1
|
Gu Y, Liu M, Niu N, Jia J, Gao F, Sun Y, Zhang Z, Dai Z, Jiao J, Zhu Z, Jia S, Xu J, Zhang Z, Xu B, Lei HM. Integrative network pharmacology and multi-omics to study the potential mechanism of Niuhuang Shangqing Pill on acute pharyngitis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119100. [PMID: 39547363 DOI: 10.1016/j.jep.2024.119100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Niuhuang Shangqing Pill (NSP) is a renowned Chinese medicine prescription listed in the Chinese Pharmacopoeia (Edition, 2020; volume 1) and is utilized in clinical practice for treating headaches and acute pharyngitis (AP) associated with "Shanghuo". Despite its widespread use, the pharmacological mechanism and bioactive components underlying NSP in treating AP remain unclear. AIM OF THE STUDY This study delved into evaluate the alleviation effect of NSP on AP and explore the mechanisms by analyzing multi-omics. MATERIALS AND METHODS UHPLC-Q Exactive Orbitrap HRMS was employed for the chemical ingredients of NSP. Multiple compositions, targets and pathways involved in the treatment of AP with NSP were predicted by network pharmacology. Additionally, wistar rat model of AP induced by capsaicin was established to evaluate the anti-AP activity of NSP in vivo. The potential mechanism of NSP to improve AP was investigated by real-time PCR, pharyngeal transcriptome analysis, non-targeted metabolomics, immunofluorescence and Western blot. RESULTS 119 compounds were identified by UHPLC-Q Exactive Orbitrap HRMS. Both clinical data of Gene Expression Omnibus (GEO) and network pharmacology demonstrated that MAPK signaling pathway and TNF signaling pathway were the critical pathway for AP treatment. In rat model of AP induced by capsaicin, NSP demonstrated the ability to reduce the levels of IL-1β, TNF-α, IL-6, CGRP, SP, PGE2, COX-2 in serum. Moreover, Transcriptomics analysis comprehensively indicated that NSP regulated the MAPK signaling pathway, TNF signaling pathway, biosynthesis of phenylalanine, tyrosine and tryptophan, arachidonic acid metabolism in AP rats. Metabolomics analysis verified that NSP could rebalance arachidonic acid metabolism, biosynthesis of phenylalanine, tyrosine and tryptophan and regulate metabolic profiles. Multi-omics Correlation analysis exhibited that the relative expression of Tnfrsf1b was significantly negatively correlated with 12(S)-HPETE. Immunofluorescence, real-time PCR and Western blot of pharyngeal tissue revealed that NSP inhibited the TNF/p38-MAPK/NF-κB signaling pathway. Additionally, in vitro study on RAW264.7 cells confirmed that NSP counteract LPS-induced inflammatory by inhibiting the TNF/p38-MAPK/NF-κB signaling pathway. Overall, NSP effectively ameliorated capsaicin-induced AP by modulating the arachidonic acid metabolism and TNF/p38-MAPK/NF-κB signaling pathway. CONCLUSION NSP effectively ameliorated capsaicin-induced AP by modulating the arachidonic acid metabolism, biosynthesis of phenylalanine, tyrosine and tryptophan, as well as the TNF/p38-MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yuhao Gu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Manting Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Nan Niu
- Scientific Research Institute of Beijing Tongrentang Co., Ltd., Beijing, 100079, China
| | - Jun Jia
- Scientific Research Institute of Beijing Tongrentang Co., Ltd., Beijing, 100079, China
| | - Feng Gao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Yangyang Sun
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medicine Science, Beijing, 100700, China
| | - Zixuan Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Ziqi Dai
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Jingyi Jiao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Zhi Zhu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Shuhe Jia
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Jiyue Xu
- Nanjing Tech University, Nanjing, 211816, China
| | - Zhaohua Zhang
- Scientific Research Institute of Beijing Tongrentang Co., Ltd., Beijing, 100079, China.
| | - Bing Xu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China.
| | - Hai Min Lei
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China.
| |
Collapse
|
2
|
Chen P, Lin C, Jin Q, Ye B, Liu X, Wang K, Zhang H, Liu J, Zhang R, Huang H, Zhang C, Li L. Investigating mechanisms of Sophora davidii (Franch.) skeels flower extract in treating LPS-induced acute pneumonia based on network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118914. [PMID: 39369925 DOI: 10.1016/j.jep.2024.118914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/29/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In TCM opinion, most of pneumonia is related to "lung heat". Sophora davidii (Franch.) Skeels flower was first documented in "Guizhou Herbal Medicine", and was recorded as having functions of clearing heat, detoxifying, and cooling blood. It can be used to treat lung heat cough. AIM OF THE STUDY To investigate main mechanisms of Sophora davidii flower extract (SDFE) in Treating LPS-induced acute Pneumonia. MATERIALS AND METHODS Acute pneumonia models on BEAS-2B cells and rats were established using LPS. The rat model was used to verified the protective effects of SDFE through HE staining, lung tissue W/D ratio assay, white blood cell count analysis, and ammonia-induced coughing test. Network pharmacology was applied to predict the active compounds, core targets and main pathways of SDFE in treating acute pneumonia. Western Blot and ELISA kits were employed to validate representative proteins in selected pathway in vivo and in vitro. RESULTS HE staining, lung tissue W/D ratio assay, white blood cell count analysis, and ammonia-induced coughing test showed SDFE could improve pathological features (leukocyte infiltration, pulmonary edema, lung injury and cough). Network pharmacology indicated MAPK/NF-κB pathway was the most relevant pathway. SDFE could significantly inhibit the expression of Fos and Jun, and the phosphorylation levels of p38, ERK, JNK, NF-κB and IκB. It also down-regulated the expression of pro-inflammatory factors (TNF-α, IL-6 and IL-1β). CONCLUSIONS SDFE can exert protective effects against acute pneumonia through the MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ping Chen
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Cheng Lin
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Qi Jin
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Baibai Ye
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Xinxu Liu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Keke Wang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Han Zhang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Jiahui Liu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Runan Zhang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Hao Huang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Chenning Zhang
- Department of Pharmacy, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441100, China.
| | - Linfu Li
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| |
Collapse
|
3
|
Sun S, Yang D, Lv J, Xia H, Mao Z, Chen X, Gao Y. Pharmacological effects of specialized pro-resolving mediators in sepsis-induced organ dysfunction: a narrative review. Front Immunol 2024; 15:1444740. [PMID: 39372413 PMCID: PMC11451296 DOI: 10.3389/fimmu.2024.1444740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/30/2024] [Indexed: 10/08/2024] Open
Abstract
Sepsis is a life-threatening syndrome of organ dysfunction, characterized by uncontrolled inflammatory response and immune dysregulation, often leading to multiple organ failure and even death. Specialized pro-resolving mediators (SPMs), which are typically thought to be formed via consecutive steps of oxidation of polyenoic fatty acids, have been shown to suppress inflammation and promote timely resolution of inflammation. They are mainly divided into four categories: lipoxins, resolvins, protectins, and maresins. The SPMs may improve the prognosis of sepsis by modulating the immune and inflammatory balance, thereby holding promise for clinical applications. However, their biosynthetic and pharmacological properties are very complex. Through a literature review, we aim to comprehensively elucidate the protective mechanisms of different SPMs in sepsis and its organ damage, in order to provide sufficient theoretical basis for the future clinical translation of SPMs.
Collapse
Affiliation(s)
- Shujun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Yang
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Lv
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Haifa Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Zhangyan Mao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yafen Gao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| |
Collapse
|
4
|
Yeh TY, Chu WJ, Huang YS. GM1 ganglioside protects against LPS-induced neuroinflammatory and oxidative responses by inhibiting the activation of Akt, TAK1 and NADPH oxidase in MG6 microglial cells. Glycobiology 2024; 34:cwad087. [PMID: 37935390 DOI: 10.1093/glycob/cwad087] [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: 09/04/2022] [Revised: 10/13/2023] [Accepted: 10/14/2023] [Indexed: 11/09/2023] Open
Abstract
GM1 is a major brain ganglioside that exerts neurotrophic, neuroprotective and antineuroinflammatory effects. The aim of this study was to obtain insights into the antineuroinflammatory mechanisms of exogenous GM1 in lipopolysaccharide (LPS)-stimulated MG6 mouse transformed microglial cell line. First, we found that GM1 prevented the LPS-induced transformation of microglia into an amoeboid-like shape. GM1 treatment inhibited LPS-induced expression of inducible nitric oxide synthase, cyclooxygenase-2 (COX-2), and proinflammatory cytokines such as TNF-α, IL-1β and IL-6 in MG6 cells. In LPS-treated mice, GM1 also reduced striatal microglia activation and attenuated COX-2 expression. Subsequent mechanistic studies showed that GM1 suppressed LPS-induced nuclear translocation of nuclear factor κB (NF-κB) and activator protein-1 (AP-1), two critical transcription factors responsible for the production of proinflammatory mediators. GM1 exhibited antineuroinflammatory properties by suppressing Akt/NF-κB signaling and the activation of mitogen-activated protein kinases (MAPKs), including p38 MAPK, extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK). Furthermore, GM1 suppressed LPS-induced activation of transforming growth factor-β-activated kinase 1 (TAK1) and NADPH oxidase 2 (NOX2), upstream regulators of the IκBα/NF-κB and MAPK/AP-1 signaling pathways. GM1 also inhibited NOX-mediated reactive oxygen species (ROS) production and protected against LPS-induced MG6 cell death, suggesting an antioxidant role of GM1. In conclusion, GM1 exerts both antineuroinflammatory and antioxidative effects by inhibiting Akt, TAK1 and NOX2 activation.
Collapse
Affiliation(s)
- Ting-Yin Yeh
- Graduate Institute of Life Sciences, National Defense Medical Center, No. 161, Sec. 6, Minquan E. Rd., Neihu Dist, Taipei City 11490, Taiwan
| | - Wen-Jui Chu
- Department of Biology and Anatomy, National Defense Medical Center, No. 161, Sec. 6, Minquan E. Rd., Neihu Dist, Taipei City 11490, Taiwan
| | - Yuahn-Sieh Huang
- Graduate Institute of Life Sciences, National Defense Medical Center, No. 161, Sec. 6, Minquan E. Rd., Neihu Dist, Taipei City 11490, Taiwan
- Department of Biology and Anatomy, National Defense Medical Center, No. 161, Sec. 6, Minquan E. Rd., Neihu Dist, Taipei City 11490, Taiwan
| |
Collapse
|
5
|
Gholami M, Sadegh M, Koroush-Arami M, Norouzi S, Arismani RJ, Asadi E, Amini M, Khodayari N. Targeting memory loss with aspirin, a molecular mechanism perspective for future therapeutic approaches. Inflammopharmacology 2023; 31:2827-2842. [PMID: 37924473 DOI: 10.1007/s10787-023-01347-1] [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: 07/19/2023] [Accepted: 09/19/2023] [Indexed: 11/06/2023]
Abstract
Acetylsalicylic acid (ASA), also known as aspirin, was discovered in 1897 as an acetylated form of salicylate. It has been widely used for its anti-inflammatory and antiplatelet effects. It is commonly used for its cardiovascular benefits and is prescribed as secondary prophylaxis after a heart attack. Furthermore, low-dose, long-term ASA is used to reduce the risk of heart attack and stroke in individuals without prior cardiovascular disease. Acetylsalicylic acid acts as a non-selective inhibitor of cyclooxygenase (COX), which inhibits the synthesis of prostaglandins and prevents pro-inflammatory cytokines. Findings suggest that targeting cytokines and growth factors could be a potential therapeutic strategy for reducing neuroinflammation and slowing down the progression of dementia. Additionally, prostaglandins contribute to synaptic plasticity and can act as retrograde messengers in synapses. Research has implicated COX-1, one of the isoforms of the enzyme, in neuroinflammation and neurodegenerative disorders. The inhibition of COX-1 might potentially prevent impairments in working memory and reduce neuroinflammation caused by beta-amyloid proteins in some conditions, such as Alzheimer's disease (AD). Cyclooxygenase-2, an inducible form of the enzyme, is expressed in cortical and hippocampal neurons and is associated with long-term synaptic plasticity. The inhibition or knockout of COX-2 has been shown to decrease long-term potentiation, a process involved in memory formation. Studies have also demonstrated that the administration of COX-2 inhibitors impairs cognitive function and memory acquisition and recall in animal models. There remains a debate regarding the effects of aspirin on dementia and cognitive decline. Although some studies suggest a possible protective effect of non-steroidal anti-inflammatory drugs, including aspirin, against the development of AD, others have shown inconsistent evidence. This review provides an overview of the effects of ASA or its active metabolite salicylate on learning, memory, and synaptic plasticity.
Collapse
Affiliation(s)
- Masoumeh Gholami
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran.
| | - Mehdi Sadegh
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Masoumeh Koroush-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Rasoul Jafari Arismani
- Department of Urologic Surgery, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Erfan Asadi
- Medical Student, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Mohammad Amini
- Medical Student, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Nahid Khodayari
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
6
|
Pan SW, Hu LS, Wang H, Li RT, He YJ, Shang Y, Dai ZL, Chen LX, Xiong W. Resolvin D1 Induces mTOR-independent and ATG5-dependent Autophagy in BV-2 Microglial Cells. Curr Med Sci 2023; 43:1096-1106. [PMID: 37924386 DOI: 10.1007/s11596-023-2787-5] [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: 02/07/2023] [Accepted: 08/03/2023] [Indexed: 11/06/2023]
Abstract
OBJECTIVE The activation state of microglia is known to occupy a central position in the pathophysiological process of cerebral inflammation. Autophagy is a catabolic process responsible for maintaining cellular homeostasis. In recent years, autophagy has been demonstrated to play an important role in neuroinflammation. Resolvin D1 (RvD1) is a promising therapeutic mediator that has been shown to exert substantial anti-inflammatory and proresolving activities. However, whether RvD1-mediated resolution of inflammation in microglia is related to autophagy regulation needs further investigation. The present study aimed to explore the effect of RvD1 on microglial autophagy and its corresponding pathways. METHODS Mouse microglial cells (BV-2) were cultured, treated with RvD1, and examined by Western blotting, confocal immunofluorescence microscopy, transmission electron microscopy, and flow cytometry. RESULTS RvD1 promoted autophagy in both BV-2 cells and mouse primary microglia by favoring the maturation of autophagosomes and their fusion with lysosomes. Importantly, RvD1 had no significant effect on the activation of mammalian target of rapamycin (mTOR) signaling. Furthermore, RvD1-induced mTOR-independent autophagy was confirmed by observing reduced cytoplasmic calcium levels and suppressed calcium/calmodulin-dependent protein kinase II (CaMK II) activation. Moreover, by downregulating ATG5, the increased phagocytic activity induced by RvD1 was demonstrated to be tightly controlled by ATG5-dependent autophagy. CONCLUSION The present work identified a previously unreported mechanism responsible for the role of RvD1 in microglial autophagy, highlighting its therapeutic potential against neuroinflammation.
Collapse
Affiliation(s)
- Shang-Wen Pan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Li-Sha Hu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Han Wang
- Department of Anesthesiology, Qingdao Women and Children's Hospital, Qingdao University, Qingdao, 266034, China
| | - Rui-Ting Li
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ya-Jun He
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhong-Liang Dai
- Department of Anesthesiology, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, 518020, China
| | - Li-Xin Chen
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Wei Xiong
- Department of Anesthesiology, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, 518020, China.
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
7
|
Sánchez-García S, Jaén RI, Fernández-Velasco M, Delgado C, Boscá L, Prieto P. Lipoxin-mediated signaling: ALX/FPR2 interaction and beyond. Pharmacol Res 2023; 197:106982. [PMID: 37925045 DOI: 10.1016/j.phrs.2023.106982] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/06/2023]
Abstract
In the aftermath of tissue injury or infection, an efficient resolution mechanism is crucial to allow tissue healing and preserve appropriate organ functioning. Pro-resolving bioactive lipids prevent uncontrolled inflammation and its consequences. Among these mediators, lipoxins were the first described and their pro-resolving actions have been mainly described in immune cells. They exert their actions mostly through formyl-peptide receptor 2 (ALX/FPR2 receptor), a G-protein-coupled receptor whose biological function is tremendously complex, primarily due to its capacity to mediate variable cellular responses. Moreover, lipoxins can also interact with alternative receptors like the cytoplasmic aryl hydrocarbon receptor, the cysteinyl-leukotrienes receptors or GPR32, triggering different intracellular signaling pathways. The available information about this complex response mediated by lipoxins is addressed in this review, going over the different mechanisms used by these molecules to stop the inflammatory reaction and avoid the development of dysregulated and chronic pathologies.
Collapse
Affiliation(s)
- Sergio Sánchez-García
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael I Jaén
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - María Fernández-Velasco
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain; Instituto de Investigación del Hospital La Paz, IdiPaz, Madrid, Spain
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain.
| | - Patricia Prieto
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
8
|
Thornton JM, Padovani CM, Rodriguez A, Spur BW, Yin K. Lipoxin A 4 promotes antibiotic and monocyte bacterial killing in established Pseudomonas aeruginosa biofilm formed under hydrodynamic conditions. FASEB J 2023; 37:e23098. [PMID: 37462621 PMCID: PMC10694838 DOI: 10.1096/fj.202300619r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023]
Abstract
Pseudomonas aeruginosa is a gram-negative, opportunistic bacteria commonly found in wounds and in lungs of immunocompromised patients. These bacteria commonly form biofilms which encapsulate the bacteria, making it difficult for antibiotics or immune cells to reach the bacterial cells. We previously reported that Lipoxin A4 (LxA4 ), a Specialized Pro-resolving Mediator, has direct effects on P. aeruginosa where it reduced biofilm formation and promoted ciprofloxacin antibiotic efficacy in a static biofilm-forming system. In the current studies, we examined the actions of LxA4 on established biofilms formed in a biofilm reactor under dynamic conditions with constant flow and shear stress. These conditions allow for biofilm growth with nutrient replenishment and for examination of bacteria within the biofilm structure. We show that LxA4 helped ciprofloxacin reduction of live/dead ratio of bacteria within the biofilm. THP-1 monocytes interacted with the biofilm to increase the number of viable bacteria within the biofilm as well as TNF-α production in the biofilm milieu, suggesting that monocyte interaction with bacterial biofilm exacerbates the inflammatory state. Pre-treatment of the THP-1 monocytes with LxA4 abolished the increase in biofilm bacteria and reduced TNF-α production. The effect of decreased biofilm bacteria was associated with increased LxA4 -induced monocyte adherence to biofilm but not increased bacteria killing suggesting that the mechanism for the reduced biofilm bacteria was due to LxA4 -mediated increase in adherence to biofilm. These results suggest that LxA4 can help antibiotic efficacy and promote monocyte activity against established P. aeruginosa biofilm formed under hydrodynamic conditions.
Collapse
Affiliation(s)
- Julianne M. Thornton
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine Stratford, NJ, USA 08084
| | - Cristina M. Padovani
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine Stratford, NJ, USA 08084
| | - Ana Rodriguez
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine Stratford, NJ, USA 08084
| | - Bernd W. Spur
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine Stratford, NJ, USA 08084
| | - Kingsley Yin
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine Stratford, NJ, USA 08084
| |
Collapse
|
9
|
Vergil Andrews JF, Selvaraj DB, Kumar A, Roshan SA, Anusuyadevi M, Kandasamy M. A Mild Dose of Aspirin Promotes Hippocampal Neurogenesis and Working Memory in Experimental Ageing Mice. Brain Sci 2023; 13:1108. [PMID: 37509038 PMCID: PMC10376986 DOI: 10.3390/brainsci13071108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Aspirin has been reported to prevent memory decline in the elderly population. Adult neurogenesis in the hippocampus has been recognized as an underlying basis of learning and memory. This study investigated the effect of aspirin on spatial memory in correlation with the regulation of hippocampal neurogenesis and microglia in the brains of ageing experimental mice. Results from the novel object recognition (NOR) test, Morris water maze (MWM), and cued radial arm maze (cued RAM) revealed that aspirin treatment enhances working memory in experimental mice. Further, the co-immunohistochemical assessments on the brain sections indicated an increased number of doublecortin (DCX)-positive immature neurons and bromodeoxyuridine (BrdU)/neuronal nuclei (NeuN) double-positive newly generated neurons in the hippocampi of mice in the aspirin-treated group compared to the control group. Moreover, a reduced number of ionized calcium-binding adaptor molecule (Iba)-1-positive microglial cells was evident in the hippocampus of aspirin-treated animals. Recently, enhanced activity of acetylcholinesterase (AChE) in circulation has been identified as an indicative biomarker of dementia. The biochemical assessment in the blood of aspirin-treated mice showed decreased activity of AChE in comparison with that of the control group. Results from this study revealed that aspirin facilitates hippocampal neurogenesis which might be linked to enhanced working memory.
Collapse
Affiliation(s)
- Jemi Feiona Vergil Andrews
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India; (J.F.V.A.); (D.B.S.); (A.K.)
| | - Divya Bharathi Selvaraj
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India; (J.F.V.A.); (D.B.S.); (A.K.)
| | - Akshay Kumar
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India; (J.F.V.A.); (D.B.S.); (A.K.)
| | - Syed Aasish Roshan
- Molecular Neuro-Gerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India; (S.A.R.); (M.A.)
| | - Muthuswamy Anusuyadevi
- Molecular Neuro-Gerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India; (S.A.R.); (M.A.)
| | - Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India; (J.F.V.A.); (D.B.S.); (A.K.)
- University Grants Commission-Faculty Recharge Programme (UGC-FRP), New Delhi 110002, India
| |
Collapse
|
10
|
Xia Q, Mao M, Zhan G, Luo Z, Zhao Y, Li X. SENP3-mediated deSUMOylation of c-Jun facilitates microglia-induced neuroinflammation after cerebral ischemia and reperfusion injury. iScience 2023; 26:106953. [PMID: 37332598 PMCID: PMC10272502 DOI: 10.1016/j.isci.2023.106953] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 04/18/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
Recent evidences have implicated that SENP3 is a deSUMOylase which possesses neuronal damage effects in cerebral ischemia. However, its role in microglia remains poorly understood. Here, we found that SENP3 was upregulated in the peri-infarct areas of mice following ischemic stroke. Furthermore, knockdown of SENP3 significantly inhibits the expression of proinflammatory cytokines and chemokines in microglial cells. Mechanistically, SENP3 can bind and then mediated the deSUMOylation of c-Jun, which activated its transcriptional activity, ultimately followed by the activation of MAPK/AP-1 signaling pathway. In addition, microglia-specific SENP3 knockdown alleviated ischemia-induced neuronal damage, and markedly diminished infract volume, ameliorated sensorimotor and cognitive function in animals subjected to ischemic stroke. These results indicated SENP3 functions as a novel regulator of microglia-induced neuroinflammation by activating the MAPK/AP-1 signaling pathway via mediating the deSUMOylation of c-Jun. Interventions of SENP3 expression or its interaction with c-Jun would be a new and promising therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Qian Xia
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Meng Mao
- Department of Anesthesiology and Perioperative Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Gaofeng Zhan
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhenzhao Luo
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xing Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
11
|
Harwood JL. Polyunsaturated Fatty Acids: Conversion to Lipid Mediators, Roles in Inflammatory Diseases and Dietary Sources. Int J Mol Sci 2023; 24:ijms24108838. [PMID: 37240183 DOI: 10.3390/ijms24108838] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are important components of the diet of mammals. Their role was first established when the essential fatty acids (EFAs) linoleic acid and α-linolenic acid were discovered nearly a century ago. However, most of the biochemical and physiological actions of PUFAs rely on their conversion to 20C or 22C acids and subsequent metabolism to lipid mediators. As a generalisation, lipid mediators formed from n-6 PUFAs are pro-inflammatory while those from n-3 PUFAs are anti-inflammatory or neutral. Apart from the actions of the classic eicosanoids or docosanoids, many newly discovered compounds are described as Specialised Pro-resolving Mediators (SPMs) which have been proposed to have a role in resolving inflammatory conditions such as infections and preventing them from becoming chronic. In addition, a large group of molecules, termed isoprostanes, can be generated by free radical reactions and these too have powerful properties towards inflammation. The ultimate source of n-3 and n-6 PUFAs are photosynthetic organisms which contain Δ-12 and Δ-15 desaturases, which are almost exclusively absent from animals. Moreover, the EFAs consumed from plant food are in competition with each other for conversion to lipid mediators. Thus, the relative amounts of n-3 and n-6 PUFAs in the diet are important. Furthermore, the conversion of the EFAs to 20C and 22C PUFAs in mammals is rather poor. Thus, there has been much interest recently in the use of algae, many of which make substantial quantities of long-chain PUFAs or in manipulating oil crops to make such acids. This is especially important because fish oils, which are their main source in human diets, are becoming limited. In this review, the metabolic conversion of PUFAs into different lipid mediators is described. Then, the biological roles and molecular mechanisms of such mediators in inflammatory diseases are outlined. Finally, natural sources of PUFAs (including 20 or 22 carbon compounds) are detailed, as well as recent efforts to increase their production.
Collapse
Affiliation(s)
- John L Harwood
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, Wales, UK
| |
Collapse
|
12
|
Inhibition of Microglial GSK3β Activity Is Common to Different Kinds of Antidepressants: A Proposal for an In Vitro Screen to Detect Novel Antidepressant Principles. Biomedicines 2023; 11:biomedicines11030806. [PMID: 36979785 PMCID: PMC10045655 DOI: 10.3390/biomedicines11030806] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/17/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Depression is a major public health concern. Unfortunately, the present antidepressants often are insufficiently effective, whilst the discovery of more effective antidepressants has been extremely sluggish. The objective of this review was to combine the literature on depression with the pharmacology of antidepressant compounds, in order to formulate a conceivable pathophysiological process, allowing proposals how to accelerate the discovery process. Risk factors for depression initiate an infection-like inflammation in the brain that involves activation microglial Toll-like receptors and glycogen synthase kinase-3β (GSK3β). GSK3β activity alters the balance between two competing transcription factors, the pro-inflammatory/pro-oxidative transcription factor NFκB and the neuroprotective, anti-inflammatory and anti-oxidative transcription factor NRF2. The antidepressant activity of tricyclic antidepressants is assumed to involve activation of GS-coupled microglial receptors, raising intracellular cAMP levels and activation of protein kinase A (PKA). PKA and similar kinases inhibit the enzyme activity of GSK3β. Experimental antidepressant principles, including cannabinoid receptor-2 activation, opioid μ receptor agonists, 5HT2 agonists, valproate, ketamine and electrical stimulation of the Vagus nerve, all activate microglial pathways that result in GSK3β-inhibition. An in vitro screen for NRF2-activation in microglial cells with TLR-activated GSK3β activity, might therefore lead to the detection of totally novel antidepressant principles with, hopefully, an improved therapeutic efficacy.
Collapse
|
13
|
dos Santos HT, Nam K, Gil D, Yellepeddi V, Baker OJ. Current experimental methods to investigate the impact of specialized pro-resolving lipid mediators on Sjögren's syndrome. Front Immunol 2023; 13:1094278. [PMID: 36713415 PMCID: PMC9878840 DOI: 10.3389/fimmu.2022.1094278] [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: 11/09/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
Sjögren's syndrome is a chronic inflammatory autoimmune disease characterized by diminished secretory function of the exocrine glands. Although extensive investigation has been done to understand Sjögren's syndrome, the causes of the disease are as yet unknown and treatments remain largely ineffective, with established therapeutic interventions being limited to use of saliva substitutes with modest effectiveness. A primary feature of Sjögren's syndrome is uncontrolled inflammation of exocrine tissues and previous studies have demonstrated that lipid-based specialized pro-resolving mediators reduce inflammation and restores tissue integrity in salivary glands. However, these studies are limited to a single specialized pro-resolving lipid mediator's family member resolvin D1 or RvD1 and its aspirin-triggered epimer, AT-RvD1. Consequently, additional studies are needed to explore the potential benefits of other members of the specialized pro-resolving lipid mediator's family and related molecules (e.g., additional resolvin subtypes as well as lipoxins, maresins and protectins). In support of this goal, the current review aims to briefly describe the range of current experimental methods to investigate the impact of specialized pro-resolving lipid mediators on Sjögren's syndrome, including both strengths and weaknesses of each approach where this information is known. With this article, the possibilities presented by specialized pro-resolving lipid mediators will be introduced to a wider audience in immunology and practical advice is given to researchers who may wish to take up this work.
Collapse
Affiliation(s)
- Harim T. dos Santos
- Bond Life Sciences Center, University of Missouri, Columbia, MO, United States,Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Kihoon Nam
- Bond Life Sciences Center, University of Missouri, Columbia, MO, United States,Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Diana Gil
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, United States,Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States,Department of Biological and Biomedical Engineering, College of Engineering, University of Missouri, Columbia, MO, United States
| | - Venkata Yellepeddi
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, United States,Department of Molecular Pharmaceutics, College of Pharmacy, University of Utah, Salt Lake City, UT, United States
| | - Olga J. Baker
- Bond Life Sciences Center, University of Missouri, Columbia, MO, United States,Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Missouri, Columbia, MO, United States,Department of Biochemistry, University of Missouri, Columbia, MO, United States,*Correspondence: Olga J. Baker,
| |
Collapse
|
14
|
Abstract
OBJECTIVE This review aims to summarize the capability of lipoxin in regulating oxidative stress. BACKGROUND Oxidative stress is defined as an imbalance between the production of free radicals and the antioxidant system, and it is associated with the existence of a large number of oxidation products, such as reactive oxygen species (ROS) and reaction nitrogen species (RNS), causing damage to human tissues through immunoinflammatory responses. Therefore, reducing oxidative stress is vital to alleviate pathological damage. Lipoxin, an acronym for lipoxygenase interaction product, is a bioactive autacoid metabolite of arachidonic acid made by various cell types. Previous studies have shown that lipoxin is associated with a variety of biological functions, including anti-inflammatory, regulating immune responses, promoting the repair of damaged cells, etc. The deficiency of lipoxin is a critical pathological mechanism in different diseases. Moreover, the ability of lipoxin to attenuate oxidative stress is noteworthy, thereby protecting the human body from diverse diseases. METHODS We searched papers from PubMed database using search terms, such as lipoxin, lipoxin A4, oxidative stress, and other relevant terms. RESULTS A total of 103 articles published over the past 20 years were identified for inclusion. We summarized the capability of lipoxin in regulating oxidative stress and mechanism. CONCLUSION Lipoxin is provided with a protective role in attenuating oxidative stress.
Collapse
|
15
|
Lu Y, Li B, Xu A, Liang X, Xu T, Jin H, Xie Y, Wang R, Liu X, Gao X, Han Y, Zeng J. NF-κB and AP-1 are required for the lipopolysaccharide-induced expression of MCP-1, CXCL1, and Cx43 in cultured rat dorsal spinal cord astrocytes. Front Mol Neurosci 2022; 15:859558. [PMID: 35966011 PMCID: PMC9368326 DOI: 10.3389/fnmol.2022.859558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
TLR4 and Cx43 signaling in dorsal spinal cord has been shown to be involved in the development of neuropathic pain. However, it is not clear whether TLR4 signaling is associated with the expression of MCP-1, CXCL1, and Cx43 in LPS (lipopolysaccharide)-treated rat dorsal spinal cord astrocytes under in vitro condition. In the present study, we found that TLR4 antagonist TAK-242 significantly inhibited LPS-induced MCP-1, CXCL1, and Cx43 expression, suggesting the role of TLR4 in response to LPS in cultured dorsal spinal cord astrocytes. Application of TAK-242 significantly blocked LPS-induced NF-κB and AP-1 activity and the expression of MCP-1, CXCL1 and Cx43. Furthermore, NF-κB inhibitor PDTC and AP-1 inhibitor SR11302 significantly blocked LPS-induced MCP-1, CXCL1, and Cx43 expression. DNA-binding activity of NF-κB, its effect on MCP-1 expression was suppressed by PDTC and SR11302. On the other hand, DNA-binding activity of AP-1, its effect on CXCL1 or Cx43 expression was also suppressed by PDTC and SR11302. In addition, PDTC was found to inhibit the nuclear translocation of AP-1 and the expression of c-Jun induced by LPS, which suggested that NF-κBp65 is essential for the AP-1 activity. Similarly, SR11302 significantly blocked LPS-induced the nuclear translocation of NF-κBp65 and the expression of NF-κBp65 induced by LPS. Pretreatment with CBX, Gap26, or Gap19 (Cx43 blockers) significantly inhibited abnormal astrocytic hemichannel opening and chemokines (MCP-1 and CXCL1) release in LPS-stimulated astrocytes. In summary, cell culture experiments revealed that LPS stimulation could evoke TLR4 signaling with the subsequent activation of NF-κB and AP-1, resulting in the expression of MCP-1, CXCL1, and Cx43. TLR4 activation increased Cx43 hemichannel, but not gap-junction activities and induced the release of the MCP-1 and CXCL1 from astrocytes via Cx43 hemichannel. These findings may help us to understand the role of astrocytic signaling in inflammatory response within dorsal spinal cord tissue.
Collapse
|
16
|
Zheng J, Zhang W, Kang P, Zheng X, He K, Bai H, Yu X. Midazolam Ameliorates Impairment of the Blood-Brain Barrier (BBB) Against LPS. Neurotox Res 2022; 40:751-762. [PMID: 35451708 DOI: 10.1007/s12640-022-00508-4] [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: 02/03/2022] [Revised: 03/27/2022] [Accepted: 04/09/2022] [Indexed: 10/18/2022]
Abstract
Central nervous system (CNS) dysfunction induced by sepsis and pathogenic microbial infections is reported to be closely associated with increased permeability of the blood-brain barrier (BBB), which is mainly mediated by the stimulation of lipopolysaccharide (LPS) on inflammatory signaling. Midazolam is a novel sedative acting on the benzodiazepine receptor, which is recently reported to exert a neuroprotective effect by inhibiting inflammation. The present study will explore the potential repair capacity of Midazolam on LPS-induced damage to the BBB. The in vivo mice model was established by intraperitoneal injection of LPS, while the in vitro model was constructed by stimulating endothelial cells utilizing LPS. We found that the increased malondialdehyde (MDA) level and reduced superoxide dismutase (SOD) activity in the brain cortices, promoted serum concentration of inflammatory factors, and elevated BBB permeability were found in the LPS group, all of which were dramatically reversed by 1 mg/kg and 2 mg/kg Midazolam. Interestingly, Midazolam increased the expression of the tight junction protein zonula occludens-1 (ZO-1). In LPS-challenged in vitro human brain microvascular endothelial cells (HBMECs), the increased concentration of inflammatory factors, reduced trans-endothelial electrical resistance (TEER) level, elevated relative value of trans-endothelial permeability, and downregulated ZO-1 were observed, all of which were pronouncedly alleviated by Midazolam, accompanied by the inhibition on the Ras homolog family member A/ Rho-kinase 2 (RhoA/ROCK-2) pathway. Furthermore, the regulatory effects of Midazolam on ZO-1 expression and the endothelial monolayer permeability in LPS-challenged HBMECs were abolished by the overexpression of RhoA. Collectively, our data imply that Midazolam ameliorated the impairment of the BBB against LPS by regulating the RhoA/ROCK2 pathway.
Collapse
Affiliation(s)
- Juyan Zheng
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Wei Zhang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Bejing, 100730, China
| | - PeiPei Kang
- Department of Anesthesiology, Affiliated Tumor Hospital of Nantong University, Nantong, 226006, China
| | - Xiaojiao Zheng
- Department of Blood Transfusion, The First Affiliated Hospital of Baotou Medical College, Baotou, 014017, China
| | - Kai He
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Hong Bai
- Department of Anesthesiology, Wuhai People's Hospital, Wuhai, 016099, China
| | - Xuerong Yu
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
17
|
Zhang XF, Luan MZ, Yan WB, Zhao FL, Hou Y, Hou GG, Meng QG. Anti-neuroinflammatory effects of novel 5,6-dihydrobenzo[h]quinazolin-2-amine derivatives in lipopolysaccharide-stimulated BV2 microglial cells. Eur J Med Chem 2022; 235:114322. [DOI: 10.1016/j.ejmech.2022.114322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 01/07/2023]
|
18
|
Zhang J, Li Z, Fan M, Jin W. Lipoxins in the Nervous System: Brighter Prospects for Neuroprotection. Front Pharmacol 2022; 13:781889. [PMID: 35153778 PMCID: PMC8826722 DOI: 10.3389/fphar.2022.781889] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/07/2022] [Indexed: 12/28/2022] Open
Abstract
Lipoxins (LXs) are generated from arachidonic acid and are involved in the resolution of inflammation and confer protection in a variety of pathological processes. In the nervous system, LXs exert an array of protective effects against neurological diseases, including ischemic or hemorrhagic stroke, neonatal hypoxia-ischemia encephalopathy, brain and spinal cord injury, Alzheimer's disease, multiple sclerosis, and neuropathic pain. Lipoxin administration is a potential therapeutic strategy in neurological diseases due to its notable efficiency and unique superiority regarding safety. Here, we provide an overview of LXs in terms of their synthesis, signaling pathways and neuroprotective evidence. Overall, we believe that, along with advances in lipoxin-related drug design, LXs will bring brighter prospects for neuroprotection.
Collapse
Affiliation(s)
- Jiayu Zhang
- Graduate School of Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Zhe Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Mingyue Fan
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Wei Jin
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
19
|
Tung YT, Wei CH, Yen CC, Lee PY, Ware LB, Huang HE, Chen W, Chen CM. Aspirin Attenuates Hyperoxia-Induced Acute Respiratory Distress Syndrome (ARDS) by Suppressing Pulmonary Inflammation via the NF-κB Signaling Pathway. Front Pharmacol 2022; 12:793107. [PMID: 35111059 PMCID: PMC8802116 DOI: 10.3389/fphar.2021.793107] [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: 10/11/2021] [Accepted: 12/16/2021] [Indexed: 12/18/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common destructive syndrome with high morbidity and mortality rates. Currently, few effective therapeutic interventions for ARDS are available. Clinical trials have shown that the effectiveness of aspirin is inconsistent. The contribution of platelets to the inflammatory response leading to the development of ARDS is increasingly recognized. The antiplatelet agent aspirin reportedly exerts a protective effect on acid- and hyperoxia-induced lung injury in murine models. Our previous study showed that pretreatment with aspirin exerts protective effects on hyperoxia-induced lung injury in mice. However, the mechanisms and therapeutic efficacy of aspirin in the posttreatment of hyperoxia-induced acute lung injury (ALI) remain unclear. In this study, we used a homozygous NF-κB-luciferase+/+ transgenic mouse model and treated mice with low-dose (25 μg/g) or high-dose (50 μg/g) aspirin at 0, 24, and 48 h after exposure to hyperoxia (inspired oxygen fraction (FiO2) > 95%). Hyperoxia-induced lung injury significantly increased the activation of NF-κB in the lung and increased the levels of macrophages infiltrating the lung and reactive oxygen species (ROS), increased the HO-1, NF-κB, TNF-α, IL-1β, and IL-4 protein levels, and reduced the CC10, SPC, eNOS, Nrp-1, and IκBα protein levels in the lung tissue. Pulmonary edema and alveolar infiltration of neutrophils were also observed in the lung tissue of mice exposed to hyperoxia. However, in vivo imaging revealed that posttreatment with aspirin reduced luciferase expression, suggesting that aspirin might reduce NF-κB activation. Posttreatment with aspirin also reduced hyperoxia-induced increases in the numbers of lung macrophages, intracellular ROS levels, and the expression of TNF-α, IL-1β, and IL-4; it also increased CC10, SPC and Nrp-1 levels compared with hyperoxia exposure alone. Lung histopathology also indicated that the aspirin posttreatment significantly reduced neutrophil infiltration and lung edema compared with hyperoxia exposure alone. Aspirin effectively induces an anti-inflammatory response in a model of hyperoxia-induced lung injury. Thus, aspirin may have potential as a novel treatment for hyperoxia-induced ALI.
Collapse
Affiliation(s)
- Yu-Tang Tung
- Department of Life Sciences and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan.,Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan.,Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Nutrition Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chi-Hsuan Wei
- Department of Life Sciences and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan.,Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chih-Ching Yen
- Department of Internal Medicine, China Medical University Hospitaland College of Health Care, China Medical University, Taichung, Taiwan
| | - Po-Ying Lee
- Department of Surgery, Division of Plastic Surgery, Cathay General Hospital, Taipei, Taiwan
| | - Lorraine B Ware
- Departments of Medicine and Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Hao-En Huang
- Nutrition Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Wei Chen
- Division of Pulmonary and Critical Care Medicine, Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan.,Rong Hsing Research Center for Translational Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| |
Collapse
|
20
|
Futokoro R, Hijioka M, Arata M, Kitamura Y. Lipoxin A4 Receptor Stimulation Attenuates Neuroinflammation in a Mouse Model of Intracerebral Hemorrhage. Brain Sci 2022; 12:brainsci12020162. [PMID: 35203926 PMCID: PMC8869920 DOI: 10.3390/brainsci12020162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is caused by the rupture of blood vessels in the brain. The excessive activation of glial cells and the infiltration of numerous inflammatory cells are observed during bleeding. Thrombin is a key molecule that triggers neuroinflammation in the ICH brain. In this study, we focused on lipoxin A4 (LXA4), an arachidonic acid metabolite that has been reported to suppress inflammation and cell migration. LXA4 and BML-111, an agonist of the LXA4 receptor/formyl peptide receptor 2 (ALX/FPR2), suppressed microglial activation; LXA4 strongly inhibited the migration of neutrophil-like cells in vitro. ALX/FPR2 was expressed on neutrophils in the ICH mouse brain and the daily administration of BML-111 attenuated the motor coordination dysfunction and suppressed the production of proinflammatory cytokines in the ICH mouse brain. On the other hand, BML-111 did not show a significant reduction in the number of microglia and neutrophils. These results suggest that systemic administration of ALX/FPR2 agonists may suppress the neuroinflammatory response of microglia and neutrophils without a change in cell numbers. Additionally, their combination with molecules that reduce cell numbers, such as modulators of leukotriene B4 signaling, may be required in future studies.
Collapse
Affiliation(s)
- Risa Futokoro
- Laboratory of Pharmacology and Neurobiology, Collage of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan; (R.F.); (M.A.); (Y.K.)
- Department of Pharmacology II, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University, 11-68 Koshien Kyuban-cho, Nishinomiya 663-8179, Japan
| | - Masanori Hijioka
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
- Correspondence: ; Tel.: +81-52-853-8196
| | - Moe Arata
- Laboratory of Pharmacology and Neurobiology, Collage of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan; (R.F.); (M.A.); (Y.K.)
| | - Yoshihisa Kitamura
- Laboratory of Pharmacology and Neurobiology, Collage of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu 525-8577, Japan; (R.F.); (M.A.); (Y.K.)
| |
Collapse
|
21
|
Irisin Protects Against LPS-Stressed Cardiac Damage Through Inhibiting Inflammation, Apoptosis, and Pyroptosis. Shock 2021; 56:1009-1018. [PMID: 34779800 DOI: 10.1097/shk.0000000000001775] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
ABSTRACT Septic cardiac dysfunction remains a clinical problem due to its high morbidity and mortality. Uncontrolled cell death and excessive inflammatory response are closely related to sepsis-induced cardiac dysfunction. Irisin has been found to play cardioprotective roles in sepsis. However, there is enough uncertainty in the mechanism of irisin-mediated cardioprotection. We hypothesized that irisin may ameliorate myocardial dysfunction via reducing cardiac apoptosis, pyroptosis, and inflammation during LPS-induced sepsis. Mice were subjected to LPS with or without irisin treatment. After stimuli of LPS, the function of myocardium was distinctly impaired, which was closely related to increased level of apoptosis (decreased expression of Bcl-2 and elevated expression of Caspase-3 and Bax), pyroptosis (increased expression of Caspase1, NLR family pyrin domain containing 3 (NLRP3), and gasdermin D) and inflammatory mediators (increased level of IL-1β, TNF-α, and IL-6). This process is consistent with increased toll-like receptor 4 (TLR4)/nuclear factor-kappa B signal, apoptotic signal, and NLRP3-mediated pyroptotic signal. Activation of apoptosis and pyroptosis enhanced the expression of proinflammatory cytokines and further exacerbated septic myocardial damage. However, irisin can inhibit the expression of TLR4 and its downstream signaling molecules and also lower the level of apoptosis and pyroptosis. Besides, similar results were also found in vitro model of LPS-induced H9c2 cardiomyocyte injury. In general, irisin suppressed inflammation, apoptosis, and pyroptosis by blocking the TLR4 and NLRP3 inflammasome signalings to mitigate myocardial dysfunction in sepsis.
Collapse
|
22
|
The Preeclamptic Environment Promotes the Activation of Transcription Factor Kappa B by P53/RSK1 Complex in a HTR8/SVneo Trophoblastic Cell Line. Int J Mol Sci 2021; 22:ijms221910200. [PMID: 34638542 PMCID: PMC8508006 DOI: 10.3390/ijms221910200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/13/2021] [Accepted: 09/17/2021] [Indexed: 12/16/2022] Open
Abstract
Preeclampsia is a pregnancy disorder associated with shallow placentation, forcing placental cells to live in hypoxic conditions. This activates the transcription factor kappa B (NFκB) in maternal and placental cells. Although the role of NFκB in preeclampsia is well documented, its mechanism of activation in trophoblastic cells has been never studied. This study investigates the mechanism of NFκB activation in a first trimester trophoblastic cell line (HTR8/SVneo) stimulated by a medium containing serum from preeclamptic (PE) or normotensive (C) women in hypoxic (2% O2) or normoxic (8% O2) conditions. The results indicate that in HTR8/SVneo cells, the most widely studied NFκB pathways, i.e., canonical, non-canonical and atypical, are downregulated in environment PE 2% O2 in comparison to C 8% O2. Therefore, other pathways may be responsible for NFκB activation. One such pathway depends on the activation of NFκB by the p53/RSK1 complex through its phosphorylation at Serine 536 (pNFκB Ser536). The data generated by our study show that inhibition of the p53/RSK1 pathway by p53-targeted siRNA results in a depletion of pNFκB Ser536 in the nucleus, but only in cells incubated with PE serum at 2% O2. Thus, the p53/RSK1 complex might play a critical role in the activation of NFκB in trophoblastic cells and preeclamptic placentas.
Collapse
|
23
|
Tylek K, Trojan E, Leśkiewicz M, Regulska M, Bryniarska N, Curzytek K, Lacivita E, Leopoldo M, Basta-Kaim A. Time-Dependent Protective and Pro-Resolving Effects of FPR2 Agonists on Lipopolysaccharide-Exposed Microglia Cells Involve Inhibition of NF-κB and MAPKs Pathways. Cells 2021; 10:cells10092373. [PMID: 34572022 PMCID: PMC8472089 DOI: 10.3390/cells10092373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023] Open
Abstract
Prolonged or excessive microglial activation may lead to disturbances in the resolution of inflammation (RoI). The importance of specialized pro-resolving lipid mediators (SPMs) in RoI has been highlighted. Among them, lipoxins (LXA4) and aspirin-triggered lipoxin A4 (AT-LXA4) mediate beneficial responses through the activation of N-formyl peptide receptor-2 (FPR2). We aimed to shed more light on the time-dependent protective and anti-inflammatory impact of the endogenous SPMs, LXA4, and AT-LXA4, and of a new synthetic FPR2 agonist MR-39, in lipopolysaccharide (LPS)-exposed rat microglial cells. Our results showed that LXA4, AT-LXA4, and MR-39 exhibit a protective and pro-resolving potential in LPS-stimulated microglia, even if marked differences were apparent regarding the time dependency and efficacy of inhibiting particular biomarkers. The LXA4 action was found mainly after 3 h of LPS stimulation, and the AT-LXA4 effect was varied in time, while MR-39′s effect was mainly observed after 24 h of stimulation by endotoxin. MR-39 was the only FPR2 ligand that attenuated LPS-evoked changes in the mitochondrial membrane potential and diminished the ROS and NO release. Moreover, the LPS-induced alterations in the microglial phenotype were modulated by LXA4, AT-LXA4, and MR-39. The anti-inflammatory effect of MR-39 on the IL-1β release was mediated through FPR2. All tested ligands inhibited TNF-α production, while AT-LXA4 and MR-39 also diminished IL-6 levels in LPS-stimulated microglia. The favorable action of LXA4 and MR-39 was mediated through the inhibition of ERK1/2 phosphorylation. AT-LXA4 and MR39 diminished the phosphorylation of the transcription factor NF-κB, while AT-LXA4 also affected p38 kinase phosphorylation. Our results suggest that new pro-resolving synthetic mediators can represent an attractive treatment option for the enhancement of RoI, and that FPR2 can provide a perspective as a target in immune-related brain disorders.
Collapse
Affiliation(s)
- Kinga Tylek
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Ewa Trojan
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Monika Leśkiewicz
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Magdalena Regulska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Natalia Bryniarska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Katarzyna Curzytek
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Enza Lacivita
- Department of Pharmacy—Drug Sciences, University of Bari, Via Orabona 4, 70125 Bari, Italy; (E.L.); (M.L.)
| | - Marcello Leopoldo
- Department of Pharmacy—Drug Sciences, University of Bari, Via Orabona 4, 70125 Bari, Italy; (E.L.); (M.L.)
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
- Correspondence: ; Tel.: +48-12-662-32-73
| |
Collapse
|
24
|
Awad MA, Ahmed ZSO, AbuBakr HO, Elbargeesy GAEFH, Moussa MHG. Fipronil induced oxidative stress in neural tissue of albino rat with subsequent apoptosis and tissue reactivity. Acta Histochem 2021; 123:151764. [PMID: 34352653 DOI: 10.1016/j.acthis.2021.151764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 07/20/2021] [Accepted: 07/27/2021] [Indexed: 11/15/2022]
Abstract
Fipronil (FIP) insecticide is extensively used in agriculture, public health and veterinary medicine. Although it is considered as a neurotoxin to insects (target organisms) and exhibits neurological signs upon vertebrates (non-target organisms) exposure, slight is known about its potential neurotoxic effects and its molecular mechanisms on vertebrates. The current study is designed to assess oxidative stress as a molecular mechanism of FIP neurotoxicity subordinated with apoptosis and neural tissue reactivity. Ten adult male albino rats received 10 mg/kg body weight fipronil technical grade by oral gavage daily for 45 days (subacute exposure). Brain neural tissue regions (hippocampus, cerebellum and caudate putamen) were processed to examine oxidative stress induced cellular macromolecular alterations as MDA, PCC and DNA fragmentation. Besides, TNF-α and Bcl-2 gene expression and immunoreactivity for caspase-3 (active form), iNOS and GFAP were evaluated. Also, histopathological assessment was conducted. We found that FIP significantly raised MDA, PCC and DNA fragmentation (p ≤ 0.05). Also, it significantly upregulated TNF-α and non-significantly down-regulated Bcl-2 gene expression (p ≤ 0.05). Further, significant increased immunoreactivity to GFAP, iNOS and caspase-3 (active form) in these brain neural tissue regions in FIP treated group was noticed (p ≤ 0.05). Histopathological findings, including alterations in the histological architecture and neuronal degeneration, were also observed in these brain regions of FIP treated group. In conclusion, we suggest the ability of FIP to induce oxidative stress mediated macromolecular alterations, leading to apoptosis and tissue reaction in these brain regions which showed variable susceptibility to FIP toxic effects.
Collapse
Affiliation(s)
- Mohamed A Awad
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| | - Zainab Sabry Othman Ahmed
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| | - Huda O AbuBakr
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| | | | - Moukhtar H G Moussa
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| |
Collapse
|
25
|
The N-Formyl Peptide Receptor 2 (FPR2) Agonist MR-39 Exhibits Anti-Inflammatory Activity in LPS-Stimulated Organotypic Hippocampal Cultures. Cells 2021; 10:cells10061524. [PMID: 34204273 PMCID: PMC8235773 DOI: 10.3390/cells10061524] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 01/25/2023] Open
Abstract
Accumulating evidence indicates a pivotal role for chronic inflammatory processes in the pathogenesis of neurodegenerative and psychiatric disorders. G protein-coupled formyl peptide receptor 2 (FPR2) mediates pro-inflammatory or anti-/pro-resolving effects upon stimulation with biased agonists. We aimed to evaluate the effects of a new FPR2 ureidopropanamide agonist, compound MR-39, on neuroinflammatory processes in organotypic hippocampal cultures (OHCs) derived from control (WT) and knockout FPR2−/− mice (KO) exposed to bacterial endotoxin (lipopolysaccharide; LPS). Higher LPS-induced cytokine expression and basal release were observed in KO FPR2 cultures than in WT cultures, suggesting that a lack of FPR2 enhances the OHCs response to inflammatory stimuli. Pretreatment with MR-39 abolished some of the LPS-induced changes in the expression of genes related to the M1/M2 phenotypes (including Il-1β, Il-6, Arg1, Il-4, Cd74, Fizz and Cx3cr1) and TNF-α, IL-1β and IL-4 release in tissue derived from WT but not KO mice. Receptor specificity was confirmed by adding the FPR2 antagonist WRW4, which abolished the abovementioned effects of MR-39. Further biochemical data showed an increase in the phospho-p65/total p65 ratio after LPS stimulation in hippocampal tissues from both WT and KO mice, and MR-39 only reversed this effect on WT OHCs. LPS also increased TRAF6 levels, which are critical for the TLR4-mediated NF-κB pro-inflammatory responses. MR-39 attenuated the LPS-evoked increase in the levels of the NLRP3 and caspase-1 proteins in WT but not KO hippocampal cultures. Since NLRP3 may be involved in the pyroptosis, a lytic type of programmed cell death in which the main role is played by Gasdermin D (GSDMD), we examined the effects of LPS and/or MR-39 on the GSDMD protein level. LPS only increased GSDMD production in the WT tissues, and this effect was ameliorated by MR-39. Collectively, this study indicates that the new FPR2 agonist efficiently abrogates LPS-induced neuroinflammation in an ex vivo model, as evidenced by a decrease in pro-inflammatory cytokine expression and release as well as the downregulation of NLRP3 inflammasome-related pathways.
Collapse
|
26
|
Aspirin using was associated with slower cognitive decline in patients with Alzheimer's disease. PLoS One 2021; 16:e0252969. [PMID: 34133445 PMCID: PMC8208559 DOI: 10.1371/journal.pone.0252969] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/26/2021] [Indexed: 11/19/2022] Open
Abstract
We aimed to examine whether the use of aspirin is associated with change in cognitive performance over time, and whether this association is modified by the cognitive stages. This study included a total of 1866 subjects, including 509 subjects with normal cognition (NC), 985 subjects with mild cognitive impairment (MCI), and 372 patients with Alzheimer’s disease (AD). In each group, we further categorized our subjects into two groups based on their aspirin using conditions: Aspirin users and non-aspirin users. Mini-Mental State Examination (MMSE) was the cognitive outcome. Linear mixed models were conducted to examine the longitudinal relationship between the use of aspirin and cognitive performance in each diagnostic group. In the cross-sectional analysis, there were no significant differences in MMSE scores between non-aspirin users and aspirin users in subjects with NC, subjects with MCI or patients with AD. In the longitudinal analysis, we detected an association of the baseline use of aspirin with cognitive decline (MMSE) over time in patients with AD, but not in the NC group or MCI group. Specifically, in AD patients, the use of aspirin at baseline was associated with slower cognitive decline over time. Our data may support an association between the use of aspirin and slower cognitive decline, while this association may be dependent on the clinical stages.
Collapse
|
27
|
Melnikov V, Tiburcio-Jimenez D, Mendoza-Hernandez MA, Delgado-Enciso J, De-Leon-Zaragoza L, Guzman-Esquivel J, Rodriguez-Sanchez IP, Martinez-Fierro ML, Lara-Esqueda A, Delgado-Enciso OG, Jacinto-Cortes I, Zaizar-Fregoso SA, Paz-Michel BA, Murillo-Zamora E, Delgado-Enciso I, Galvan-Salazar HR. Improve cognitive impairment using mefenamic acid non-steroidal anti-inflammatory therapy: additional beneficial effect found in a controlled clinical trial for prostate cancer therapy. Am J Transl Res 2021; 13:4535-4543. [PMID: 34150033 PMCID: PMC8205720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/20/2021] [Indexed: 06/12/2023]
Abstract
Inflammation is an essential component of prostate cancer (PCa), and mefenamic acid has been reported to decrease its biochemical progression. The current standard therapy for PCa is androgen deprivation therapy (ADT), which has side effects such as cognitive dysfunction, risk of Alzheimer's disease, and dementia. Published results of in vitro tests and animal models studies have shown that mefenamic acid could be used as a neuroprotector. Objective: Examine the therapeutic potential of mefenamic acid in cognitive impairment used in a controlled clinical trial. Clinical trial phase II was conducted on patients undergoing ADT for PCa. Two groups of 14 patients were included. One was treated with a placebo, while the other received mefenamic acid 500 mg PO every 12hrs for six months. The outcome was evaluated through the Mini-Mental State Examination (MMSE) score at six months. At the beginning of the study, both groups had similar MMSE scores (mefenamic acid vs. placebo: 26.0±2.5 vs. 27.0±2.6, P=0.282). The mefenamic acid group improved its MMSE score after six months compared with the placebo group (27.7±1.8 vs. 25.5±4.2, P=0.037). Treatment with mefenamic acid significantly increases the probability of maintained or raised cognitive function compared to placebo (92% vs. 42.9%, RR=2.2, 95% CI: 1.16-4.03, NNT=2.0, 95% CI: 1.26-4.81, P=0.014). Furthermore, 42.9% of the placebo group patients had relevant cognitive decline (a 2-point decrease in the MMSE score), while in patients treated with mefenamic acid, cognitive impairment was not present. This study is the first conducted on humans that suggests that mefenamic acid protects against cognitive decline.
Collapse
Affiliation(s)
- Valery Melnikov
- Department of Molecular Medicine, School of Medicine, University of ColimaColima 28040, Mexico
| | - Daniel Tiburcio-Jimenez
- Department of Molecular Medicine, School of Medicine, University of ColimaColima 28040, Mexico
| | | | - Josuel Delgado-Enciso
- Department of Research, Foundation for Cancer Ethics, Education and Research of The Cancerology State InstituteColima, Mexico
| | - Luis De-Leon-Zaragoza
- Department of Research, Cancerology State Institute, Colima State Health ServicesColima, Mexico
| | - Jose Guzman-Esquivel
- Department of Research, General Hospital of Zone No. 1 IMSSVilla de Alvarez, Colima, Mexico
| | - Iram P Rodriguez-Sanchez
- Molecular and Structural Physiology Laboratory, School of Biological Sciences, Autonomous University of Nuevo LeonMonterrey, Nuevo Leon, Mexico
| | - Margarita L Martinez-Fierro
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Autonomous University of ZacatecasZacatecas, Mexico
| | - Agustin Lara-Esqueda
- Department of Research, Cancerology State Institute, Colima State Health ServicesColima, Mexico
| | - Osiris G Delgado-Enciso
- Department of Molecular Medicine, School of Medicine, University of ColimaColima 28040, Mexico
- Department of Research, Foundation for Cancer Ethics, Education and Research of The Cancerology State InstituteColima, Mexico
| | - Ivan Jacinto-Cortes
- Department of Research, Cancerology State Institute, Colima State Health ServicesColima, Mexico
| | - Sergio A Zaizar-Fregoso
- Department of Molecular Medicine, School of Medicine, University of ColimaColima 28040, Mexico
| | - Brenda A Paz-Michel
- Department of Molecular Medicine, School of Medicine, University of ColimaColima 28040, Mexico
| | | | - Ivan Delgado-Enciso
- Department of Molecular Medicine, School of Medicine, University of ColimaColima 28040, Mexico
- Department of Research, Cancerology State Institute, Colima State Health ServicesColima, Mexico
| | - Hector R Galvan-Salazar
- Department of Research, Cancerology State Institute, Colima State Health ServicesColima, Mexico
- Department of Research, General Hospital of Zone No. 1 IMSSVilla de Alvarez, Colima, Mexico
| |
Collapse
|
28
|
Liu J, Xu J, Mi Y, Yang Y, Li Q, Zhou D, Wei K, Chen G, Li N, Hou Y. Pterostilbene alleviates cerebral ischemia and reperfusion injury in rats by modulating microglial activation. Food Funct 2021; 11:5432-5445. [PMID: 32490497 DOI: 10.1039/d0fo00084a] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ischemic stroke is a severe neurological disease without known effective therapy. Microglia-mediated neuroinflammation plays an important role in ischemic stroke. Therefore, finding a safe and effective microglial activation inhibitor might lead to an effective therapeutic strategy against ischemic stroke. In this project, our goal was to explore both the mechanism and effect of pterostilbene in MCAO/R rats. The potential effect of pterostilbene on ischemic stroke was tested using MCAO/R rats and its effect on microglial activation was tested in LPS-stimulated BV-2 cells. In vivo, pterostilbene decreased the neurological scores, brain water content and infarct volume in MCAO/R rats. Pterostilbene increased the number of mature neurons, decreased the number of activated microglia, and reduced iNOS and IL-1β mRNA expression. Pterostilbene inhibited phosphorylated-IκBα expression, thus promoting IκBα expression and inhibiting ROS overexpression. In vitro, pterostilbene inhibited the expression of inflammatory cytokines and suppressed NAPDH activity as well as activation of both the NF-κB pathway and ROS production. To our knowledge, our study is the first to demonstrate that pterostilbene-mediated alleviation of cerebral ischemia and reperfusion injury in rats may be correlated with the inhibition of the ROS/NF-κB-mediated inflammatory pathway in microglia, indicating the potential for the use of pterostilbene as a candidate therapeutic compound for ischemic stroke.
Collapse
Affiliation(s)
- Jingyu Liu
- College of Life and Health Sciences, Northeastern University, Shenyang, China. and Key Laboratory of Data Analytics and Optimization for Smart Industry, Northeastern University, Ministry of Education, Shenyang, China
| | - Jikai Xu
- College of Life and Health Sciences, Northeastern University, Shenyang, China. and Key Laboratory of Data Analytics and Optimization for Smart Industry, Northeastern University, Ministry of Education, Shenyang, China
| | - Yan Mi
- College of Life and Health Sciences, Northeastern University, Shenyang, China. and Key Laboratory of Data Analytics and Optimization for Smart Industry, Northeastern University, Ministry of Education, Shenyang, China
| | - Yanqiu Yang
- College of Life and Health Sciences, Northeastern University, Shenyang, China.
| | - Qing Li
- College of Life and Health Sciences, Northeastern University, Shenyang, China.
| | - Di Zhou
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China.
| | - Kun Wei
- School of Chemical Science and Technology, Yunnan University, Kunming, China
| | - Gang Chen
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China.
| | - Ning Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China.
| | - Yue Hou
- College of Life and Health Sciences, Northeastern University, Shenyang, China. and Key Laboratory of Data Analytics and Optimization for Smart Industry, Northeastern University, Ministry of Education, Shenyang, China
| |
Collapse
|
29
|
Jaén RI, Sánchez-García S, Fernández-Velasco M, Boscá L, Prieto P. Resolution-Based Therapies: The Potential of Lipoxins to Treat Human Diseases. Front Immunol 2021; 12:658840. [PMID: 33968061 PMCID: PMC8102821 DOI: 10.3389/fimmu.2021.658840] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/07/2021] [Indexed: 02/05/2023] Open
Abstract
Inflammation is an a physiological response instead an essential response of the organism to injury and its adequate resolution is essential to restore homeostasis. However, defective resolution can be the precursor of severe forms of chronic inflammation and fibrosis. Nowadays, it is known that an excessive inflammatory response underlies the most prevalent human pathologies worldwide. Therefore, great biomedical research efforts have been driven toward discovering new strategies to promote the resolution of inflammation with fewer side-effects and more specificity than the available anti-inflammatory treatments. In this line, the use of endogenous specialized pro-resolving mediators (SPMs) has gained a prominent interest. Among the different SPMs described, lipoxins stand out as one of the most studied and their deficiency has been widely associated with a wide range of pathologies. In this review, we examined the current knowledge on the therapeutic potential of lipoxins to treat diseases characterized by a severe inflammatory background affecting main physiological systems, paying special attention to the signaling pathways involved. Altogether, we provide an updated overview of the evidence suggesting that increasing endogenously generated lipoxins may emerge as a new therapeutic approach to prevent and treat many of the most prevalent diseases underpinned by an increased inflammatory response.
Collapse
Affiliation(s)
- Rafael I. Jaén
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | | | - María Fernández-Velasco
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de investigación del Hospital la Paz, IdiPaz, Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Prieto
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
30
|
Fish Hydrolysate Supplementation Containing n-3 Long Chain Polyunsaturated Fatty Acids and Peptides Prevents LPS-Induced Neuroinflammation. Nutrients 2021; 13:nu13030824. [PMID: 33801489 PMCID: PMC7998148 DOI: 10.3390/nu13030824] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/18/2021] [Accepted: 02/26/2021] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation constitutes a normal part of the brain immune response orchestrated by microglial cells. However, a sustained and uncontrolled production of proinflammatory factors together with microglial activation contribute to the onset of a chronic low-grade inflammation, leading to neuronal damage and cognitive as well as behavioral impairments. Hence, limiting brain inflammatory response and improving the resolution of inflammation could be particularly of interest to prevent these alterations. Dietary n-3 long chain polyunsaturated fatty acids (LC-PUFAs) and low molecular weight peptides are good candidates because of their immunomodulatory and proresolutive properties. These compounds are present in a fish hydrolysate derived from marine-derived byproducts. In this study, we compared the effect of an 18-day supplementation with this fish hydrolysate to a supplementation with docosahexaenoic acid (DHA) on lipopolysaccharide (LPS)-induced inflammation in mice. In response to peripherally injected LPS, the fish hydrolysate supplementation decreased the hippocampal mRNA expression of the proinflammatory cytokines IL-6 (p < 0.001), IL-1β (p = 0.0008) and TNF-α (p < 0.0001), whereas the DHA supplementation reduced only the expression of IL-6 (p = 0.004). This decline in proinflammatory cytokine expressions was associated with an increase in the protein expression of IκB (p = 0.014 and p = 0.0054 as compared to the DHA supplementation and control groups, respectively) and to a modulation of microglial activation markers in the hippocampus. The beneficial effects of the fish hydrolysate could be due in part to the switch of the hippocampal oxylipin profile towards a more anti-inflammatory profile as compared to the DHA supplementation. Thus, the valorization of fish byproducts seems very attractive to prevent and counteract neuroinflammation.
Collapse
|
31
|
Vasefi M, Ghaboolian-Zare E, Abedelwahab H, Osu A. Environmental toxins and Alzheimer's disease progression. Neurochem Int 2020; 141:104852. [PMID: 33010393 DOI: 10.1016/j.neuint.2020.104852] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/05/2020] [Accepted: 09/18/2020] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, which causes progressive memory loss and cognitive decline. Effective strategies to treat or prevent remains one of the most challenging undertakings in the medical field. AD is a complex and multifactorial disease that involves several risk factors. Aging and genetic factors both play important roles in the onset of the AD, however; certain environmental factors have been reported to increase the risk of AD. Chronic exposure to toxins has been seen as an environmental factor that may increase the risk of developing a neurodegenerative disease such as AD. Exposure to metals and biotoxins produced by bacteria, molds, and viruses may contribute to the cognitive decline and pathophysiology associated with AD. Toxins may contribute to the pathology of the disease through various mechanisms such as deposition of amyloid-beta (Aβ) plaques and tangles in the brain, induction of apoptosis, inflammation, or oxidative damage. Here, we will review how toxins affect brain physiology with a focus on mechanisms by which toxins may contribute to the development and progression of AD. A better understanding of these mechanisms may help contribute towards the development of an effective strategy to slow the progression of AD.
Collapse
Affiliation(s)
- Maryam Vasefi
- Department Biology, Lamar University, Beaumont, TX, United States.
| | | | | | - Anthony Osu
- Department Biology, Lamar University, Beaumont, TX, United States
| |
Collapse
|
32
|
Zhou Y, You H, Zhang A, Jiang X, Pu Z, Xu G, Zhao M. Lipoxin A4 attenuates uric acid-activated, NADPH oxidase-dependent oxidative stress by interfering with translocation of p47phox in human umbilical vein endothelial cells. Exp Ther Med 2020; 20:1682-1692. [PMID: 32765680 PMCID: PMC7388524 DOI: 10.3892/etm.2020.8812] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 04/17/2020] [Indexed: 12/20/2022] Open
Abstract
LipoxinA4 (LXA4) is a well-known key mediator of endogenous anti-inflammation and of the resolution of inflammation. Considerable oxidative stress occurs during inflammation due to the generation of reactive oxidative species (ROS). Moreover, high levels of uric acid (UA) contribute to endothelial cell dysfunction, which can promote disease-related morbidity, and NADPH oxidase-derived ROS are crucial regulatory factors in these responses. However, LXA4 also has the potential to reduce oxidative stress. The aim of the present study was to examine whether LXA4 could suppress UA-induced oxidative stress in human umbilical vein endothelial cells (HUVECs) and to investigate its mechanisms of action in vitro. HUVECs were incubated with or without LXA4, followed by the addition of UA. ROS levels were then measured using 2,7-dichlorodihydrofluorescein diacetate and lucigenin-enhanced chemiluminescence was used to evaluate NADPH oxidase activity. p47phox or p22phox small interfering (si)RNA were transfected into HUVECs and protein levels of p47phox were detected using western blot analysis. LXA4 significantly inhibited UA-induced generation of ROS to the same extent as the NADPH oxidase inhibitor, diphenyleneiodonium chloride. Notably, transfection of p47phox siRNA attenuated the generation of ROS and the activation of NADPH oxidase. Cells transfected with p22phox siRNA demonstrated a significant reduction in the expression of p47phox on the membrane. Further experiments demonstrated that LXA4 interfered with the transfer of p47phox from the cytoplasm to the cell membrane. These findings suggested that LXA4 inhibited the release of NADPH oxidase derived ROS in HUVECs stimulated by UA. A potential mechanism of action underlying this effect could be LXA4-mediated suppression of NADPH oxidase activity, leading to inhibition of p47phox translocation from the cytoplasm to the cell membrane.
Collapse
Affiliation(s)
- You Zhou
- Department of Medical Laboratory, Central Hospital of Suining, Suining, Sichuan 629100, P.R. China
| | - Hui You
- Department of Ophthalmology, Central Hospital of Suining, Suining, Sichuan 629100, P.R. China
| | - Aijie Zhang
- Basic Medical Laboratory, Central Hospital of Suining, Suining, Sichuan 629100, P.R. China
| | - Xingliang Jiang
- Department of Medical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Zheyan Pu
- Department of Medical Laboratory, Central Hospital of Suining, Suining, Sichuan 629100, P.R. China
| | - Guoqiang Xu
- Department of Medical Laboratory, Central Hospital of Suining, Suining, Sichuan 629100, P.R. China
| | - Mingcai Zhao
- Department of Medical Laboratory, Central Hospital of Suining, Suining, Sichuan 629100, P.R. China
| |
Collapse
|
33
|
Fan X, Li J, Deng X, Lu Y, Feng Y, Ma S, Wen H, Zhao Q, Tan W, Shi T, Wang Z. Design, synthesis and bioactivity study of N-salicyloyl tryptamine derivatives as multifunctional agents for the treatment of neuroinflammation. Eur J Med Chem 2020; 193:112217. [PMID: 32182488 DOI: 10.1016/j.ejmech.2020.112217] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/03/2020] [Accepted: 03/07/2020] [Indexed: 12/12/2022]
Abstract
Because of the complex etiology in neuroinflammatory process, the design of multifunctional agents is a potent strategy to cure neuroinflammatory diseases including AD and PD. Herein, based on the combination principles, 23 of N-salicyloyl tryptamine derivatives as multifunctional agents were designed and their new application for anti-neuroinflammation was disclosed. In cyclooxygenase assay, two compounds 3 and 16 displayed extremely preferable COX-2 inhibition than N-salicyloyl tryptamine. In LPS-induced C6 and BV2 cell models, some compounds decreased the production of proinflammatory mediators NO, PGE2, TNF-α, iNOS, COX-2 and ROS, while increased the production of IL-10. Among them, compound 3 and 16 showed approximately six-fold better inhibition on nitric oxide production than N-salicyloyl tryptamine in C6. Besides, compounds 3, 13 and 16 attenuated the activation of BV2 and C6 cells. More importantly, in vivo, compounds 3 and 16 reduced GFAP and Iba-1 levels in the hippocampus, and displayed neuroprotection in Nissl staining. Besides, both compounds 3 and 16 had high safety (LD50 > 1000 mg/kg). Longer plasma half-life of compounds 3 and 16 than melatonin supported combination strategy. All these results demonstrated that N-salicyloyl tryptamine derivatives are potential anti-neuroinflammation agents for the treatment of neurodegenerative disorder.
Collapse
Affiliation(s)
- Xiaohong Fan
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Junfang Li
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Xuemei Deng
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Yingmei Lu
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Yiyue Feng
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Shumeng Ma
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Huaixiu Wen
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810000, China
| | - Quanyi Zhao
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Tao Shi
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China.
| | - Zhen Wang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
34
|
Liu Z, Lu S, Zhang J, Chen X, Mickymaray S. Limonin: A triterpenoid exerts protective effect during lipopolysaccharide stimulated inflammation in BV2 microglial cells. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_304_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
35
|
Tong W, Chen X, Song X, Chen Y, Jia R, Zou Y, Li L, Yin L, He C, Liang X, Ye G, Lv C, Lin J, Yin Z. Resveratrol inhibits LPS-induced inflammation through suppressing the signaling cascades of TLR4-NF-κB/MAPKs/IRF3. Exp Ther Med 2019; 19:1824-1834. [PMID: 32104238 PMCID: PMC7027153 DOI: 10.3892/etm.2019.8396] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022] Open
Abstract
Resveratrol (Res) is a natural compound that possesses anti-inflammatory properties. However, the protective molecular mechanisms of Res against lipopolysaccharide (LPS)-induced inflammation have not been fully studied. In the present study, RAW264.7 cells were stimulated with LPS in the presence or absence of Res, and the subsequent modifications to the LPS-induced signaling pathways caused by Res treatment were examined. It was identified that Res decreased the mRNA levels of Toll-like receptor 4 (TLR4), myeloid differentiation primary response protein MyD88, TIR domain-containing adapter molecule 2, which suggested that Res may inhibit the activation of the TLR4 signaling pathway. It suppressed the expression levels of total and phosphorylated TLR4, NF-κB inhibitor, p38 mitogen-activated protein kinase (MAPK), c-Jun N-terminal kinase, extracellular signal-regulated kinase 1/2 and interferon (IFN) regulatory factor 3 (IRF3) proteins. Following treatment with Res or specific inhibitors, the production of pro-inflammatory mediators including tumor necrosis factor-α, interleukin (IL)-6, IL-8 and IFN-β were decreased and the expression of anti-inflammatory mediator IL-10 was increased. These results suggested that Res may inhibit the signaling cascades of NF-κB, MAPKs and IRF3, which modulate pro-inflammatory cytokines. In conclusion, Res exhibited a therapeutic effect on LPS-induced inflammation through suppression of the TLR4-NF-κB/MAPKs/IRF3 signaling cascades.
Collapse
Affiliation(s)
- Wenzhi Tong
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Xiangxiu Chen
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Yaqin Chen
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Renyong Jia
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Yuanfeng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Lizi Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Changliang He
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Xiaoxia Liang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Gang Ye
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Cheng Lv
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Juchun Lin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, P.R. China
| |
Collapse
|
36
|
Yan JJ, Du GH, Qin XM, Gao L. Baicalein attenuates the neuroinflammation in LPS-activated BV-2 microglial cells through suppression of pro-inflammatory cytokines, COX2/NF-κB expressions and regulation of metabolic abnormality. Int Immunopharmacol 2019; 79:106092. [PMID: 31863920 DOI: 10.1016/j.intimp.2019.106092] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/08/2019] [Accepted: 11/28/2019] [Indexed: 12/25/2022]
Abstract
Baicalein (5,6,7-trihydroxyflavone), isolated from the root of traditional Chinese herb Scutellaria baicalensis Georgi, has anti-inflammatory and anti-oxidative activities. This study explored the protective and modulatory mechanisms of baicalein on neuroinflammation, oxidative stress and metabolic abnormality in lipopolysaccharide (LPS)-activated BV-2 cells. Our results demonstrated that treatment with baicalein remarkably restrained the production of pro-inflammatory factors including nitric oxide (NO), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in LPS-activated BV-2 cells. Moreover, baicalein significantly inhibited reactive oxygen species (ROS) production, decreased cyclooxygenase-2 (COX-2) and nuclear factor-b (NF-κB)/p65 expression. 1H NMR metabolomics analysis revealed that 12 differential metabolites were regulated by baicalein, implicated in alanine, aspartate and glutamate metabolism, glutathione metabolism, arginine and proline metabolism, D-glutamine and D-glutamate metabolism. In conclusion, these results indicated that baicalein has protective and modulatory effects on neuroinflammation and oxidative stress in LPS-activated BV-2 cells.
Collapse
Affiliation(s)
- Jiao-Jiao Yan
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China; College of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Guan-Hua Du
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China; Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China.
| | - Li Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
37
|
Zhang YY, Liu X, Zhang X, Zhang J. Shikonin improve sepsis-induced lung injury via regulation of miRNA-140-5p/TLR4-a vitro and vivo study. J Cell Biochem 2019; 121:2103-2117. [PMID: 31769066 DOI: 10.1002/jcb.28199] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 11/15/2018] [Indexed: 12/24/2022]
Abstract
Shikonin is an anti-inflammatory agent extracted from natural herbs. The aim of this study is to explain the treatment effects and mechanism of Shikonin in acute lung injury induced by sepsis. In this study, first, we evaluate different Shikonin concentrations for the anti-inflammation of acute lung injury induced by sepsis in an in vivo study. On the basis of the results, we confirm that 50.0 mg/kg was the best therapeutic Shikonin concentration. As a second step, we discuss the mechanism of Shikonin by a vitro cell experiment. Finaly, we validate that Shikonin has effective treatment effects on acute lung injury via regulation of microRNA-140-5p/toll-like receptor 4 (miRNA-140-5p/TLR4) in the in vivo study. The results of vitro and vivo study showed that Shikonin could improve acute lung injury induced by sepsis. The mechanism might be correlation miRNA-140-5p expression increasing, and regulated targeted gene TLR4, with TLR4 expression depressing, the downstream myeloid differentiation protein 88 and nuclear factor κB proteins expression were suppressed. In conclusion, Shikonin improved sepsis induced lung injury by regulation miRNA-140-5p/TLR4.
Collapse
Affiliation(s)
- Ying Ying Zhang
- Department of Hand and Foot Surgery, Linyi Central Hospital, Linyi, China
| | - Xue Liu
- Department of Respiration, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaojie Zhang
- Surgical Intensive Care Unit, Shandong Provincial Chest Hospital, Jinan, China
| | - Jingjing Zhang
- Respiratory and Critical Medical Ward 7, Shandong Provincial Chest Hospital, Jinan, China
| |
Collapse
|
38
|
Gil‐Villa AM, Alvarez AM, Velásquez‐Berrío M, Rojas‐López M, Cadavid J AP. Role of aspirin‐triggered lipoxin A4, aspirin, and salicylic acid in the modulation of the oxidative and inflammatory responses induced by plasma from women with pre‐eclampsia. Am J Reprod Immunol 2019; 83:e13207. [DOI: 10.1111/aji.13207] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/19/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Aura María Gil‐Villa
- Grupo Reproducción Departamento de Microbiología y Parasitología Facultad de Medicina Universidad de Antioquia Medellín Colombia
| | - Angela M. Alvarez
- Grupo Reproducción Departamento de Microbiología y Parasitología Facultad de Medicina Universidad de Antioquia Medellín Colombia
- Red Iberoamericana de Alteraciones Vasculares Asociadas a Transtornos del Embarazo (RIVA‐TREM) Chillán Chile
| | - Manuela Velásquez‐Berrío
- Grupo Reproducción Departamento de Microbiología y Parasitología Facultad de Medicina Universidad de Antioquia Medellín Colombia
| | - Mauricio Rojas‐López
- Grupo de Inmunología Celular e Inmunogenética – Unidad de Citometría de Flujo Sede de Investigación Universitaria (SIU) Universidad de Antioquia Medellín Colombia
| | - Angela P. Cadavid J
- Grupo Reproducción Departamento de Microbiología y Parasitología Facultad de Medicina Universidad de Antioquia Medellín Colombia
- Red Iberoamericana de Alteraciones Vasculares Asociadas a Transtornos del Embarazo (RIVA‐TREM) Chillán Chile
| |
Collapse
|
39
|
Liu Y, Lin J, Wu X, Guo X, Sun H, Yu B, Shen J, Bai J, Chen Z, Yang H, Geng D, Mao H. Aspirin-Mediated Attenuation of Intervertebral Disc Degeneration by Ameliorating Reactive Oxygen Species In Vivo and In Vitro. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7189854. [PMID: 31781346 PMCID: PMC6874877 DOI: 10.1155/2019/7189854] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 09/04/2019] [Accepted: 09/14/2019] [Indexed: 12/13/2022]
Abstract
Intervertebral disc (IVD) degeneration (IDD) is a major cause of low back pain. The pathogenesis of IDD is associated with the disturbance of reactive oxygen species (ROS) equilibrium, inflammation, and matrix loss. Aspirin is a nonsteroidal anti-inflammatory drug that effectively inhibits inflammation and oxidative stress and has been widely used for the treatment of back pain. Therefore, we hypothesize that aspirin reverses the IDD process via antioxidative and anti-inflammatory effects on the AMPK signaling pathway. In vitro, aspirin diminished cellular oxygen free radicals (ROS, nitric oxide (NO)) and inflammatory cytokines (interleukin- (IL-) 1β and IL-6 and tumor necrosis factor alpha (TNF-α)) induced by lipopolysaccharides (LPS) in nucleus pulposus cells (NPCs). We found that aspirin preserved the extracellular matrix (ECM) content of collagen type II (COL2) and aggrecan while inhibiting the expression of matrix-degenerating enzymes, including matrix metalloproteinase 3 and 13 (MMP-3 and MMP-13) and A disintegrin and metalloproteinase with thrombospondin motifs 4 and 5 (ADAMTS-4, ADAMTS-5). Aspirin significantly promoted the ratios of p-AMPK to AMPK and p-ACC to ACC expression in NPCs. Furthermore, pretreatment with the AMPK inhibitor compound C abrogated the antioxidant effects of aspirin. In vivo, an IDD model was established in Sprague-Dawley rats via percutaneous disc puncture with the 20-gauge needle on levels 8-9 and 9-10 of the coccygeal vertebrae. Imaging assessment showed that after aspirin treatment, improvements in disc height index (DHI) ranged from 1.22-fold to 1.54-fold and nucleus pulposus signal strength improved from 1.26-fold to 1.33-fold. Histological analysis showed that aspirin treatment prevented the loss of COL2 and decreased MMP-3 and MMP-13, inducible NO synthase (iNOS), cyclooxygenase-2 (COX-2), IL-1β, and TNF-α expression in the IVD tissues. These results suggest that treatment with aspirin could reverse the IDD process via the AMPK signaling pathway, which provides new insights into the potential clinical applications of aspirin, particularly for IDD treatment.
Collapse
Affiliation(s)
- Yu Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jiayi Lin
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiexing Wu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiaobin Guo
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Houyi Sun
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Binqing Yu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jining Shen
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jiaxiang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Zhanghuan Chen
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Haiqing Mao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
40
|
Joffre C, Rey C, Layé S. N-3 Polyunsaturated Fatty Acids and the Resolution of Neuroinflammation. Front Pharmacol 2019; 10:1022. [PMID: 31607902 PMCID: PMC6755339 DOI: 10.3389/fphar.2019.01022] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/12/2019] [Indexed: 12/22/2022] Open
Abstract
In the past few decades, as a result of their anti-inflammatory properties, n-3 long chain polyunsaturated fatty acids (n-3 LC-PUFAs), have gained greater importance in the regulation of inflammation, especially in the central nervous system (in this case known as neuroinflammation). If sustained, neuroinflammation is a common denominator of neurological disorders, including Alzheimer’s disease and major depression, and of aging. Hence, limiting neuroinflammation is a real strategy for neuroinflammatory disease therapy and treatment. Recent data show that n-3 LC-PUFAs exert anti-inflammatory properties in part through the synthesis of specialized pro-resolving mediators (SPMs) such as resolvins, maresins and protectins. These SPMs are crucially involved in the resolution of inflammation. They could be good candidates to resolve brain inflammation and to contribute to neuroprotective functions and could lead to novel therapeutics for brain inflammatory diseases. This review presents an overview 1) of brain n-3 LC-PUFAs as precursors of SPMs with an emphasis on the effect of n-3 PUFAs on neuroinflammation, 2) of the formation and action of SPMs in the brain and their biological roles, and the possible regulation of their synthesis by environmental factors such as inflammation and nutrition and, in particular, PUFA consumption.
Collapse
Affiliation(s)
- Corinne Joffre
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France.,Université de Bordeaux 2, Bordeaux, France
| | - Charlotte Rey
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France.,Université de Bordeaux 2, Bordeaux, France.,ITERG, Nutrition Health and Lipid Biochemistry Department, Canéjan, France
| | - Sophie Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France.,Université de Bordeaux 2, Bordeaux, France
| |
Collapse
|
41
|
Akhmetzyanova E, Kletenkov K, Mukhamedshina Y, Rizvanov A. Different Approaches to Modulation of Microglia Phenotypes After Spinal Cord Injury. Front Syst Neurosci 2019; 13:37. [PMID: 31507384 PMCID: PMC6718713 DOI: 10.3389/fnsys.2019.00037] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 07/29/2019] [Indexed: 01/04/2023] Open
Abstract
Microglial cells, which are highly plastic, immediately respond to any change in the microenvironment by becoming activated and shifting the phenotype toward neurotoxicity or neuroprotection. The polarization of microglia/macrophages after spinal cord injury (SCI) seems to be a dynamic process and can change depending on the microenvironment, stage, course, and severity of the posttraumatic process. Effective methods to modulate microglia toward a neuroprotective phenotype in order to stimulate neuroregeneration are actively sought for. In this context, available approaches that can selectively impact the polarization of microglia/macrophages regulate synthesis of trophic factors and cytokines/chemokines in them, and their phagocytic function and effects on the course and outcome of SCI are discussed in this review.
Collapse
Affiliation(s)
- Elvira Akhmetzyanova
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Konstantin Kletenkov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Yana Mukhamedshina
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Albert Rizvanov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
42
|
Ba F, Zhou X, Zhang Y, Wu C, Xu S, Wu L, Li J, Yin Y, Gu X. Lipoxin A4 ameliorates alveolar fluid clearance disturbance in lipopolysaccharide-induced lung injury via aquaporin 5 and MAPK signaling pathway. J Thorac Dis 2019; 11:3599-3608. [PMID: 31559067 DOI: 10.21037/jtd.2019.08.86] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background A characteristic of acute lung injury (ALI) is the inflammatory damage of alveolar fluid transport. Lipoxins are endogenous lipids involving in the resolution of inflammation. It is found that lipoxin A4 (LXA4) has the distinct properties to improve the anti-edema and pro-resolution function in inflammation. Since aquaporins (AQPs) have essential roles in the integrity of barrier function during fluid transport, especially AQP5 in the maintaining of the epithelium permeability, the current study is aimed to evaluate the potential role of LXA4 in regulating alveolar fluid clearance (AFC) during fluid transport and the corresponding change of AQP5 in the lung. Methods ALI was induced by the lipopolysaccharide (LPS) intraperitoneal injection, and LXA4 treatment was given 8 hours after LPS administration. We investigated changes in the capacity of AFC, pro-inflammatory cytokine concentrations in bronchoalveolar lavage fluid (BALF) and the severity of ALI. Then AQP5 expression in lung tissue and potential regulatory pathways in LPS-induced ALI was explored. Results LXA4 treatment was found to inhibit AFC capacity, inflammatory cytokine release, partially, alleviate ALI severity, and restored AQP5 expression partially. Additionally, we found that LXA4 played a protective role by the inhibition of the phosphorylation of p38 and JNK. Conclusions In summary, our results suggest that LXA4 plays a protective role in lipopolysaccharide-induced ALI by restoring AFC capacity and upregulating AQP5 expression and inhibiting the phosphorylation of p38 and JNK. These findings suggest potential new mechanism of LXA4 as anti-inflammation therapy for the impairment of alveolar fluid transport in ALI.
Collapse
Affiliation(s)
- Fang Ba
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiaoming Zhou
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yingqi Zhang
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Cen Wu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Shenqian Xu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Liqin Wu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jiayang Li
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yan Yin
- Institute of Respiratory Disease, First Hospital of China Medical University, Shenyang 110004, China
| | - Xiu Gu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
43
|
Wei F, Gong W, Wang J, Yang Y, Liu J, Wang Y, Cao J. Role of the lipoxin A4 receptor in the development of neutrophil extracellular traps in Leishmania infantum infection. Parasit Vectors 2019; 12:275. [PMID: 31142352 PMCID: PMC6542009 DOI: 10.1186/s13071-019-3530-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 05/23/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Neutrophils play an immunomodulatory role through the release of neutrophil extracellular traps (NETs). NETs are released in response to Leishmania infection, but the mechanism of NET extrusion has not been elucidated. The lipoxin A4 receptor on neutrophils is crucial for the inflammatory response and immune regulation of many diseases, including Leishmania infection. Therefore, in the present study, we tried to explore whether Leishmania infantum promastigotes stimulate neutrophil activation and NET release via activating the lipoxin A4 receptor. RESULTS Leishmania infantum promastigotes stimulated neutrophil activity, but blocking of the lipoxin A4 receptor with its antagonist Boc prior to L. infantum stimulation abrogated these effects. Neutrophils showed citrullinated histone H3 expression and simultaneous NET extrusion on L. infantum stimulation, but a decline in both was observed on blocking of the lipoxin A4 receptor. Moreover, differentiated HL-60 cells with lipoxin A4 receptor silencing showed a decrease in citrullinated histone H3 expression as compared to the unsilenced HL-60 samples on stimulation with promastigotes. CONCLUSIONS Leishmania infantum promastigotes altered the characteristics of neutrophils and induced NET extrusion by activating the lipoxin A4 receptor. The lipoxin A4 receptor may have potential as a therapeutic target in relation to NET extrusion in the treatment of leishmaniasis, but its mechanisms of action need to be explored in more depth.
Collapse
Affiliation(s)
- Furong Wei
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Wenci Gong
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Junyun Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Yuetao Yang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Jianxiu Liu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Yanjuan Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China.,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China.,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China.,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, China. .,Chinese Center for Tropical Diseases Research, Shanghai, 200025, China. .,WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, China. .,National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, 200025, China. .,Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025, China.
| |
Collapse
|
44
|
Meng J, Chen Y, Zhang C. Protective impacts of long noncoding RNA taurine-upregulated 1 against lipopolysaccharide-evoked injury in MRC-5 cells through inhibition of microRNA-127. J Cell Biochem 2019; 120:14928-14935. [PMID: 31017717 DOI: 10.1002/jcb.28755] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Pneumonia is a respiratory disease, which is triggered by pathogenic microorganisms or physical/chemical factors. Increasing evidence confirmed the vital impacts of long noncoding RNAs on various inflammatory diseases. Nonetheless, the influence of taurine-upregulated 1 (TUG1) in pneumonia remains vague. The research tried to disclose the protective impacts of TUG1 against lipopolysaccharide (LPS)-evoked injury in MRC-5 cells. METHODS MRC-5 cells were disposed with LPS to construct pulmonary injury model. Then, pc-TUG1 vector was transfected into MRC-5 cells and the influence of overexpressed TUG1 in cell viability, apoptosis, and pro-inflammatory cytokines in LPS-disposed cells were evaluated. The correlation between TUG1 and microRNA (miR)-127 was estimated via utilizing real-time quantitative polymerase chain reaction (RT-qPCR), meanwhile whether miR-127 affected the impacts of TUG1 on LPS-injured MRC-5 cells was explored. Besides, NF-κB and p38MAPK pathways were evaluated to understand the dormant mechanisms. RESULTS LPS administration apparently evoked inflammatory injury in MRC-5 cells by restraining cell viability, accelerating apoptosis, and enhancing TNF-α and IL-6 productions. But, TUG1 lightened LPS-evoked pro-inflammatory response in MRC-5 cells. In addition, miR-127 was repressed by overexpressed TUG1, meanwhile the protective impacts of TUG1 against LPS-evoked inflammatory injury in MRC-5 cells were overturned by overexpressed miR-127. Finally, we disclosed that TUG1 hindered the activation of NF-κB and p38MAPK pathways via restraining miR-127. CONCLUSIONS These explorations testified that taurine-upregulated 1 (TUG1) protected MRC-5 cells against lipopolysaccharide (LPS)-evoked inflammatory injury via hindering miR-127/NF-κB/p38MAPK axis.
Collapse
Affiliation(s)
- Jie Meng
- Department of Pediatrics, Jining No.1 People's Hospital, Jining, China
| | - Yan Chen
- Department of Pediatrics, Jining No.1 People's Hospital, Jining, China
| | - Cunxue Zhang
- Department of Pediatrics, Jining No.1 People's Hospital, Jining, China
| |
Collapse
|
45
|
Zhang Y, Li C, Zhou C, Hong P, Zhang Y, Sun S, Qian ZJ. 2'-Hydroxy-5'-methoxyacetophenone attenuates the inflammatory response in LPS-induced BV-2 and RAW264.7 cells via NF-κB signaling pathway. J Neuroimmunol 2019; 330:143-151. [PMID: 30884275 DOI: 10.1016/j.jneuroim.2019.03.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/11/2019] [Accepted: 03/11/2019] [Indexed: 02/07/2023]
Abstract
Seahorse has been used as a traditional medicine in Southeast Asian countries for a long time. A compound, 2'-Hydroxy-5'-Methoxyacetophenone (2H5M) isolated from seahorse, Hippocampus kuda, was tested for its anti-inflammatory effect in lipopolysaccharides (LPS)-stimulated BV-2 cells and RAW264.7 cells. MTT assay indicated that 2H5M has no cytotoxicity on two kinds of cells. The concentration of nitric oxide (NO) measured by Griess Reaction System showed that 2H5M could significantly inhibit the NO concentration. The ELISA results showed that 2H5M could suppress the secretion of TNF-α in a dose-dependent manner. Moreover, western blot analysis was utilized to measure the protein levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) signaling pathways. Electrophoretic mobility shift assay (EMSA) demonstrated that 2H5M reduced NF-κB DNA binding activity. Furthermore, the molecular docking study showed that 2H5M can form active sites with NF-κB. Collectively, these results indicated that 2H5M possesses anti-inflammatory effects and may have a potential application in inflammatory disorders in the future.
Collapse
Affiliation(s)
- YuanYuan Zhang
- School of Chemistry and Environment of Chemistry and Environment, Guangdong Ocean University, Zhanjiang 524088, China; College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - ChengYong Li
- School of Chemistry and Environment of Chemistry and Environment, Guangdong Ocean University, Zhanjiang 524088, China; Shenzhen Institute of Guangdong Ocean University, Shenzhen 518108, China
| | - ChunXia Zhou
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; Shenzhen Institute of Guangdong Ocean University, Shenzhen 518108, China
| | - PengZhi Hong
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yi Zhang
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - ShengLi Sun
- School of Chemistry and Environment of Chemistry and Environment, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhong-Ji Qian
- School of Chemistry and Environment of Chemistry and Environment, Guangdong Ocean University, Zhanjiang 524088, China; Shenzhen Institute of Guangdong Ocean University, Shenzhen 518108, China.
| |
Collapse
|
46
|
Wu J, Ding DH, Li QQ, Wang XY, Sun YY, Li LJ. Lipoxin A4 Regulates Lipopolysaccharide-Induced BV2 Microglial Activation and Differentiation via the Notch Signaling Pathway. Front Cell Neurosci 2019; 13:19. [PMID: 30778288 PMCID: PMC6369213 DOI: 10.3389/fncel.2019.00019] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 01/16/2019] [Indexed: 12/16/2022] Open
Abstract
Inflammatory responses contribute to the pathogenesis of various neurological diseases, and microglia plays an important role in the process. Activated microglia can differentiate into the pro-inflammatory, tissue-damaging M1 phenotype or the anti-inflammatory, tissue-repairing M2 phenotype. Regulating microglia differentiation, hence limiting a harmful response, might help improve the prognosis of inflammation-related nervous system diseases. The present study aimed 1. to observe the anti-inflammatory effect of lipoxin A4 (LXA4) on the inflammatory response associated to lipopolysaccharide (LPS)-induced microglia activation, 2. to clarify that LXA4 modulates the activation and differentiation of microglia induced by LPS stimulation, 3. to determine whether LXA4 regulates the activation and differentiation of microglia through the Notch signaling pathway, 4. to provide a foundation for the use of LXA4 for the treatment of inflammatory related neurological diseases. To construct a model of cellular inflammation, immortalized murine BV2 microglia cells were provided 200 ng/ml LPS. To measure the mRNA and protein levels of inflammatory factors (interleukin [IL]-1β, IL-10, and tumor necrosis factor [TNF]-α) and M1 and M2 microglia markers (inducible nitric oxide synthase [iNOS], cluster of differentiation [CD]32, arginase [Arg]1, and CD206), we performed quantitative reverse transcription polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA), immunofluorescence, or flow cytometry. To determine the mRNA and protein levels of Notch signaling components (Notch1, Hes1, and Hes5), we performed qRT-PCR and western blot. LXA4 inhibits the expression of Notch1 and Hes1 associated with M1 type microglial differentiation and decreases the M1 type microglia marker iNOS and related inflammatory factors IL-1β and TNF-α. Moreover, LXA4 upregulates the expression of the M2-associated Hes5, as well as the expression of the M2 microglia marker Arg1 and the associated inflammatory factor IL-10. These effects are blocked by the administration of the γ-secretase inhibitor DAPT, a specific blocker of the Notch signaling pathway. LXA4 inhibits the microglia activation induced by LPS and the differentiation into M1 type with pro-inflammatory effect, while promoting the differentiation to M2 type with anti-inflammatory effect. LXA4 downregulates the inflammatory mediators IL-1β, TNF-α, and iNOS, while upregulating the anti-inflammatory mediator IL-10, which acts through the Notch signaling pathway.
Collapse
Affiliation(s)
- Jun Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dan-Hua Ding
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qian-Qian Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin-Yu Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu-Ying Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lan-Jun Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
47
|
Pharmacodynamic Effect of Luteolin Micelles on Alleviating Cerebral Ischemia Reperfusion Injury. Pharmaceutics 2018; 10:pharmaceutics10040248. [PMID: 30501051 PMCID: PMC6320772 DOI: 10.3390/pharmaceutics10040248] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 12/15/2022] Open
Abstract
Oxidative stress and inflammation are important mechanisms of cerebral ischemia reperfusion (IR) injury. Luteolin (Lu), one of the major active components in the classical Tibetan prescription, which has been used in the treatment of cardiovascular diseases since 700 BC, has potential for IR injury therapy. Its hydrophobicity has impeded its further applications. In this study, we first prepared Lu micelles (M-Lu) by self-assembling with an amphiphilic copolymer via the thin film hydration method to improve the dispersion of Lu in water. The obtained M-Lu was about 30 nm, with a narrow particle size distribution, and a 5% (w/w) of Lu. The bioavailability of the micelles was further evaluated in vitro and in vivo. Compared to free Lu, M-Lu had a better penetration efficiency, which enhanced its therapeutic effect in IR injury restoration. M-Lu further strengthened the protection of nerve cells through the nuclear factor-κ-gene binding κ (NF-κB) and mitogen-activated protein kinases (MAPK) pathways and inhibited the apoptosis of cells by adjusting the expression of B-cell lymphoma-2 (Bcl-2) and Bcl-2 associated X protein (Bax) in the case of oxidative stress damage. M-Lu induced stem cells to differentiate into neuron-like cells to promote the repair and regeneration of neurons. The results of in vivo pharmacodynamics of Lu on occlusion of the middle cerebral artery model further demonstrated that M-Lu better inhibited inflammation and the oxidative stress response by the down-regulation of the inflammatory cytokine, including tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6, and the up-regulation of the activity of anti-oxidant kinase, such as superoxide dismutase (SOD) and glutathione peroxidase (GSH-px), which further ameliorated the degree of IR injury. The M-Lu could be a new strategy for IR injury therapy.
Collapse
|
48
|
Youssef M, Ibrahim A, Akashi K, Hossain MS. PUFA-Plasmalogens Attenuate the LPS-Induced Nitric Oxide Production by Inhibiting the NF-kB, p38 MAPK and JNK Pathways in Microglial Cells. Neuroscience 2018; 397:18-30. [PMID: 30496826 DOI: 10.1016/j.neuroscience.2018.11.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 12/29/2022]
Abstract
The special lipids plasmalogens (Pls) were reported to be reduced in the neurodegenerative brains such as Alzheimer's disease where a marked increase of glial activation is often observed. We previously found that a reduction of brain Pls can enhance the glial activation in murine brains. However, the detailed role of Pls in the prevention of glial activation was mostly elusive. Here we report that the Pls, extracted from scallop (sPls), significantly inhibited the inducible form of nitric oxide synthase (NOS2) and the production of NO in LPS (lipopolysaccharide)-activated microglial cells. We also observed that the polyunsaturated docosahexaenoic acid (DHA)-containing Pls but not the monounsaturated oleic acid-containing Pls attenuated the NOS2 induction. In addition, sPls blocked the activation of nuclear factor (NF)-kB and mitogen-activated protein kinases (MAPKs) e.g., JNK and p38 MAPK, thereby attenuated the nuclear translocation of NF-kB subunit, p65, and activator protein-1 (AP-1) proteins (c-Fos and c-Jun). Interestingly, LPS treatments suppressed the expression of Pls synthesizing enzymes, glycerone phosphate O-acyltransferase (GNPAT) and alkylglycerone phosphate synthase (AGPS) in the microglial cells by the p38MAPK and JNK pathways. Furthermore, the knockdown of GNPAT and AGPS genes by sh-RNAs accelerated the LPS-induced activation of p38MAPK and JNK, resulting in the increased production of NO. These findings suggested that a decrease of brain Pls can activate the NF-kB, p38MAPK and JNK pathways to induce a prolonged microglial activation which may downplay the neuroprotective events in the brains of neurodegenerative diseases.
Collapse
Affiliation(s)
- Mohammed Youssef
- Department of Neuroinflammation and Brain Fatigue Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582 Japan; Department of Animal Physiology, Veterinary Medicine Faculty, South Valley University, Qena 83523 Egypt
| | - Ahmed Ibrahim
- Department of Poultry Diseases, Veterinary Medicine Faculty, South Valley University, Qena 83523 Egypt
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Faculty of Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Md Shamim Hossain
- Department of Neuroinflammation and Brain Fatigue Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582 Japan.
| |
Collapse
|
49
|
Ji YD, Luo ZL, Chen CX, Li B, Gong J, Wang YX, Chen L, Yao SL, Shang Y. BML-111 suppresses TGF-β1-induced lung fibroblast activation in vitro and decreases experimental pulmonary fibrosis in vivo. Int J Mol Med 2018; 42:3083-3092. [PMID: 30280199 PMCID: PMC6202103 DOI: 10.3892/ijmm.2018.3914] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 09/04/2018] [Indexed: 01/07/2023] Open
Abstract
Pulmonary fibrosis is an aggressive end‑stage disease. Transforming growth factor‑β1 (TGF‑β1) mediates lung fibroblast activation and is essential for the progress of pulmonary fibrosis. BML‑111, a lipoxinA4 (LXA4) receptor (ALX) agonist, has been reported to possess anti‑fibrotic properties. The present study aimed to elucidate whether BML‑111 inhibits TGF‑β1‑induced mouse embryo lung fibroblast (NIH3T3 cell line) activation in vitro and bleomycin (BLM)‑induced pulmonary fibrosis in vivo. In vitro experiments demonstrated that BML‑111 treatment inhibits TGF‑β1‑induced NIH3T3 cell viability and the expression of smooth muscle α actin (α‑SMA), fibronectin and total collagen. Furthermore, this suppressive effect was associated with mothers against decapentaplegic homolog (Smad)2/3, extracellular signal‑regulated kinase (ERK) and Akt phosphorylation interference. In vivo experiments revealed that BML‑111 treatment markedly improved survival rate and ameliorated the destruction of lung tissue structure. It also reduced interleukin‑1β (IL‑1β), tumor necrosis factor‑α (TNF‑α) and TGF‑β1 expression in the BLM intratracheal mouse model. In addition, the expression ofα‑SMA and extracellular matrix (ECM) deposition (total collagen, hydroxyproline and fibronectin) were also suppressed following BML‑111 treatment. However, BOC‑2, an antagonist of ALX, partially weakened the effects of BML‑111. In conclusion, these results indicated that BML‑111 inhibits TGF‑β1‑induced fibroblasts activation and alleviates BLM‑induced pulmonary fibrosis. Therefore, BML‑111 may be used as a potential therapeutic agent for pulmonary fibrosis treatment.
Collapse
Affiliation(s)
- Yu-Dong Ji
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhen-Long Luo
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Chun-Xiu Chen
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Bo Li
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jie Gong
- Department of Critical Care Medicine, Institute of Anesthesiology and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Ya-Xin Wang
- Department of Critical Care Medicine, Institute of Anesthesiology and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lin Chen
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shang-Long Yao
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - You Shang
- Department of Critical Care Medicine, Institute of Anesthesiology and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
50
|
Pan S, Wu Y, Pei L, Li S, Song L, Xia H, Wang Y, Yu Y, Yang X, Shu H, Zhang J, Yuan S, Shang Y. BML-111 Reduces Neuroinflammation and Cognitive Impairment in Mice With Sepsis via the SIRT1/NF-κB Signaling Pathway. Front Cell Neurosci 2018; 12:267. [PMID: 30186119 PMCID: PMC6110933 DOI: 10.3389/fncel.2018.00267] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 08/02/2018] [Indexed: 01/31/2023] Open
Abstract
Sepsis is a life-threatening state of organ dysfunction caused by infection and which can induce severe neurological disorders that lead to neuroinflammation and cognitive impairment. Inflammation has been reported to cause neuronal apoptosis in sepsis, which can finally lead to cognitive impairment. Previous studies have suggested that BML-111 can exhibit anti-inflammatory and proresolution activities. Additionally, silent information regulator 1 (SIRT1) can inhibit the NF-κB signaling pathway in an inflammation state. However, the role of the SIRT1/NF-κB signaling pathway in the protective effects of BML-111 against sepsis-induced neuroinflammation and cognitive impairment remains unclear. This study aimed to determine the effects of BML-111 on neuroinflammation and cognitive impairment induced by sepsis. Male C57BL/6J mice were subjected to cecal ligation and puncture (CLP) or a sham operation. BML-111 was administered via intracerebroventricular injection (0.1 mg/kg) immediately after CLP. Boc-2 (50 μg/kg) was administered intracerebroventricularly 30 min before CLP, and EX527 (10 μg) was administered every 2 days for a total of three times before CLP, also intracerebroventricularly. Some of the surviving mice underwent open-field, novel-object-recognition, and fear-conditioning behavioral tests at 7 days after surgery. Some of the other surviving mice were killed at 24 h after surgery to assess synaptic damage (PSD95 and Synapsin1), markers of inflammation [tumor necrosis factor alpha (TNF-α) and interleukin (IL)-1β], cytoplasmic p65, nuclear p65, Ac- NF-κB and SIRT1. At 48 h after CLP, TUNEL and glia-activation by immunofluorescence investigations were performed on a separate cohort of surviving animals. The results suggested that sepsis resulted in cognitive impairment, which was accompanied by the decreased the expression of PSD95 and Synapsin1, increased amount of TUNEL-positive cells and the activation of glias, increased production of TNF-α and IL-1β, increased expression of nuclear p65, Ac- NF-κB, and decreased expression of SIRT1 and cytoplasmic p65. It is especially notable that these abnormalities could be reduced by BML-111 treatment. EX527, an SIRT1 inhibitor, abolished the effects of BML-111. These results demonstrate that BML-111 can reduce the neuroinflammation and cognitive impairment induced by sepsis via SIRT/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Shangwen Pan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Pei
- Department of Neurobiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengnan Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Limin Song
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haifa Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaxin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Yu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobo Yang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huaqing Shu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiancheng Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiying Yuan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|