Ther 2022:102131. [PMID: 35551994 DOI: 10.1016/j.pupt.2022.102131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 04/07/2022] [Accepted: 05/01/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Acute lung injury (ALI) is a debilitating condition in clinics. Dexmedetomidine (Dex) is known for its anti-apoptotic and anti-inflammatory properties. This study attempted to investigate the protective mechanism of Dex in ALI mice. METHODS Mice were pretreated with Dex before model establishment by tracheal injection of lipopolysaccharide (LPS). Pulmonary function indexes and wet-to-dry (W/D) ratio were measured. Pulmonary pathological changes were observed through HE staining, CD31+-positive mouse pulmonary microvascular endothelial cells (MPMVECs) were counted through immunofluorescence staining, and apoptosis was detected through TUNEL staining. miR-152-3p mimic, sh-DUSP1, or p38 MAPK inhibitor was delivered into MPMVECs, followed by combined treatment of Dex and LPS. miR-152-3p expression, apoptosis, levels of apoptosis- and MAPK/NF-κB pathway-associated proteins, and inflammatory factors were measured through RT-qPCR, flow cytometry, Western blot, and ELISA. The binding relationship of miR-152-3p and DUSP1 was verified through bioinformatics software and dual-luciferase assay. ALI mouse model was established after injection of miR-152-3p antagomir. RESULTS Dex improved ALI mouse pulmonary function and mitigated injury in mice and MPMVECs. miR-125-3p overexpression or sh-DUSP1 partially abolished the protection of Dex on MPMVECs. miR-152-3p targeted DUSP1. sh-DUSP1 partially averted the protection of Dex on MPMVECs. Dex inhibited the activation of the MAPK/NF-κB pathway in MPMVECs mediated by LPS, which was partially reversed by sh-DUSP1. The p38 MAPK inhibitor SB203580 antagonized the protective effect of Dex on MPMVECs mediated by sh-DUSP1. Similarly, downregulation of miR-152-3p mitigated ALI via the DUSP1/MAPK/NF-κB axis in vivo. CONCLUSION Dex relieved ALI in mice via the DUSP1/MAPK/NF-κB axis by down-regulating miR-152-3p.
Collapse
Affiliation(s)
- Jieran Han
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Xiao Liu
- Department of Anesthesiology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471003, China
| | - Ling Wang
- Department of Anesthesiology, 989 Hospital of JOINT Logistic Support Force of PLA, Luoyang, 471031, China.
| |
Collapse
|
13
|
Li Y, Wu B, Hu C, Hu J, Lian Q, Li J, Ma D. The role of the vagus nerve on dexmedetomidine promoting survival and lung protection in a sepsis model in rats. Eur J Pharmacol 2022; 914:174668. [PMID: 34863997 DOI: 10.1016/j.ejphar.2021.174668] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/27/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Sepsis often results in acute lung injury (ALI). Dexmedetomidine (Dex) was reported to protect cells and organs due to its direct cellular effects. This study aims to investigate the role of vagus nerves on Dex induced lung protection in lipopolysaccharide (LPS)-induced ALI rats. METHODS The bilateral cervical vagus nerve of male Sprague-Dawley rats was sectioned or just exposed as sham surgery. After LPS administration, Dex antagonist yohimbine (YOH) and/or Dex was injected intraperitoneally to rats with or without vagotomy. The severity of ALI was determined with survival curve analysis and lung pathological scores. The plasma concentrations of interleukin 1 beta (IL-1β), tumor necrosis factor-alpha (TNF-α), catecholamine and acetylcholine were measured with enzyme-linked immunosorbent assay. RESULTS The median survival time of LPS-induced ALI rats was prolonged by Dex (22 h, 95% CI, [24.46, 92.20]) vs. 14 h, 95% CI, [14.60, 89.57] of the LPS control group, P < 0.05), and the ALI score was reduced by Dex (6.5, 95% CI, [5.23, 8.10] vs. 11.5, 95% CI, [10.23, 13.10] in the LPS group, P < 0.01). However, these protective effects were significantly decreased by either YOH administration or vagotomy. Dex decreased LPS-induced IL-1β, TNF-α, and catecholamine but increased acetylcholine in blood serum; these effects of Dex was partially abolished by vagotomy. CONCLUSIONS Our data suggested that Dex increased vagal nerve tone that partially contributed to its anti-inflammatory and lung-protective effects. The indirect anti-inflammation and direct cytoprotection of Dex are likely through high vagal nerve tone and α2-adrenoceptor activation, respectively.
Collapse
Affiliation(s)
- Yumo Li
- Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Binbin Wu
- Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Cong Hu
- Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Jie Hu
- Department of Anesthesiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Qingquan Lian
- Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun Li
- Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom.
| |
Collapse
|
14
|
Jiang Z, Tan J, Yuan Y, Shen J, Chen Y. Semaglutide ameliorates lipopolysaccharide-induced acute lung injury through inhibiting HDAC5-mediated activation of NF-κB signaling pathway. Hum Exp Toxicol 2022; 41:9603271221125931. [PMID: 36075570 DOI: 10.1177/09603271221125931] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND As a life-threatening respiratory syndrome, acute lung injury (ALI) is characterized by uncontrollable inflammatory activities. Semaglutide (SEM) has been identified as an effective anti-inflammatory drug in a variety of diseases. This study intended to explore the functional effect and potential mechanisms of SEM in ALI. METHODS Lipopolysaccharide (LPS) was used to construct an in vivo ALI model based on Sprague-Dawley (SD) rats and an in vitro ALI model based on human pulmonary artery endothelial cells (HPAECs). Hematoxylin & eosin (H&E) staining and ELISA were applied to evaluate the histopathological changes in pulmonary tissues and detect TNF-α and IL-6 levels. RT-qPCR and Western blotting were used to measure gene and protein expressions in pulmonary tissues and cells. HPAEC viability and apoptosis were evaluated by CCK-8 method and flow cytometry methods. RESULTS Semaglutide pretreatment significantly mitigated pulmonary injury, reduced TNF-α and IL-6 production, and led to a decrease in cleaved caspase-3 level and an increase in Bcl-2 level, suggesting SEM could ameliorate LPS-induced ALI in rats. In vitro, SEM increased the proliferative capability and mitigated inflammation and apoptosis in LPS-stimulated HPAECs. In addition, SEM inhibited HDAC5-mediated NF-κB signaling pathway in HPAECs. HDAC5 overexpression or NF-κB signaling activation could partly impair SEM-mediated protective effects against LPS-induced damage to HPAECs. CONCLUSION Semaglutide restrains LPS-induced ALI by inhibiting HDAC5/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zeyu Jiang
- Department of Anesthesiology, 117850The First People's Hospital of Changzhou, P.R. China
| | - Jinyi Tan
- Department of Anesthesiology, Changzhou Children's Hospital, P.R. China
| | - Yan Yuan
- Department of Anesthesiology, 117850The First People's Hospital of Changzhou, P.R. China
| | - Jiang Shen
- Department of Anesthesiology, 117850The First People's Hospital of Changzhou, P.R. China
| | - Yan Chen
- Department of Anesthesiology, 117850The First People's Hospital of Changzhou, P.R. China
| |
Collapse
|
15
|
He R, Yuan X, Lv X, Liu Q, Tao L, Meng J. Caveolin-1 negatively regulates inflammation and fibrosis in silicosis. J Cell Mol Med 2021; 26:99-107. [PMID: 34889029 PMCID: PMC8742238 DOI: 10.1111/jcmm.17045] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/30/2021] [Accepted: 10/24/2021] [Indexed: 11/28/2022] Open
Abstract
Inhalation of crystalline silica causes silicosis, the most common and serious occupational disease, which is characterized by progressive lung inflammation and fibrosis. Recent studies revealed the anti-inflammatory and anti-fibrosis role of Caveolin-1 (Cav-1) in lung, but this role in silicosis has not been investigated. Thus, this study evaluated Cav-1 regulatory effects in silicosis. It was found that Cav-1 levels were significantly reduced in the lung from silicosis patients and silicotic mice. The silicosis models were established in C57BL/6 (wild-type) and Cav-1 deficiency (Cav-1-/- ) mice, and Cav-1-/- mice displayed wider alveolar septa, increased collagen deposition and more silicotic nodules. The mice peritoneal-derived macrophages were used to explore the role of Cav-1 in silica-induced inflammation, which plays a central role in mechanism of silicosis. Cav-1 inhibited silica-induced infiltration of inflammatory cells and secretion of inflammatory factors in vitro and in vivo, partly by downregulating NF-κB pathway. Additionally, silica uptake and expression of 4-hydroxynonenal in silicotic mice were observed, and it was found that Cav-1 absence triggered excessive silica deposition, causing a stronger oxidative stress response. These findings demonstrate the protective effects of Cav-1 in silica-induced lung injury, suggesting its potential therapeutic value in silicosis.
Collapse
Affiliation(s)
- RongLing He
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
| | - XiangNing Yuan
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China.,Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Lv
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China.,Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - QingXiang Liu
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
| | - LiJian Tao
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China.,Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Meng
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
| |
Collapse
|
16
|
Sun H, Hu H, Xu X, Fang M, Tao T, Liang Z. Protective effect of dexmedetomidine in cecal ligation perforation-induced acute lung injury through HMGB1/RAGE pathway regulation and pyroptosis activation. Bioengineered 2021; 12:10608-10623. [PMID: 34747306 PMCID: PMC8810048 DOI: 10.1080/21655979.2021.2000723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/02/2022] Open
Abstract
Dexmedetomidine (DEX) has been reported to attenuate cecal ligation perforation (CLP)-stimulated acute lung injury (ALI) by downregulating HMGB1 and RAGE. This study aimed to further investigate the specific mechanisms of RAGE and its potential-related mechanisms of DEX on ALI models in vitro and in vivo. The in vitro and in vivo ALI models were established by lipopolysaccharide treatment in MLE-12 cells and CLP in mice, respectively. The effect of DEX on pathological alteration was investigated by HE staining. Thereafter, the myeloperoxidase (MPO) activity and inflammatory cytokine levels were respectively detected to assess the lung injury of mice using commercial kits. The expression levels of HMGB1, RAGE, NF-κB, and pyroptosis-related molecules were detected by RT-qPCR and Western blot. HE staining showed that lung injury, increased inflammatory cell infiltration, and lung permeability was found in the ALI mice, and DEX treatment significantly attenuated lung tissue damage induced by CLP. The MPO activity and inflammatory cytokines (TNF-α, IL-1β, and NLRP3) levels were also significantly reduced after DEX treatment compared with those in the ALI mice. Moreover, DEX activated the HMGB1/RAGE/NF-κB pathway and upregulated the pyroptosis-related proteins. However, the protective DEX effect was impaired by RAGE overexpression in ALI mice and MLE-12 cells. Additionally, DEX treatment significantly suppressed HMGB1 translocation from the nucleus region to the cytoplasm, and this effect was reversed by RAGE overexpression. These findings suggested that DEX may be a useful ALI treatment, and the protective effects on ALI mice may be through the inhibition of HMGB1/RAGE/NF-κB pathway and cell pyroptosis.
Collapse
Affiliation(s)
- Huaqin Sun
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Hongyi Hu
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaoping Xu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Laboratory Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Mingsun Fang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Laboratory Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Tao Tao
- Department of Anesthesiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhehao Liang
- Department of Ultrasound, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
17
|
Zhou HH, Zhang YM, Zhang SP, Xu QX, Tian YQ, Li P, Cao D, Zheng YQ. Suppression of PTRF Alleviates Post-Infectious Irritable Bowel Syndrome via Downregulation of the TLR4 Pathway in Rats. Front Pharmacol 2021; 12:724410. [PMID: 34690766 PMCID: PMC8529073 DOI: 10.3389/fphar.2021.724410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Accumulating evidence suggests that the polymerase I and transcript release factor (PTRF), a key component of the caveolae structure on the plasma membrane, plays a pivotal role in suppressing the progression of colorectal cancers. However, the role of PTRF in the development of functional gastrointestinal (GI) disorders remains unclear. Post-infectious irritable bowel syndrome (PI-IBS) is a common functional GI disorder that occurs after an acute GI infection. Here, we focused on the role of PTRF in the occurrence of PI-IBS and investigated the underlying mechanisms. Methods: Lipopolysaccharide (LPS) (5 μg/ml) was used to induce inflammatory injury in human primary colonic epithelial cells (HCoEpiCs). Furthermore, a rat model of PI-IBS was used to study the role of PTRF. Intestinal sensitivity was assessed based on the fecal water content. A two-bottle sucrose intake test was used to evaluate behavioral changes. Furthermore, shRNA-mediated knockdown of PTRF was performed both in vitro and in vivo. We detected the expression of PTRF in colonic mucosal tissues through immunohistochemistry (IHC), western blotting (WB), and immunofluorescence (IF) analysis. Luciferase activity was quantified using a luciferase assay. Co-localization of PTRF and Toll-like receptor 4 (TLR4) was detected using IF analysis. The activation of the signaling pathways downstream of TLR4, including the iNOs, p38, extracellular signal-regulated kinase (ERK), and c-Jun N-terminal kinase (JNK) pathways, was detected via WB. The levels of NO, IL-1β, IL-6, and TNF-α were measured using enzyme-linked immunosorbent assays. Results: LPS significantly induced PTRF expression and signaling downstream of TLR4, including p38, ERK, and JNK pathways, in HCoEpiCs. Moreover, shRNA-mediated knockdown of PTRF in HCoEpiCs significantly decreased the phosphorylation of JNK, ERK, and p38 and iNOS expression. In PI-IBS rats, the lack of PTRF not only reduced fecal water content and suppressed depressive behavior but also increased the body weight. Furthermore, we found a strong co-localization pattern for PTRF and TLR4. Consistently, the lack of PTRF impaired TLR4 signaling, as shown by the decreased levels of p-JNK, p-ERK, and p-p38, which are upstream factors involved in iNOS expression. Conclusion: PTRF promoted PI-IBS and stimulated TLR4 signaling both in vitro and in vivo. The results of this study not only enlighten the pathogenesis of PI-IBS but also help us understand the biological activity of PTRF and provide an important basis for the clinical treatment of PI-IBS by targeting PTRF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Di Cao
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Teaching and Research Section of Traditional Chinese Medicine, School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Yong-qiu Zheng
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Teaching and Research Section of Traditional Chinese Medicine, School of Pharmacy, Wannan Medical College, Wuhu, China
| |
Collapse
|
18
|
Zhang C, Zhao Y, Yang X. Azilsartan attenuates lipopolysaccharide-induced acute lung injury via the Nrf2/HO-1 signaling pathway. Immunol Res 2021; 70:97-105. [PMID: 34608599 DOI: 10.1007/s12026-021-09240-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/25/2021] [Indexed: 11/30/2022]
Abstract
Acute lung injury (ALI) is a severe complication of sepsis and hemorrhagic shock with high morbidity. In the present study, the protective effect of Azilsartan on lipopolysaccharide (LPS)-induced ALI in mice was investigated to explore the potential therapeutic property of Azilsartan for the treatment of ALI. LPS was used to induce an ALI model in mice. Hematoxylin-eosin (HE) staining sections were then evaluated for the pathological state of lung tissues. Bronchoalveolar lavage fluid (BALF) protein concentration, wet/dry weight ratios of lung tissues, and pulmonary myeloperoxidase (MPO) activity were detected to determine the degree of pulmonary injury. The number of total cells, macrophages, and neutrophils in BALF were counted using a hemocytometer to illustrate the inflammatory cell infiltration. The lung function was monitored using a spirometer. The concentrations of interleukin-1β (IL-1β), monocyte chemoattractant protein-1 (MCP-1), and interleukin-8 (IL-8) were determined using enzyme-linked immunosorbent assay (ELISA). Oxidative stress was evaluated by the superoxide dismutase (SOD) activity, glutathione (GSH), and malondialdehyde (MDA) concentrations in the lung tissue. The expressions of nuclear erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) were determined using Western blot analysis. Azilsartan therapy alleviated LPS-induced lung tissue damage, increased BALF protein concentration, lung wet to dry weight ratio, MPO activity, and macrophage and neutrophils infiltration. Also, Azilsartan ameliorated the production of inflammatory factors (IL-1β, MCP-1, and IL-8). Azilsartan ameliorated LPS-impaired lung SOD activity, the GSH concentration, and the MDA concentration. Mechanistically, Azilsartan activated the LPS-impaired Nrf2/HO-1 signaling pathway. Azilsartan therapy attenuates LPS-induced ALI via the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Chengshi Zhang
- Department of Respiratory, Punan Hospital, Pudong New Area, Shanghai, 200125, China
| | - Yunfeng Zhao
- Department of Respiratory, Punan Hospital, Pudong New Area, Shanghai, 200125, China
| | - Xiaorong Yang
- Department of Endocrinology, Punan Hospital, No.279, Linyi Road, Pudong New Area, Shanghai, 200125, China.
| |
Collapse
|
19
|
Li G, Xu M, Wang H, Qi X, Wang X, Li Y, Sun J, Li Y. MicroRNA-146a overexpression alleviates intestinal ischemia/reperfusion-induced acute lung injury in mice. Exp Ther Med 2021; 22:937. [PMID: 34335886 PMCID: PMC8290461 DOI: 10.3892/etm.2021.10369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/01/2021] [Indexed: 12/31/2022] Open
Abstract
Previous studies have shown that microRNAs (miRs), such as miR-146a play an important role in the pathogenesis of intestinal ischemia/reperfusion (I/R)-induced injury; however, the role of miR-146a in intestinal I/R-induced acute lung injury has not been elucidated. An intestinal I/R-induced injury mouse model was established in the present study by clamping the superior mesenteric artery and expression levels of miR-146a in intestinal and lung tissue samples were evaluated using reverse transcription-quantitative PCR (RT-qPCR). Intestinal and lung histopathological characteristics in mice with intestinal I/R-induced injury were assessed by hematoxylin and eosin staining, and mRNA and protein expression levels in intestinal and lung tissue samples were evaluated using RT-qPCR and western blotting, respectively. miR-146a expression was significantly downregulated in the intestinal and lung tissue samples of mice with intestinal I/R-induced injury. Intestinal I/R injury-induced histopathological changes in the lung and intestines, and pulmonary edema in mice transduced with an adenoviral miR-146a-overexpression vector (the miR-146a overexpression group) were alleviated. mRNA expression levels of TNF-α, IL-1β, IFN-γ and TGF-β1, and protein expression levels of TNF receptor-associated factor 6, phosphorylated-p65 NF-κB, cleaved caspase-3 and cleaved caspase-9 in lung and intestinal tissue samples were downregulated in I/R-miR-146a-overexpressing mice, compared with those from the I/R-negative control group. Thus, the present study identified that pre-treatment with the miR-146a overexpression vector alleviated intestinal I/R-induced acute lung injury in mice.
Collapse
Affiliation(s)
- Gehui Li
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Min Xu
- Department of Anesthesiology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei 441000, P.R. China
| | - Hao Wang
- Department of Food Safety, Market Supervision Administration of Shenzhen Municipality, Shenzhen, Guangdong 518040, P.R. China
| | - Xiaofei Qi
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Xiaoguang Wang
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Yong Li
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Jing Sun
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Yuantao Li
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| |
Collapse
|
20
|
Shi J, Yu T, Song K, Du S, He S, Hu X, Li X, Li H, Dong S, Zhang Y, Xie Z, Li C, Yu J. Dexmedetomidine ameliorates endotoxin-induced acute lung injury in vivo and in vitro by preserving mitochondrial dynamic equilibrium through the HIF-1a/HO-1 signaling pathway. Redox Biol 2021; 41:101954. [PMID: 33774474 PMCID: PMC8027777 DOI: 10.1016/j.redox.2021.101954] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/08/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Increasing lines of evidence identified that dexmedetomidine (DEX) exerted protective effects against sepsis-stimulated acute lung injury via anti-inflammation, anti-oxidation and anti-apoptosis. However, the mechanisms remain unclear. Herein, we investigated whether DEX afforded lung protection by regulating the process of mitochondrial dynamics through the HIF-1a/HO-1 pathway in vivo and in vitro. Using C57BL/6J mice exposed to lipopolysaccharide, it was initially observed that preemptive administration of DEX (50μg/kg) alleviated lung pathologic injury, reduced oxidative stress indices (OSI), improved mitochondrial dysfunction, upregulated the expression of HIF-1α and HO-1, accompanied by shifting the dynamic course of mitochondria into fusion. Moreover, HO-1-knockout mice or HO-1 siRNA transfected NR8383 cells were pretreated with HIF-1α stabilizer DMOG and DEX to validate the effect of HIF-1a/HO-1 pathway on DEX-mediated mitochondrial dynamics in a model of endotoxin-induced lung injury. We found that pretreatment with DEX and DMOG distinctly relieved lung injury, decreased the levels of mitochondrial ROS and mtDNA, reduced OSI, increased nuclear accumulation of HIF-1a and HO-1 protein in wild type mice but not HO-1 KO mice. Similar observations were recapitulated in NC siRNA transfected NR8383 cells after LPS stimulation but not HO-1 siRNA transfected cells. Concertedly, DEX reversed the impaired mitochondrial morphology in LPS stimulated-wild type mice or NC siRNA transfected NR8383 cells, upregulated the expression of mitochondrial fusion protein, while downregulated the expression of fission protein in HIF-1a/HO-1 dependent pathway. Altogether, our data both in vivo and in vitro certified that DEX treatment ameliorated endotoxin-induced acute lung injury by preserving the dynamic equilibrium of mitochondrial fusion/fission through the regulation of HIF-1a/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Jia Shi
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Tianxi Yu
- Department of Sanitary Inspection and Quarantine, Kunming Medical University, YunNan, China
| | - Kai Song
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Shihan Du
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Simeng He
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Nankai University, Tianjin, China
| | - Xinxin Hu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Xiangyun Li
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Haibo Li
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Shuan Dong
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Yuan Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Zilei Xie
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Cui Li
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Jianbo Yu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
21
|
Dexmedetomidine attenuates lipopolysaccharide-induced acute liver injury in rats by inhibiting caveolin-1 downstream signaling pathway. Biosci Rep 2021; 41:227822. [PMID: 33558888 PMCID: PMC7938455 DOI: 10.1042/bsr20204279] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/27/2022] Open
Abstract
Objective: The aim of the present study is to investigate the anti-injury and anti-inflammatory effects of dexmedetomidine (Dex) in acute liver injury induced by lipopolysaccharide (LPS) in Sprague–Dawley rats and its possible mechanism. Methods: The acute liver injury model of male rats was established by injecting LPS into tail vein. The mean arterial pressure (MAP) of rats was recorded at 0–7 h, and lactic acid was detected at different time points. Wet/dry weight ratio (W/D) was calculated. Pathological changes of rat liver were observed by HE staining. ALT and AST levels in serum were detected. The activities of myeloperoxidase (MPO) and superoxide dismutase (SOD) in liver tissue homogenate and the levels of IL-1β and IL-18 in serum were detected by ELISA. Protein levels of Caveolin-1 (Cav-1), TLR-4 and NLRP3 in liver tissue were tested by immunohistochemistry method. The expression of Cav-1, TLR-4 and NLRP3 mRNA in liver tissue was detected by quantitative polymerase chain reaction (qPCR) to explore its related mechanism. Results: Compared with NS group, serum lactic acid, W/D of liver tissue, MPO, SOD, IL-1β and IL-18 were significantly increased and MAP decreased significantly in LPS group and D+L group. However, compared with NS group, D group showed no significant difference in various indicators. Compared with LPS group, MPO, SOD, IL-1β and IL-18 were significantly decreased and MAP was significantly increased in D+L group. D+L group could significantly increase the level of Cav-1 protein and decrease the level of TLR-4 and NLRP3 protein in liver tissue caused by sepsis. The expression of Cav-1 mRNA was significantly up-regulated and the expression of TLR-4 and NLRP3 mRNA was inhibited in D+L group. Conclusion: Dex pretreatment protects against LPS-induced actue liver injury via inhibiting the activation of the NLRP3 signaling pathway by up-regulating the expression of Cav-1 by sepsis.
Collapse
|
22
|
Liao Q, Chen W, Tong Z, Xue M, Gu T, Yuan Y, Song Z, Tao Z. Shufeng Jiedu capsules protect rats against LPS-induced acute lung injury via activating NRF2-associated antioxidant pathway. Histol Histopathol 2021; 36:317-324. [PMID: 33346364 DOI: 10.14670/hh-18-293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Shufeng Jiedu capsule (SFJDC) is a traditional Chinese medicine, which has been used for the treatment of respiratory infections for more than thirty years in Hunan (China). SFJDC protected rats against LPS-induced acute lung injury (ALI); however, the molecular mechanisms underlying the therapeutic effects of SFJDC remain unclear. Therefore, this study aimed at analyzing the major anti-inflammatory compounds of SFJDC and exploring the underlying molecular mechanisms. SFJDC dissolved in water was fingerprinted by UPLC/Q-TOF. Inflammation response was assessed by histopathological examination and ELISA assay. Arterial blood gases were also analyzed to evaluate the function of rat lungs. The expression levels of Kelch-like ECH-associating protein 1 (Keap1), Nrf2, heme oxygenase-1 (HO1), Cullin 3 (CUL3) and NQO1 were analyzed by Western blotting. Results indicated that SFJDC alleviated inflammation response by reducing the level of inflammatory cytokines, and upregulation of glutathione-S-transferase (GST) and superoxide dismutase (SOD) in lung tissues. Furthermore, SFJDC suppressed LPS-induced upregulation of Keap 1 and CUL3 in rat lungs. The expression of NRF2 HO1 and NQO1 were further upregulated by SFJDC in the presence of LPS, indicating that SFJDC might activate the NRF2-associated antioxidant pathway. In conclusion, SFJDC treatment may protect the rat lungs from LPS by alleviating the inflammation response via NRF2-associated antioxidant pathway.
Collapse
Affiliation(s)
- Qingwu Liao
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenan Chen
- Emergency Department, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhufeng Tong
- Department of General Practice, Yijishan Hospital of Wannan Medical College, Anhui, China
| | - Mingming Xue
- Emergency Department, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianwen Gu
- Emergency Department, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Yuan
- Geriatrics Department, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenju Song
- Emergency Department, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhengang Tao
- Emergency Department, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
23
|
Xue H, Li M. Protective effect of pterostilbene on sepsis-induced acute lung injury in a rat model via the JAK2/STAT3 pathway. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1452. [PMID: 33313197 PMCID: PMC7723647 DOI: 10.21037/atm-20-5814] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Bacterial infection is one of the most common causes of sepsis, with acute lung injury (ALI) being a related complication. Pterostilbene (PTS) is extracted from blueberries, peanuts, and grapes, and has numerous pharmacologic activities. The aim of the present study was to explore the underlying role of PTS protects against sepsis-mediated ALI. Methods We established a sepsis model induced by cecal ligation and puncture (CLP) in rats. The rats were randomly divided into five groups (n=5 each): sham group, CLP group, Dexmedetomidine group (Dex, 50 µg/kg) and PTS groups (25 and 50 mg/kg). Twenty-hours hours after CLP, PTS was intraperitoneally injected for 14 continuous days. The rats were killed, and blood and lung tissue were collected for pathological analysis and mRNA and protein detection. Results Our findings showed that PTS reduced the wet/dry ratio and ameliorated sepsis-induced pulmonary fibrosis (PF), which was associated with improvement of pathological damage in lung tissues. We also observed the inhibitory effect of PTS on apoptosis and release of inflammatory cytokines (i.e., tumor necrosis factor-α, interleukin-6, and monocyte chemotactic protein 1). In addition, PTS markedly suppressed the phosphorylation levels of Janus kinase-2 (JAK2) and signal transducer and activator of transcription 3 (STAT3). Conclusions Our results indicated that PTS inhibited the PF, apoptosis, and inflammatory response via the JAK2/STAT3 pathway in a sepsis-induced ALI rat model, providing a candidate for drug therapy of sepsis-induced ALI.
Collapse
Affiliation(s)
- Hua Xue
- Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
24
|
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: 2.3] [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
|
25
|
Hu Q, Wang Q, Han C, Yang Y. Sufentanil attenuates inflammation and oxidative stress in sepsis-induced acute lung injury by downregulating KNG1 expression. Mol Med Rep 2020; 22:4298-4306. [PMID: 33000200 PMCID: PMC7533471 DOI: 10.3892/mmr.2020.11526] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
The present study aimed to investigate the effects of sufentanil on sepsis-induced acute lung injury (ALI), and identify the potential molecular mechanisms underlying its effect. In order to achieve this, a rat sepsis model was established. Following treatment with sufentanil, the lung wet/dry (W/D) weight ratio was calculated. Histopathological analysis was performed via hematoxylin and eosin staining. Levels of inflammatory factors in bronchoalveolar lavage fluid were determined via ELISA. Furthermore, malondialdehyde (MDA) content and the activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px) in tissue homogenates were assessed using commercial kits. Western blot analysis was performed to determine kininogen-1 (KNG1) protein expression. In addition, alveolar epithelial type II cells (AEC II) were stimulated with lipopolysaccharide (LPS) to mimic ALI. The levels of inflammation and oxidative stress were evaluated following overexpression of KNG1. Protein expression levels of nuclear factor-κB (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling were determined via western blot analysis. The results of the present study demonstrated that sufentanil alleviated histopathological injury and the W/D ratio in lung tissue. Following treatment with sufentanil, levels of inflammatory factors also decreased, accompanied by decreased concentrations of MDA, and increased activities of SOD, CAT and GSH-Px. Notably, KNG1 was decreased in lung tissues following treatment with sufentanil. Furthermore, overexpression of KNG1 attenuated the inhibitory effects of sufentanil on LPS-induced inflammation and oxidative stress in AEC II. Sufentanil markedly downregulated NF-κB expression, while upregulating Nrf2 and HO-1 expression levels, which was reversed following overexpression of KNG1. Taken together, the results of the present study suggested that sufentanil may alleviate inflammation and oxidative stress in sepsis-induced ALI by downregulating KNG1 expression.
Collapse
Affiliation(s)
- Quan Hu
- Intensive Care Unit, The First People's Hospital, Wuhan, Hubei 430200, P.R. China
| | - Qin Wang
- Department of Pathology, Hubei Women and Children Health Care Hospital, Wuhan, Hubei 430200, P.R. China
| | - Chuangang Han
- Department of Anesthesiology, The First People's Hospital, Wuhan, Hubei 430200, P.R. China
| | - Yan Yang
- Department of Anesthesiology, The First People's Hospital, Wuhan, Hubei 430200, P.R. China
| |
Collapse
|
26
|
Zhu Y, Sun A, Meng T, Li H. RETRACTED: Protective role of long noncoding RNA CRNDE in myocardial tissues from injury caused by sepsis through the microRNA-29a/SIRT1 axis. Life Sci 2020; 255:117849. [PMID: 32473249 DOI: 10.1016/j.lfs.2020.117849] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/19/2020] [Accepted: 05/25/2020] [Indexed: 12/17/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of Editor-in-Chief and the corresponding author. The corresponding author confessed that the paper was outsourced to a third-party company, who could not guarantee the integrity of the data.
Collapse
Affiliation(s)
- Yan Zhu
- Department of ICU, Zaozhuang Municipal Hospital, Zaozhuang 277102, Shandong, PR China
| | - Aichen Sun
- Department of Orthopaedics, Zaozhuang Municipal Hospital, Zaozhuang 277102, Shandong, PR China.
| | - Taocheng Meng
- Department of ICU, Zaozhuang Municipal Hospital, Zaozhuang 277102, Shandong, PR China
| | - Haolan Li
- Department of Infectious Diseases, Zaozhuang Municipal Hospital, Zaozhuang 277102, Shandong, PR China
| |
Collapse
|
27
|
GDF-15 prevents lipopolysaccharide-mediated acute lung injury via upregulating SIRT1. Biochem Biophys Res Commun 2020; 526:439-446. [DOI: 10.1016/j.bbrc.2020.03.103] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/18/2020] [Indexed: 02/06/2023]
|
28
|
Zhang X, Yue X, Cui Y, Zhao Z, Huang Y, Cai S, Wang G, Wang W, Hugh S, Pan X, Wu C, Tan W. A Systematic Safety Evaluation of Nanoporous Mannitol Material as a Dry-Powder Inhalation Carrier System. J Pharm Sci 2020; 109:1692-1702. [PMID: 31987851 DOI: 10.1016/j.xphs.2020.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/28/2019] [Accepted: 01/15/2020] [Indexed: 12/14/2022]
Abstract
For carrier-based dry-powder inhaler (DPI) formulations, the adhesion between carrier particles and active pharmaceutical ingredients (API) particles have a significant influence on the aerosolization performance of the API-carrier complexes and the desired detachment of the API for efficient pulmonary delivery. In our previous study, nanoporous mannitol material was successfully fabricated as carriers by a one-step nonorganic solvent spray drying method with the thermal degradation of ammonium carbonate. These carriers were shown to achieve excellent aerosolization performance. In addition, no residue of ammonium carbonate was detected on the powder surface. However, the safety of nanoporous mannitol carriers (Nano-PMCs) during pulmonary administration/delivery was still unknown because the lung is vulnerable to the inhaled particles. To address this question, the present study was conducted to construct a systematic safety evaluation for DPIs carriers to investigate the safety of Nano-PMCs in the whole inhalation, which would make up for the lack of detailed and standardized method in this field. In vitro safety evaluation was carried out using respiratory and pulmonary cytotoxicity tests, hemolysis assay, and ciliotoxicity test. In vivo safety evaluation was studied by measuring inflammatory indicators in the bronchoalveolar lavage fluid, assessing the pulmonary function and observing pulmonary pathological changes. Nano-PMCs showed satisfactory biocompatibility on respiratory tracts and lungs in vitro and in vivo. It was suggested that Nano-PMCs were safe for intrapulmonary delivery and potential as DPI carriers.
Collapse
Affiliation(s)
- Xuejuan Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006 Guangdong, P. R. China; School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong, P. R. China
| | - Xiao Yue
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong, P. R. China
| | - Yingtong Cui
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong, P. R. China
| | - Ziyu Zhao
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong, P. R. China
| | - Ying Huang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong, P. R. China; College of Pharmacy, Jinan University, Guangzhou, 511443 Guangdong, P. R. China.
| | - Shihao Cai
- College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712
| | - Guanlin Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong, P. R. China
| | - Wenhao Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong, P. R. China
| | - Smyth Hugh
- College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong, P. R. China.
| | - Chuanbin Wu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong, P. R. China; College of Pharmacy, Jinan University, Guangzhou, 511443 Guangdong, P. R. China
| | - Wen Tan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006 Guangdong, P. R. China
| |
Collapse
|