1
|
Jana B, Całka J, Bulc M, Kawka D. The regulatory action of acetylcholine and its receptors on B4 and C4 leukotriene formation in the porcine endometrium after experimental inflammogenic Escherichia coli infection. J Vet Res 2024; 68:571-581. [PMID: 39776692 PMCID: PMC11702257 DOI: 10.2478/jvetres-2024-0066] [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: 06/21/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Introduction Endometritis is a very common pathology in animals which changes endometrial leukotriene (LT) formation and muscarinic 2 and 3 receptor subtypes (M2R/M3R) and α-7 nicotinic acetylcholine (ACh) receptor (α-7 nAChR) expression patterns. With the relationship between ACh, its receptors and LT production remaining unclear, the role of M2R, M3R and α-7 nAChR in action of ACh on the 5-lipoxygenase (5-LO), LTA4 hydrolase (LTAH) and LTC4 synthase (LTCS) protein abundances in the inflamed porcine endometrium and on the tissue secretion of LTB4 and LTC4 were studied. Material and Methods On day three of the oestrous cycle in gilts aged 7-8 months, 50 mL of either saline solution (control group, n = 5) or an E. coli suspension at 109 colony-forming units/mL (E. coli group, n = 5), was injected into each uterine horn. Endometrial explants obtained eight days later, were incubated with ACh alone, antagonists of M2R, M3R and α-7 nAChR alone, or with ACh together with particular antagonists for 16 h. Enzyme abundances in endometrial tissue were estimated by Western blotting, and LT concentrations in medium by ELISA. Results Severe acute endometritis developed in the E. coli group. In the endometrial explants from both groups, ACh elevated 5-LO, LTAH and LTCS protein abundances and LTB4 and LTC4 release. In the E. coli group, ACh-induced 5-LO and LTCS abundances and LTB4 release were increased versus the control group. In both groups, the M3R antagonist with ACh reduced all ACh-stimulated enzyme abundances and LT release in comparison to the abundances and release mediated by ACh alone. This effect on LTCS protein abundance and LTB4 release was also produced by the M2R antagonist with ACh in the E. coli group. Compared to the effect of ACh alone, exposure of the E. coli group endometrium to the α-7 nAChR antagonist with ACh led to a rise in LTAH and LTCS protein abundances and LTB4 and LTC4 secretion. Conclusion In the inflamed pig endometrium, ACh increased 5-LO, LTAH and LTCS protein abundances and LTB4 and LTC4 release by M3R, and LTCS protein abundance and LTB4 release also by M2R. By interaction with α-7 nAChR, ACh reduced LTAH and LTCS protein abundances and the release of these LTs. Thus, in an indirect manner, ACh can affect LT-controlled processes.
Collapse
Affiliation(s)
- Barbara Jana
- Division of Reproductive Biology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, 10-748Olsztyn, Poland
| | - Jarosław Całka
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-718Olsztyn, Poland
| | - Michał Bulc
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-718Olsztyn, Poland
| | - Dominika Kawka
- Division of Reproductive Biology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, 10-748Olsztyn, Poland
| |
Collapse
|
2
|
Hou Z, Yang F, Zhang Q, Wang Y, Liu J, Liang F. Targeting the PI3K/AKT signaling pathway with PNU120596 protects against LPS-induced acute lung injury. J Pharm Pharmacol 2024; 76:1508-1520. [PMID: 39288376 DOI: 10.1093/jpp/rgae076] [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: 11/13/2023] [Accepted: 06/04/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVES This study investigated the potential therapeutic benefits of PNU120596, a positive allosteric modulator of the α7 nicotinic acetylcholine receptor (α7nAChR), in mitigating acute lung injury (ALI) induced by lipopolysaccharide (LPS) in a mouse model. Specifically, we sought to examine the impact of PNU120596 on the PI3K/AKT signaling pathway in the context of ALI. METHODS ALI was induced in mice by LPS administration, and the protective effects of PNU120596 were assessed. Lung injury, lung function, and the inflammatory response were evaluated. Additionally, the activation of the PI3K/AKT signaling pathway was examined, along with the levels of inflammatory factors and oxidative stress markers. KEY FINDINGS PNU120596 significantly ameliorated LPS-induced lung injury, improved lung function, and reduced the inflammatory response in the mouse model of ALI. Furthermore, we observed that PNU120596 inhibited the activation of the PI3K/AKT signaling pathway, which was associated with decreased levels of inflammatory factors and oxidative stress markers. CONCLUSIONS PNU120596 exhibits promising therapeutic potential for the treatment of acute lung injury, potentially by targeting the PI3K/AKT signaling pathway. These findings suggest that modulation of the α7 nicotinic acetylcholine receptor with PNU120596 may offer a viable strategy for the management of ALI, warranting further investigation and potential clinical applications.
Collapse
Affiliation(s)
- Zixin Hou
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Fengrui Yang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
- Department of Anesthesiology, General Hospital, Hunan University of Medicine, Huaihua 418000, PR China
| | - Qiang Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Yuxia Wang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Junwen Liu
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Feng Liang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| |
Collapse
|
3
|
Zhang S, Zhang C, Yan H, Yang L, Shi N, Liu C, Chen Y. Sacral Nerve Stimulation Alleviates Intestinal Inflammation Through Regulating the Autophagy of Macrophages and Activating the Inflammasome Mediated by a Cholinergic Antiinflammatory Pathway in Colitis Rats. Neuromodulation 2024; 27:302-311. [PMID: 36740464 DOI: 10.1016/j.neurom.2023.01.005] [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: 10/23/2022] [Revised: 12/16/2022] [Accepted: 01/07/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is characterized by chronic progressive intestinal inflammation. Sacral nerve stimulation (SNS) ameliorates colon inflammation caused by IBD. The aim of this study was to investigate the antiinflammatory benefits of SNS in colitis rats and explore the roles of the cholinergic antiinflammatory pathway, macrophage autophagy, and nucleotide oligomerization domain-like receptor thermal protein domain associated protein 3 (NLRP3) inflammatory bodies. MATERIALS AND METHODS Rats were divided into four groups: healthy control, dextran sulfate sodium (DSS), DSS + sham-SNS, and DSS + SNS groups. An electrode was surgically placed in the right sacral nerve (S3) for stimulation. The disease activity index (DAI) score was recorded each day, and the degree of inflammatory injury was evaluated using hematoxylin and eosin staining. The alpha7 nicotinic acetylcholine receptor (α7nAChR) and autophagy- and NLRP3-related factors were assessed using immunofluorescence staining and Western blotting. RESULTS The DSS group showed a higher DAI score, colon shortening, upregulated proinflammatory action, and colon damage, and the DSS + SNS group showed significantly improved symptoms. The number of α7nAChR+ cells and the expression level of autophagy decreased in the DSS group but increased in the DSS + SNS group. Conversely, the DSS group showed increased activation of NLRP3 inflammatory bodies, whereas the DSS + SNS group showed decreased activation of NLRP3 inflammatory bodies. CONCLUSION In this study, SNS ameliorated colon inflammation by enhancing macrophage autophagy and inhibiting the activation of NLRP3 inflammatory bodies, which may be related to the opening of the cholinergic antiinflammatory pathway.
Collapse
Affiliation(s)
- Shuhui Zhang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Can Zhang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Hui Yan
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lijuan Yang
- Cancer Research Laboratory, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ning Shi
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chengxia Liu
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yan Chen
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
4
|
Fu H, Liang X, Tan W, Hu X. Unraveling the protective mechanisms of Chuanfangyihao against acute lung injury: Insights from experimental validation. Exp Ther Med 2023; 26:535. [PMID: 37869635 PMCID: PMC10587870 DOI: 10.3892/etm.2023.12234] [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: 06/25/2023] [Accepted: 08/21/2023] [Indexed: 10/24/2023] Open
Abstract
Chuanfangyihao (CFYH) is an effective treatment for acute lung injury (ALI) in clinical practice; however, its underlying mechanism of action remains unclear. Therefore, the aim of the present study was to elucidate the pharmacological mechanism of action of CFYH in ALI through experimental validation. First, a rat model of ALI was established using lipopolysaccharide (LPS). Next, the pathological changes in the lungs of the rats and the pathological damage were scored. The wet/dry weight ratios were measured, and ROS content was detected using flow cytometry. ELISA was used to examine IL-6, TNF-α, IL-1β, IL-18, and LDH levels. Immunohistochemistry was used to detect Beclin-1 and NLRP3 expression. Western blotting was performed to analyze the expression of HMGB1, RAGE, TLR4, NF-κB p65, AMPK, p-AMPK, mTOR, p-mTOR, Beclin-1, LC3-II/I, p62, Bcl-2, Bax, Caspase-3, Caspase-1, and GSDMD-NT. The mRNA levels of HMGB1, RAGE, AMPK, mTOR, and HIF-1α were determined using reverse transcription quantitative PCR. CFYH alleviated pulmonary edema and decreased the expression of IL-6, TNF-α, TLR4, NF-κB p65, HMGB1/RAGE, ROS, and HIF-1α. In addition, pretreatment with CFYH reversed ALI-induced programmed cell death. In conclusion, CFYH alleviates LPS-induced ALI, and these findings provide a preliminary clarification of the predominant mechanism of action of CFYH in ALI.
Collapse
Affiliation(s)
- Hongfang Fu
- Infectious Disease Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Xiao Liang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Wanying Tan
- Infectious Disease Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Xiaoyu Hu
- Infectious Disease Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
5
|
Huo K, Xu J, Ma K, Wang J, Wei M, Zhang M, Guo Q, Qu Q. Loganin attenuates neuroinflammation after ischemic stroke and fracture by regulating α7nAChR-mediated microglial polarization. ENVIRONMENTAL TOXICOLOGY 2023; 38:926-940. [PMID: 36637150 DOI: 10.1002/tox.23738] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 06/17/2023]
Abstract
Fracture in acute stage of ischemic stroke can increase inflammatory response and enhance stroke injury. Loganin alleviates the symptoms of many inflammatory diseases through its anti-inflammatory effect, but its role in ischemic stroke and fracture remains to be explored. Here, mice were handled with permanent middle cerebral artery occlusion (pMCAO) followed by tibial fracture 1 day later to establish a pMCAO+fracture model. Loganin or Methyllycaconitine (MLA, a specific a7nAchR inhibitor) were intragastrically administered 2 or 0.5 h before pMCAO, respectively. And mouse motor function and infarct volume were evaluated 3 days after pMCAO. We found that loganin alleviated the neurological deficit, cerebral infarction volume, and neuronal apoptosis (NeuN+ TUNEL+ ) in mice with pMCAO+fracture. And loganin suppressed pMCAO+fracture-induced neuroinflammation by promoting M2 microglia polarization (Iba1+ CD206+ ) and inhibiting M1 microglia polarization (Iba1+ CD11b+ ). While administration with MLA reversed the protective effect of loganin on pMCAO+fracture-induced neurological deficit and neuroinflammation. Next, LPS was used to stimulate BV2 microglia to simulate pMCAO+fracture-induced inflammatory microenvironment in vitro. Loganin facilitated the transformation of LPS-stimulated BV2 cells from M1 pro-inflammatory state (CD11b+ ) to M2 anti-inflammatory state (CD206+ ), which was antagonized by treatment with MLA. And loganin induced autophagy activation in LPS-stimulated BV2 cells by activating a7nAchR. Moreover, treatment with rapamycin (an autophagy activator) neutralized the inhibitory effect of MLA on loganin induced transformation of BV2 cells to M2 phenotype. Furthermore, BV2 cells were treated with LPS, LPS + loganin, LPS + loganin+MLA, or LPS + loganin+MLA+ rapamycin to obtain conditioned medium (CM) for stimulating primary neurons. Loganin reduced the damage of primary neurons caused by LPS-stimulated BV2 microglia through activating a7nAchR and inducing autophagy activation. In conclusion, loganin played anti-inflammatory and neuroprotective roles in pMCAO + fracture mice by activating a7nAchR, enhancing autophagy and promoting M2 polarization of microglia.
Collapse
Affiliation(s)
- Kang Huo
- Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Center of brain health, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Jing Xu
- Department of Emergency, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Kaige Ma
- Department of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, People's Republic of China
| | - Jianyi Wang
- Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Meng Wei
- Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Meng Zhang
- Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Qinyue Guo
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Qiumin Qu
- Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| |
Collapse
|
6
|
Tian F, Lei J, Ni Y, Zhong D, Xie N, Ma J, Wang H, Si S, Wu Y, Jiang T. Regulation of CD18 stability by SIGIRR-modulated ubiquitination: new insights into the relationship between innate immune response and acute lung injury. FEBS J 2022; 290:2721-2743. [PMID: 36527283 DOI: 10.1111/febs.16708] [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: 01/28/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 12/23/2022]
Abstract
Inappropriate accumulation of alveolar macrophages (AMs) and subsequent excessive production of immune responses play critical roles in the pathogenesis of acute lung injury (ALI), but the core negative regulators governing innate signalling in AMs are ill defined. We have previously shown that single immunoglobin IL-1 receptor-related protein (SIGIRR), a negative regulator of IL-1 receptor and Toll-like receptor signalling, inhibits lipopolysaccharide (LPS)-induced inflammatory responses in AMs. To address the biological relevance of SIGIRR in vivo, we generated a murine ALI model via intratracheal instillation of LPS. Intriguingly, SIGIRR expression was observed to be decreased in resident and recruited macrophages during ALI. This decrease was associated with parallel induction in CD18 protein levels in LPS-challenged lung tissues. Through intranasal injection of SIGIRR lentiviral particles studies, we showed that the overexpression of SIGIRR attenuated recruitment of macrophages and neutrophils, decreased production of inflammatory cytokines and ameliorated pathological changes in lungs. Whilst exploring the basis for this phenotype, SIGIRR was found to be coexpressed with CD18 in AMs, and SIGIRR potentiated the instability of CD18 protein via enhancement of its ubiquitination and proteasome degradation. Conversely, by using CD18-/- mice, we further observed that CD18 deletion completely abolished the therapeutic effects of overexpression of SIGIRR on LPS-induced ALI. Mover, overexpression of CD18 in AMs promoted adhesion to ECM components, enhanced TLR4-mediated inflammasome activation and thereby potentiated IL-1β production. These data collectively identify SIGIRR/CD18 as a key negative regulatory circuit maintaining innate immune homeostasis in AMs along the pathogenesis of ALI.
Collapse
Affiliation(s)
- Feng Tian
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jie Lei
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yunfeng Ni
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Daixing Zhong
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Nianlin Xie
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jun Ma
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Haiqiang Wang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Shaokui Si
- Department of Respiration, Third Hospital of Baoji, Baoji, China
| | - Yumei Wu
- Department of Pharmacy, Air Force Medical University, Xi'an, China
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
7
|
Su Y, Zhang W, Zhang R, Yuan Q, Wu R, Liu X, Wuri J, Li R, Yan T. Activation of Cholinergic Anti-Inflammatory Pathway Ameliorates Cerebral and Cardiac Dysfunction After Intracerebral Hemorrhage Through Autophagy. Front Immunol 2022; 13:870174. [PMID: 35812436 PMCID: PMC9260497 DOI: 10.3389/fimmu.2022.870174] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/23/2022] [Indexed: 12/12/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) is the devastating subtype of stroke with cardiovascular complications, resulting in high rates of mortality and morbidity with the release of inflammatory factors. Previous studies have demonstrated that activation of α7nAChR can reduce immune and inflammation-related diseases by triggering the cholinergic anti-inflammatory pathway (CAIP). α7nAChR mediates protection from nervous system inflammation through AMPK-mTOR-p70S6K-associated autophagy. Therefore, the purpose of this study is to explore whether the activation of α7nAChR improves cerebral and cardiac dysfunction after ICH through autophagy. Methods Male C57BL/6 mice were randomly divided into five groups (1): Control + saline (2), ICH+ saline (3), ICH + PNU-282987 (4), ICH+ PNU-282987 + MLA (5), ICH + PNU-282987 + 3-MA. The neurological function was evaluated at multiple time points. Brain water content was measured at 3 days after ICH to assess the severity of brain edema. PCR, immunofluorescence staining, and Western Blot were performed at 7 days after ICH to detect inflammation and autophagy. Picro-Sirius Red staining was measured at 30 days after ICH to evaluate myocardial fibrosis, echocardiography was performed at 3 and 30 days to measure cardiac function. Results Our results indicated that the PNU-282987 reduced inflammatory factors (MCP-1, IL-1β, MMP-9, TNF-α, HMGB1, TLR2), promoted the polarization of macrophage/microglia into anti-inflammatory subtypes(CD206), repaired blood-brain barrier injury (ZO-1, Claudin-5, Occludin), alleviated acute brain edema and then recovered neurological dysfunction. Echocardiography and PSR indicated that activation of α7nAChR ameliorated cardiac dysfunction. Western Blot showed that activation of α7nAChR increased autophagy protein (LC3, Beclin) and decreased P62. It demonstrated that the activation of α7nAChR promotes autophagy and then recovers brain and heart function after ICH. Conclusions In conclusion, PNU-282987 promoted the cerebral and cardiac functional outcomes after ICH in mice through activated α7nAChR, which may be attributable to promoting autophagy and then reducing inflammatory reactions after ICH.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Tao Yan
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma, Neurorepair, and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| |
Collapse
|
8
|
Tan Y, Chu Z, Shan H, Zhangsun D, Zhu X, Luo S. Inflammation Regulation via an Agonist and Antagonists of α7 Nicotinic Acetylcholine Receptors in RAW264.7 Macrophages. Mar Drugs 2022; 20:md20030200. [PMID: 35323499 PMCID: PMC8955479 DOI: 10.3390/md20030200] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 02/01/2023] Open
Abstract
The α7 nicotinic acetylcholine receptor (nAChR) is widely distributed in the central and peripheral nervous systems and is closely related to a variety of nervous system diseases and inflammatory responses. The α7 nAChR subtype plays a vital role in the cholinergic anti-inflammatory pathway. In vivo, ACh released from nerve endings stimulates α7 nAChR on macrophages to regulate the NF-κB and JAK2/STAT3 signaling pathways, thereby inhibiting the production and release of downstream proinflammatory cytokines and chemokines. Despite a considerable level of recent research on α7 nAChR-mediated immune responses, much is still unknown. In this study, we used an agonist (PNU282987) and antagonists (MLA and α-conotoxin [A10L]PnIA) of α7 nAChR as pharmacological tools to identify the molecular mechanism of the α7 nAChR-mediated cholinergic anti-inflammatory pathway in RAW264.7 mouse macrophages. The results of quantitative PCR, ELISAs, and transcriptome analysis were combined to clarify the function of α7 nAChR regulation in the inflammatory response. Our findings indicate that the agonist PNU282987 significantly reduced the expression of the IL-6 gene and protein in inflammatory macrophages to attenuate the inflammatory response, but the antagonists MLA and α-conotoxin [A10L]PnIA had the opposite effects. Neither the agonist nor antagonists of α7 nAChR changed the expression level of the α7 nAChR subunit gene; they only regulated receptor function. This study provides a reference and scientific basis for the discovery of novel α7 nAChR agonists and their anti-inflammatory applications in the future.
Collapse
Affiliation(s)
- Yao Tan
- Medical School, Guangxi University, Nanning 530004, China; (Y.T.); (H.S.)
| | - Zhaoli Chu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (Z.C.); (D.Z.)
| | - Hongyu Shan
- Medical School, Guangxi University, Nanning 530004, China; (Y.T.); (H.S.)
| | - Dongting Zhangsun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (Z.C.); (D.Z.)
| | - Xiaopeng Zhu
- Medical School, Guangxi University, Nanning 530004, China; (Y.T.); (H.S.)
- Correspondence: (X.Z.); (S.L.)
| | - Sulan Luo
- Medical School, Guangxi University, Nanning 530004, China; (Y.T.); (H.S.)
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (Z.C.); (D.Z.)
- Correspondence: (X.Z.); (S.L.)
| |
Collapse
|
9
|
Yu YY, Li XQ, Hu WP, Cu SC, Dai JJ, Gao YN, Zhang YT, Bai XY, Shi DY. Self-developed NF-κB inhibitor 270 protects against LPS-induced acute kidney injury and lung injury through improving inflammation. Biomed Pharmacother 2022; 147:112615. [PMID: 35026488 DOI: 10.1016/j.biopha.2022.112615] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/02/2022] [Accepted: 01/02/2022] [Indexed: 12/11/2022] Open
Abstract
Sepsis-induced acute kidney injury (AKI) and acute lung injury (ALI) have high morbidity and mortality, with no effective clinically available drugs. Anti-inflammation is effective strategy in the therapy of AKI and ALI. NF-κB is a target for the development of anti‑inflammatory agents. The purpose of the study is to evaluate the effect of 270, self-developed NF-κB inhibitor, in LPS-induced AKI and ALI. LPS-induced macrophages were used to examine the anti-inflammation activity of 270 in vitro. Sepsis-induced AKI and ALI mice models were established by intraperitoneal injection of LPS (10 mg/kg) for 24 h. Oral administration 270 for 14 days before LPS stimulation. Plasma, kidney and lung tissues were collected and used for histopathology, biochemical assay, ELISA, RT-PCR, and western blot analyses. In vitro, we showed that 270 suppressed the inflammation response in LPS-induced RAW 264.7 macrophages and bone marrow derived macrophages. In vivo, we found that 270 ameliorated LPS-induced AKI and ALI, as evidenced by improving various pathological changes, reducing the expression of pro-inflammation genes, blocking the activation of NF-κB and JNK pathways, attenuating the elevated myeloperoxidase (MPO) activity and malondialdehyde (MDA) content, ameliorating the activated ER stress, reversing the inhibition effect on autophagy in kidney and lung tissues, and alleviating the enhanced plasma level of creatinine (Crea), blood urea nitrogen (BUN) and pro-inflammation cytokines. Our investigations provides evidence that NF-κB inhibitor 270 is a potential drug that against LPS-induced AKI and ALI in the future.
Collapse
Affiliation(s)
- Yan-Yan Yu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266200 China
| | - Xiang-Qian Li
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266200 China
| | - Wen-Peng Hu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266200 China
| | - Shi-Chao Cu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Jia-Jia Dai
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266200 China
| | - Ya-Nan Gao
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266200 China
| | - Yi-Ting Zhang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266200 China
| | - Xiao-Yi Bai
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266200 China
| | - Da-Yong Shi
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266200 China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China.
| |
Collapse
|
10
|
Cao F, Wang C, Long D, Deng Y, Mao K, Zhong H. Network-Based Integrated Analysis of Transcriptomic Studies in Dissecting Gene Signatures for LPS-Induced Acute Lung Injury. Inflammation 2021; 44:2486-2498. [PMID: 34462829 PMCID: PMC8405180 DOI: 10.1007/s10753-021-01518-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/07/2021] [Indexed: 10/26/2022]
Abstract
Acute lung injury (ALI) is a type of serious clinical syndrome leading to morbidity and mortality. However, the precise pathogenesis of ALI remains elusive. Here, we implemented an integrative meta-analysis of six GEO microarray studies with 76 samples in the ALI mouse model. A total of 958 differentially expressed genes (DEGs) were identified in LPS relative to normal samples. Then, a network-based meta-analysis was used to mine core DEGs and to unfold the interactions among these genes. We found that Ebi3 was the top upregulated genes in the LPS-induced ALI. GO, KEGG, and GSEA analyses were performed for functional annotation. qRT-PCR revealed augmented expression of six candidate genes (Stat1, Syk, Jak3, Rac2, Ripk1, and Traf6) in the established ALI mouse model with LPS exposure. Taken together, our study investigated comprehensively hub DEGs and their networks for LPS-stimulated ALI, which might afford an additional approach to determine biomarkers and therapeutic targets and explore the molecular pathophysiology toward ALI.
Collapse
Affiliation(s)
- Fang Cao
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, Huichuan District, 149 Dalian Road, Zunyi, Guizhou, 563003 China
| | - Chunyan Wang
- Department of Gastroenterology, Sichuan Provincial Peoples Hospital, University of Electronic Science and Technology, Chengdu, 610000 Sichuan China
| | - Danling Long
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000 Hubei China
| | - Yujuan Deng
- School of Computer Science and Engineering, Shijiazhuang University, Shijiazhuang, Hebei China
| | - Kaimin Mao
- Department of Critical Care Medicine, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, 200127 China
| | - Hua Zhong
- College of Life Sciences, Wuhan University, Wuhan, 430072 Hubei China
| |
Collapse
|
11
|
Comprehensive Bioinformatics Analysis of Lipopolysaccharide-Induced Altered Autophagy in Acute Lung Injury and Construction of Underlying Competing Endogenous RNA Regulatory Mechanism. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6831770. [PMID: 34722769 PMCID: PMC8553468 DOI: 10.1155/2021/6831770] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/18/2021] [Accepted: 09/18/2021] [Indexed: 01/18/2023]
Abstract
Background Acute lung injury (ALI) is a fatal syndrome frequently induced by lipopolysaccharide (LPS) released from the bacterial cell wall. LPS could also trigger autophagy of lung bronchial epithelial cell to relieve the inflammation, while the overwhelming LPS would impair the balance of autophagy consequently inducing serious lung injury. Methods We observed the autophagy variation of 16HBE, human bronchial epithelial cell, under exposure to different concentrations of LPS through western blot, immunofluorescence staining, and electron microscopy. Eight strands of 16HBE were divided into two groups upon 1000 ng/ml LPS stimulation or not, which were sent to be sequenced at whole transcriptome. Subsequently, we analyzed the sequencing data in functional enrichment, pathway analysis, and candidate gene selection and constructed a hsa-miR-663b-related competing endogenous RNA (ceRNA) network. Results We set a series of concentrations of LPS to stimulate 16HBE and observed the variation of autophagy in related protein expression and autophagosome count. We found that the effective concentration of LPS was 1000 ng/ml at 12 hours of exposure and sequenced the 1000 ng/ml LPS-stimulated 16HBE. As a result, a total of 750 differentially expressed genes (DEGs), 449 differentially expressed lncRNAs (DElncRNAs), 76 differentially expressed circRNAs (DEcircRNAs), and 127 differentially expressed miRNAs (DEmiRNAs) were identified. We constructed the protein-protein interaction (PPI) network to visualize the interaction between DEGs and located 36 genes to comprehend the core discrepancy between LPS-stimulated 16HBE and the negative control group. In combined analysis of differentially expressed RNAs (DERNAs), we analyzed all the targeted relationships of ceRNA in DERNAs and figured hsa-miR-663b as a central mediator in the ceRNA network to play when LPS induced the variation of autophagy in 16HBE. Conclusion Our research indicated that the hsa-miR-663b-related ceRNA network may contribute to the key regulatory mechanism in LPS-induced changes of autophagy and ALI.
Collapse
|
12
|
de Melo GD, Lazarini F, Larrous F, Feige L, Kornobis E, Levallois S, Marchio A, Kergoat L, Hardy D, Cokelaer T, Pineau P, Lecuit M, Lledo P, Changeux J, Bourhy H. Attenuation of clinical and immunological outcomes during SARS-CoV-2 infection by ivermectin. EMBO Mol Med 2021; 13:e14122. [PMID: 34170074 PMCID: PMC8350903 DOI: 10.15252/emmm.202114122] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
The devastating pandemic due to SARS-CoV-2 and the emergence of antigenic variants that jeopardize the efficacy of current vaccines create an urgent need for a comprehensive understanding of the pathophysiology of COVID-19, including the contribution of inflammation to disease. It also warrants for the search of immunomodulatory drugs that could improve disease outcome. Here, we show that standard doses of ivermectin (IVM), an anti-parasitic drug with potential immunomodulatory activities through the cholinergic anti-inflammatory pathway, prevent clinical deterioration, reduce olfactory deficit, and limit the inflammation of the upper and lower respiratory tracts in SARS-CoV-2-infected hamsters. Whereas it has no effect on viral load in the airways of infected animals, transcriptomic analyses of infected lungs reveal that IVM dampens type I interferon responses and modulates several other inflammatory pathways. In particular, IVM dramatically reduces the Il-6/Il-10 ratio in lung tissue and promotes macrophage M2 polarization, which might account for the more favorable clinical presentation of IVM-treated animals. Altogether, this study supports the use of immunomodulatory drugs such as IVM, to improve the clinical condition of SARS-CoV-2-infected patients.
Collapse
Affiliation(s)
| | | | - Florence Larrous
- Lyssavirus Epidemiology and Neuropathology UnitInstitut PasteurParisFrance
| | - Lena Feige
- Lyssavirus Epidemiology and Neuropathology UnitInstitut PasteurParisFrance
| | - Etienne Kornobis
- Biomics Technological PlatformCenter for Technological Resources and Research (C2RT)Institut PasteurParisFrance
- Bioinformatics and Biostatistics HubComputational Biology DepartmentInstitut PasteurParisFrance
| | | | - Agnès Marchio
- Nuclear Organization and Oncogenesis UnitInstitut PasteurParisFrance
| | - Lauriane Kergoat
- Lyssavirus Epidemiology and Neuropathology UnitInstitut PasteurParisFrance
| | - David Hardy
- Experimental Neuropathology UnitInstitut PasteurParisFrance
| | - Thomas Cokelaer
- Biomics Technological PlatformCenter for Technological Resources and Research (C2RT)Institut PasteurParisFrance
- Bioinformatics and Biostatistics HubComputational Biology DepartmentInstitut PasteurParisFrance
| | - Pascal Pineau
- Nuclear Organization and Oncogenesis UnitInstitut PasteurParisFrance
| | - Marc Lecuit
- Biology of Infection UnitInstitut PasteurInserm U1117ParisFrance
- Division of Infectious Diseases and Tropical MedicineInstitut ImagineUniversité de ParisNecker‐Enfants Malades University HospitalAP‐HPParisFrance
| | | | | | - Hervé Bourhy
- Lyssavirus Epidemiology and Neuropathology UnitInstitut PasteurParisFrance
| |
Collapse
|
13
|
Pinheiro NM, Banzato R, Tibério I, Prado MAM, Prado VF, Hamouda AK, Prado CM. Acute Lung Injury in Cholinergic-Deficient Mice Supports Anti-Inflammatory Role of α7 Nicotinic Acetylcholine Receptor. Int J Mol Sci 2021; 22:ijms22147552. [PMID: 34299169 PMCID: PMC8303767 DOI: 10.3390/ijms22147552] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Background: The lung cholinergic pathway is important for controlling pulmonary inflammation in acute lung injury, a condition that is characterized by a sudden onset and intense inflammation. This study investigated changes in the expression levels of nicotinic and muscarinic acetylcholine receptors (nAChR and mAChR) in the lung during acute lung injury. (2) Methods: acute lung injury (ALI) was induced in wild-type and cholinergic-deficient (VAChT-KDHOM) mice using intratracheal lipopolysaccharide (LPS) instillation with or without concurrent treatment with nicotinic ligands. Bronchoalveolar lavage fluid was collected to evaluate markers of inflammation, and then the lung was removed and processed for isolation of membrane fraction and determination of acetylcholine receptors level using radioligand binding assays. (3) Results: LPS-induced increase in lung inflammatory markers (e.g., neutrophils and IL-1β) was significantly higher in VAChT-KDHOM than wild-type mice. In contrast, LPS treatment resulted in a significant increase in lung’s α7 nicotinic receptor level in wild-type, but not in VAChT-KDHOM mice. However, treatment with PNU 282987, a selective α7 nicotinic receptor agonist, restored VAChT-KDHOM mice’s ability to increase α7 nicotinic receptor levels in response to LPS-induced acute lung injury and reduced lung inflammation. LPS also increased muscarinic receptors level in VAChT-KDHOM mice, and PNU 282987 treatment reduced this response. (4) Conclusions: Our data indicate that the anti-inflammatory effects of the lung cholinergic system involve an increase in the level of α7 nicotinic receptors. Pharmacological agents that increase the expression or the function of lung α7 nicotinic receptors have potential clinical uses for treating acute lung injury.
Collapse
Affiliation(s)
- Nathalia M. Pinheiro
- Department of Bioscience, Federal University of Sao Paulo, Santos 11015-020, SP, Brazil;
- College of Pharmacy, University of Texas at Tyler, Tyler, TX 75799, USA;
| | - Rosana Banzato
- Department of Medicine, School of Medicine, University of Sao Paulo, Sao Paulo 01246-903, SP, Brazil; (R.B.); (I.T.); (V.F.P.)
| | - Iolanda Tibério
- Department of Medicine, School of Medicine, University of Sao Paulo, Sao Paulo 01246-903, SP, Brazil; (R.B.); (I.T.); (V.F.P.)
| | - Marco A. M. Prado
- Molecular Medicine Group, Robarts Research Institute, London, ON N6A 5B7, Canada;
- Department of Physiology & Pharmacology, University of Western Ontario, London, ON N6A 5B7, Canada
- Department of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 5B7, Canada
| | - Vânia F. Prado
- Department of Medicine, School of Medicine, University of Sao Paulo, Sao Paulo 01246-903, SP, Brazil; (R.B.); (I.T.); (V.F.P.)
- Molecular Medicine Group, Robarts Research Institute, London, ON N6A 5B7, Canada;
| | - Ayman K. Hamouda
- College of Pharmacy, University of Texas at Tyler, Tyler, TX 75799, USA;
| | - Carla M. Prado
- Department of Bioscience, Federal University of Sao Paulo, Santos 11015-020, SP, Brazil;
- Correspondence: ; Tel.: +55-13-3229-0118
| |
Collapse
|
14
|
Bricher Choque PN, Vieira RP, Ulloa L, Grabulosa C, Irigoyen MC, De Angelis K, Ligeiro De Oliveira AP, Tracey KJ, Pavlov VA, Consolim-Colombo FM. The Cholinergic Drug Pyridostigmine Alleviates Inflammation During LPS-Induced Acute Respiratory Distress Syndrome. Front Pharmacol 2021; 12:624895. [PMID: 34017249 PMCID: PMC8129580 DOI: 10.3389/fphar.2021.624895] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 04/13/2021] [Indexed: 01/12/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a critical illness complication that is associated with high mortality. ARDS is documented in severe cases of COVID-19. No effective pharmacological treatments for ARDS are currently available. Dysfunctional immune responses and pulmonary and systemic inflammation are characteristic features of ARDS pathogenesis. Recent advances in our understanding of the regulation of inflammation point to an important role of the vagus-nerve-mediated inflammatory reflex and neural cholinergic signaling. We examined whether pharmacological cholinergic activation using a clinically approved (for myasthenia gravis) cholinergic drug, the acetylcholinesterase inhibitor pyridostigmine alters pulmonary and systemic inflammation in mice with lipopolysaccharide (LPS)-induced ARDS. Male C57Bl/6 mice received one intratracheal instillation of LPS or were sham manipulated (control). Both groups were treated with either vehicle or pyridostigmine (1.5 mg/kg twice daily, 3 mg/day) administered by oral gavage starting at 1 h post-LPS and euthanized 24 h after LPS administration. Other groups were either sham manipulated or received LPS for 3 days and were treated with vehicle or pyridostigmine and euthanized at 72 h. Pyridostigmine treatment reduced the increased total number of cells and neutrophils in the bronchoalveolar lavage fluid (BALF) in mice with ARDS at 24 and 72 h. Pyridostigmine also reduced the number of macrophages and lymphocytes at 72 h. In addition, pyridostigmine suppressed the levels of TNF, IL-1β, IL-6, and IFN-γ in BALF and plasma at 24 and 72 h. However, this cholinergic agent did not significantly altered BALF and plasma levels of the anti-inflammatory cytokine IL-10. Neither LPS nor pyridostigmine affected BALF IFN-γ and IL-10 levels at 24 h post-LPS. In conclusion, treatments with the cholinergic agent pyridostigmine ameliorate pulmonary and systemic inflammatory responses in mice with endotoxin-induced ARDS. Considering that pyridostigmine is a clinically approved drug, these findings are of substantial interest for implementing pyridostigmine in therapeutic strategies for ARDS.
Collapse
Affiliation(s)
- Pamela Nithzi Bricher Choque
- Laboratory of Pulmonary Immunology, Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
| | - Rodolfo P. Vieira
- Post-graduation Program in Bioengineering and in Biomedical Engineering, Universidade Brasil, São Paulo, Brazil
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), São Paulo, Brazil
- Federal University of São Paulo (UNIFESP), Post-graduation Program in Sciences of Human Movement and Rehabilitation, São Paulo, Brazil
- Departament of Physiology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Luis Ulloa
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Caren Grabulosa
- Laboratory of Pulmonary Immunology, Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
| | - Maria Claudia Irigoyen
- Hypertension Unit, Heart Institute (INCOR), Medical School of University of São Paulo, São Paulo, Brazil
| | - Katia De Angelis
- Departament of Physiology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Ana Paula Ligeiro De Oliveira
- Laboratory of Pulmonary Immunology, Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
| | - Kevin J. Tracey
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Valentin A. Pavlov
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Fernanda Marciano Consolim-Colombo
- Laboratory of Pulmonary Immunology, Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
- Hypertension Unit, Heart Institute (INCOR), Medical School of University of São Paulo, São Paulo, Brazil
| |
Collapse
|
15
|
Yang Y, Li Q, Tan F, Zhang J, Zhu W. Mechanism of IL-8-induced acute lung injury through pulmonary surfactant proteins A and B. Exp Ther Med 2020; 19:287-293. [PMID: 31853301 PMCID: PMC6909794 DOI: 10.3892/etm.2019.8192] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/01/2019] [Indexed: 12/03/2022] Open
Abstract
This study explored how interleukin-8 (IL-8) causes acute lung injury (ALI) through pulmonary surfactant protein A (SP-A) and surfactant protein B (SP-B). Serum was collected from 53 ALI patients and further 56 healthy subjects who underwent physical examination. The IL-8, SP-A, and SP-B levels were determined using enzyme-linked immunosorbent assay (ELISA). An ALI model was constructed using lipopolysaccharide (LSP)-induced normal A549 cells. siRNA was employed to interfere with the expression of IL-8, SP-A and SP-B. Western blot analysis was carried out to determine the protein levels, and MTT assay to determine the cell activity. In addition, co-immunoprecipitation (Co-IP) assay was used to verify the interaction between IL-8, SP-A and SP-B. ALI patients showed high expression of serum IL-8, and low expression of SP-A and SP-B, and IL-8 was negatively correlated with SP-A and SP-B, respectively. LSP-induced normal A549 cells showed increased expression of IL-8 and decreased expression of SP-A and SP-B. Silencing IL-8 led to increased expression levels of SP-A, SP-B and Bcl2, decreased expression levels of caspase-9, caspase-3, Bax, TNF-α, IL-17 and IL-1β, reduced cell apoptosis rate, and enhanced cell viability. Silencing SP-A and SP-B resulted in increased expression of IL-8, caspase-9, caspase-3, Bax, TNF-α, IL-17 and IL-1β, and decreased expression of Bcl2. Co-IP assay revealed that IL-8 could interact with SP-A and SP-B, respectively. IL-8 induces apoptosis by inhibiting SP-A and SP-B, and intensifies cellular inflammatory reaction, leading eventually to ALI.
Collapse
Affiliation(s)
- Yinong Yang
- Medical School, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| | - Qing Li
- Nursing School, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| | - Feng Tan
- School of Stomatology, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| | - Jun Zhang
- Medical School, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| | - Wu Zhu
- Medical School, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| |
Collapse
|
16
|
Guo X, Zhang Y, Lu C, Qu F, Jiang X. Protective effect of hyperoside on heart failure rats via attenuating myocardial apoptosis and inducing autophagy. Biosci Biotechnol Biochem 2019; 84:714-724. [PMID: 31797747 DOI: 10.1080/09168451.2019.1685369] [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
Heart failure (HF) is one of the most severe heart conditions, which lacks effective therapies. Therefore, it is necessary to develop more efficient drugs for HF. In this study, we investigated the cardioprotective effects of hyperoside against the pathological progression of HF. Thoracic aortic constriction (TAC) was performed to induce HF in rats. Hyperoside treatment improved cardiac function, decreased cardiomyocyte cross-sectional area and heart weight to body weight (HW/BW) ratio in HF rats. Moreover, hyperoside administration repressed apoptosis as evidenced by changing apoptosis-related protein levels, and promoted autophagy in TAC rats and angiotensin II (AngII)-induced H9C2 cells. Inhibition of autophagy by 3-methyladenine (3-MA) attenuated the beneficial effect of hyperoside against apoptosis in H9C2 cells. In summary, these data confirm that hyperoside effectively alleviates HF via suppressing apoptosis and inducing autophagy, which provides evidence that hyperoside may serve as a promising natural drug for treating HF.
Collapse
Affiliation(s)
- Xiao Guo
- Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao, People's Republic of China
| | - Yongtao Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Changhong Lu
- Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao, People's Republic of China
| | - Fengxia Qu
- Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao, People's Republic of China
| | - Xianyan Jiang
- Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao, People's Republic of China
| |
Collapse
|