1
|
Cao L, Du M, Cai M, Feng Y, Miao J, Sun J, Song J, Du B. Neutrophil membrane-coated nanoparticles for targeted delivery of toll-like receptor 4 siRNA ameliorate LPS-induced acute lung injury. Int J Pharm 2025; 668:124960. [PMID: 39551221 DOI: 10.1016/j.ijpharm.2024.124960] [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: 06/13/2024] [Revised: 10/29/2024] [Accepted: 11/13/2024] [Indexed: 11/19/2024]
Abstract
Pulmonary delivery of small interfering RNAs (siRNAs) is an effective treatment for acute lung injury (ALI), which can modulate the expression of pro-inflammatory cytokines and alleviate the symptoms of ALI. However, the rapid degradation of siRNA in vivo and its limited ability to target and validate cells are important challenges it faces in clinical practice. In this work, we developed neutrophil membrane-coated Poly (lactic-co-glycolic acid) nanoparticles loaded with TLR4 siRNA (si-TLR4) (Neutrophil-NP-TLR4), which can target both inflammatory and macrophage cells to alleviate the pulmonary inflammation in lipopolysaccharide (LPS)-induced ALI mice. These Neutrophil-NP-TLR4 effectively reduce the TNF-α and IL-1β expressions both in vitro and in vivo. Meanwhile, they also reduced the expression of TLR4, and its downstream genes including TNF receptor-associated factor 6 (TRAF6), X-linked inhibitor of apoptosis protein (XIAP), and Nuclear Factor kappa-B (NF-κB), but elevated the levels of Aquaporin 1 (AQP1) and Aquaporin 5 (AQP5). Moreover, the Neutrophil-NP-TLR4 precisely targets the inflammatory site to attenuate the lung injury without causing toxicity to normal tissue. This system provides a promising approach to effective delivery of siRNA to precisely treat the ALI.
Collapse
Affiliation(s)
- Liang Cao
- Department of ICU, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Min Du
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Mengmeng Cai
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Yan Feng
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Juanjuan Miao
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Jiafeng Sun
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Jie Song
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Boxiang Du
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China.
| |
Collapse
|
2
|
Yue M, Shang W, Zhang J, Chen R, Wei L, Wang H, Meng M, Zhang M, Liu Q. The polysaccharide from purple sweet potato (Ipomoea batatas (L.) Lam) alleviates lipopolysaccharide-induced acute lung injury in mice via the VIP/cAMP/PKA/AQPs signaling pathway. Int J Biol Macromol 2024; 282:137428. [PMID: 39522908 DOI: 10.1016/j.ijbiomac.2024.137428] [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: 04/16/2024] [Revised: 10/15/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The polysaccharide (PSP) from purple sweet potato has great potential for regulating apoptosis, but its regulatory role in acute lung injury (ALI) is unknown. METHODS The objective of this study was to investigate the protective effect of PSP on lipopolysaccharide (LPS)-induced ALI in mice and lung epithelial A549 cells and its mechanism. Moreover, subacute toxicity evaluation of PSP was carried out on ICR mice. RESULTS The results showed that compared with the ALI group, PSP significantly reduced the total protein content, wet-to-dry (W/D) ratio, the number of neutrophils, lymphocytes, and monocytes. Moreover, PSP was able to reduce cell apoptosis, the levels of macrophage inflammatory protein-2 (MIP-2), intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-α (TNF-α), malondialdehyde (MDA) and myeloperoxidase (MPO) and increase the level of superoxide (SOD). In addition, PSP could up-regulate the levels of VIP, cAMP, p-PKA/PKA and AQP1 in mice and A549 cells. And PSP exhibited no apparent adverse effects on the mice. CONCLUSIONS PSP had a protective effect on LPS-induced ALI in mice and lung epithelial A549 cells, which may be related to the inflammatory response and via VIP/cAMP/PKA/AQPs signaling pathway. Thus, PSP may be a promising pharmacologic agent for ALI therapy.
Collapse
Affiliation(s)
- Maokui Yue
- Emergency Medicine, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - Wenli Shang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - Junli Zhang
- Emergency Medicine, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - Ran Chen
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - Li Wei
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - HaiDong Wang
- Shandong First Medical University, No.2, Yingsheng East Road, Taian 271000, China
| | - Meng Meng
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Science and Engineering, China
| | - Min Zhang
- Emergency Medicine, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China.
| | - Qinghua Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China.
| |
Collapse
|
3
|
Hou Z, Yang F, Zhang Q, Wang Y, Liu J, Liang F. Targeting the PI3K/AKT signaling pathway with PNU120596 protects against LPS-induced acute lung injury. J Pharm Pharmacol 2024; 76:1508-1520. [PMID: 39288376 DOI: 10.1093/jpp/rgae076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 06/04/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVES This study investigated the potential therapeutic benefits of PNU120596, a positive allosteric modulator of the α7 nicotinic acetylcholine receptor (α7nAChR), in mitigating acute lung injury (ALI) induced by lipopolysaccharide (LPS) in a mouse model. Specifically, we sought to examine the impact of PNU120596 on the PI3K/AKT signaling pathway in the context of ALI. METHODS ALI was induced in mice by LPS administration, and the protective effects of PNU120596 were assessed. Lung injury, lung function, and the inflammatory response were evaluated. Additionally, the activation of the PI3K/AKT signaling pathway was examined, along with the levels of inflammatory factors and oxidative stress markers. KEY FINDINGS PNU120596 significantly ameliorated LPS-induced lung injury, improved lung function, and reduced the inflammatory response in the mouse model of ALI. Furthermore, we observed that PNU120596 inhibited the activation of the PI3K/AKT signaling pathway, which was associated with decreased levels of inflammatory factors and oxidative stress markers. CONCLUSIONS PNU120596 exhibits promising therapeutic potential for the treatment of acute lung injury, potentially by targeting the PI3K/AKT signaling pathway. These findings suggest that modulation of the α7 nicotinic acetylcholine receptor with PNU120596 may offer a viable strategy for the management of ALI, warranting further investigation and potential clinical applications.
Collapse
Affiliation(s)
- Zixin Hou
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Fengrui Yang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
- Department of Anesthesiology, General Hospital, Hunan University of Medicine, Huaihua 418000, PR China
| | - Qiang Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Yuxia Wang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Junwen Liu
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Feng Liang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| |
Collapse
|
4
|
Duan H, Wang W, Li S, Li H, Khan GJ, Ma Y, Liu F, Zhai K, Hu H, Wei Z. The potential mechanism of
Isodon suzhouensis against COVID-19 via EGFR/TLR4 pathways. FOOD SCIENCE AND HUMAN WELLNESS 2024; 13:3245-3255. [DOI: 10.26599/fshw.2023.9250011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
5
|
Zhang X, Lv D, Li B, Ding Y. Inhaled aerosolized algal polysaccharides: A novel and reliable strategy for treating pneumonia through inflammation and oxidative stress inhibition. Int Immunopharmacol 2024; 137:112532. [PMID: 38908087 DOI: 10.1016/j.intimp.2024.112532] [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: 02/28/2024] [Revised: 06/02/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Sepsis-associated acute lung injury (ALI) poses a significant threat, characterized by inflammation and oxidative damage. Effective drugs targeting these aspects with reliable drug delivery systems are vital for ALI management. This study aimed to evaluate the influence of algal polysaccharides (APs) with aerosolized drug delivery in ALI mice and clarify the underlying mechanism. To induce the sepsis-associated acute lung injury (ALI) model, mice were administered intraperitoneal injections of 10 mg/kg LPS for 48 h in vivo. ALI mice received APs via atomization to arrive at different sites within the lungs. Lung tissue samples and bronchoalveolar lavage fluid (BALF) were collected to access lung injury parameters. Concurrently, western blotting, H&E staining, and immunofluorescence (IF) were applied to investigate the specific impact of APs on ALI. The results showed that APs protect lung tissue against ALI by inhibiting inflammation and mitigating oxidative stress-induced damage. This study highlights promising avenues for ALI intervention using natural compounds with anti-inflammatory and antioxidant properties.
Collapse
Affiliation(s)
- XingXing Zhang
- Department of Neurology, Changshu No. 2 People's Hospital (Affiliated Changshu Hospital of Nantong University), Changshu 215500, Jiangsu, China; School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Demin Lv
- Department of Traumatic Orthopedics, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu, China
| | - Bingxia Li
- Department of Stomatology, The 359th Hospital of the People's Liberation Army of China, Zhenjiang 212001, Jiangsu, China
| | - Yuting Ding
- Department of Neurology, Changshu No. 2 People's Hospital (Affiliated Changshu Hospital of Nantong University), Changshu 215500, Jiangsu, China.
| |
Collapse
|
6
|
Wang K, Wang Y, Zhang T, Chang B, Fu D, Chen X. The Role of Intravenous Anesthetics for Neuro: Protection or Toxicity? Neurosci Bull 2024:10.1007/s12264-024-01265-4. [PMID: 39153174 DOI: 10.1007/s12264-024-01265-4] [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: 12/15/2023] [Accepted: 03/15/2024] [Indexed: 08/19/2024] Open
Abstract
The primary intravenous anesthetics employed in clinical practice encompass dexmedetomidine (Dex), propofol, ketamine, etomidate, midazolam, and remimazolam. Apart from their established sedative, analgesic, and anxiolytic properties, an increasing body of research has uncovered neuroprotective effects of intravenous anesthetics in various animal and cellular models, as well as in clinical studies. However, there also exists conflicting evidence pointing to the potential neurotoxic effects of these intravenous anesthetics. The role of intravenous anesthetics for neuro on both sides of protection or toxicity has been rarely summarized. Considering the mentioned above, this work aims to offer a comprehensive understanding of the underlying mechanisms involved both in the central nerve system (CNS) and the peripheral nerve system (PNS) and provide valuable insights into the potential safety and risk associated with the clinical use of intravenous anesthetics.
Collapse
Affiliation(s)
- Kaixin Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Bingcheng Chang
- The Second Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550003, China
| | - Daan Fu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China.
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China.
| |
Collapse
|
7
|
Liu FC, Yang YH, Liao CC, Lee HC. Xanthoxylin Attenuates Lipopolysaccharide-Induced Lung Injury through Modulation of Akt/HIF-1α/NF-κB and Nrf2 Pathways. Int J Mol Sci 2024; 25:8742. [PMID: 39201430 PMCID: PMC11354527 DOI: 10.3390/ijms25168742] [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: 05/29/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Xanthoxylin, a bioactive phenolic compound extracted from the traditional herbal medicine Penthorum Chinense Pursh, is renowned for its anti-inflammatory effects. While previous studies have highlighted the anti-inflammatory and antioxidant properties of Xanthoxylin, its precise mechanisms, particularly concerning immune response and organ protection, remain underexplored. This study aimed to elucidate the effects of Xanthoxylin on inflammation and associated signaling pathways in a mouse model of lipopolysaccharide (LPS)-induced acute lung injury (ALI). ALI was induced via intratracheal administration of LPS, followed by intraperitoneal injections of Xanthoxylin at doses of 1, 2.5, 5, and 10 mg/kg, administered 30 min post-LPS exposure. Lung tissues were harvested for analysis 6 h after LPS challenge. Xanthoxylin treatment significantly mitigated lung tissue damage, pathological alterations, immune cell infiltration, and the production of pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). Additionally, Xanthoxylin modulated the expression of key proteins in the protein kinase B (Akt)/hypoxia-inducible factor 1-alpha (HIF-1α)/nuclear factor-kappa B (NF-κB) signaling pathway, as well as nuclear factor erythroid 2-related factor 2 (Nrf2) and oxidative markers such as superoxide dismutase (SOD) and malondialdehyde (MDA) in the context of LPS-induced injury. This study demonstrates that Xanthoxylin exerts protective and anti-inflammatory effects by down-regulating and inhibiting the Akt/HIF-1α/NF-κB pathways, suggesting its potential as a therapeutic target for the prevention and treatment of ALI or acute respiratory distress syndrome (ARDS).
Collapse
Affiliation(s)
- Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan; (F.-C.L.); (C.-C.L.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Yuan-Han Yang
- Department of Chinese Acupuncture and Traumatology, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
| | - Chia-Chih Liao
- Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan; (F.-C.L.); (C.-C.L.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Hung-Chen Lee
- Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan; (F.-C.L.); (C.-C.L.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
8
|
Yu C, Huang Y, Xie J, Duan C, Liu S, Zhao W, Wang Y, Zhuang R, Li J, Yin W. HMGB1 released from pyroptotic vascular endothelial cells promotes immune disorders in exertional heatstroke. Int J Hyperthermia 2024; 41:2378867. [PMID: 39117343 DOI: 10.1080/02656736.2024.2378867] [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: 06/08/2023] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Exertional heatstroke (EHS) mainly occurs in healthy young people with rapid onset and high mortality. EHS immune disorders can cause systemic inflammatory responses and multiple organ failure; however, the underlying mechanisms remain unclear. As high mobility group box 1 (HMGB1) is a prototypical alarmin that activates inflammatory and immune responses, this study aimed to investigate the effect and mechanism of HMGB1 in the pathogenesis of EHS. METHODS Peripheral blood mononuclear cell (PBMC) transcriptome sequencing of healthy volunteers, classical heatstroke patients, and EHS patients was performed. A mouse model of EHS was established and murine tissue damage was evaluated by H&E staining. HMGB1 localization and release were visualized using immunofluorescence staining. Human umbilical vein endothelial cells (HUVECs) and THP-1 cells were co-cultured to study the effects of HMGB1 on macrophages. A neutralizing anti-HMGB1 antibody was used to evaluate the efficacy of EHS treatment in mice. RESULTS Plasma and serum HMGB1 levels were significantly increased in EHS patients or mice. EHS-induced endothelial cell pyroptosis promoted HMGB1 release in mice. HMGB1 derived from endothelial cell pyroptosis enhanced macrophage pyroptosis, resulting in immune disorders under EHS conditions. Administration of anti-HMGB1 markedly alleviated tissue injury and systemic inflammatory responses after EHS. CONCLUSIONS The release of HMGB1 from pyroptotic endothelial cells after EHS promotes pyroptosis of macrophages and systemic inflammatory response, and HMGB1-neutralizing antibody therapy has good application prospects for EHS.
Collapse
Affiliation(s)
- Chaoping Yu
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- Department of Emergency, Air Force Hospital of Eastern Theater Command, Nanjing, China
| | - Yang Huang
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiangang Xie
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chujun Duan
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shanshou Liu
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei Zhao
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yutong Wang
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Junjie Li
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wen Yin
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
9
|
Wu W, He Y, Lin D, Zhang G, Zhang X, Zhang N, Xie T, Wei H. Dexmedetomidine mitigates lipopolysaccharide-induced acute lung injury by modulating heat shock protein A12B to inhibit the toll-like receptor 4/nuclear factor-kappa B signaling pathway. Chem Biol Interact 2024; 398:111112. [PMID: 38901789 DOI: 10.1016/j.cbi.2024.111112] [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: 04/03/2024] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS): Life-threatening medical conditions characterized by high morbidity and mortality rates, where the inflammatory process plays a crucial role in lung tissue damage, especially in models induced by lipopolysaccharide (LPS). Heat shock protein A12B (HSPA12B) has strong anti-infammatory properties However, it is unknown whether increased HSPA12B is protective against LPS-induced ALI. And Dexmedetomidine (DEX) is a potent α2-adrenergic receptor (α2-AR) agonist that has been shown to protect against sepsis-induced lung injury, however, the underlying mechanisms of this protection are not fully understood. This study utilized bioinformatics analysis and an LPS-induced ALI model to explore how DEX alleviates lung injury by modulating HSPA12B and inhibiting the Toll-like receptor 4/nuclear factor-kappa B (TLR4/NF-κB) signaling pathway. Results indicate that HSPA12B overexpression and DEX pre-treatment markedly mitigated LPS-induced lung injury, which was evaluated by the deterioration of histopathology, histologic scores, the W/D weight ratio, and total protein expression, tumor necrosis factor-alpha (TNF-α), and interleukin-1β (IL-1β) in the BALF, and the levels of NO, MDA,SOD and MPO in the lung. Moreover, HSPA12B overexpression and DEX pre-treatment significantly reduces lung injury and inflammation levels by upregulating HSPA12B and inhibiting the activation of the TLR4/NF-κB signaling pathway. On the contrary, when the expression of HSPA12B is inhibited, the protective effect of DEX pre-treatment on lung tissue is significantly weakened.In summary, our research demonstrated that the increased expression of AAV-mediated HSPA12B in the lungs of mice inhibits acute inflammation and suppresses the activation of TLR4/NF-κB pathway in a murine model of LPS-induced ALI. DEX could enhance HSPA12B and inhibit the initiation and development of inflammation through down-regulating TLR4/NF-κB pathway.These findings highlight the potential of DEX as a therapeutic agent for treating ALI and ARDS, offering new strategies for clinical intervention.
Collapse
Affiliation(s)
- Weifang Wu
- The Department of Anesthesiology, Fuzhou First General Hospital Affiliated with Fujian Medical University, Fuzhou, 350001, Fujian, China; The Third Clinical Medical College of Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Yi He
- The Third Clinical Medical College of Fujian Medical University, Fuzhou, 350122, Fujian, China; The Department of Anesthesiology, Affiliated Nanping First Hospital, Fujian Medical University, Nanping, 353000, Fujian, China
| | - Duoduo Lin
- The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Guifei Zhang
- The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Xutao Zhang
- The Third Clinical Medical College of Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Nanwen Zhang
- The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, China.
| | - Tingliang Xie
- The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, China; The School of Medical, Minjiang Teachers College, Fuzhou, 350108, Fujian, China.
| | - Haixiang Wei
- The Third Clinical Medical College of Fujian Medical University, Fuzhou, 350122, Fujian, China; The Department of Anesthesiology, Affiliated Nanping First Hospital, Fujian Medical University, Nanping, 353000, Fujian, China.
| |
Collapse
|
10
|
Su Q, Yang SP, Guo JP, Rong YR, Sun Y, Chai YR. Epigallocatechin-3-gallate ameliorates lipopolysaccharide-induced acute thymus involution in mice via AMPK/Sirt1 pathway. Microbiol Immunol 2024; 68:281-293. [PMID: 38886542 DOI: 10.1111/1348-0421.13159] [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: 01/05/2024] [Revised: 05/08/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024]
Abstract
The thymus, a site to culture the naïve T lymphocytes, is susceptible to atrophy or involution due to aging, inflammation, and oxidation. Epigallocatechin-3-gallate (EGCG) has been proven to possess anti-inflammatory, antioxidant, and antitumor activity. Here, we investigate the effects of EGCG on thymic involution induced by lipopolysaccharide (LPS), an endotoxin derived from Gram-negative bacteria. The methodology included an in vivo experiment on female Kunming mice exposed to LPS and EGCG. Morphological assessment of thymic involution, immunohistochemical detection, and thymocyte subsets analysis by flow cytometry were further carried out to evaluate the potential role of EGCG on the thymus. As a result, we found that EGCG alleviated LPS-induced thymic atrophy, increased mitochondrial membrane potential and superoxide dismutase levels, and decreased malondialdehyde and reactive oxygen species levels. In addition, EGCG pre-supplement restored the ratio of thymocyte subsets, the expression of autoimmune regulator, sex-determining region Y-box 2, and Nanog homebox, and reduced the number of senescent cells and collagen fiber deposition. Western blotting results indicated that EGCG treatment elevated LPS-induced decrease in pAMPK, Sirt1 protein expression. Collectively, EGCG relieved thymus architecture and function damaged by LPS via regulation of AMPK/Sirt1 signaling pathway. Our findings may provide a new strategy on protection of thymus from involution caused by LPS by using EGCG. And EGCG might be considered as a potential agent for the prevention and treatment of thymic involution.
Collapse
Affiliation(s)
- Qing Su
- Department of Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, PR China
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Shu-Ping Yang
- Department of Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, PR China
- School of Medical Technology, Sanquan College of Xinxiang Medical University, Xinxiang, Henan, PR China
| | - Jun-Ping Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Yi-Ren Rong
- Department of Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Yun Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Yu-Rong Chai
- Department of Histology and Embryology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, PR China
| |
Collapse
|
11
|
Qiu H, Shao Z, Wen X, Qu D, Liu Z, Chen Z, Zhang X, Ding X, Zhang L. HMGB1/TREM2 positive feedback loop drives the development of radioresistance and immune escape of glioblastoma by regulating TLR4/Akt signaling. J Transl Med 2024; 22:688. [PMID: 39075517 PMCID: PMC11287841 DOI: 10.1186/s12967-024-05489-w] [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: 04/22/2024] [Accepted: 07/04/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Radioresistance and immune escape are crucial reasons for unsatisfactory therapeutic effects of glioblastoma (GBM). Although triggering receptor expressed on myeloid cells-2 (TREM2) involved in forming immunosuppressive microenvironment, but the underlying mechanism and its roles in mediating cancer radioresistance remain unclear, moreover, the efficient delivery of drugs targeting TREM2 to GBM encounters serious challenges. Hence, this study aimed to elucidate the effect and mechanisms of targeted TREM2 silencing on reversing the radioresistance and immune escape of GBM aided by a glutathione-responsive biomimetic nanoparticle (NP) platform. METHODS Radioresistant GBM cell lines and TREM2 stable knockdown GBM cell lines were firstly established. RNA sequencing, colony formation assay, western blot, enzyme-linked immunosorbent assay and co-immunoprecipitation assay were used to detect the molecular mechanisms of TREM2 in regulating the radioresistance and immune escape of GBM. The glutathione-responsive biomimetic NP, angiopep-2 (A2)- cell membrane (CM)-NP/siTREM2/spam1, was then constructed to triply and targeted inhibit TREM2 for in vivo study. Orthotopic GBM-bearing mouse models were established to evaluate the anti-GBM effect of TREM2 inhibition, multiplex immunofluorescence assay was conducted to detect the infiltration of immune cells. RESULTS TREM2 was a regulator in accelerating the radioresistance and immune escape of GBM through participating in DNA damage repair and forming a positive feedback loop with high mobility group box 1 (HMGB1) to cascade the activation of Toll-like receptor 4 (TLR4)/protein kinase B (Akt) signaling. A2-CM-NP/siTREM2/spam1 was successfully synthesized with excellent passive targeting, active targeting and homologous targeting, and the in vivo results exhibited its remarkable anti-GBM therapeutic effect through promoting the infiltration of type 1 helper T cells and CD8+T cells, reducing the infiltration of type 2 helper T cells and regulatory T cells, repolarizing macrophages to M1-type, and decreasing the secretion of pro-tumor and immunosuppressive cytokines. CONCLUSIONS Targeting TREM2 therapy is a promising avenue for optimizing radiotherapy and immunotherapy to improve the prognosis of GBM patients.
Collapse
Affiliation(s)
- Hui Qiu
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, 221000, Jiangsu, China
| | - Zhiying Shao
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Xin Wen
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, 221000, Jiangsu, China
| | - Debao Qu
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, 221000, Jiangsu, China
| | - Zhengyang Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Ziqin Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Xinyan Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Xin Ding
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China.
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, 221000, Jiangsu, China.
| | - Longzhen Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China.
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, No. 9 Kunpeng North Road, Xuzhou, 221000, Jiangsu, China.
| |
Collapse
|
12
|
Ciftel S, Mercantepe F, Mercantepe T, Ciftel E, Klisic A. Dexmedetomidine on the interplay of IL-6 and STAT3 pathways in adrenal gland damage-induced scalding burns in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03300-7. [PMID: 39042159 DOI: 10.1007/s00210-024-03300-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/14/2024] [Indexed: 07/24/2024]
Abstract
Scalding burns are a common form of thermal injury that often leads to systemic complications. Pro-inflammatory cytokines like interleukin-6 (IL-6) and the activation of signal transducer and activator of transcription 3 (STAT3) pathways have been linked to the pathophysiology of organ damage caused by burns. This study aimed to investigate the potential therapeutic effects of dexmedetomidine, an α2-adrenergic receptor agonist with anti-inflammatory properties, on the interplay of IL-6 and STAT3 pathways in adrenal gland damage following scalding burns in rats. Twenty-eight rats were divided randomly into four groups. Rats in group 1 (n=7, control) were given only 0.9% intraperitoneal (i.p.) NaCl. Rats in group 2 (n=7, DEX) were exposed to 25°C water for 17 s on day 1 and received 100 mcg/kg/day dexmedetomidine i.p. for 3 days; for rats in group 3 (n=7, Burn), boiling water of 94°C was applied inside for 17 s. Rats in group 4 (n=7, Burn+DEX) were exposed to 94°C water for 17 s and received 100 mcg/kg/day dexmedetomidine i.p. for 3 days. Adrenal gland tissues were histopathological examined, and STAT3, IL-6, and TUNEL staining were performed using immunohistochemically. Our results revealed that scalding burns increased IL-6 and STAT3 expression in the adrenal glands of rats. Histological analysis demonstrated that dexmedetomidine administration ameliorated adrenal gland damage and reduced inflammatory cell infiltration. Our findings suggest that dexmedetomidine protects the adrenal glands in scalding burns. This protection appears to be mediated, at least in part, by its modulation of IL-6 and STAT3 pathways.
Collapse
Affiliation(s)
- Serpil Ciftel
- Department of Endocrinology and Metabolism, Erzurum Regional Training and Research Hospital, Erzurum, Turkey
| | - Filiz Mercantepe
- Department of Endocrinology and Metabolism, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, 53010, Turkey.
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Enver Ciftel
- Department of Endocrinology and Metabolism, Sivas Numune Hospital, Sivas, Turkey
| | - Aleksandra Klisic
- University of Montenegro-Faculty of Medicine, Podgorica, Montenegro
- Center for Laboratory Diagnostics, Primary Health Care Center, Podgorica, Montenegro
| |
Collapse
|
13
|
Zhang W, Zhong Y, Wang Z, Tang F, Zheng C. Apple polysaccharide improves age-matched cognitive impairment and intestinal aging through microbiota-gut-brain axis. Sci Rep 2024; 14:16215. [PMID: 39003416 PMCID: PMC11246462 DOI: 10.1038/s41598-024-67132-4] [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: 03/29/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024] Open
Abstract
The Apple polysaccharides (AP), extracted from the fruit of apple, has been used to treat multiple pathological diseases. In this study, we evaluated the effects of AP on cognitive impairment and intestinal aging in naturally aging mice. As a result, it was found that AP could improve spatial learning and memory impairment in aging mice through the Morris water maze experiment. Additionally, AP intervention can upregulate the expression of nerve growth factor (BDNF), postsynaptic marker (PSD95), and presynaptic marker (SYP) proteins. Moreover, AP can enhance total antioxidant capacity, reduce the level of pro-inflammatory cytokine, and inhibit the activation of the NF-κB signaling pathway, exerting anti-inflammatory and antioxidant functions. And the administration of AP restored intestinal mucosal barrier function, reduced the expression of aging and apoptosis related proteins. The administration of AP also altered the gut microbiota of mice. At the genus level, AP decreased the abundance of Helicobacter and Bilophila, while increased the abundance of Lactobacillus and Bacteroides. In summary, these data demonstrate that AP treatment can alleviate cognitive impairment, oxidative stress, and inflammatory reactions, repair the intestinal mucosal barrier, reduce intestinal aging, and alter specific microbial characteristics, ultimately improving the health of the elderly.
Collapse
Affiliation(s)
- Wenming Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, 330000, Nanchang, Jiangxi, People's Republic of China
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Yuchun Zhong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, 330000, Nanchang, Jiangxi, People's Republic of China
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Zhuoya Wang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Furui Tang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Cihua Zheng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, 330000, Nanchang, Jiangxi, People's Republic of China.
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China.
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
14
|
Shaikh TB, Chandra Y, Andugulapati SB, Sistla R. Vistusertib improves pulmonary inflammation and fibrosis by modulating inflammatory/oxidative stress mediators via suppressing the mTOR signalling. Inflamm Res 2024; 73:1223-1237. [PMID: 38789791 DOI: 10.1007/s00011-024-01894-5] [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: 03/25/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
INTRODUCTION Inflammation and oxidative stress are key factors in the development of pulmonary fibrosis (PF) by promoting the differentiation of fibroblasts through modulating various pathways including Wnt/β-catenin, TGF-β and mTOR signalling. OBJECTIVE AND METHODS This study aimed to evaluate the effects and elucidate the mechanisms of vistusertib (VSB) in treating pulmonary inflammation/fibrosis, specifically by targeting the mTOR pathway using various in vitro and in vivo models. RESULTS Lipopolysaccharide (LPS)-induced inflammation model in macrophages (RAW 264.7), epithelial (BEAS-2B) and endothelial (HMVEC-L) cells revealed that treatment with VSB significantly reduced the IL-6, TNF-α, CCL2, and CCL7 expression. TGF-β induced differentiation was also significantly reduced upon VSB treatment in fibrotic cells (LL29 and DHLF). Further, bleomycin-induced inflammation and fibrosis models demonstrated that treatment with VSB significantly ameliorated the severe inflammation, and lung architectural distortion, by reducing the inflammatory markers expression/levels, inflammatory cells and oxidative stress indicators. Further, fibrosis model results exhibited that, VSB treatment significantly reduced the α-SMA, collagen and TGF-β expressions, improved the lung architecture and restored lung functions. CONCLUSION Overall, this study uncovers the anti-inflammatory/anti-fibrotic effects of VSB by modulating the mTOR activation. Although VSB was tested for lung fibrosis, it can be tested for other fibrotic disorders to improve the patient's survival and quality of life.
Collapse
Affiliation(s)
- Taslim B Shaikh
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India
| | - Yogesh Chandra
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India
| | - Sai Balaji Andugulapati
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India.
| | - Ramakrishna Sistla
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India.
| |
Collapse
|
15
|
Liu Z, Wei J, Sun H, Xu L. Plumbagin ameliorates LPS-induced acute lung injury by regulating PI3K/AKT/mTOR and Keap1-Nrf2/HO-1 signalling pathways. J Cell Mol Med 2024; 28:e18386. [PMID: 38990057 PMCID: PMC11238321 DOI: 10.1111/jcmm.18386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/17/2024] [Accepted: 04/27/2024] [Indexed: 07/12/2024] Open
Abstract
Acute lung injury (ALI) is a major pathophysiological problem characterized by severe inflammation, resulting in high morbidity and mortality. Plumbagin (PL), a major bioactive constituent extracted from the traditional Chinese herb Plumbago zeylanica, has been shown to possess anti-inflammatory and antioxidant pharmacological activities. However, its protective effect on ALI has not been extensively studied. The objective of this study was to investigate the protective effect of PL against ALI induced by LPS and to elucidate its possible mechanisms both in vivo and in vitro. PL treatment significantly inhibited pathological injury, MPO activity, and the wet/dry ratio in lung tissues, and decreased the levels of inflammatory cells and inflammatory cytokines TNF-α, IL-1β, IL-6 in BALF induced by LPS. In addition, PL inhibited the activation of the PI3K/AKT/mTOR signalling pathway, increased the activity of antioxidant enzymes CAT, SOD, GSH and activated the Keap1/Nrf2/HO-1 signalling pathway during ALI induced by LPS. To further assess the association between the inhibitory effects of PL on ALI and the PI3K/AKT/mTOR and Keap1/Nrf2/HO-1 signalling, we pretreated RAW264.7 cells with 740Y-P and ML385. The results showed that the activation of PI3K/AKT/mTOR signalling reversed the protective effect of PL on inflammatory response induced by LPS. Moreover, the inhibitory effects of PL on the production of inflammatory cytokines induced by LPS also inhibited by downregulating Keap1/Nrf2/HO-1 signalling. In conclusion, the results indicate that the PL ameliorate LPS-induced ALI by regulating the PI3K/AKT/mTOR and Keap1-Nrf2/HO-1 signalling, which may provide a novel therapeutic perspective for PL in inhibiting ALI.
Collapse
Affiliation(s)
- Zhengjia Liu
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Jiahui Wei
- Department of RespiratoryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Hongbin Sun
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Lei Xu
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
16
|
Ding X, Gao X, Ren A, Xu J, Jiang X, Liang X, Xie K, Zhou Y, Hu C, Huang D. Sevoflurane enhances autophagy via Rac1 to attenuate lung ischaemia‒reperfusion injury. Chem Biol Interact 2024; 397:111078. [PMID: 38815668 DOI: 10.1016/j.cbi.2024.111078] [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: 03/26/2024] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Sevoflurane can attenuate lung ischaemia‒reperfusion injury (LIRI). However, the protective mechanism is unclear. In this study, we developed a LIRI model in vivo that animals (SD, n = 15) were subjected to the administration of 2.2 % sevoflurane 30 min before the onset of left pulmonary artery clamping for 45 min, which was then followed by 60 min of reperfusion treatment. Then, transcriptome sequencing was used to analyse lung tissues. Autophagy inhibition (3-MA) and Rac1-overexpression transfection plasmids were used in BEAS-2B cells, and BEAS-2B cells were subjected to hypoxia reoxygenation (H/R) and sevoflurane treatment. In both animal tissue and cells, inflammatory cytokines and apoptotic and autophagy molecules were measured by quantitative real-time PCR, western blotting and immunostaining. As a result, decreased arterial partial oxygen and damage to the histological structure of lung tissues were observed in LIRI model rats, and these effects were reversed by sevoflurane treatment. Activation of inflammation (elevated IL-1β, IL-6, and TNF-α) and apoptosis (elevated cleaved caspase3/caspase3 and Bax, degraded expression of Bcl2) and inhibition of autophagy (elevated P62, degraded expression of Beclin1 and LC3-II/LC3I) in the model group were ameliorated by sevoflurane. Transcriptome sequencing indicated that the PI3K/Akt pathway regulated by Rac1 plays an important role in LIRI. Furthermore, overexpression of Rac1 in a cell line inhibited the protective effect of sevoflurane in LIRI. Autophagy inhibition (3-MA) also prevented the protective effect of sevoflurane on inflammation and apoptosis. As shown in the present study, sevoflurane enhances autophagy via Rac1/PI3K/AKT signalling to attenuate lung ischaemia‒reperfusion injury.
Collapse
Affiliation(s)
- Xian Ding
- Department of Anesthesiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Xiang Gao
- Department of Anesthesiology, The Affiliated Fujian Maternity and Child Health Hospital of Fujian Medical University, 350001, China
| | - Aolin Ren
- Department of Anesthesiology and Pain Medicine, Jiangnan University Medical Center, Wuxi No.2 People's Hospital, 214002, China
| | - Jingjing Xu
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 214023, China
| | - Xuliang Jiang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, 200030, China
| | - Xiao Liang
- Department of Anesthesiology and Pain Medicine, Jiangnan University Medical Center, Wuxi No.2 People's Hospital, 214002, China
| | - Kangjie Xie
- Department of Anesthesiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Research Center for Neuro-Oncology Interaction, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, China
| | - Yan Zhou
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 214023, China
| | - Chunxiao Hu
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 214023, China
| | - Dongxiao Huang
- Department of Anesthesiology and Pain Medicine, Jiangnan University Medical Center, Wuxi No.2 People's Hospital, 214002, China.
| |
Collapse
|
17
|
Yang H, Zhao Y, Chen Y, Yang T, Dou X, Li J, Yang G, Feng G, Fang H, Fan H, Zhang S. Dexmedetomidine Alleviates Acute Stress-Induced Acute Kidney Injury by Attenuating Inflammation and Oxidative Stress via Inhibiting the P2X 7R/NF-κB/NLRP3 Pathway in Rats. Inflammation 2024:10.1007/s10753-024-02065-8. [PMID: 38896231 DOI: 10.1007/s10753-024-02065-8] [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: 04/25/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
This study aimed to investigate the potential protective effects of Dexmedetomidine (DEX) against acute kidney injury (AKI) induced by acute stress (AS). Wistar rats were divided into five groups: Control, DEX, AS, AS + DEX, and AS + A438079. The results showed that AS led to AKI by increasing inflammatory biomarkers and oxidative stress-related indicators. The acute stress model in rats was successfully established. Renal function, histopathology, oxidative stress, and inflammation were assessed. Localization of P2X7 receptor (P2X7R) was determined by immunofluorescence. Additionally, the key inflammatory proteins of the P2X7R/NF-κB/NLRP3 signaling pathway were measured by Western blotting. DEX significantly improved kidney function, alleviated kidney injury, and reduced oxidative stress and inflammation. DEX inhibited the activation of the P2X7R, decreased the expression of NF-κB, NLRP3 inflammasome, and Caspase-1, and inhibited the expression of interleukin-1β (IL-1β) and tumor necrosis factor α (TNFα). Furthermore, DEX also alleviated AS-induced AKI by inhibiting the excessive production of reactive oxygen species (ROS) and reducing oxidative stress. In conclusion, DEX attenuates AS-induced AKI by mitigating inflammation and oxidative stress through the inhibition of the P2X7R/NF-κB/NLRP3 pathway in rats.
Collapse
Affiliation(s)
- Haotian Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Academy of Agricultural Science Branch of Animal Husbandry and Veterinary Branch, Qiqihar, China
| | - Yuan Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yongping Chen
- College of Veterinary Medicine, Agricultural University, Qingdao, China
| | - Tianyuan Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinyi Dou
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Junfeng Li
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Guiyan Yang
- Department of Pathology and Laboratory Medicine, Davis Health, University of California, Sacramento, CA, USA
| | - Guofeng Feng
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hao Fang
- College of Optoelectronic Engineering, Chongqing University, Chongqing, China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Shuai Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
18
|
Gu W, Zeng Q, Wang X, Jasem H, Ma L. Acute Lung Injury and the NLRP3 Inflammasome. J Inflamm Res 2024; 17:3801-3813. [PMID: 38887753 PMCID: PMC11182363 DOI: 10.2147/jir.s464838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Acute lung injury (ALI) manifests through harm to the capillary endothelium and alveolar epithelial cells, arising from a multitude of factors, leading to scattered interstitial alterations, pulmonary edema, and subsequent acute hypoxic respiratory insufficiency. Acute lung injury (ALI), along with its more serious counterpart, acute respiratory distress syndrome (ARDS), carry a fatality rate that hovers around 30-40%. Its principal pathological characteristic lies in the unchecked inflammatory reaction. Currently, the main strategies for treating ALI are alleviation of inflammation and prevention of respiratory failure. Concerning the etiology of ALI, NLRP3 Inflammasome is essential to the body's innate immune response. The composition of this inflammasome complex includes NLRP3, the pyroptosis mediator ASC, and pro-caspase-1. Recent research has reported that the inflammatory response centered on NLRP3 inflammasomes plays a key part in inflammation in ALI, and may hence be a prospective candidate for therapeutic intervention. In the review, we present an overview of the ailment characteristics of acute lung injury along with the constitution and operation of the NLRP3 inflammasome within this framework. We also explore therapeutic strategies targeting the NLRP3 inflammasome to combat acute lung injury.
Collapse
Affiliation(s)
- Wanjun Gu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Qi Zeng
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Xin Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Huthaifa Jasem
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Ling Ma
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
19
|
Sun Q, Kamath P, Sun Y, Liang M, Wu L, Chang E, Chen Q, Alam A, Liu Y, Zhao H, Ma D. Dexmedetomidine attenuates lipopolysaccharide-induced renal cell fibrotic phenotypic changes by inhibiting necroinflammation via activating α 2-adrenoceptor: A combined randomised animal and in vitro study. Biomed Pharmacother 2024; 174:116462. [PMID: 38513598 DOI: 10.1016/j.biopha.2024.116462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Acute kidney injury (AKI) was reported to be one of the initiators of chronic kidney disease (CKD) development. Necroinflammation may contribute to the progression from AKI to CKD. Dexmedetomidine (Dex), a highly selective α2-adrenoreceptor (AR) agonist, has cytoprotective and "anti-" inflammation effects. This study was designed to investigate the anti-fibrotic properties of Dex in sepsis models. METHODS C57BL/6 mice were randomly treated with an i.p. injection of lipopolysaccharides (LPS) (10 mg/kg) alone, LPS with Dex (25 μg/kg), or LPS, Dex and Atipamezole (Atip, an α2-adrenoreceptor antagonist) (500 μg/kg) (n=5/group). Human proximal tubular epithelial cells (HK2) were also cultured and then exposed to LPS (1 μg/ml) alone, LPS and Dex (1 μM), transforming growth factor-beta 1 (TGF-β1) (5 ng/ml) alone, TGF-β1 and Dex, with or without Atip (100 μM) in culture media. Epithelial-mesenchymal transition (EMT), cell necrosis, necroptosis and pyroptosis, and c-Jun N-terminal kinase (JNK) phosphorylation were then determined. RESULTS Dex treatment significantly alleviated LPS-induced AKI, myofibroblast activation, NLRP3 inflammasome activation, and necroptosis in mice. Atip counteracted its protective effects. Dex attenuated LPS or TGF-β1 induced EMT and also prevented necrosis, necroptosis, and pyroptosis in response to LPS stimulation in the HK2 cells. The anti-EMT effects of Dex were associated with JNK phosphorylation. CONCLUSIONS Dex reduced EMT following LPS stimulation whilst simultaneously inhibiting pyroptosis and necroptosis via α2-AR activation in the renal tubular cells. The "anti-fibrotic" and cytoprotective properties and its clinical use of Dex need to be further studied.
Collapse
Affiliation(s)
- Qizhe Sun
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Priyanka Kamath
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Yibing Sun
- Department of Anaesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Min Liang
- Department of Anaesthesiology, the First Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Lingzhi Wu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Enqiang Chang
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Qian Chen
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Azeem Alam
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Yi Liu
- Department of Anaesthesiology, Shanxi Province Cancer Hospital (Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University), Shanxi Province, China
| | - Hailin Zhao
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK; Perioperative and Systems Medicine Laboratory, National Clinical Research Center for Child Health, Children's hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
20
|
Nguyen TU, Hurh S, In S, Nguyen LP, Cho M, Mykhailova K, Kim HR, Ham BJ, Choi Y, Kim WK, Hwang JI. SP-8356 inhibits acute lung injury by suppressing inflammatory cytokine production and immune cell infiltration. Int Immunopharmacol 2024; 131:111847. [PMID: 38518593 DOI: 10.1016/j.intimp.2024.111847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/08/2024] [Accepted: 03/09/2024] [Indexed: 03/24/2024]
Abstract
This study investigated the anti-inflammatory and protective properties of SP-8356, a synthetic derivative of (1S)-(-)-verbenone, in a mouse model of LPS-induced acute lung injury (ALI). By targeting intracellular signaling pathways and inflammatory responses, SP-8356 demonstrated a potent ability to attenuate deleterious effects of proinflammatory stimuli. Specifically, SP-8356 effectively inhibited the activation of crucial signaling molecules such as NF-κB and Akt, and subsequently dampened the expression of inflammatory cytokines in various lung cellular components. Intervention with SP-8356 treatment also preserved the structural integrity of the epithelial and endothelial barriers. By reducing immune cell infiltration into inflamed lung tissue, SP-8356 exerted a broad protective effect against ALI. These findings position SP-8356 as a promising therapeutic candidate for pulmonary inflammatory diseases that cause ALI.
Collapse
Affiliation(s)
- Thai-Uy Nguyen
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Sunghoon Hurh
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Soyeon In
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Lan Phuong Nguyen
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Minyeong Cho
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Kateryna Mykhailova
- Department of Biotechnology, College of Life Sciences Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Hong-Rae Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Byung-Joo Ham
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea; Department of Psychiatry, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Yongseok Choi
- Department of Biotechnology, College of Life Sciences Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Won-Ki Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea; Institute for Inflammation Control, Korea University, Seoul 02841, Republic of Korea.
| | - Jong-Ik Hwang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
21
|
Fan A, Gao M, Tang X, Jiao M, Wang C, Wei Y, Gong Q, Zhong J. HMGB1/RAGE axis in tumor development: unraveling its significance. Front Oncol 2024; 14:1336191. [PMID: 38529373 PMCID: PMC10962444 DOI: 10.3389/fonc.2024.1336191] [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: 11/10/2023] [Accepted: 02/15/2024] [Indexed: 03/27/2024] Open
Abstract
High mobility group protein 1 (HMGB1) plays a complex role in tumor biology. When released into the extracellular space, it binds to the receptor for advanced glycation end products (RAGE) located on the cell membrane, playing an important role in tumor development by regulating a number of biological processes and signal pathways. In this review, we outline the multifaceted functions of the HMGB1/RAGE axis, which encompasses tumor cell proliferation, apoptosis, autophagy, metastasis, and angiogenesis. This axis is instrumental in tumor progression, promoting tumor cell proliferation, autophagy, metastasis, and angiogenesis while inhibiting apoptosis, through pivotal signaling pathways, including MAPK, NF-κB, PI3K/AKT, ERK, and STAT3. Notably, small molecules, such as miRNA-218, ethyl pyruvate (EP), and glycyrrhizin exhibit the ability to inhibit the HMGB1/RAGE axis, restraining tumor development. Therefore, a deeper understanding of the mechanisms of the HMGB1/RAGE axis in tumors is of great importance, and the development of inhibitors targeting this axis warrants further exploration.
Collapse
Affiliation(s)
- Anqi Fan
- College of Life Science, Yangtze University, Jingzhou, Hubei, China
| | - Mengxiang Gao
- College of Life Science, Yangtze University, Jingzhou, Hubei, China
| | - Xuhuan Tang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengya Jiao
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chenchen Wang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingying Wei
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
22
|
Wang LL, Mai YZ, Zheng MH, Yan GH, Jin JY. A single fluorescent probe to examine the dynamics of mitochondria-lysosome interplay and extracellular vesicle role in ferroptosis. Dev Cell 2024; 59:517-528.e3. [PMID: 38272028 DOI: 10.1016/j.devcel.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/21/2023] [Accepted: 01/05/2024] [Indexed: 01/27/2024]
Abstract
Ferroptosis is a non-apoptotic form of cell death characterized by iron-dependent lipid peroxidation and glutathione (GSH) depletion. Despite recent advances, challenges remain in understanding the bidirectional interactions or interplay between organelles during ferroptosis. In this study, we aimed to understand the interplay between mitochondria (Mito) and lysosomes (Lyso) in cell homeostasis and ferroptosis. For this purpose, we designed a single fluorescent probe that marks GSH in Mito and hypochlorous acid (HOCl) in Lyso with two distinct emissions. Using this dual-targeted single fluorescent probe (9-morphorino pyronine), we detected Mito-Lyso interplay in ferroptosis. We disclosed differences in Mito-Lyso interplay depending on the induction of ferroptosis. Although erastin treatment decreased GSH, RSL3 triggered a HOCl burst, and FIN56- and FINO2-induced ferroptosis increased GSH and HOCl. Additionally, we showed that only extracellular vesicles generated during erastin-induced ferroptosis could spontaneously move and dock to neighboring cells, resulting in accelerated cell death.
Collapse
Affiliation(s)
- Ling-Li Wang
- Research Centre of Chemical Biology, Yanbian University, Yanji 133002, Jilin, China
| | - Yu-Zhuo Mai
- Research Centre of Chemical Biology, Yanbian University, Yanji 133002, Jilin, China
| | - Ming-Hua Zheng
- Research Centre of Chemical Biology, Yanbian University, Yanji 133002, Jilin, China.
| | - Guang-Hai Yan
- Department of Anatomy, Histology, and Embryology, Jilin Key Laboratory of Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, China.
| | - Jing-Yi Jin
- Research Centre of Chemical Biology, Yanbian University, Yanji 133002, Jilin, China.
| |
Collapse
|
23
|
Pan F, Shu Q, Xie H, Zhao L, Wu P, Du Y, Lu J, He Y, Wang X, Peng H. Protective effects of triptolide against oxidative stress in retinal pigment epithelium cells via the PI3K/AKT/Nrf2 pathway: a network pharmacological method and experimental validation. Aging (Albany NY) 2024; 16:3955-3972. [PMID: 38393691 PMCID: PMC10929812 DOI: 10.18632/aging.205570] [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: 09/08/2023] [Accepted: 01/11/2024] [Indexed: 02/25/2024]
Abstract
PURPOSE Among aging adults, age-related macular degeneration (AMD), is a prevalent cause of blindness. Nevertheless, its progression may be halted by antioxidation in retinal pigment epithelium (RPE). The primary effective constituent of Tripterygium wilfordii Hook. F., triptolide (TP), has demonstrated anti-inflammatory, antiproliferative, and antioxidant properties. The mechanics of the protective effect of triptolide against the oxidative damage in retinal pigment epithelial (RPE) were assessed in this study. METHODS ARPE-19 cells were pretreated with TP, and then exposed to sodium iodate (SI). First, cell viability was assessed using CCK-8. Subsequently, we measured indicators for cell oxidation including reactive oxygen species (ROS), catalase (CAT), superoxide dismutase (SOD), and malondialdehyde (MDA). Then, we used network pharmacological analysis and molecular docking to explore the signaling pathway of TP. Last, we used western blot, ELISA, and immunofluorescence assays to clarify the potential mechanistic pathways. RESULTS The network pharmacology data suggested that TP may inhibit AMD by regulating the PI3K/Akt signaling pathway. Experimental results showed that the potential mechanism is that it regulates the PI3K/Akt pathway and promotes Nrf2 phosphorylation and activation, thereby raising the level of antioxidant factors (HO-1, NQO1) and reducing the generation of ROS, which inhibit oxidative damage. CONCLUSION Our findings suggested that the effect of TP on SI-exposed RPE cells principally relies on the regulation of oxidative stress through the PI3K/Akt/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Fuying Pan
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, China
| | - Qinxin Shu
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, China
| | - Hao Xie
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, China
| | - Long Zhao
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, China
| | - Ping Wu
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, China
| | - Yong Du
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, China
| | - Jing Lu
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, China
| | - Yuxia He
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, China
| | - Xing Wang
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, China
| | - Hui Peng
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, China
| |
Collapse
|
24
|
Li C, Wang Y, Zhao X, Li J, Wang H, Ren Y, Sun H, Zhu X, Song Q, Wang J. Comparative Analysis of Intestinal Inflammation and Microbiota Dysbiosis of LPS-Challenged Piglets between Different Breeds. Animals (Basel) 2024; 14:665. [PMID: 38473050 DOI: 10.3390/ani14050665] [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: 01/22/2024] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Post-weaning diarrhea is common in piglets, causing huge economic losses worldwide. Associations between LPS challenge, intestinal inflammation, and microbiota have been reported in Duroc × Landrace × Yorkshire (DLY) crossbred pigs. However, the effects of LPS challenge in other breeds remain unclear. In the current study, we performed a comprehensive comparative analysis of the effects of LPS challenge on jejunal mucosal morphology, jejunal microbial composition, and serum indexes in two pig breeds: DLY and Heigai, an indigenous Chinese breed. The results showed that LPS caused considerable damage to the mucosal morphology, enhanced serum levels of inflammatory cytokines and the intestinal permeability index, and lowered the antioxidant capacity index. LPS challenge also changed the microbial composition and structure of the jejunum, significantly increased the abundances of Escherichia-Shigella in DLY pigs, and decreased those of Gemella and Saccharimonadales in Heigai pigs. Furthermore, LPS challenge triggered functional changes in energy metabolism and activities related to the stress response in the jejunal bacterial community, alleviating the inflammatory response in Heigai pigs. This study also revealed that Heigai pigs had a weaker immune response to LPS challenge than DLY pigs, and identified several genera related to the breed-specific phenotypes of Heigai pigs, including Gemella, Saccharimonadales, Clostridia_UCG_014, Terrisporobacter, and Dielma. Our collective findings uncovered differences between Heigai and DLY pigs in intestinal inflammation and microbiota dysbiosis induced by LPS challenge, providing a theoretical basis for unraveling the mechanism of intestinal inflammation in swine and proposing microbial candidates involved in the resistance to diarrhea in piglets.
Collapse
Affiliation(s)
- Chao Li
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Jinan 250100, China
- Hebei Veterinary Biotechnology Innovation Center, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071000, China
| | - Yanping Wang
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Jinan 250100, China
| | - Xueyan Zhao
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Jinan 250100, China
| | - Jingxuan Li
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Jinan 250100, China
| | - Huaizhong Wang
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Jinan 250100, China
| | - Yifan Ren
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Jinan 250100, China
| | - Houwei Sun
- Zaozhuang Heigai Pigs Breeding Co., Ltd., Zaozhuang 277100, China
| | - Xiaodong Zhu
- Zaozhuang Heigai Pigs Breeding Co., Ltd., Zaozhuang 277100, China
| | - Qinye Song
- Hebei Veterinary Biotechnology Innovation Center, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071000, China
| | - Jiying Wang
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Jinan 250100, China
| |
Collapse
|
25
|
Cho S, Choi HJ, Song GY, Bae JS. Therapeutic effects of hederacolchiside A1 on particulate matter-induced pulmonary injury. Toxicon 2024; 241:107650. [PMID: 38360299 DOI: 10.1016/j.toxicon.2024.107650] [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: 09/21/2023] [Revised: 01/21/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
Particulate matter (PM) comprises a hazardous mixture of inorganic and organic particles that carry health risks. Inhaling fine PM particles with a diameter of ≤2.5 μm (PM2.5) can promote significant lung damage. Hederacolchiside A1 (HA1) exhibits notable in vivo antitumor effects against various solid tumors. However, our understanding of its therapeutic potential for individuals with PM2.5-induced lung injuries remains limited. Here, we explored the protective properties of HA1 against lung damage caused by PM2.5 exposure. HA1 was administered to the mice 30 min after intratracheal tail vein injection of PM2.5. Various parameters, such as changes in lung tissue wet/dry (W/D) weight ratio, total protein/total cell ratio, lymphocyte counts, inflammatory cytokine levels in bronchoalveolar lavage fluid (BALF), vascular permeability, and histology, were assessed in mice exposed to PM2.5. Our data showed that HA1 mitigated lung damage, reduced the W/D weight ratio, and suppressed hyperpermeability caused by PM2.5 exposure. Moreover, HA1 effectively decreased plasma levels of inflammatory cytokines in those exposed to PM2.5, including tumor necrosis factor-α, interleukin-1β, and nitric oxide, while also lowering the total protein concentration in BALF and successfully alleviating PM2.5-induced lymphocytosis. Furthermore, HA1 significantly decreased the expression levels of toll-like receptor 4 (TLR4), myeloid differentiation primary response (MyD) 88, and autophagy-related proteins LC3 II and Beclin 1 but increased the protein phosphorylation of the mammalian target of rapamycin (mTOR). The anti-inflammatory characteristics of HA1 highlights its potential as a promising therapeutic agent for mitigating PM2.5-induced lung injuries by modulating the TLR4-MyD88 and mTOR-autophagy pathways.
Collapse
Affiliation(s)
- Sanghee Cho
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hui Ji Choi
- College of Pharmacy, Chungnam National University, Daejon 34134, Republic of Korea
| | - Gyu Yong Song
- College of Pharmacy, Chungnam National University, Daejon 34134, Republic of Korea.
| | - Jong-Sup Bae
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
26
|
Yarmohammadi F, Hesari M, Shackebaei D. The Role of mTOR in Doxorubicin-Altered Cardiac Metabolism: A Promising Therapeutic Target of Natural Compounds. Cardiovasc Toxicol 2024; 24:146-157. [PMID: 38108960 DOI: 10.1007/s12012-023-09820-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/09/2023] [Indexed: 12/19/2023]
Abstract
Doxorubicin (DOX) is commonly used for the treatment of various types of cancer, however can cause serious side effects, including cardiotoxicity. The mechanisms involved in DOX-induced cardiac damage are complex and not yet fully understood. One mechanism is the disruption of cardiac metabolism, which can impair cardiac function. The mammalian target of rapamycin (mTOR) is a key regulator of cardiac energy metabolism, and dysregulation of mTOR signaling has been implicated in DOX-induced cardiac dysfunction. Natural compounds (NCs) have been shown to improve cardiac function in vivo and in vitro models of DOX-induced cardiotoxicity. This review article explores the protective effects of NCs against DOX-induced cardiac injury, with a focus on their regulation of mTOR signaling pathways. Generally, the modulation of mTOR signaling by NCs represents a promising strategy for decreasing the cardiotoxic effects of DOX.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahvash Hesari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Dareuosh Shackebaei
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
27
|
Abdelgawad FAM, El-Hawary SS, El-Kader EMA, Alshehri SA, Rabeh MA, El-Mosallamy AEMK, Salama A, El Gedaily RA. Phytochemical Elucidation and Effect of Maesa indica (Roxb.) Sweet on Alleviation of Potassium Dichromate-Induced Pulmonary Damage in Rats. PLANTS (BASEL, SWITZERLAND) 2024; 13:338. [PMID: 38337870 PMCID: PMC10857331 DOI: 10.3390/plants13030338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/08/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024]
Abstract
Maesa indica (Roxb.) Sweet is one of the well-known traditionally-used Indian plants. This plant is rich in secondary metabolites like phenolic acids, flavonoids, alkaloids, glycosides, saponins, and carbohydrates. It contains numerous therapeutically active compounds like palmitic acid, chrysophanol, glyceryl palmitate, stigmasterol, β-sitosterol, dodecane, maesaquinone, quercetin 3-rhaminoside, rutin, chlorogenic acid, catechin, quercetin, nitrendipine, 2,3-dihydroxypropyl octadeca-9,12-dienoate, kiritiquinon, and β-thujone. The Maesa indica plant has been reported to have many biological properties including antidiabetic, anticancer, anti-angiogenic, anti-leishmanial, antioxidant, radical scavenging, antibacterial, antiviral, and anti-coronavirus effects. One purpose of the current study was to investigate the leaves' metabolome via Triple-Time-of-Flight-Liquid-Chromatography-Mass Spectrometry (T-TOF LC/MS/MS) to identify the chemical constituents of the Maesa indica ethanolic extract (ME). Another purpose of this study was to explore the protective effect of ME against potassium dichromate (PD)-induced pulmonary damage in rats. Rats were assigned randomly into four experimental groups. Two different doses of the plant extract, (25 and 50 mg/kg), were administered orally for seven consecutive days before PD instillation injection. Results of our study revealed that ME enhanced cellular redox status as it decreased lipid peroxidation marker, MDA and elevated reduced glutathione (GSH). In addition, ME upregulated the cytoprotective signaling pathway PI3K/AKT. Moreover, ME administration ameliorated histopathological anomalies induced by PD. Several identified metabolites, such as chlorogenic acid, quercetin, apigenin, kaempferol, luteolin, and rutin, had previously indicated lung-protective effects, possibly through an antioxidant effect and inhibition of oxidative stress and inflammatory mediators. In conclusion, our results indicated that ME possesses lung-protective effects, which may be the result of its antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
| | - Seham S. El-Hawary
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Giza 11562, Egypt;
| | - Essam M. Abd El-Kader
- Department of Timber Trees Research, Horticultural Research Institute (ARC), Giza 12619, Egypt;
| | - Saad Ali Alshehri
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62251, Saudi Arabia; (S.A.A.); (M.A.R.)
| | - Mohamed Abdelaaty Rabeh
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62251, Saudi Arabia; (S.A.A.); (M.A.R.)
| | | | - Abeer Salama
- Department of Pharmacology, National Research Centre, Cairo 12622, Egypt; (A.E.M.K.E.-M.); (A.S.)
| | - Rania A. El Gedaily
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Giza 11562, Egypt;
| |
Collapse
|
28
|
Huang Q, Le Y, Li S, Bian Y. Signaling pathways and potential therapeutic targets in acute respiratory distress syndrome (ARDS). Respir Res 2024; 25:30. [PMID: 38218783 PMCID: PMC10788036 DOI: 10.1186/s12931-024-02678-5] [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: 09/30/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common condition associated with critically ill patients, characterized by bilateral chest radiographical opacities with refractory hypoxemia due to noncardiogenic pulmonary edema. Despite significant advances, the mortality of ARDS remains unacceptably high, and there are still no effective targeted pharmacotherapeutic agents. With the outbreak of coronavirus disease 19 worldwide, the mortality of ARDS has increased correspondingly. Comprehending the pathophysiology and the underlying molecular mechanisms of ARDS may thus be essential to developing effective therapeutic strategies and reducing mortality. To facilitate further understanding of its pathogenesis and exploring novel therapeutics, this review provides comprehensive information of ARDS from pathophysiology to molecular mechanisms and presents targeted therapeutics. We first describe the pathogenesis and pathophysiology of ARDS that involve dysregulated inflammation, alveolar-capillary barrier dysfunction, impaired alveolar fluid clearance and oxidative stress. Next, we summarize the molecular mechanisms and signaling pathways related to the above four aspects of ARDS pathophysiology, along with the latest research progress. Finally, we discuss the emerging therapeutic strategies that show exciting promise in ARDS, including several pharmacologic therapies, microRNA-based therapies and mesenchymal stromal cell therapies, highlighting the pathophysiological basis and the influences on signal transduction pathways for their use.
Collapse
Affiliation(s)
- Qianrui Huang
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jie Fang Avenue, Wuhan, 430030, China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Avenue, Wuhan, 430030, China
| | - Yue Le
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China
| | - Shusheng Li
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jie Fang Avenue, Wuhan, 430030, China.
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Avenue, Wuhan, 430030, China.
| | - Yi Bian
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jie Fang Avenue, Wuhan, 430030, China.
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Avenue, Wuhan, 430030, China.
| |
Collapse
|
29
|
Hu Q, Liu H, Wang R, Yao L, Chen S, Wang Y, Lv C. Capsaicin Attenuates LPS-Induced Acute Lung Injury by Inhibiting Inflammation and Autophagy Through Regulation of the TRPV1/AKT Pathway. J Inflamm Res 2024; 17:153-170. [PMID: 38223422 PMCID: PMC10787572 DOI: 10.2147/jir.s441141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/03/2024] [Indexed: 01/16/2024] Open
Abstract
Purpose Acute lung injury (ALI) is a severe pulmonary disease characterized by damage to the alveoli and pulmonary blood vessels, leading to severe impairment of lung function. Studies on the effect of capsaicin (8-methyl-N-geranyl-6-nonamide, CAP) on lipopolysaccharide (LPS)-induced ALI in bronchial epithelial cells transformed with Ad12-SV40 2B (BEAS-2B) are still limited. This study aimed to investigate the effect and specific mechanism by which CAP improves LPS-induced ALI. Methods The present study investigated the effect of CAP and the potential underlying mechanisms in LPS-induced ALI in vitro and vivo via RNA sequencing, Western blotting (WB), quantitative real-time reverse transcription PCR (qRT‒PCR), enzyme-linked immunosorbent assay (ELISA), and transmission electron microscopy (TEM). The TRPV1 inhibitor AMG9810 and the AKT agonist SC79 were used to confirm the protective effect of the TRPV1/AKT axis against ALI. The autophagy agonist rapamycin (Rapa) and the autophagy inhibitors 3-methyladenine (3-MA) and bafilomycin A1 (Baf-A1) were used to clarify the characteristics of LPS-induced autophagy. Results Our findings demonstrated that CAP effectively suppressed inflammation and autophagy in LPS-induced ALI, both in vivo and in vitro. This mechanism involves regulation by the TRPV1/AKT signaling pathway. By activating TRPV1, CAP reduces the expression of P-AKT, thereby exerting its anti-inflammatory and inhibitory effects on pro-death autophagy. Furthermore, prior administration of CAP provided substantial protection to mice against ALI induced by LPS, reduced the lung wet/dry ratio, decreased proinflammatory cytokine expression, and downregulated LC3 expression. Conclusion Taken together, our results indicate that CAP protects against LPS-induced ALI by inhibiting inflammatory responses and autophagic death through the TRPV1/AKT signaling pathway, presenting a novel strategy for ALI therapy.
Collapse
Affiliation(s)
- Qin Hu
- Emergency and Trauma College, Hainan Medical University, Haikou, People’s Republic of China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, People’s Republic of China
| | - Haoran Liu
- Emergency and Trauma College, Hainan Medical University, Haikou, People’s Republic of China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, People’s Republic of China
| | - Ruiyu Wang
- Emergency Medicine Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Li Yao
- Emergency Medicine Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Shikun Chen
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yang Wang
- Emergency Medicine Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Chuanzhu Lv
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, People’s Republic of China
- Emergency Medicine Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, People’s Republic of China
| |
Collapse
|
30
|
Kamal MM, El-Abhar HS, Abdallah DM, Ahmed KA, Aly NES, Rabie MA. Mirabegron, dependent on β3-adrenergic receptor, alleviates mercuric chloride-induced kidney injury by reversing the impact on the inflammatory network, M1/M2 macrophages, and claudin-2. Int Immunopharmacol 2024; 126:111289. [PMID: 38016347 DOI: 10.1016/j.intimp.2023.111289] [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: 07/12/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023]
Abstract
The β3-adrenergic receptor (β3-AR) agonism mirabegron is used to treat overactive urinary bladder syndrome; however, its role against acute kidney injury (AKI) is not unveiled, hence, we aim to repurpose mirabegron in the treatment of mercuric chloride (HgCl2)-induced AKI. Rats were allocated into normal, normal + mirabegron, HgCl2 untreated, HgCl2 + mirabegron, and HgCl2 + the β3-AR blocker SR59230A + mirabegron. The latter increased the mRNA of β3-AR and miR-127 besides downregulating NF-κB p65 protein expression and the contents of its downstream targets iNOS, IL-4, -13, and -17 but increased that of IL-10 to attest its anti-inflammatory capacity. Besides, mirabegron downregulated the protein expression of STAT-6, PI3K, and ERK1/2, the downstream targets of the above cytokines. Additionally, it enhanced the transcription factor PPAR-α but turned off the harmful hub HNF-4α/HNF-1α and the lipid peroxide marker MDA. Mirabegron also downregulated the CD-163 protein expression, which besides the inhibited correlated cytokines of M1 (NF-κB p65, iNOS, IL-17) and M2 (IL-4, IL-13, CD163, STAT6, ERK1/2), inactivated the macrophage phenotypes. The crosstalk between these parameters was echoed in the maintenance of claudin-2, kidney function-related early (cystatin-C, KIM-1, NGAL), and late (creatinine, BUN) injury markers, besides recovering the microscopic structures. Nonetheless, the pre-administration of SR59230A has nullified the beneficial effects of mirabegron on the aforementioned parameters. Here we verified that mirabegron can berepurposedto treat HgCl2-induced AKI by activating the β3-AR. Mirabegron signified its effect by inhibiting inflammation, oxidative stress, and the activated M1/M2 macrophages, events that preserved the proximal tubular tight junction claudin-2 via the intersection of several trajectories.
Collapse
Affiliation(s)
- Mahmoud M Kamal
- Research Institute of Medical Entomology, General Organization for Teaching Hospitals and Institutes, Cairo, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt (FUE), 11835 Cairo, Egypt
| | - Dalaal M Abdallah
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, 11562 Cairo, Egypt.
| | - Kawkab A Ahmed
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Nour Eldin S Aly
- Research Institute of Medical Entomology, General Organization for Teaching Hospitals and Institutes, Cairo, Egypt
| | - Mostafa A Rabie
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, 11562 Cairo, Egypt; Faculty of Pharmacy and Drug Technology, Egyptian Chinese University (ECU), 19346, Egypt
| |
Collapse
|
31
|
Zhang Y, Li J, Deng H, Wan H, Xu P, Wang J, Liu R, Tang T. High mobility group box 1 knockdown inhibits EV71 replication and attenuates cell pyroptosis through TLR4/NF-κB/NLRP3 axis. J Biochem Mol Toxicol 2024; 38:e23620. [PMID: 38229319 DOI: 10.1002/jbt.23620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 08/07/2023] [Accepted: 12/14/2023] [Indexed: 01/18/2024]
Abstract
Enterovirus 71 (EV71) is a major causative agent of hand, foot, and mouth disease (HFMD) in children. Nowadays, there are still no effective antiviral drugs for EV71 infection. High mobility group box 1 (HMGB1) is reported to be highly expressed in HFMD patients. However, the role and underlying mechanism of HMGB1 in EV71-associated HFMD are still unclear. HMGB1 expression was detected using RT-qPCR and western blot assays. Loss- and gain-function experiments were performed to evaluate the effects of HMGB1 on EV71-infected cells. The virus titer was examined by TCID50. CCK-8 and flow cytometry assays were applied to detect the cell viability and cell cycle. Oxidative stress was determined by relative commercial kits. HMGB1 level was elevated in the serum of EV71-infected patients with HFMD and EV71-induced RD cells. EV71 infection induced the transfer of HMGB1 from the nucleus into the cytoplasm. HMGB1 knockdown inhibited virus replication, viral protein (VP1) expression and promoted antiviral factor expression. In addition, the inhibition of HMGB1 improved cell viability, protected against S phase arrest, and inhibited EV71-induced cell injury and oxidative stress, whereas HMGB1 overexpression showed the opposite effects. In terms of mechanism, HMGB1 overexpression activated the TLR4/NF-κB/NLRP3 signaling pathway and promoted cell pyroptosis. The inhibition of TLR4 and NF-κB reversed the effects of HMGB1 overexpression on virus replication, oxidative stress, and pyroptosis. In conclusion, HMGB1 knockdown inhibits EV71 replication and attenuates pyroptosis through TLR4/NF-κB/NLRP3 axis.
Collapse
Affiliation(s)
- Yufeng Zhang
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Jing Li
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Huiling Deng
- Department of Pediatrics, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Han Wan
- Department of General Surgery, Xi'an No. 3 Hospital, Xi'an, Shaanxi, China
| | - Pengfei Xu
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Jun Wang
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Ruiqing Liu
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Tiantian Tang
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
32
|
Weng W, Wang X, Cui Y. Artesunate Alleviates Chronic Hyperoxia-induced Bronchopulmonary Dysplasia by Suppressing NF-κB Pathway in Neonatal Mice. Comb Chem High Throughput Screen 2024; 27:2681-2690. [PMID: 37861045 DOI: 10.2174/0113862073246710231002042239] [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: 02/23/2023] [Revised: 07/29/2023] [Accepted: 08/30/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a chronic lung condition that occurs in premature infants who undergo prolonged mechanical ventilation and oxygen therapy. Existing treatment methods have shown limited efficacy, highlighting the urgent need for new therapeutic strategies. Artesunate (AS) is a compound known for its potential anti-inflammatory properties, and studies have shown its protective effects against acute lung injury. However, its impact on BPD and the underlying mechanisms remain unclear. OBJECTIVE To investigate the effect and underlying mechanism of AS on chronic hyperoxiainduced BPD in neonatal mice. METHOD Full-term C57BL/6J mice were randomly assigned to the Air+lactate Ringer's solution (L/R) group, O2 + L/R group, and O2 + AS group. Analysis was performed using assay methods such as ELISA, RT-qPCR, hematoxylin-eosin staining, and Western blotting. RESULTS Compared with the O2+L/R group, the expression of inflammatory factors in the serum, tissue, and BALF of the O2+AS group was significantly reduced, the lung function of the mice was improved, and the inflammatory infiltrates were significantly alleviated. AS inhibited the mRNA expression of inflammatory factors in mice. We found that the expression of nuclear p65 and cytoplasmic p-IκBα in the NF-κB pathway was inhibited after adding AS. CONCLUSION AS ameliorated chronic hyperoxia-induced BPD in neonatal mice probably by inhibiting the expression of NF-κB pathway and inflammatory factors.
Collapse
Affiliation(s)
- Wenbo Weng
- Yuyao Health Training School, Ningbo, Zhejiang, China
| | - Xiaoying Wang
- Department of Pediatrics, Jiande First People's Hospital, Hangzhou, Zhejiang, China
| | - Yifei Cui
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
33
|
Chao-shun W, Xiao-Li W. The impacts of SphK1 on inflammatory response and oxidative stress in LPS-induced ALI/ARDS. EUR J INFLAMM 2023. [DOI: 10.1177/1721727x231158310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
As severe conditions, acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) threaten human health. Inflammation and oxidative stress play a vital role in the pathogenesis of ALI/ARDS. Sphingosine kinase 1 (SphK1) significantly contributes to mediating inflammatory responses. Nevertheless, the impact of SphK1 on lipopolysaccharide (LPS)-triggered ALI/ARDS remains largely undetermined. In our current work, we explored the impact of SphK1 on ALI/ARDS using a mouse model. We studied whether it could reduce LPS-triggered inflammatory response and oxidative stress by suppressing SphK1 in ALI/ARDS. The mice were treated with the inhibitor of SphK1 (N,N-dimethylsphingosine, DMS) before intraperitoneal injection of LPS. Moreover, we assessed the survival rate, and several parameters, such as the lung wet/dry (W/D) ratio, myeloperoxidase (MPO) activity, superoxide dismutase (SOD) activity, malondialdehyde (MDA) content, and the release of inflammatory cytokines. Western blotting analysis was adopted to evaluate the levels of phosphoinositide 3-kinase (PI3K)/serine/threonine kinase (AKT) pathways. We showed that the inhibitor of SphK1 not only ameliorated LPS-stimulated lung histopathological changes and W/D ratio of lung tissue but also elevated the survival rate, the SOD activity and decreased the MDA content, MPO activity, interleukin-6 (IL-6) and tumor necrosis factor-ɑ (TNF-ɑ) production by regulating the PI3K/AKT signaling pathway in lung tissue. Taken together, SphK1 played an essential role in inflammatory responses and oxidative stress. The underlying mechanism might be linked to the activation and up-regulation of the PI3K/AKT signaling pathway in LPS-triggered ALI/ARDS.
Collapse
Affiliation(s)
- Wei Chao-shun
- Medical College of Jishou University, Jishou, P. R. China
| | - Wang Xiao-Li
- Medical College of Jishou University, Jishou, P. R. China
| |
Collapse
|
34
|
Yang Q, Li M, Hou Y, He H, Sun S. High-mobility group box 1 emerges as a therapeutic target for asthma. Immun Inflamm Dis 2023; 11:e1124. [PMID: 38156383 PMCID: PMC10739362 DOI: 10.1002/iid3.1124] [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: 08/01/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/30/2023] Open
Abstract
High-mobility group box 1 (HMGB1) is a highly conserved nonhistone nuclear protein found in the calf thymus and participates in a variety of intracellular processes such as DNA transcription, replication and repair. In the cytoplasm, HMGB1 promotes mitochondrial autophagy and is involved in in cellular stress response. Once released into the extracellular, HMGB1 becomes an inflammatory factor that triggers inflammatory responses and a variety of immune responses. In addition, HMGB1 binding with the corresponding receptor can activate the downstream substrate to carry out several biological effects. Meanwhile, HMGB1 is involved in various signaling pathways, such as the HMGB1/RAGE pathway, HMGB1/NF-κB pathway, and HMGB1/JAK/STAT pathway, which ultimately promote inflammation. Moreover, HMGB1 may be involved in the pathogenesis of asthma by regulating downstream signaling pathways through corresponding receptors and mediates a number of signaling pathways in asthma, such as HMGB1/TLR4/NF-κB, HMGB1/RAGE, HMGB1/TGF-β, and so forth. Accordingly, HMGB1 emerges as a therapeutic target for asthma.
Collapse
Affiliation(s)
- Qianni Yang
- Department of Pulmonary and Critical Care MedicineFirst Affiliated Hospital, Kunming Medical UniversityKunmingChina
- 2021 Class 2 of AnesthesiologyKunming Medical UniversityKunmingChina
| | - Min Li
- Department of Pulmonary and Critical Care MedicineFirst Affiliated Hospital, Kunming Medical UniversityKunmingChina
| | - Yunjiao Hou
- Department of Pulmonary and Critical Care MedicineFirst Affiliated Hospital, Kunming Medical UniversityKunmingChina
| | - Huilin He
- Department of Pulmonary and Critical Care MedicineFirst Affiliated Hospital, Kunming Medical UniversityKunmingChina
| | - Shibo Sun
- Department of Pulmonary and Critical Care MedicineFirst Affiliated Hospital, Kunming Medical UniversityKunmingChina
| |
Collapse
|
35
|
He X, Sun Y, Lu X, Yang F, Li T, Deng C, Song J, Huang X. Assessment of the anti-inflammatory mechanism of quercetin 3,7-dirhamnoside using an integrated pharmacology strategy. Chem Biol Drug Des 2023; 102:1534-1552. [PMID: 37806949 DOI: 10.1111/cbdd.14346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/22/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023]
Abstract
Pouzolzia zeylanica (L.) Benn. is a Chinese herbal medicine widely used for its anti-inflammatory and pus-removal properties. To explore its potential anti-inflammatory mechanism, quercetin 3,7-dirhamnoside (QDR), the main flavonoid component of P. zeylanica (L.) Benn., was extracted and purified. The potential anti-inflammatory targets of QDR were predicted using network analysis. These potential targets were verified using molecular docking, molecular dynamics simulations, and in vitro experiments. Consequently, 342 potential anti-inflammatory QDR targets were identified. By analyzing the intersection between the protein-protein interaction and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, we identified several potential protein targets of QDR, including RAC-alpha serine/threonine-protein kinase (AKT1), Ras-related C3 botulinum toxin substrate 1 (RAC1), nitric oxide synthase 3 (NOS3), serine/threonine-protein kinase mTOR (mTOR), epidermal growth factor receptor (EGFR), growth factor receptor-bound protein 2 (GRB2), and endothelin-1 receptor (EDNRA). QDR has anti-inflammatory activity and regulates immune responses and apoptosis through chemokines, Phosphatidylinositol 3-kinase 3(PI3K)/AKT, cAMP, T-cell receptor, and Ras signaling pathways. Molecular docking analysis showed that QDR has good binding abilities with AKT1, mTOR, and NOS3. In addition, molecular dynamics simulations demonstrated that the protein-ligand complex systems formed between QDR and AKT1, mTOR, and NOS3 have high dynamic stability, and their protein-ligand complex systems possess strong binding ability. In RAW264.7 macrophages, QDR significantly inhibited lipopolysaccharides (LPS)-induced inducible nitric oxide synthase expression, nitric oxide (NO) release and the generation of proinflammatory cytokines IL-6, IL-1β, and TNF-α. QDR downregulated the expression of p-AKT1(Ser473)/AKT1 and p-mTOR (Ser2448)/mTOR, and upregulated the expression of NOS3, Rictor, and Raptor. This indicates that the anti-inflammatory mechanisms of QDR involve regulation of AKT1 and mTOR to prevent apoptosis and of NOS3 which leads to the release of endothelial NO. Thus, our study elucidated the potential anti-inflammatory mechanism of QDR, the main flavonoid found in P. zeylanica (L.) Benn.
Collapse
Affiliation(s)
- Xinqian He
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongzhi Sun
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaomeng Lu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fan Yang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ting Li
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changsheng Deng
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianping Song
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin'an Huang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
36
|
Dong L, Jiang N, Bai J, Li Y, Song Z, Liu X, Zhang C. Neuroprotective Effects of Dammarane Sapogenins Against lipopolysaccharide-induced Cognitive Impairment, Neuroinflammation and Synaptic Dysfunction. Neurochem Res 2023; 48:3525-3537. [PMID: 37490197 DOI: 10.1007/s11064-023-03997-7] [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: 02/23/2023] [Revised: 04/11/2023] [Accepted: 07/15/2023] [Indexed: 07/26/2023]
Abstract
Neuroinflammation is a critical driver in the pathogenesis and progression of neurodegenerative disorders. Dammarane sapogenins (DS), a deglycosylated product of ginsenoside, possess a variety of potent biological activities. The present study aimed to explore the neuroprotective effects of DS in a rat model of neuroinflammation induced by intracerebroventricular injection of lipopolysaccharide (LPS). Our study revealed that DS pretreatment effectively improved LPS-induced associative learning and memory impairments in the active avoidance response test and spatial learning and memory in Morris water maze test. DS also remarkably inhibited LPS-induced neuroinflammation by suppressing microglia overactivation, pro-inflammatory cytok ine release (TNF-α and IL-1β) and reducing neuronal loss in the CA1 and DG regions of the hippocampus. Importantly, pretreatment with DS reversed LPS-induced upregulation of HMGB1 and TLR4 and inhibited their downstream NF-κB signaling activation, as evidenced by increased IκBα and decreased p-NF-κB p65 levels. Furthermore, DS ameliorated LPS-induced synaptic dysfunction by decreasing MMP-9 and increasing NMDAR1 expression in the hippocampus. Taken together, this study suggests that DS could be a promising treatment for preventing cognitive impairments caused by neuroinflammation.
Collapse
Affiliation(s)
- Liming Dong
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Ning Jiang
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Jie Bai
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yiman Li
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Zhihui Song
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xinmin Liu
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Chao Zhang
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
37
|
He H, Zhu M, Lyu Y, Yuan Y, Qi Y. Effects and possible mechanisms of dexmedetomidine on post-operative cognitive dysfunction. Chin Med J (Engl) 2023; 136:2392-2394. [PMID: 36914953 PMCID: PMC10538912 DOI: 10.1097/cm9.0000000000002372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Indexed: 03/15/2023] Open
Affiliation(s)
- Huijuan He
- Department of Anesthesiology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang 315000, China
| | - Manhua Zhu
- Department of Anesthesiology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang 315000, China
| | - Yupeng Lyu
- Department of Anesthesiology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang 315000, China
| | - Yuan Yuan
- Department of Critical Care Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang 315000, China
| | - Yong Qi
- Department of Anesthesiology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang 315000, China
| |
Collapse
|
38
|
Hong QL, Ding YH, Chen JY, Shi SS, Liang RS, Tu XK. Schisandrin B Protects against Ischemic Brain Damage by Regulating PI3K/AKT Signaling in Rats. Chin J Integr Med 2023; 29:885-894. [PMID: 37357242 DOI: 10.1007/s11655-023-3596-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2022] [Indexed: 06/27/2023]
Abstract
OBJECTIVE To explore the effect and mechanism of schisandrin B (Sch B) in the treatment of cerebral ischemia in rats. METHODS The cerebral ischemia models were induced by middle cerebral artery occlusion (MCAO) and reperfusion. Sprague-Dawley rats were divided into 6 groups using a random number table, including sham, MCAO, MCAO+Sch B (50 mg/kg), MCAO+Sch B (100 mg/kg), MCAO+Sch B (100 mg/kg)+LY294002, and MCAO+Sch B (100 mg/kg)+wortmannin groups. The effects of Sch B on pathological indicators, including neurological deficit scores, cerebral infarct volume, and brain edema, were subsequently studied. Tissue apoptosis was identified by terminal transferase-mediated dUTP nick end-labeling (TUNEL) staining. The protein expressions involved in apoptosis, inflammation response and oxidative stress were examined by immunofluorescent staining, biochemical analysis and Western blot analysis, respectively. The effect of Sch B on phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling was also explored. RESULTS Sch B treatment decreased neurological deficit scores, cerebral water content, and infarct volume in MCAO rats (P<0.05 or P<0.01). Neuronal nuclei and TUNEL staining indicated that Sch B also reduced apoptosis in brain tissues, as well as the Bax/Bcl-2 ratio and caspase-3 expression (P<0.01). Sch B regulated the production of myeloperoxidase, malondialdehyde, nitric oxide and superoxide dismutase, as well as the release of cytokine interleukin (IL)-1 β and IL-18, in MCAO rats (P<0.05 or P<0.01). Sch B promoted the phosphorylation of PI3K and AKT. Blocking the PI3K/AKT signaling pathway with LY294002 or wortmannin reduced the protective effect of Sch B against cerebral ischemia (P<0.05 or P<0.01). CONCLUSIONS Sch B reduced apoptosis, inflammatory response, and oxidative stress of MCAO rats by modulating the PI3K/AKT pathway. Sch B had a potential for treating cerebral ischemia.
Collapse
Affiliation(s)
- Quan-Long Hong
- Department of Neurology, The First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yi-Hang Ding
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jing-Yi Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Song-Sheng Shi
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Ri-Sheng Liang
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Xian-Kun Tu
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| |
Collapse
|
39
|
Yimam M, Horm T, O’Neal A, Jiao P, Hong M, Brownell L, Jia Q, Lin M, Gauthier A, Wu J, Venkat Mateti K, Yang X, Dial K, Zefi S, Mantell LL. A Standardized Botanical Composition Mitigated Acute Inflammatory Lung Injury and Reduced Mortality through Extracellular HMGB1 Reduction. Molecules 2023; 28:6560. [PMID: 37764336 PMCID: PMC10538186 DOI: 10.3390/molecules28186560] [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: 07/29/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
HMGB1 is a key late inflammatory mediator upregulated during air-pollution-induced oxidative stress. Extracellular HMGB1 accumulation in the airways and lungs plays a significant role in the pathogenesis of inflammatory lung injury. Decreasing extracellular HMBG1 levels may restore innate immune cell functions to protect the lungs from harmful injuries. Current therapies for air-pollution-induced respiratory problems are inadequate. Dietary antioxidants from natural sources could serve as a frontline defense against air-pollution-induced oxidative stress and lung damage. Here, a standardized botanical antioxidant composition from Scutellaria baicalensis and Acacia catechu was evaluated for its efficacy in attenuating acute inflammatory lung injury and sepsis. Murine models of disorders, including hyperoxia-exposed, bacterial-challenged acute lung injury, LPS-induced sepsis, and LPS-induced acute inflammatory lung injury models were utilized. The effect of the botanical composition on phagocytic activity and HMGB1 release was assessed using hyperoxia-stressed cultured macrophages. Analyses, such as hematoxylin-eosin (HE) staining for lung tissue damage evaluation, ELISA for inflammatory cytokines and chemokines, Western blot analysis for proteins, including extracellular HMGB1, and bacterial counts in the lungs and airways, were performed. Statistically significant decreases in mortality (50%), proinflammatory cytokines (TNF-α, IL-1β, IL-6) and chemokines (CINC-3) in serum and bronchoalveolar lavage fluid (BALF), and increased bacterial clearance from airways and lungs; reduced airway total protein, and decreased extracellular HMGB1 were observed in in vivo studies. A statistically significant 75.9% reduction in the level of extracellular HMGB1 and an increase in phagocytosis were observed in cultured macrophages. The compilations of data in this report strongly suggest that the botanical composition could be indicated for oxidative-stress-induced lung damage protection, possibly through attenuation of increased extracellular HMGB1 accumulation.
Collapse
Affiliation(s)
- Mesfin Yimam
- Unigen Inc., 2121 South State Street, Suite #400, Tacoma, WA 98405, USA; (T.H.); (A.O.); (P.J.); (M.H.); (L.B.); (Q.J.)
| | - Teresa Horm
- Unigen Inc., 2121 South State Street, Suite #400, Tacoma, WA 98405, USA; (T.H.); (A.O.); (P.J.); (M.H.); (L.B.); (Q.J.)
| | - Alexandria O’Neal
- Unigen Inc., 2121 South State Street, Suite #400, Tacoma, WA 98405, USA; (T.H.); (A.O.); (P.J.); (M.H.); (L.B.); (Q.J.)
| | - Ping Jiao
- Unigen Inc., 2121 South State Street, Suite #400, Tacoma, WA 98405, USA; (T.H.); (A.O.); (P.J.); (M.H.); (L.B.); (Q.J.)
| | - Mei Hong
- Unigen Inc., 2121 South State Street, Suite #400, Tacoma, WA 98405, USA; (T.H.); (A.O.); (P.J.); (M.H.); (L.B.); (Q.J.)
| | - Lidia Brownell
- Unigen Inc., 2121 South State Street, Suite #400, Tacoma, WA 98405, USA; (T.H.); (A.O.); (P.J.); (M.H.); (L.B.); (Q.J.)
| | - Qi Jia
- Unigen Inc., 2121 South State Street, Suite #400, Tacoma, WA 98405, USA; (T.H.); (A.O.); (P.J.); (M.H.); (L.B.); (Q.J.)
| | - Mosi Lin
- College of Pharmacy and Health Sciences, St John’s University, Queens, NY 11439, USA; (M.L.); (A.G.); (J.W.); (K.V.M.); (X.Y.); (K.D.); (S.Z.); (L.L.M.)
| | - Alex Gauthier
- College of Pharmacy and Health Sciences, St John’s University, Queens, NY 11439, USA; (M.L.); (A.G.); (J.W.); (K.V.M.); (X.Y.); (K.D.); (S.Z.); (L.L.M.)
| | - Jiaqi Wu
- College of Pharmacy and Health Sciences, St John’s University, Queens, NY 11439, USA; (M.L.); (A.G.); (J.W.); (K.V.M.); (X.Y.); (K.D.); (S.Z.); (L.L.M.)
| | - Kranti Venkat Mateti
- College of Pharmacy and Health Sciences, St John’s University, Queens, NY 11439, USA; (M.L.); (A.G.); (J.W.); (K.V.M.); (X.Y.); (K.D.); (S.Z.); (L.L.M.)
| | - Xiaojian Yang
- College of Pharmacy and Health Sciences, St John’s University, Queens, NY 11439, USA; (M.L.); (A.G.); (J.W.); (K.V.M.); (X.Y.); (K.D.); (S.Z.); (L.L.M.)
| | - Katelyn Dial
- College of Pharmacy and Health Sciences, St John’s University, Queens, NY 11439, USA; (M.L.); (A.G.); (J.W.); (K.V.M.); (X.Y.); (K.D.); (S.Z.); (L.L.M.)
| | - Sidorela Zefi
- College of Pharmacy and Health Sciences, St John’s University, Queens, NY 11439, USA; (M.L.); (A.G.); (J.W.); (K.V.M.); (X.Y.); (K.D.); (S.Z.); (L.L.M.)
| | - Lin L. Mantell
- College of Pharmacy and Health Sciences, St John’s University, Queens, NY 11439, USA; (M.L.); (A.G.); (J.W.); (K.V.M.); (X.Y.); (K.D.); (S.Z.); (L.L.M.)
| |
Collapse
|
40
|
Maisat W, Han X, Koutsogiannaki S, Soriano SG, Yuki K. Differential effects of dexmedetomidine on Gram-positive and Gram-negative bacterial killing and phagocytosis. Int Immunopharmacol 2023; 120:110327. [PMID: 37201408 PMCID: PMC10330683 DOI: 10.1016/j.intimp.2023.110327] [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: 03/07/2023] [Revised: 05/01/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
Dexmedetomidine is a commonly used sedative in perioperative and intensive care settings with purported immunomodulatory properties. Since its effects on immune functions against infections have not been extensively studied, we tested the effects of dexmedetomidine on Gram-positive [Staphylococcus aureus and Enterococcus faecalis] and Gram-negative bacteria [Escherichia coli], and on effector functions of human monocytes THP-1 cells against them. We evaluated phagocytosis, reactive oxygen species (ROS) formation, and CD11b activation, and performed RNA sequencing analyses. Our study revealed that dexmedetomidine improved Gram-positive but mitigated Gram-negative bacterial phagocytosis and killing in THP-1 cells. The attenuation of Toll-like receptor 4 (TLR4) signaling by dexmedetomidine was previously reported. Thus, we tested TLR4 inhibitor TAK242. Similar to dexmedetomidine, TAK242 reduced E. coli phagocytosis but enhanced CD11b activation. The reduced TLR4 response potentially increases CD11b activation and ROS generation and subsequently enhances Gram-positive bacterial killing. Conversely, dexmedetomidine may inhibit the TLR4-signaling pathway and mitigate the alternative phagocytosis pathway induced by TLR4 activation through LPS-mediated Gram-negative bacteria, resulting in worsened bacterial loads. We also examined another α2 adrenergic agonist, xylazine. Because xylazine did not affect bacterial clearance, we proposed that dexmedetomidine may have an off-target effect on bacterial killing process, potentially involving crosstalk between CD11b and TLR4. Despite its potential to attenuate inflammation, we provide a novel insight into potential risks of dexmedetomidine use during Gram-negative infections, highlighting the differential effect of dexmedetomidine on Gram-positive and Gram-negative bacteria.
Collapse
Affiliation(s)
- Wiriya Maisat
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA; Department of Anesthesiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Xiaohui Han
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA
| | - Sophia Koutsogiannaki
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA; Department of Immunology, Harvard Medical School, Boston, USA
| | - Sulpicio G Soriano
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA
| | - Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA; Department of Immunology, Harvard Medical School, Boston, USA.
| |
Collapse
|
41
|
Yang FY, Zheng YT. Ultrasound Alleviates Lipopolysaccharide-Induced Colonic Damage. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38082993 DOI: 10.1109/embc40787.2023.10340959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic inflammation in the intestinal tract. There is currently no effective cure for IBD. The aim of this study was to evaluate the protective effect of low-intensity pulsed ultrasound (LIPUS) on lipopolysaccharide (LPS)-induced intestinal damage in a C57BL/6 mouse model. Colonic inflammation was induced by LPS injection (0.75 mg/kg, i.p.) for 7 days. A 1.0 MHz ultrasound transducer was used with a duty cycle of 5% and a repetition frequency of 1 Hz. LIPUS was applied to the abdominal region for 15 min/day from days 1 to 6 at both intensity of 0.5 W/cm2 or 1.0 W/cm2. Colonic samples were collected for macroscopic and westerm blotting analysis. First, the optimal dose of LPS for experiments was investigated. Our results demonstrated that LIPUS alleviates colonic damage by reducing colon shortening and increasing the levels of tight junction proteins such as Occludin and ZO-1. These findings show that abdominal LIPUS stimulation may be a novel therapeutic strategy for IBD through enhancement of tight junction protein levels and attenuation of colonic length.
Collapse
|
42
|
Zhang Z, Zheng Y, Chen N, Xu C, Deng J, Feng X, Liu W, Ma C, Chen J, Cai T, Xu Y, Wang S, Cao Y, Ge G, Jia C, Cao Y. San Huang Xiao Yan recipe modulates the HMGB1-mediated abnormal inflammatory microenvironment and ameliorates diabetic foot by activating the AMPK/Nrf2 signalling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154931. [PMID: 37364421 DOI: 10.1016/j.phymed.2023.154931] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/27/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Diabetic foot (DF) is one of the serious complications of diabetes and lacks of therapeutic drugs. Abnormal and chronic inflammation promoting foot infection and wound healing delay are the main pathogenesis of DF. The traditional prescription San Huang Xiao Yan Recipe (SHXY) has been used in the clinical treatment of DF for several decades as approved hospital experience prescription and showed remarkable therapeutic effect, but the mechanisms by which SHXY treats DF are still unclear. PURPOSE Objectives of this study were to investigate SHXY anti-inflammatory effect on DF and explore the molecular mechanism for SHXY. METHODS We detected the effects of SHXY on DF in C57 mouse and SD rat DF models. Animal blood glucose, weight and wound area were detected every week. Serum inflammatory factors were detected by ELISA. H&E and Masson's trichrome were used to observe tissue pathology. Single-cell sequencing data reanalysis revealed the role of M1 macrophages in DF. Venn analysis showed the co-target genes between DF M1 macrophages and compound-disease network pharmacology. Western blotting was used to explored target protein expression. Meanwhile, RAW264.7 cells were treated with drug-containing serum of SHXY to further unravel the roles of target proteins during high glucose-induced inflammation in vitro. The Nrf2 inhibitor ML385 was used on RAW 264.7 cells to further explore the relationship between Nrf2, AMPK and HMGB1. The main components of SHXY were analysed by HPLC. Finally, the treatment effect of SHXY on DF were detected on rat DF model. RESULTS In vivo, SHXY can ameliorate inflammatory, accelerate wound healing and upregulate expression of Nrf2, AMPK and downregulate of HMGB1. Bioinformatic analysis showed that M1 macrophages were the main inflammatory cell population in DF. Moreover, the Nrf2 downstream proteins HO-1 and HMGB1 were potential DF therapeutic targets for SHXY. In vitro, we also found that SHXY increased AMPK and Nrf2 protein levels and downregulated HMGB1 expression in RAW264.7 cells. Inhibiting the expression of Nrf2 impaired the inhibition effect of SHXY on HMGB1. SHXY promoted Nrf2 translocation into the nucleus and increased the phosphorylation of Nrf2. SHXY also inhibited HMGB1 extracelluar release under high glucose. In rat DF models, SHXY also exhibited significant anti-inflammatory effect. CONCLUSION The SHXY activated AMPK/Nrf2 pathway to suppress abnormal inflammation on DF via inhibiting HMGB1 expression. These findings provide novel insight into the mechanisms by which SHXY treats DF.
Collapse
Affiliation(s)
- Zhihui Zhang
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China.
| | - Yihan Zheng
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China
| | - Nan Chen
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China
| | - Chenqin Xu
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China
| | - Jie Deng
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China
| | - Xia Feng
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China
| | - Wei Liu
- Department of Pharmacy, The SATCM Third Grade Laboratory of Traditional Chinese Medicine Preparations, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Chao Ma
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China
| | - Jian Chen
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China
| | - Tongkai Cai
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China
| | - Yicheng Xu
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China
| | - Song Wang
- Pharmacy Department, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yemin Cao
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China
| | - Guangbo Ge
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Chenglin Jia
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China.
| | - Yongbing Cao
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 303 Changyang Road, Shanghai 200082, China.
| |
Collapse
|
43
|
Mercantepe F, Tumkaya L, Mercantepe T, Akyildiz K, Ciftel S, Yilmaz A. The Effects of Dexmedetomidine on Abdominal Aortic Occlusion-Induced Ovarian Injury via Oxidative Stress and Apoptosis. Cells Tissues Organs 2023; 212:554-566. [PMID: 37339613 DOI: 10.1159/000531613] [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: 03/28/2023] [Accepted: 06/14/2023] [Indexed: 06/22/2023] Open
Abstract
Ischemia/reperfusion (I/R) induced ovarian damage is caused by various diseases such as ovarian torsion, ovarian transplantation, cardiovascular surgery, sepsis, or intra-abdominal surgery. I/R-related oxidative damage can impair ovarian functions, from oocyte maturation to fertilization. This study investigated the effects of dexmedetomidine (DEX), which has been shown to exhibit antiapoptotic, anti-inflammatory, and antioxidant effects, on ovarian I/R injury. We designed four study groups: group 1 (n = 6): control group; group 2 (n = 6): only DEX group; group 3 (n = 6): I/R group; group 4 (n = 6): I/R + DEX group. Then, ovarian samples were taken and examined histologically and immunohistochemically, and tissue malondialdehyde (MDA) and glutathione (GSH) levels were measured. In the I/R group MDA levels, caspase-3, NF-κB/p65, 8-OHdG positivity, and follicular degeneration, edema, and inflammation were increased compared to the control group (p = 0.000). In addition, GSH levels were significantly decreased in the I/R group compared to the control group (p = 0.000). On the other hand, in the I/R + DEX treatment group MDA levels, caspase-3, NF-κB/p65, 8-OHdG positivity, follicular degeneration, edema, and inflammation findings were decreased than in the I/R group (p = 0.000, p = 0.005, p = 0.005, p = 0.001, p = 0.005, respectively). However, GSH levels increased significantly in the I/R + DEX treatment group compared to the I/R group (p = 0.000). DEX protects against ovarian I/R injury through antioxidation and by suppressing inflammation and apoptosis.
Collapse
Affiliation(s)
- Filiz Mercantepe
- Department of Endocrinology and Metabolism Diseases, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Levent Tumkaya
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Kerimali Akyildiz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Serpil Ciftel
- Department of Endocrinology and Metabolism Diseases, Erzurum Regional Education and Research Hospital, Erzurum, Turkey
| | - Adnan Yilmaz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
44
|
Li YL, Qin SY, Li Q, Song SJ, Xiao W, Yao GD. Jinzhen Oral Liquid alleviates lipopolysaccharide-induced acute lung injury through modulating TLR4/MyD88/NF-κB pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154744. [PMID: 36934667 DOI: 10.1016/j.phymed.2023.154744] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 02/12/2023] [Accepted: 03/04/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Acute lung injury (ALI) has the attribution of excessive inflammation of the lung. Jinzhen oral liquid (JO), a famous Chinese recipe used to treat ALI, has a favorable therapeutic effect on ALI. However, its anti-inflammatory mechanism has not been extensively studied. PURPOSE This study was to elucidate the effects of JO on lipopolysaccharide (LPS)-induced ALI and its molecular mechanism. METHODS An ALI model was established by intratracheal instillation of LPS (2 mg/50 μl). The open field experiment was carried out to explore the spontaneous movement and exploratory behavior of ALI mice. Cytokines levels concentrations (IL-6, IL-10 and TNF-α) were determined by enzyme-linked immunosorbent assay (ELISA). Network pharmacology was used to predict the mechanism of JO against ALI. Immunofluorescence, co-immunoprecipitation, fluorescence resonance energy transfer (FRET), Western blot and RT-PCR were used to verify the molecular mechanisms of JO. RESULTS The in vivo results suggested that JO (1, 2, 4 g/kg) dose-dependently improved the exercise performance of mice and reduced the lung W/D weight ratio as well as the production of IL-6 and TNF-α, but increased the release of IL-10 in the ALI group. The network pharmacological analysis demonstrated that the Toll-like receptor (TLR) pathway might be the fundamental action mechanisms of JO against ALI. Immunofluorescence staining and co-immunoprecipitation analysis showed that JO decreased the expression levels of TLR4 and MyD88 and reduced their interaction in the lung tissue of ALI mice. Meanwhile, JO decreased nuclear translocation and phosphorylation of NF-κB P65. The results from cellular experiments were in line with those in vivo. The FRET experiment also confirmed that JO disturbed the interaction of TLR4 and MyD88. Subsequently, we also found that the six indicative components of JO have the similar therapeutic effect as JO. CONCLUSIONS In summary, we suggested that JO suppressed the TLR4/MyD88/NF-κB signaling pathway, thus inhibiting LPS-induced ALI in vitro and in vivo. The clarified mechanism provided an important theoretical basis and a novel treatment strategy for the ALI treatment of JO.
Collapse
Affiliation(s)
- Ya-Ling Li
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shu-Yan Qin
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Qian Li
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, China.
| | - Guo-Dong Yao
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, China.
| |
Collapse
|
45
|
Ye S, Mahmood DFD, Ma F, Leng L, Bucala R, Vera PL. Urothelial Oxidative Stress and ERK Activation Mediate HMGB1-Induced Bladder Pain. Cells 2023; 12:1440. [PMID: 37408274 PMCID: PMC10217556 DOI: 10.3390/cells12101440] [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: 04/17/2023] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 07/07/2023] Open
Abstract
Activation of intravesical protease activated receptors-4 (PAR4) results in bladder pain through the release of urothelial macrophage migration inhibitory factor (MIF) and high mobility group box-1 (HMGB1). We aimed to identify HMGB1 downstream signaling events at the bladder that mediate HMGB1-induced bladder pain in MIF-deficient mice to exclude any MIF-related effects. We studied whether oxidative stress and ERK activation are involved by examining bladder tissue in mice treated with intravesical disulfide HMGB1 for 1 h and analyzed with Western blot and immunohistochemistry. HMGB1 intravesical treatment increased urothelium 4HNE and phospho-ERK1/2 staining, suggesting that HMGB1 increased urothelial oxidative stress and ERK activation. Furthermore, we examined the functional roles of these events. We evaluated lower abdominal mechanical thresholds (an index of bladder pain) before and 24 h after intravesical PAR4 or disulfide HMGB1. Intravesical pre-treatments (10 min prior) included: N-acetylcysteine amide (NACA, reactive oxygen species scavenger) and FR180204 (FR, selective ERK1/2 inhibitor). Awake micturition parameters (voided volume; frequency) were assessed at 24 h after treatment. Bladders were collected for histology at the end of the experiment. Pre-treatment with NACA or FR significantly prevented HMGB1-induced bladder pain. No significant effects were noted on micturition volume, frequency, inflammation, or edema. Thus, HMGB1 activates downstream urothelial oxidative stress production and ERK1/2 activation to mediate bladder pain. Further dissection of HMGB1 downstream signaling pathway may lead to novel potential therapeutic strategies to treat bladder pain.
Collapse
Affiliation(s)
- Shaojing Ye
- Lexington VA Health Care System, Research & Development, Lexington, KY 40502, USA
| | - Dlovan F. D. Mahmood
- Lexington VA Health Care System, Research & Development, Lexington, KY 40502, USA
| | - Fei Ma
- Lexington VA Health Care System, Research & Development, Lexington, KY 40502, USA
| | - Lin Leng
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | - Richard Bucala
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | - Pedro L. Vera
- Lexington VA Health Care System, Research & Development, Lexington, KY 40502, USA
- Department of Physiology, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
46
|
Zhou Y, Jin T, Gao M, Luo Z, Mutahir S, Shi C, Xie T, Lin L, Xu J, Liao Y, Chen M, Deng H, Zheng M, Shan J. Aqueous extract of Platycodon grandiflorus attenuates lipopolysaccharide-induced apoptosis and inflammatory cell infiltration in mouse lungs by inhibiting PI3K/Akt signaling. Chin Med 2023; 18:36. [PMID: 37016413 PMCID: PMC10071731 DOI: 10.1186/s13020-023-00721-z] [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: 11/22/2022] [Accepted: 02/06/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI), an acute inflammatory lung disease, can cause a rapid inflammatory response in clinic, which endangers the patient's life. The components of platycodon grandiflorum, such as platycodins have a wide range of pharmacological activities such as expectorant, anti-apoptotic, anti-inflammatory, anti-tumor and anti-oxidant properties, and can be used for improving human immunity. Previous studies have shown that aqueous extract of platycodon grandiflorum (PAE) has a certain protective effect on ALI, but the main pharmacodynamic components and the mechanism of action are not clear. METHODS The anti-inflammatory properties of PAE were studied using the lipopolysaccharide (LPS)-induced ALI animal model. Hematoxylin and eosin stains were used to assess the degree of acute lung damage. Changes in RNA levels of pro-inflammatory cytokines in the lungs were measured using quantitative RT-qPCR. The potential molecular mechanism of PAE preventing ALI was predicted by lipidomics and network pharmacology. To examine the anti-apoptotic effects of PAE, TdT-mediated dUTP nick-end labelling (TUNEL) was employed to determine apoptosis-related variables. The amounts of critical pathway proteins and apoptosis-related proteins were measured using Western blotting. RESULTS Twenty-six chemical components from the PAE were identified, and their related pathways were obtained by the network pharmacology. Combined with the analysis of network pharmacology and literature, it was found that the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (AKT) signaling pathway is related to ALI. The results of lipidomics show that PAE alleviates ALI via regulating lung lipids especially phosphatidylinositol (PI). Finally, the methods of molecular biology were used to verify the mechanism of PAE. It can be found that PAE attenuates the inflammatory response to ALI by inhibiting apoptosis through PI3K/Akt signaling pathway. CONCLUSION The study revealed that the PAE attenuates lipopolysaccharide-induced apoptosis and inflammatory cell infiltration in mouse lungs by inhibiting PI3K/Akt signaling. Furthermore, our findings provide a novel strategy for the application of PAE as a potential agent for preventing patients with ALI.
Collapse
Affiliation(s)
- Yang Zhou
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Wuhu Fanchang District People's Hospital, Wuhu, 241200, China
| | - Tianzi Jin
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mingtong Gao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zichen Luo
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Sadaf Mutahir
- Department of Chemistry, University of Sialkot, Sialkot, 51300, Pakistan
| | - Chen Shi
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tong Xie
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lili Lin
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jianya Xu
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yingzhao Liao
- Department of Pediatrics, Shenzhen Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Shenzhen, 518033, China
| | - Ming Chen
- Jiangsu Suzhong Pharmaceutical Research Institute Co. Ltd, Nanjing, 210031, China
| | - Haishan Deng
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Min Zheng
- Department of Pediatrics, Shenzhen Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Shenzhen, 518033, China.
| | - Jinjun Shan
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
47
|
Zhang X, Hu Y, Yang T, Qian X, Hu W, Li G. Penazaphilones J–L, Three New Hydrophilic Azaphilone Pigments from Penicillium sclerotiorum cib-411 and Their Anti-Inflammatory Activity. Molecules 2023; 28:molecules28073146. [PMID: 37049911 PMCID: PMC10095951 DOI: 10.3390/molecules28073146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Penazaphilones J–L (1–3), three new hydrophilic azaphilone pigments, as well as six known compounds, were discovered from the filamentous fungus Penicillium sclerotiorum cib-411. Compounds 1–3 were structurally elucidated by the detailed interpretation of their 1D and 2D NMR spectroscopic data. Compound 1 is an unprecedented hybrid of an azaphilone and a glycerophosphate choline. Compounds 2 and 3 each contain an intact amino acid moiety. The bioassay showed that compound 3 exhibited significant anti-inflammatory activity. Concretely, compound 3 significantly suppressed the NO production, the expression levels of COX-2, IL-6, IL-1β, and iNOS mRNA in LPS-stimulated RAW264.7 cells. Moreover, treatment of compound 3 prevented the translocation of NF-κB through inhibiting the phosphorylation of PI3K, PDK1, Akt, and GSK-3β. Thus, the inhibition of compound 3 against LPS-induced inflammation should rely on its inactivation on NF-κB.
Collapse
Affiliation(s)
- Xia Zhang
- Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Yeye Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Tao Yang
- Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Xueqing Qian
- Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Weicheng Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Guoyou Li
- Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| |
Collapse
|
48
|
Tian Y, Zhu CL, Li P, Li HR, Liu Q, Deng XM, Wang JF. Nicotinamide Mononucleotide Attenuates LPS-Induced Acute Lung Injury With Anti-Inflammatory, Anti-Oxidative and Anti-Apoptotic Effects. J Surg Res 2023; 283:9-18. [PMID: 36347171 DOI: 10.1016/j.jss.2022.09.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 08/23/2022] [Accepted: 09/18/2022] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Nicotinamide mononucleotide (NMN) is a nucleotide that is commonly recognized for its role as an intermediate of nicotinamide adenine dinucleotide (NAD+) biosynthesis with multiple pharmacological effects. The purpose of this study was to evaluate the protective effect of nicotinamide mononucleotide (NMN) against lipopolysaccharide (LPS)-induced acute lung injury (ALI). METHODS We investigated the effect of NMN on ALI-induced inflammatory response, oxidative stress, and cell apoptosis. The ALI mouse model was performed by injecting LPS intratracheally at a dose of 10 mg/kg in 50 μL saline. Flow cytometry was used to detect neutrophil infiltration in bronchoalveolar lavage fluid (BALF), and ELISA was used to detect the contents of inflammatory cytokines TNF-α, IL-1β and IL-6 in BALF. Oxidative stress was evaluated by determining the superoxide dismutase (SOD) activity and malondialdehyde (MDA) content in lung tissue. ROS formation was analyzed by immunofluorescence. Western blotting was performed to detect apoptotic levels and p38MAPK/NF-κB phosphorylation levels in lung tissue. RESULTS In the ALI mouse model, NMN showed a significant therapeutic effect compared to the LPS group. NMN attenuated the pathological damage and cell apoptosis in lung tissue, decreased the levels of TNF-α, IL-1β, and IL-6 in BALF, and reduced the number of total cells and neutrophils in BALF. In addition, NMN attenuated the LPS-induced elevation of dry-to-wet ratio, MDA content, p38 MAPK and p65 NF-κB phosphorylation levels, and the SOD activity was increased by NMN treatment. CONCLUSIONS In conclusion, the present study showed that NMN exerted a protective effect on LPS-induced ALI with anti-inflammatory, antioxidative, and antiapoptotic effects.
Collapse
Affiliation(s)
- Ye Tian
- Department of Anesthesiology, The Sixth Medical Centre of General Hospital of PLA, Beijing, China
| | - Cheng-Long Zhu
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Peng Li
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hui-Ru Li
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qiang Liu
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Ming Deng
- Changhai Hospital, Naval Medical University, Shanghai, China.
| | - Jia-Feng Wang
- Changhai Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
49
|
Li J, Wang W, Yuan Y, Cui X, Bian H, Wen H, Zhang X, Yu H, Wu H. Pinellia ternata lectin induces inflammation through TLR4 receptor and mediates PI3K/Akt/mTOR axis to regulate NF-κB signaling pathway. Toxicology 2023; 486:153430. [PMID: 36669722 DOI: 10.1016/j.tox.2023.153430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/19/2023]
Abstract
Pinellia ternata, a widely used traditional Chinese medicine, contains a strong mucosal irritant that is connected with Pinellia ternata lectin (PTL) in its tubers. The purpose of this study was to explore the mechanisms by which PTL induces inflammation. We found that in RAW264.7 cells, PTL activated the PI3K/Akt/mTOR and NF-κB pathways, which resulted in the release of proinflammatory cytokines. Flow cytometry and laser confocal microscopy analysis showed that FITC-labeled PTL bound to the macrophages' surface. Based on kinetic analyses and protein-protein docking simulations, PTL was shown to bind toll-like receptor 4 (TLR4).it was demonstrated that PTL binds highly to Toll-like receptor 4 (TLR4). TLR4 knock-down or knockout resulted in a decrease in both cytokine release and PI3K/Akt/mTOR and NF-κB pathway activation in PTL-stimulated macrophages or mice. RNA-seq analysis showed that genes involved in the PI3K/Akt/mTOR signaling pathway were strongly upregulated in response to PTL stimulation, confirming that the PI3K/Akt/mTOR pathway is linked to the inflammatory effect of PTL in RAW264.7 cells. These findings reveal that PTL can mediate inflammation through TLR4 and activating the PI3K/Akt/mTOR to regulate NF-κB signaling pathways.
Collapse
Affiliation(s)
- Jinfei Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei Wang
- Department of Chinese Medicine and Pharmacy, School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Yuan Yuan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaobing Cui
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Huimin Bian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hongmei Wen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xingde Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hongli Yu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing 210023, China; State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Hao Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing 210023, China; State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
50
|
Bhat AA, Gupta G, Singh SK, Yadav HK, Saini M, Salfi R, Singh SK, Dua K. Nanotechnology-based advancements in NF-κB pathway inhibition for the treatment of inflammatory lung diseases. Nanomedicine (Lond) 2023; 17:2209-2213. [PMID: 36802843 DOI: 10.2217/nnm-2022-0220] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, 302017, Jaipur, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, 302017, Jaipur, India.,Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 602105, India.,Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, 248007, India
| | - Santosh Kumar Singh
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, 302017, Jaipur, India
| | - Hemant Ks Yadav
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, 302017, Jaipur, India
| | - Mahendra Saini
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, 302017, Jaipur, India
| | - Roshan Salfi
- Deccan School of Pharmacy, Darussalam, Aghapura, Nampally, Hyderabad, 500001, India.,Yenepoya Pharmacy College & Research Centre, Yenepoya University, Mangalore, Karnataka, 575023, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India.,Faculty of Health, Australian Research Center in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Center in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| |
Collapse
|