1
|
Ma J, Wang Z, Sun Y, Zheng R, Tan H, Zhang H, Jin Z, Wu Y, Sun Z. Phillyrin: A potential therapeutic agent for osteoarthritis via modulation of NF-κB and Nrf2 signaling pathways. Int Immunopharmacol 2024; 141:112960. [PMID: 39159565 DOI: 10.1016/j.intimp.2024.112960] [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: 02/26/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
Osteoarthritis (OA) is the predominant cause of disability among elderly people worldwide and is characterized by cartilage degeneration and excessive bone formation. Phillyrin, derived from forsythia, is a key extract renowned for its pronounced antibacterial and anti-inflammatory effects. Forsythia, deeply integrated into traditional Oriental medicine, has historically been utilized for its various pharmacological effects, including antibacterial, anti-inflammatory, and hepato-protective properties. Nevertheless, the anti-inflammatory impact of phillyrin on the progression of osteoarthritis remains enigmatic. The objective of this research was to assess the anti-inflammatory and anti-aging properties of phillyrin in mouse chondrocytes induced by IL-1β, as well as to elucidate the fundamental mechanisms underlying the phenomenon at play. Additionally, the investigation extends to observing the impact of phillyrin by establishing a murine osteoarthritic model. The ultimate goal was to identify phillyrin as a potential antiosteoarthritic agent. This investigation employs a multifaceted approach. Initially, key action targets of phillyrin, along with its probable action pathways, were identified by molecular docking and network pharmacological techniques. These findings were subsequently confirmed through both in vivo and in vitro studies. Network pharmacological analysis revealed NFE2L2 (NRF2), NFKB1, TLR4, and SERPING1 as pivotal candidate targets for the treatment of osteoarthritis with phillyrin. Molecular docking revealed hydrogen bond interactions between phillyrin and Arg415, Arg483, Ser508, and Asn387 on the Nrf2 receptor, while electrostatic interactions occurred with residues Arg415 and Arg380. Experiments conducted in vitro indicated that phillyrin preconditioning hindered the IL-1β-induced expression of proinflammatory factors which included TNF-α, COX-2, IL-6, and iNOS. Furthermore, phillyrin counteracts the IL-1β-induced degradation of aggrecan and collagen II within the extracellular matrix (ECM). This protective action is caused by the inhibition of the NF-κB pathway by phillyrin. Additionally, the mitigation of chondrocyte aging by phillyrin was observed. Our investigation revealed that phillyrin mitigates inflammation and counteracts cartilage degeneration in osteoarthritis (OA) patients by suppressing inflammation in chondrocytes and impeding aging through suppression of the NF-κB pathway.
Collapse
Affiliation(s)
- Jiawei Ma
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325088, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou 325035, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Ze Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325088, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou 325035, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Yun Sun
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325088, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou 325035, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Rukang Zheng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325088, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou 325035, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Hongye Tan
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325088, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou 325035, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Hanwen Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325088, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou 325035, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Zebin Jin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325088, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou 325035, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Yaosen Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325088, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou 325035, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China.
| | - Zeming Sun
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325088, Zhejiang Province, China; Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou 325035, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China.
| |
Collapse
|
2
|
Qi LFR, Liu Y, Liu S, Xiang L, Liu Z, Liu Q, Zhao JQ, Xu X. Phillyrin promotes autophagosome formation in A53T-αSyn-induced Parkinson's disease model via modulation of REEP1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155952. [PMID: 39178680 DOI: 10.1016/j.phymed.2024.155952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/30/2024] [Accepted: 08/10/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND The preservation of autophagosome formation presents a promising strategy for tackling neurological disorders, such as Parkinson's disease (PD). Mitochondria-associated endoplasmic reticulum (ER) membranes (MAM) serve not only as a focal point linked to various neurological disorders but also play a crucial role in supporting the biogenesis of autophagosomes. PURPOSE This investigation aimed to elucidate the neuroprotective properties of phillyrin against PD and its underlying mechanisms in promoting autophagosome formation. METHODS ER and mitochondria co-localization was assessed via fluorescent staining. Annexin V-fluorescein isothiocyanate (FITC) fluorescence was employed to quantify accessible cardiolipin (CL) on mitochondrial surfaces. The levels of CL within the MAM fraction of SH-SY5Y cells were evaluated using a CL probe assay kit. Monodansylcadaverine staining was utilized to detect autophagosome formation in SH-SY5Y cells. In an A53T-alpha-synuclein (αSyn)-induced PD mouse model, the anti-PD properties of phillyrin were assessed using open field, pole climbing, and rotarod tests, as well as immunohistochemistry staining of TH+ neurons in the brain sections. RESULTS In A53T-αSyn-treated SH-SY5Y cells, phillyrin facilitated autophagosome formation by suppressing CL externalization and restoring MAM integrity. Phillyrin enhanced the localization of receptor expression-enhancing protein 1 (REEP1) within MAM and mitochondria, bolstering MAM formation. Increased REEP1 levels in mitochondria, attributed to phillyrin, enhanced the interaction between REEP1 and NDPK-D, thereby reducing CL externalization. Furthermore, phillyrin exhibited a dose-dependent enhancement of motor function in mice, accompanied by an increase in the abundance of dopaminergic neurons within the substantia nigra. CONCLUSIONS These findings illuminate phillyrin's ability to enhance MAM formation through upregulation of REEP1 expression within MAM, while concurrently attenuating CL externalization via the REEP1-NDPK-D interaction. These mechanisms bolster autophagosome biogenesis, offering resilience against A53T-αSyn-induced PD. Thus, our study advances the understanding of phillyrin's complex mechanisms and underscores its potential as a therapeutic approach for PD, opening new avenues in natural product pharmacology.
Collapse
Affiliation(s)
- Li-Feng-Rong Qi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.
| | - Yuci Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shuai Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lin Xiang
- Department of Translational Medicine Research Institute, Jiangsu Yifengrong Biotechnology Co., Ltd., Nanjing, Jiangsu, China
| | - Zhiyuan Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qingling Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Jin-Quan Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Xiaojun Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health) & School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Pharmacy, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China; Center for Innovative Traditional Chinese Medicine Target and New Drug Research, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, China.
| |
Collapse
|
3
|
Xu X, Lu Y, Shen R, Fang L. Phillyrin inhibits oxidative stress and neutrophil extracellular trap formation through the KEAP1/NRF2 pathway in gouty arthritis. Immunol Res 2024:10.1007/s12026-024-09548-8. [PMID: 39436625 DOI: 10.1007/s12026-024-09548-8] [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: 07/13/2024] [Accepted: 09/25/2024] [Indexed: 10/23/2024]
Abstract
Gouty arthritis (GA) is an inflammatory disorder characterized by deposition of monosodium urate (MSU) crystal in joints. Phillyrin, a natural compound with anti-inflammatory properties, shows promise in mitigating inflammatory responses. This study investigates the therapeutic potential of phillyrin in GA and explores its mechanisms of action. GA was induced in mice via intraarticular MSU injection, and joint inflammation, inflammatory cell infiltration, and their level in serum/tissue were assessed. Key proteins in the NF-κB and NLRP3 pathways were examined using western blot analysis. The impact of phillyrin on oxidative stress, neutrophil extracellular trap (NET) formation, and neutrophil accumulation was evaluated by measuring CD11b + Ly6G + cells, MPO, CitH3, extracellular DNA ratio, and oxidative stress markers. In vitro studies assessed the effects of phillyrin on oxidative stress, cell viability, cytokine production, and NET formation in MSU-treated neutrophils. The KEAP1/NRF2 pathway's role was analyzed using ML385, an NRF2 inhibitor. Phillyrin significantly reversed MSU-induced ankle swelling and inflammatory cell infiltration in joint tissues. It suppressed pro-inflammatory cytokines and proteins in the NF-κB and NLRP3 pathways. Phillyrin reduced neutrophil infiltration, evidenced by lower MPO activity and NET formation, marked by reduced CitH3 expression. In vitro, phillyrin inhibited inflammatory marker expression and NET formation without affecting cell viability. It also restored antioxidant enzyme levels and reduced ROS production, regulating the KEAP1/NRF2 pathway, enhancing NRF2 expression and stability. These effects were reversed by NRF2 inhibition with ML385. Phillyrin alleviates GA by reducing joint inflammation, inhibiting NET formation, and suppressing oxidative stress through NRF2 modulation.
Collapse
Affiliation(s)
- Xiangfeng Xu
- Department of Rheumatology and Immunology, Zhoushan Hospital, Wenzhou Medical University, No.739 Dingshen Road Lincheng New District, Zhoushan, 316021, Zhejiang, China
| | - Yao Lu
- Department of Rheumatology and Immunology, Zhoushan Hospital, Wenzhou Medical University, No.739 Dingshen Road Lincheng New District, Zhoushan, 316021, Zhejiang, China
| | - Rong Shen
- Department of Geriatrics, Yueyang Hospital of Integrated Traditional Chinese and Wesstern Medicine, Shanghai University of Traditional Chinese Medicine, Hongkou District, No. 110 Ganhe Road, Shanghai, 200437, China.
| | - Li Fang
- Department of Rheumatology and Immunology, Zhoushan Hospital, Wenzhou Medical University, No.739 Dingshen Road Lincheng New District, Zhoushan, 316021, Zhejiang, China.
| |
Collapse
|
4
|
Zhang J, Tian L, Cao G, Yin Z, Wang S, Zhao C, Yang H. AnGong NiuHuang (AGNH) pill attenuated traumatic brain injury through regulating NF-κB/Nlrp3 axis and glycerophospholipid metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155798. [PMID: 38936259 DOI: 10.1016/j.phymed.2024.155798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/20/2024] [Accepted: 06/02/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Traumatic brain injury (TBI), especially neuroinflammation after TBI persists for a long time and causes significant neurodegenerative pathologies and neuropsychiatric problems. PURPOSE In this study, the neuroprotective effect of AnGong NiuHuang (AGNH) on TBI was investigated and the mechanism was revealed by integrating multiple omics. METHODS The rats with TBI were administrated with AGNH for 5 consecutive days and the effect was evaluated by using modified neurologic severity score (mNSS), brain edema, H&E staining, Nissl staining and TUNEL staining. The mechanism was revealed by using RNA sequencing (RNA-seq) and metabolomic analysis. The inflammatory factors, apoptosis-related proteins and identified vital targets were validated by enzyme-linked immunosorbent assay, western blotting and immunofluorescence staining. RESULTS Administration of AGNH decreased mNSS, brain edema, brain structure damage, but increased Nissl body density in the rats with TBI. Additionally, AGNH reduced IL-1β, IL-17A, TNF-α, MMP9, MCP-1, IL-6, Bax and TUNEL staining,but elevated Bcl2 level. Integrating transcriptomic analysis and metabolomic analysis identified vital targets and critical metabolic pathways. Importantly, AGNH treatment reduced the expression of TLR4, MYD88, NLRP3, BTK, IL-18 and Caspase 1 as well as glycerophospholipid metabolism-related protein AGPAT2 and PLA2G2D, and decreased the nuclear translocation of NF-κB p65 in the brain of TBI rats. Additionally, AGNH increased phosphatidylcholine (PC), phosphatidylglycerol (PG), phosphatidylserine (PS), phosphatidylethanolamine (PE), but decreased 1-acyl-sn-glycero-3-phosphocholine (LysoPC) in the metabolic pathway of glycerophospholipid metabolism. CONCLUSION Taken together, AGNH inhibited NF-κB/NLRP3 axis to suppress neuroinflammation, cell apoptosis and pyroptosis, and improved metabolic pathways of glycerophospholipid metabolism after TBI.
Collapse
Affiliation(s)
- Jingjing Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Chinese Institute for Brain Research, Beijing 102206, China.
| | - Liangliang Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Guangzhao Cao
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhiru Yin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shicong Wang
- Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou, Fujian, 363000, China
| | - Chen Zhao
- Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou, Fujian, 363000, China
| | - Hongjun Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
5
|
Pearson A, Koprivica M, Eisenbaum M, Ortiz C, Browning M, Vincennie T, Tinsley C, Mullan M, Crawford F, Ojo J. PPARγ activation ameliorates cognitive impairment and chronic microglial activation in the aftermath of r-mTBI. J Neuroinflammation 2024; 21:194. [PMID: 39097742 PMCID: PMC11297749 DOI: 10.1186/s12974-024-03173-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/12/2024] [Indexed: 08/05/2024] Open
Abstract
Chronic neuroinflammation and microglial activation are key mediators of the secondary injury cascades and cognitive impairment that follow exposure to repetitive mild traumatic brain injury (r-mTBI). Peroxisome proliferator-activated receptor-γ (PPARγ) is expressed on microglia and brain resident myeloid cell types and their signaling plays a major anti-inflammatory role in modulating microglial responses. At chronic timepoints following injury, constitutive PPARγ signaling is thought to be dysregulated, thus releasing the inhibitory brakes on chronically activated microglia. Increasing evidence suggests that thiazolidinediones (TZDs), a class of compounds approved from the treatment of diabetes mellitus, effectively reduce neuroinflammation and chronic microglial activation by activating the peroxisome proliferator-activated receptor-γ (PPARγ). The present study used a closed-head r-mTBI model to investigate the influence of the TZD Pioglitazone on cognitive function and neuroinflammation in the aftermath of r-mTBI exposure. We revealed that Pioglitazone treatment attenuated spatial learning and memory impairments at 6 months post-injury and reduced the expression of reactive microglia and astrocyte markers in the cortex, hippocampus, and corpus callosum. We then examined whether Pioglitazone treatment altered inflammatory signaling mechanisms in isolated microglia and confirmed downregulation of proinflammatory transcription factors and cytokine levels. To further investigate microglial-specific mechanisms underlying PPARγ-mediated neuroprotection, we generated a novel tamoxifen-inducible microglial-specific PPARγ overexpression mouse line and examined its influence on microglial phenotype following injury. Using RNA sequencing, we revealed that PPARγ overexpression ameliorates microglial activation, promotes the activation of pathways associated with wound healing and tissue repair (such as: IL10, IL4 and NGF pathways), and inhibits the adoption of a disease-associated microglia-like (DAM-like) phenotype. This study provides insight into the role of PPARγ as a critical regulator of the neuroinflammatory cascade that follows r-mTBI in mice and demonstrates that the use of PPARγ agonists such as Pioglitazone and newer generation TZDs hold strong therapeutic potential to prevent the chronic neurodegenerative sequelae of r-mTBI.
Collapse
Affiliation(s)
- Andrew Pearson
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA.
- The Open University, Walton Hall, Kents Hill, Milton Keynes, MK7 6AA, UK.
| | - Milica Koprivica
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Max Eisenbaum
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Walton Hall, Kents Hill, Milton Keynes, MK7 6AA, UK
| | - Camila Ortiz
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Walton Hall, Kents Hill, Milton Keynes, MK7 6AA, UK
| | | | - Tessa Vincennie
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Cooper Tinsley
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Michael Mullan
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Fiona Crawford
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Walton Hall, Kents Hill, Milton Keynes, MK7 6AA, UK
- James A. Haley Veterans' Hospital, 13000 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Joseph Ojo
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Walton Hall, Kents Hill, Milton Keynes, MK7 6AA, UK
- James A. Haley Veterans' Hospital, 13000 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| |
Collapse
|
6
|
Zhang M, Han X, Yan L, Fu Y, Kou H, Shang C, Wang J, Liu H, Jiang C, Wang J, Cheng T. Inflammatory response in traumatic brain and spinal cord injury: The role of XCL1-XCR1 axis and T cells. CNS Neurosci Ther 2024; 30:e14781. [PMID: 38887195 PMCID: PMC11183917 DOI: 10.1111/cns.14781] [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: 01/12/2024] [Revised: 04/29/2024] [Accepted: 05/11/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) and spinal cord injury (SCI) are acquired injuries to the central nervous system (CNS) caused by external forces that cause temporary or permanent sensory and motor impairments and the potential for long-term disability or even death. These conditions currently lack effective treatments and impose substantial physical, social, and economic burdens on millions of people and families worldwide. TBI and SCI involve intricate pathological mechanisms, and the inflammatory response contributes significantly to secondary injury in TBI and SCI. It plays a crucial role in prolonging the post-CNS trauma period and becomes a focal point for a potential therapeutic intervention. Previous research on the inflammatory response has traditionally concentrated on glial cells, such as astrocytes and microglia. However, increasing evidence highlights the crucial involvement of lymphocytes in the inflammatory response to CNS injury, particularly CD8+ T cells and NK cells, along with their downstream XCL1-XCR1 axis. OBJECTIVE This review aims to provide an overview of the role of the XCL1-XCR1 axis and the T-cell response in inflammation caused by TBI and SCI and identify potential targets for therapy. METHODS We conducted a comprehensive search of PubMed and Web of Science using relevant keywords related to the XCL1-XCR1 axis, T-cell response, TBI, and SCI. RESULTS This study examines the upstream and downstream pathways involved in inflammation caused by TBI and SCI, including interleukin-15 (IL-15), interleukin-12 (IL-12), CD8+ T cells, CD4+ T cells, NK cells, XCL1, XCR1+ dendritic cells, interferon-gamma (IFN-γ), helper T0 cells (Th0 cells), helper T1 cells (Th1 cells), and helper T17 cells (Th17 cells). We describe their proinflammatory effect in TBI and SCI. CONCLUSIONS The findings suggest that the XCL1-XCR1 axis and the T-cell response have great potential for preclinical investigations and treatments for TBI and SCI.
Collapse
Affiliation(s)
- Mingkang Zhang
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xiaonan Han
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Liyan Yan
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yikun Fu
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hongwei Kou
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Chunfeng Shang
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Hongjian Liu
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Chao Jiang
- Department of NeurologyPeople's Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Tian Cheng
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
7
|
Zhou C, Yan L, Xu J, Hamezah HS, Wang T, Du F, Tong X, Han R. Phillyrin: an adipose triglyceride lipase inhibitor supported by molecular docking, dynamics simulation, and pharmacological validation. J Mol Model 2024; 30:68. [PMID: 38347278 DOI: 10.1007/s00894-024-05875-7] [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: 08/13/2023] [Accepted: 02/06/2024] [Indexed: 02/15/2024]
Abstract
CONTEXT Adipose triglyceride lipase (ATGL), a key enzyme responsible for lipolysis, catalyzes the first step of lipolysis and converts triglycerides to diacylglycerols and free fatty acids (FFA). Our previous work suggested that phillyrin treatment improves insulin resistance in HFD-fed mice, which was associated with ATGL inhibition. In this study, using docking simulation, we explored the binding pose of phillyrin and atglistatin (a mouse ATGL inhibitor) to ATGL in mouse. From the docking results, the interactions with Ser47 and Asp166 were speculated to have caused phillyrin to inhibit ATGL in mice. Further, molecular dynamics simulation of 100 ns and MM-GBSA were conducted for the protein-ligand complex, which indicated that the system was stable and that phillyrin displayed a better affinity to ATGL than did atglistatin throughout the simulation period. Moreover, the results of pharmacological validation were consistent with those of the in silico simulations. In summary, our study illustrates the potential of molecular docking to accurately predict the binding protein produced by AlphaFold and suggests that phillyrin is a potential small molecule that targets and inhibits ATGL enzymatic activity. METHODS The ATGL-predicted protein structure, verified by PROCHECK, was determined using AlphaFold. Molecular docking, molecular dynamics simulation, and prime molecular mechanic-generalized born surface area were performed using LigPrep, Desmond, and prime MM-GBSA modules of Schrödinger software release 2021-2, respectively. For pharmacological validation, immunoblotting was performed to assess ATGL protein expression. The fluorescence intensity and glycerol concentration were quantified to evaluate the efficiency of phillyrin in inhibiting ATGL.
Collapse
Affiliation(s)
- Chenyu Zhou
- School of Pharmacy, Anhui University of Chinese Medicine, Xinzhan District, Hefei, 230012, China
| | - Lanmeng Yan
- School of Pharmacy, Anhui University of Chinese Medicine, Xinzhan District, Hefei, 230012, China
| | - Jing Xu
- School of Life Sciences, Anhui University of Chinese Medicine, Xinzhan District, Hefei, 230012, China
| | | | - Tongsheng Wang
- School of Life Sciences, Anhui University of Chinese Medicine, Xinzhan District, Hefei, 230012, China
| | - Fangping Du
- Jinzhai County Jinshanzhai Edible and Pharmaceutical Fungi Plantation Co. Ltd., Lu'an, 237300, Jinzhai, China
| | - Xiaohui Tong
- School of Life Sciences, Anhui University of Chinese Medicine, Xinzhan District, Hefei, 230012, China.
- Functional Activity and Resource Utilization on Edible and Medicinal Fungi Joint Laboratory of Anhui Province, Lu'an, 237300, China.
| | - Rongchun Han
- School of Pharmacy, Anhui University of Chinese Medicine, Xinzhan District, Hefei, 230012, China.
- Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Xinzhan District, Hefei, 230012, China.
- Joint Research Center for Chinese Herbal Medicine of Anhui of IHM, Anhui University of Chinese Medicine, Xinzhan District, Hefei, 230012, China.
| |
Collapse
|
8
|
Zhang L, Lang F, Feng J, Wang J. Review of the therapeutic potential of Forsythiae Fructus on the central nervous system: Active ingredients and mechanisms of action. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117275. [PMID: 37797873 DOI: 10.1016/j.jep.2023.117275] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/20/2023] [Accepted: 10/03/2023] [Indexed: 10/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine has gained significant attention in recent years owing to its multi-component, multi-target, and multi-pathway advantages in treating various diseases. Forsythiae Fructus, derived from the dried fruit of Forsythia suspensa (Thunb.) Vahl, is one such traditional Chinese medicine with numerous in vivo and ex vivo therapeutic effects, including anti-inflammatory, antibacterial, and antiviral properties. Forsythiae Fructus contains more than 200 chemical constituents, with forsythiaside, forsythiaside A, forsythiaside B, isoforsythiaside, forsythin, and phillyrin being the most active ingredients. Forsythiae Fructus exerts neuroprotective effects by modulating various pathways, including oxidative stress, anti-inflammation, NF-κB signaling, 2-AG, Nrf2 signaling, acetylcholinesterase, PI3K-Akt signaling, ferroptosis, gut-brain axis, TLR4 signaling, endoplasmic reticulum stress, PI3K/Akt/mTOR signaling, and PPARγ signaling pathway. AIM OF THE STUDY This review aims to highlight the potential therapeutic effects of Forsythiae Fructus on the central nervous system and summarize the current knowledge on the active ingredients of Forsythiae Fructus and their effects on different pathways involved in neuroprotection. MATERIALS AND METHODS In this review, we conducted a comprehensive search of databases (PubMed, Google Scholar, Web of Science, China Knowledge Resource Integrated, local dissertations and books) up until June 2023 using key terms such as Forsythia suspensa, Forsythiae Fructus, forsythiaside, isoforsythiaside, forsythin, phillyrin, Alzheimer's disease, Parkinson's disease, ischemic stroke, intracerebral hemorrhage, traumatic brain injury, aging, and herpes simplex virus encephalitis. RESULTS Our findings indicate that Forsythiae Fructus and its active ingredients own therapeutic effects on the central nervous system by modulating various pathways, including oxidative stress, anti-inflammation, NF-κB signaling, 2-AG, Nrf2 signaling, acetylcholinesterase, PI3K-Akt signaling, ferroptosis, the gut-brain axis, TLR4 signaling, endoplasmic reticulum stress, PI3K/Akt/mTOR signaling, and PPARγ signaling pathway. CONCLUSION Forsythiae Fructus and its active ingredients have demonstrated promising neuroprotective properties. Future in vivo and clinical studies of Forsythiae Fructus and its active ingredients should be conducted to establish precise dosage and standard guidelines for a more effective application in the treatment of neurological disorders.
Collapse
Affiliation(s)
- Leying Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, China
| | - Fenglong Lang
- Department of Neurology, Fushun Central Hospital, Fushun, Liaoning Province, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, China
| | - Jue Wang
- Department of Neurology, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, China.
| |
Collapse
|
9
|
Ning JW, Zang CX, Shang MY, Bao XQ, Zhang D. Natural products and their derivatives alleviating cerebral white matter lesions. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024; 26:146-153. [PMID: 38419338 DOI: 10.1080/10286020.2024.2301988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/02/2024] [Indexed: 03/02/2024]
Abstract
White matter lesions (WMLs), characterized by focal demyelination or myelination disorders, are commonly present in cerebral small vessel disease and various neurological diseases. Multiple etiologies lead to WMLs. However, there is no specific therapy or effective drugs for relieving WMLs. Natural products and their derivatives originate from bacterial, fungal, plant, and marine animal sources, many of which have multiple therapeutic targets. Compared to single target compounds, natural products and their derivatives are promising to be developed as better drugs to attenuate WMLs. Thus, this review attempts to summarize the status of natural products and their derivatives (2010-to date) alleviating cerebral white matter lesions for the discovery of new drugs.
Collapse
Affiliation(s)
- Jing-Wen Ning
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Cai-Xia Zang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mei-Yu Shang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiu-Qi Bao
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Dan Zhang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
10
|
Li Z, Zheng Y, Liu K, Liang Y, Lu J, Li Q, Zhao B, Liu X, Li X. Lignans as multi-targeted natural products in neurodegenerative diseases and depression: Recent perspectives. Phytother Res 2023; 37:5599-5621. [PMID: 37669911 DOI: 10.1002/ptr.8003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 07/09/2023] [Accepted: 08/19/2023] [Indexed: 09/07/2023]
Abstract
As the global population ages, the treatment of neurodegenerative diseases is becoming more and more important. There is an urgent need to discover novel drugs that are effective in treating neurological diseases. In recent years, natural products and their biological activities have gained widespread attention. Lignans are a class of metabolites extensively present in Chinese herbal medicine and possess good pharmacological effects. Latest studies have demonstrated their neuroprotective pharmacological activity in preventing acute/chronic neurodegenerative diseases and depression. In this review, the pharmacological effects of these disorders, the pharmacokinetics, safety, and clinical trials of lignans were summarized according to the scientific literature. These results proved that lignans mainly exert antioxidant and anti-inflammatory activities. Anti-apoptosis, regulation of nervous system functions, and modulation of synaptic signals are also potential effects. Despite the substantial evidence of the neuroprotective potential of lignans, it is not sufficient to support their use in the clinical management. Our study suggests that lignans can be used as prospective agents for the treatment of neurodegenerative diseases and depression, with a view to informing their further development and utilization.
Collapse
Affiliation(s)
- Zhibei Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kai Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Youdan Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiuxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bolin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xing Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
11
|
Li C, Wu M, Zhang H, Zhu X, Fu L, Wang S, Lu M, Zhong D, Ding Y. Safety, tolerability and pharmacokinetics of forsythin in healthy subjects: a double-blinded, placebo-controlled single-dose and multiple-dose escalation and food effect study. Ann Med 2023; 55:2274512. [PMID: 37980573 PMCID: PMC10836277 DOI: 10.1080/07853890.2023.2274512] [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: 08/18/2023] [Accepted: 10/18/2023] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Forsythin, an active compound from Forsythiae Fructus, has the potential to treat the common cold and influenza through its antipyretic-analgesic, anti-inflammatory and antiviral effects. The safety, tolerability and pharmacokinetic (PK) profile of forsythin were evaluated in healthy Chinese subjects. METHODS This phase 1a study included three parts: double-blind, randomized, placebo-controlled single-ascending-dose (SAD) (50, 100, 200, 400, 600 or 800 mg), food effect investigation (100 mg) and multiple-ascending-dose (MAD) (50, 100 or 200 mg TID for 5 days). RESULTS Forsythin is safe and tolerable in healthy Chinese subjects. The rates of adverse events (AEs) in the forsythin cohort were similar to those in the placebo cohort. Forsythin is well-absorbed after single or multiple doses and is extensively metabolized. The primary metabolites were aglycone M1, M1 sulphate (M2) and M1 glucuronide (M7). Exposure to forsythin (100 mg) was higher after food intake by approximately 1.4-fold, whereas M2 and M7 did not change. The steady state was reached around three days in the MAD study. Forsythin, M2 and M7 accumulation on day 5 was 1, 3 and 2, respectively. CONCLUSIONS The safety and PK profiles of forsythin support further evaluation of its efficacy in individuals with the common cold or influenza.
Collapse
Affiliation(s)
- Cuiyun Li
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Changchun, China
| | - Min Wu
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Changchun, China
| | - Hong Zhang
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Changchun, China
| | - Xiaoxue Zhu
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Changchun, China
| | - Li Fu
- Dalian Fusheng Institute of Natural Medicine, Dalian, China
| | - Shuo Wang
- Dalian Fusheng Institute of Natural Medicine, Dalian, China
| | - Mingming Lu
- Dalian Fusheng Institute of Natural Medicine, Dalian, China
| | - Dafang Zhong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yanhua Ding
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Changchun, China
| |
Collapse
|
12
|
Zhang S, Sun F, Zhu J, Qi J, Wang W, Liu Z, Li W, Liu C, Liu X, Wang N, Song X, Zhang D, Qi D, Wang X. Phillyrin ameliorates influenza a virus-induced pulmonary inflammation by antagonizing CXCR2 and inhibiting NLRP3 inflammasome activation. Virol J 2023; 20:262. [PMID: 37957672 PMCID: PMC10644626 DOI: 10.1186/s12985-023-02219-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023] Open
Abstract
Influenza is an acute viral respiratory illness with high morbidity rates worldwide. Excessive pulmonary inflammation is the main characteristic of lethal influenza A virus (IAV) infections. Therapeutic options for managing influenza are limited to vaccines and some antiviral medications. Phillyrin is one of the major bioactive components of the Chinese herbal medicine Forsythia suspensa, which has the functions of sterilization, heat clearing and detoxification. In this work, the effect and mechanism of phillyrin on H1N1 influenza (PR8)-induced pneumonia were investigated. We reported that phillyrin (15 mg/kg) treatment after viral challenge significantly improved the weight loss, ameliorated pulmonary inflammation and inhibited the accumulation of multiple cytokines and chemokines in bronchoalveolar lavage fluid on 7 days post infection (dpi). In vitro, phillyrin suppressed influenza viral replication (Matrixprotein and nucleoprotein messenger RNA level) and reduced influenza virus-induced cytopathic effect (CPE). Furthermore,chemokine receptor CXCR2 was confirmed to be markedly inhibited by phillyrin. Surface plasmon resonance results reveal that phillyrin exhibits binding affinity to CXCR2, having a binding affinity constant (KD) value of 1.858e-5 M, suggesting that CXCR2 is a potential therapeutic target for phillyrin. Moreover, phillyrin inhibited the mRNA and protein expression levels of Caspase1, ASC and NLRP3 in the lungs of mice with H1N1-induced pneumonia.This study reveals that phillyrin ameliorates IAV-induced pulmonary inflammation by antagonizing CXCR2 and inhibiting NLRP3 inflammasome activation partly.
Collapse
Affiliation(s)
- Shanyu Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Fengzhi Sun
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Jinlu Zhu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jianhong Qi
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 211198, China
| | - Wenjing Wang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Ziming Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wenqian Li
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Chuanguo Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xuehuan Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Nonghan Wang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xinyu Song
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Dan Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Dongmei Qi
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Xiaolong Wang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
13
|
Liu F, Cao L, Hu S, Ye H, Wu Q, Wu L. Muscone promotes functional recovery by facilitating microglia polarization into M2 phenotype through PPAR-γ pathway after ischemic stroke. Cell Immunol 2023; 386:104704. [PMID: 36921554 DOI: 10.1016/j.cellimm.2023.104704] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 01/04/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
Exploring regimens to facilitate microglia transformation from M1 to M2 phenotype is a feasible strategy to suppress neuroinflammation, therefore reinforcing functional recovery after ischemic stroke. Muscone easily crosses the blood brain barrier (BBB) and distributes throughout the brain. Here, the results illustrated the administration of 8 mg/kg muscone promoted functional recovery through reducing the infarct volume by 2,3,5-triphenyltetrazolium chloride (TTC) staining after ischemic stroke in mice. Then, the expression of pro-inflammatory factors, such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6), was significantly decreased, whereas the level of anti-inflammatory agents including C-X-C Motif Chemokine Ligand 1 (CXCL1), transforming growth factor-β (TGF-β) and interleukin-10 (IL-10) was obviously elevated in penumbra with the treatment of 8 mg/kg muscone using real-time fluorescence quantitative polymerase chain reaction (RT-qPCR), western blot and enzyme-linked immunosorbent assay (ELISA) tests. Subsequently, the results showed the application of muscone upregulated the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ) to facilitate microglia transformation into M2 phenotype using RT-qPCR, western blot and immunofluorescence analysis. Collectively, the present study provides evidence for our hypothesis that muscone intensifies microglia transformation into M2 phenotype via activating PPAR-γ signaling pathway in penumbra after ischemic stroke. These findings demonstrate muscone is a promising candidate for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Fei Liu
- Department of Neurology, Taikang Tongji (Wuhan) Hospital, Wuhan, Hubei 430050, PR China
| | - Liwei Cao
- Department of Neurology, Taikang Tongji (Wuhan) Hospital, Wuhan, Hubei 430050, PR China
| | - Shejing Hu
- Department of Neurology, Taikang Tongji (Wuhan) Hospital, Wuhan, Hubei 430050, PR China
| | - Hongxiang Ye
- Department of Neurology, Taikang Tongji (Wuhan) Hospital, Wuhan, Hubei 430050, PR China
| | - Qiang Wu
- Department of Neurology, Taikang Tongji (Wuhan) Hospital, Wuhan, Hubei 430050, PR China
| | - Le Wu
- Department of Neurology, Taikang Tongji (Wuhan) Hospital, Wuhan, Hubei 430050, PR China.
| |
Collapse
|
14
|
Hui Y, Zhao H, Shi L, Zhang H. Traumatic Brain Injury-Mediated Neuroinflammation and Neurological Deficits are Improved by 8-Methoxypsoralen Through Modulating PPARγ/NF-κB Pathway. Neurochem Res 2023; 48:625-640. [PMID: 36319778 DOI: 10.1007/s11064-022-03788-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/08/2022] [Accepted: 10/07/2022] [Indexed: 01/18/2023]
Abstract
8-Methoxypsoralen (8-MOP) has anti-inflammatory, antioxidant and tissue-repairing abilities. Here, we probed the function and mechanism of 8-MOP in traumatic brain injury (TBI). The in-vivo TBI model was constructed in Sprague-Dawley (SD) rats using controlled cortical impact (CCI) surgery. In parallel, BV2 microglia and HT22 neurons were activated by lipopolysaccharide (LPS) to establish an in-vitro model. The modified neurological score (mNSS) and the Morris water maze experiment were employed to evaluate the rats' neurological functions. The rats' brain edema was assessed by the dry and wet method, and neuronal apoptosis in damaged brain tissues was monitored by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) and Nissl's staining. Immunohistochemistry (IHC) was applied to verify Iba1-microglial activation in brain lesions of rats. The expression of inflammatory cytokines in BV2 microglia and HT22 neurons in the injured lesion of TBI rats was examined by the enzyme-linked immunosorbent assay (ELISA). The levels of iNOS, COX2, TLR4, PPARγ, STAT3, and NF-κB in brain lesions, BV2 microglia and HT22 neurons were compared by Western blot. As a result, 8-MOP administration reduced inflammation and LPS-induced neuronal damage in BV2 microglia. In vivo, 8-MOP treatment relieved neurological deficits in TBI rats, improved cognitive, learning and motor functions and mitigated brain edema and neuroinflammation induced by TBI. Furthermore, LPS or TBI activated the NF-κB and STAT3 pathways and repressed the PPARγ expression. However, 8-MOP treatment attenuated NF-κB and STAT3 phosphorylation and elevated PPARγ levels. Hence, 8-MOP exerts neuroprotective and anti-inflammatory effects in TBI rats by modulating the PPARγ/NF-κB pathway.
Collapse
Affiliation(s)
- Yuzuo Hui
- Department of Neurosurgery, Liaocheng People's Hospital, 67 Dongchang Road, Liaocheng, 252000, Shandong, China
| | - Han Zhao
- Department of Neurosurgery, Taian Central Hospital, Taian, 271000, Shandong, China
| | - Lei Shi
- Shandong Rongjun General Hospital, Ward 7, Jinan, 250000, Shandong, China
| | - Haitao Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, 67 Dongchang Road, Liaocheng, 252000, Shandong, China.
| |
Collapse
|
15
|
Kumar S, Mehan S, Narula AS. Therapeutic modulation of JAK-STAT, mTOR, and PPAR-γ signaling in neurological dysfunctions. J Mol Med (Berl) 2023; 101:9-49. [PMID: 36478124 DOI: 10.1007/s00109-022-02272-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/10/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
The cytokine-activated Janus kinase (JAK)-signal transducer and activator of transcription (STAT) cascade is a pleiotropic pathway that involves receptor subunit multimerization. The mammalian target of rapamycin (mTOR) is a ubiquitously expressed serine-threonine kinase that perceives and integrates a variety of intracellular and environmental stimuli to regulate essential activities such as cell development and metabolism. Peroxisome proliferator-activated receptor-gamma (PPARγ) is a prototypical metabolic nuclear receptor involved in neural differentiation and axon polarity. The JAK-STAT, mTOR, and PPARγ signaling pathways serve as a highly conserved signaling hub that coordinates neuronal activity and brain development. Additionally, overactivation of JAK/STAT, mTOR, and inhibition of PPARγ signaling have been linked to various neurocomplications, including neuroinflammation, apoptosis, and oxidative stress. Emerging research suggests that even minor disruptions in these cellular and molecular processes can have significant consequences manifested as neurological and neuropsychiatric diseases. Of interest, target modulators have been proven to alleviate neuronal complications associated with acute and chronic neurological deficits. This research-based review explores the therapeutic role of JAK-STAT, mTOR, and PPARγ signaling modulators in preventing neuronal dysfunctions in preclinical and clinical investigations.
Collapse
Affiliation(s)
- Sumit Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India.
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| |
Collapse
|
16
|
Xing XR, Luo LP, Li YL, Guo YW, Wang J, Qin J. Role of activating the nuclear factor kappa B signaling pathway in the development of septic cardiomyopathy in rats with sepsis. Technol Health Care 2023; 31:1671-1681. [PMID: 37092189 DOI: 10.3233/thc-220471] [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] [Indexed: 04/25/2023]
Abstract
BACKGROUND Despite advances in the treatment of sepsis over time, this condition remains both a serious threat and a cause of death among critical patients. OBJECTIVE This study aimed to explore the role of the nuclear factor kappa B (NF-κB) signaling pathway in the development of septic cardiomyopathy in rats with sepsis. METHOD A total of 32 Sprague Dawley rats were randomized into a sham operation group and three groups with sepsis, which were tested at one of the following time-points: 3, 6, or 12 h. Each group included eight rats. Sepsis models were created via cecal ligation and puncture procedures. All the study rats had the following cardiac parameters and serum levels measured at either 3, 6, or 12 h after the operation (according to their assigned group): heart rate, left ventricular systolic pressure (LVSP), maximum rate of left ventricular pressure rise (+dP/dtmax) and fall (-dP/dtmax), tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β), interleukin 6 (IL-6), and cardiac troponin I (cTnI). The myocardium of the left ventricle was collected and subjected to hematoxylin and eosin staining to observe the changes in pathological morphology. The expression of toll-like receptor 4 (TLR4) and NF-κB in the myocardium were detected by western blot analysis. RESULTS Compared with the sham operation group, the rats in the sepsis subgroups exhibited significantly lower values for all the cardiac parameters measured, including the heart rate (sham operation group = 386.63 ± 18.62 beats per minute [bpm], sepsis 3-h group = 368.38 ± 12.55 bpm, sepsis 6-h group = 341.75 ± 17.05 bpm, sepsis 12-h group = 302.13 ± 21.15 bpm), LVSP (sham operation group = 125.50 ± 11.45 mmHg, sepsis 3-h group = 110.88 ± 7.51 mmHg, sepsis 6-h group = 100.00 ± 15.06 mmHg, sepsis 12-h group = 91.38 ± 14.73 mmHg), +dp/dtmax (sham operation group = 7137.50 ± 276.44 mm Hg/sec, sepsis 3-h group = 5745.00 ± 346.16 mm Hg/sec, sepsis 6-h group = 4360.00 ± 312.04 mm Hg/sec, sepsis 12-h group = 2871.25 ± 443.99 mm Hg/sec), and -dp/dtmax (sham operation group = 6363.75 ± 123.86 mm Hg/sec, sepsis 3-h group = 6018.75 ± 173.49 mm Hg/sec, sepsis 6-h group = 5350.00 ± 337.89 mm Hg/sec, sepsis 12-h group = 4085.00 ± 326.76 mm Hg/sec). They also displayed significantly higher levels of serum cytokines, including TNF-α (sham operation group = 14.72 ± 2.90 pg/mL, sepsis 3-h group = 34.90 ± 4.79 pg/mL, sepsis 6-h group = 24.91 ± 2.57 pg/mL, sepsis 12-h group 22.06 ± 3.11 pg/mL), IL-1β (sham operation group = 42.25 ± 16.91, 3-h group = 112.25 ± 13.77, sepsis 6-h group = 207.90 ± 22.64, sepsis 12-h group = 157.18 ± 23.06), IL-6 (sham operation group = 39.89 ± 5.74, sepsis 3-h group = 78.27 ± 9.31, sepsis 6-h group = 123.75 ± 13.11, sepsis 12-h group = 93.21 ± 8.96), and cTnI (sham operation group = 0.07 ± 0.03 ng/mL, sepsis 3-h group = 0.18 ± 0.06 ng/mL, sepsis 6-h group = 0.67 ± 0.19 ng/mL, sepsis = 12-h group 1.28 ± 0.10 ng/mL). The rats in the sepsis groups exhibited pathological changes in the myocardium, which deteriorated gradually over time. The animals in all the sepsis groups exhibited significantly higher levels of TLR4 and NF-κB protein expression compared with the sham group. The TLR4 protein expressions were 0.376 in the sham operation group, 0.534 in the sepsis 3-h group, 0.551 in the sepsis 6-h group, and 0.719 in the sepsis 12-h group. The NF-κB protein expressions were 0.299 in the sham operation group, 0.488 in the sepsis 3-h group, 0.516 in the sepsis 6-h group, and 0.636 in the sepsis 12-h group. CONCLUSION Sepsis can lead to myocardial injury and cardiac dysfunction. This may be related to the activation of the NF-κB intracellular signal transduction pathway and the release of inflammatory factors as a result of lipopolysaccharides acting on TLR4 during the onset of sepsis.
Collapse
|
17
|
Liu J, Li J, Yang S, She Y, Li X, Jia Y. Phillyrin Inhibits Isoproterenol-Induced Cardiac Hypertrophy Via P38 and NF-κB Pathways. Nat Prod Commun 2023. [DOI: 10.1177/1934578x221144581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cardiac hypertrophy (CH) is the main compensatory response to chronic heart stress and often progresses to a decompensation state potentially leading to heart failure. Phillyrin (PHI) is a novel compound derived from Forsythia, which has shown anti-inflammatory and anti-virus activities as well as renal protective effects on diabetic nephropathy. Therefore, we investigated the effects of PHI on CH induced by isoproterenol (ISO). Cardiac hypertrophy was induced by ISO in vivo, and the H9C2 cells were treated with ISO. PHI treatment alleviated CH in isoproterenol-induced mice in 7 and 14 days. Echocardiography showed that the PHI improved ISO-induced CH heart function and structure. PHI significantly decreased heart weight/body weight (HW/BW) and heart weight/tibia length (HW/TL) ratios and improved left ventricular (LV) function in ISO-treated mice. Hematoxylin and eosin staining revealed cardiomyocyte areas of the ISO group were significantly increased, and PHI was significantly reduced at 7 and 14 days, PHI-100 groups showed significantly better improvements than PHI-50. Sirius red staining indicated PHI significantly decreased collagen deposition in heart cross-sections induced by ISO, and PHI repressed ISO-induced cTn-I and NT-proBNP expression in mouse serum. In vitro data from H9C2 cells showed that PHI decreased cell areas and total cell protein levels in cells induced by ISO, whereas ANP, BNP, IL-6, and IL-1β expression was significantly inhibited by PHI. Also, PHI simultaneously inhibited P65 and P38 phosphorylation in vivo and in vitro. In conclusion, this study demonstrated the protective effect of PHI on CH in in vivo and in vitro, and this effect was related to the suppression of inflammation through the activation of the P38/NF-κB pathway.
Collapse
Affiliation(s)
- Juanjuan Liu
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, ChongQing, China
| | - Jiahang Li
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, ChongQing, China
| | - Shengqian Yang
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, ChongQing, China
| | - Yuanting She
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, ChongQing, China
| | - Xiaohui Li
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, ChongQing, China
| | - Yi Jia
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, ChongQing, China
| |
Collapse
|
18
|
Fang Z, Wei L, Lv Y, Wang T, Hamezah HS, Han R, Tong X. Phillyrin restores metabolic disorders in mice fed with high-fat diet through inhibition of interleukin-6-mediated basal lipolysis. Front Nutr 2022; 9:956218. [PMID: 36276810 PMCID: PMC9581271 DOI: 10.3389/fnut.2022.956218] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
The function of white adipose tissue as an energy reservoir is impaired in obesity, leading to lipid spillover and ectopic lipid deposition. Adipose tissue inflammation can reduce the efficacy of lipid storage in adipocytes by augmenting basal lipolysis through producing interleukin-6 (IL-6). Therefore, pharmacological compounds targeting adipose tissue inflammation or IL-6 signaling might have the potential to combat obesity. This study aims to investigate the impact of Phillyrin, which is frequently used for treating respiratory infections in clinics in China, on obesity-related metabolic dysfunctions. Firstly, a mouse model of diet-induced obesity is used to assess the pharmacological applications of Phillyrin on obesity in vivo. Secondly, ex vivo culture of adipose tissue explants is utilized to investigate actions of Phillyrin on IL-6-linked basal lipolysis. Thirdly, a mouse model of IL-6 injection into visceral adipose tissue is explored to confirm the anti-basal lipolytic effect of Phillyrin against IL-6 in vivo. The results show that Phillyrin treatment reduces circulating level of glycerol, decreases hepatic steatosis and improves insulin sensitivity in obese mice. Meanwhile, Phillyrin attenuates obesity-related inflammation and IL-6 production in adipose tissue in obese mice. Furthermore, Phillyrin treatment results in resistance to IL-6-induced basal lipolysis in adipose tissue through suppressing expression of adipose triglyceride lipase (ATGL) both in vivo and in vitro. Collectively, these findings suggest that Phillyrin can restrain lipid efflux from inflamed adipose tissue in obesity by inhibiting IL-6-initiated basal lipolysis and ATGL expression, and thus is a potential candidate in the treatment of obesity-associated complications.
Collapse
Affiliation(s)
- Zhizheng Fang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Lu Wei
- School of Life Sciences, Hainan University, Haikou, China
| | - Yanping Lv
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Tongsheng Wang
- School of Life Sciences, Anhui University of Chinese Medicine, Hefei, China
| | | | - Rongchun Han
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China,*Correspondence: Rongchun Han,
| | - Xiaohui Tong
- School of Life Sciences, Anhui University of Chinese Medicine, Hefei, China,Xiaohui Tong,
| |
Collapse
|
19
|
Li YF, Ren X, Zhang L, Wang YH, Chen T. Microglial polarization in TBI: Signaling pathways and influencing pharmaceuticals. Front Aging Neurosci 2022; 14:901117. [PMID: 35978950 PMCID: PMC9376354 DOI: 10.3389/fnagi.2022.901117] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) is a serious disease that threatens life and health of people. It poses a great economic burden on the healthcare system. Thus, seeking effective therapy to cure a patient with TBI is a matter of great urgency. Microglia are macrophages in the central nervous system (CNS) and play an important role in neuroinflammation. When TBI occurs, the human body environment changes dramatically and microglia polarize to one of two different phenotypes: M1 and M2. M1 microglia play a role in promoting the development of inflammation, while M2 microglia play a role in inhibiting inflammation. How to regulate the polarization direction of microglia is of great significance for the treatment of patients with TBI. The polarization of microglia involves many cellular signal transduction pathways, such as the TLR-4/NF-κB, JAK/STAT, HMGB1, MAPK, and PPAR-γ pathways. These provide a theoretical basis for us to seek therapeutic drugs for the patient with TBI. There are several drugs that target these pathways, including fingolimod, minocycline, Tak-242 and erythropoietin (EPO), and CSF-1. In this study, we will review signaling pathways involved in microglial polarization and medications that influence this process.
Collapse
Affiliation(s)
| | | | | | - Yu-Hai Wang
- Department of Neurosurgery, The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, China
| | - Tao Chen
- Department of Neurosurgery, The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, China
| |
Collapse
|
20
|
Junshun Z, Gao M, Bao Y. Antibacterial activity of phillyrin and its effect on the preservation of minced pork. J FOOD PROCESS PRES 2022. [DOI: 10.1111/jfpp.16904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Zhang Junshun
- School of Forestry Northeast Forestry University Harbin PR China
| | - Mingkun Gao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology China Agricultural University Beijing PR China
| | - Yihong Bao
- School of Forestry Northeast Forestry University Harbin PR China
- Key Laboratory of Forest Food Resources Utilization of Heilongjiang Province Harbin PR China
| |
Collapse
|
21
|
Ding W, Cai C, Zhu X, Wang J, Jiang Q. Parthenolide ameliorates neurological deficits and neuroinflammation in mice with traumatic brain injury by suppressing STAT3/NF-κB and inflammasome activation. Int Immunopharmacol 2022; 108:108913. [PMID: 35729839 DOI: 10.1016/j.intimp.2022.108913] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/15/2022] [Accepted: 05/27/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) triggers a set of complex inflammation that results in secondary injury. Parthenolide (PTN) is a sesquiterpene lactone extracted from the herb Tanacetum parthenium (Feverfew) and has potent anti-inflammatory, anti-apoptosis and anti-oxidative stress effects in the central nervous system (CNS)-related diseases. This study focuses on investigating the potential neuroprotective effect of PTN on TBI and the related mechanism. METHODS Bv2 microglia, primary microglia were stimulated by LPS, and HT22 neuron cells were stimulated by OGD/R, and they were treated with different doses of PTN. The expression profiles of pro-inflammatory cytokines, proteins, oxidative stress mediators, STAT3/NF-κB pathway, inflammasomes were detected. Forty male/female C57BL/6 mice were randomly divided into the sham, PTN, TBI, and TBI + PTN groups (10 mice per group). A mouse TBI model was set up with a controlled cortical impact (CCI) device. The modified nerve severity score (mNSS) was implemented to check short-term neurological impairment in mice, and the mice's memory and learning were assessed by the Morris water maze test. The water content in the mice's brains was measured by the dry-wet method. Hematoxylin-eosin (H&E) staining, Nissl staining and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay were applied for neuronal apoptosis. RESULTS PTN dramatically alleviated LPS-induced inflammation in microglia, and OGD-mediated neuronal apoptosis and oxidative stress. In addition, PTN repressed LPS- or OGD-modulated STAT3/NF-κB and NLR family pyrin domain containing 1 (NLRP1), NLRP3, NLR family CARD domain containing 4 (NLRC4) inflammasomes activation. Administering the STAT3 inhibitor Stattic or NF-κB inhibitor Bay 11-7082 attenuated PTN-mediated effects. In vivo, PTN treatment relieved neural function deficits, brain edema and neuron apoptosis and improved the memory and learning function of TBI mice. Additionally, PTN impeded microglial activation and reduced the production of pro-inflammatory cytokines in brain lesions of TBI mice. Furthermore, PTN hindered STAT3/NF-κB and inflammasome activation. CONCLUSION PTN can curb microglial activation and neuron apoptosis by dampening the STAT3/NF-κB pathway, thus exerting neuroprotective effects in TBI mice.
Collapse
Affiliation(s)
- Wei Ding
- Department of Neurosurgery, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 430000, China; Department of Neurosurgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Chen Cai
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaomin Zhu
- Department of Neurology, Guangxi University of Chinese Medicine, Nanning 530200 Guangxi, China
| | - Jing Wang
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Qian Jiang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
22
|
Zhou C, Lu M, Cheng J, Rohani ER, Hamezah HS, Han R, Tong X. Review on the Pharmacological Properties of Phillyrin. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123670. [PMID: 35744798 PMCID: PMC9231344 DOI: 10.3390/molecules27123670] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/01/2022] [Accepted: 06/05/2022] [Indexed: 11/16/2022]
Abstract
Phillyrin is an effective lignan glycoside extracted from a traditional Chinese medicine Forsythia suspensa (Thunb.) Vahl (Oleaceae). It mainly exists in the roots, stems, leaves and fruits of the plant, with the highest content in the leaves. In terms of its medicinal application, there are a large number of experimental data proving its pharmacological effects in vitro and in animal models, such as anti-inflammatory, anti-obesity, anti-tumor, etc. Furthermore, pharmacokinetic experiments have also shown phillyrin's high effectiveness and low toxicity. Despite more than one thousand studies in the literature on phillyrin retrievable from Web of Science, PubMed, and CNKI, few reviews on its pharmacological activities have been presented conclusively. In this paper, we aimed to summarize the pharmacological and pharmacokinetic characteristics of phillyrin from the current literature, focusing on its anti-inflammatory, anti-aging, antiviral, antibacterial, hepatoprotective and anti-cancer effects, hoping to come up with new insights for its application as well as future studies.
Collapse
Affiliation(s)
- Chenyu Zhou
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (C.Z.); (M.L.); (J.C.); (R.H.)
| | - Mengya Lu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (C.Z.); (M.L.); (J.C.); (R.H.)
| | - Jialei Cheng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (C.Z.); (M.L.); (J.C.); (R.H.)
| | - Emelda Rosseleena Rohani
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia; (E.R.R.); (H.S.H.)
| | - Hamizah Shahirah Hamezah
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia; (E.R.R.); (H.S.H.)
| | - Rongchun Han
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (C.Z.); (M.L.); (J.C.); (R.H.)
| | - Xiaohui Tong
- School of Life Sciences, Anhui University of Chinese Medicine, Hefei 230012, China
- Correspondence: ; Tel.: +86-551-6812-9171; Fax: +86-551-6812-9028
| |
Collapse
|
23
|
Tang K, Zhong B, Luo Q, Liu Q, Chen X, Cao D, Li X, Yang S. Phillyrin attenuates norepinephrine-induced cardiac hypertrophy and inflammatory response by suppressing p38/ERK1/2 MAPK and AKT/NF-kappaB pathways. Eur J Pharmacol 2022; 927:175022. [DOI: 10.1016/j.ejphar.2022.175022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 12/24/2022]
|
24
|
Shao F, Wang X, Wu H, Wu Q, Zhang J. Microglia and Neuroinflammation: Crucial Pathological Mechanisms in Traumatic Brain Injury-Induced Neurodegeneration. Front Aging Neurosci 2022; 14:825086. [PMID: 35401152 PMCID: PMC8990307 DOI: 10.3389/fnagi.2022.825086] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/21/2022] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the most common diseases in the central nervous system (CNS) with high mortality and morbidity. Patients with TBI usually suffer many sequelae in the life time post injury, including neurodegenerative disorders such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). However, the pathological mechanisms connecting these two processes have not yet been fully elucidated. It is important to further investigate the pathophysiological mechanisms underlying TBI and TBI-induced neurodegeneration, which will promote the development of precise treatment target for these notorious neurodegenerative consequences after TBI. A growing body of evidence shows that neuroinflammation is a pivotal pathological process underlying chronic neurodegeneration following TBI. Microglia, as the immune cells in the CNS, play crucial roles in neuroinflammation and many other CNS diseases. Of interest, microglial activation and functional alteration has been proposed as key mediators in the evolution of chronic neurodegenerative pathology following TBI. Here, we review the updated studies involving phenotypical and functional alterations of microglia in neurodegeneration after injury, survey key molecules regulating the activities and functional responses of microglia in TBI pathology, and explore their potential implications to chronic neurodegeneration after injury. The work will give us a comprehensive understanding of mechanisms driving TBI-related neurodegeneration and offer novel ideas of developing corresponding prevention and treatment strategies for this disease.
Collapse
Affiliation(s)
- Fangjie Shao
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qun Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Qun Wu,
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
- Jianmin Zhang,
| |
Collapse
|
25
|
Hu C, Zhang S, Chen Q, Wang R. Ovatodiolide protects ischemia-reperfusion-induced neuronal injury via microglial neuroinflammation via mediating SIRT1/NF-κB pathway. Brain Res Bull 2022; 180:97-107. [PMID: 34968641 DOI: 10.1016/j.brainresbull.2021.12.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 11/30/2021] [Accepted: 12/22/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Ovatodiolide (OVA), a bioactive substance extracted from the bioactive component of Anisomeles indica, is reported to be endowed with anti-inflammatory properties. Nonetheless, its function in ischemia-reperfusion (I/R)-induced neurological deficits and microglial inflammation remains unclear. METHOD A middle cerebral artery occlusion (MCAO) model was set up in SD rats, which were then dealt with varying doses of OVA. The rats' neurological functions were estimated at diverse periods postoperatively. The dry and wet method, triphenyl tetrazolium chloride (TTC) staining, and Nissl's staining were conducted to measure brain edema, cerebral infarction area and neuronal damage, respectively. Immunohistochemistry (IHC) was performed to detect neuronal apoptosis and microglial activation, and the profiles of inflammatory factors in the cerebral tissues were estimated by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). In-vitro assays were implemented on HT22 neuronal cells and BV2 microglia to elaborate the effect of OVA against oxygen-glucose deprivation (OGD)-mediated effects. RESULTS OVA relieved HT22 cell apoptosis and eased inflammation in BV2 microglia, which were induced by OGD. OVA mitigated NF-κB phosphorylation in BV2 cells, whereas boosted SIRT1 expression. However, inhibiting SIRT1 abolished the anti-inflammatory effects of OVA in BV2 microglia under OGD stimulation. The condition medium (CM) of OGD-treated BV2 cells enhanced HT22 cell apoptosis and damage. OVA treatment in BV2 cells relieved BV2-mediated injury on HT22 cells, which was reversed by SIRT1 inhibitor. In-vivo results revealed that OVA dose-dependently attenuated I/R rats' neurological deficits, reduced brain edema, cerebral infarction area, neuronal apoptosis and microglial overactivation. Additionally, OVA inactivated the NF-κB pathway and up-regulated SIRT1 in the I/R rat model. CONCLUSION OVA prevented rats from brain I/R damage by hampering neuronal apoptosis and microglial inflammation via the SIRT1-NF-κB pathway. DATA AVAILABILITY The data sets used and analyzed during the current study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Chaojun Hu
- Department of Emergency, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Shan Zhang
- Department of Neurology, The People's Hospital of Leshan, Leshan 614000, Sichuan, China
| | - Qian Chen
- Department of Neurology, The First People's Hospital of Ziyang, Ziyang 641300, Sichuan, China
| | - Rong Wang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China.
| |
Collapse
|
26
|
Li H, Wang N, Xu Y, Chang X, Ke J, Yin J. Upregulating microRNA-373-3p promotes apoptosis and inhibits metastasis of hepatocellular carcinoma cells. Bioengineered 2022; 13:1304-1319. [PMID: 34983307 PMCID: PMC8805941 DOI: 10.1080/21655979.2021.2014616] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies in the digestive system. Abnormal miR-373-3p and TFAP4 expressions are critical in many malignant tumors, but it is unclear whether they work in the context of HCC. qRT-PCR measured miR-373-3p expression in HCC tissues and adjacent normal tissues. Flow cytometry and Western blot analyzed cell apoptosis. EMT, Transwell, and wound healing assay examined HCC cell migration and EMT, respectively. Western blot determined the profile of TFAP4/PI3K/AKT. IHC detected Ki67, E-cadherin, and vimentin in the tumor tissues. Moreover, the downstream target of miR-373-3p was predicted using the database. Dual luciferase activity assay and RIP verified the binding correlation between TFAP4 and miR-373-3p. In HCC tissues and cell lines, miR-373-3p was downregulated, and its overexpression stepped up HCC cell apoptosis and suppressed migration and EMT. Furthermore, miR-373-3p overexpression elevated Bax and caspase 3 expressions and attenuated Bcl2’s level. A xenograft tumor experiment in nude mice unveiled that miR-373-3p overexpression dampened tumor growth and proliferation. miR-373-3p cramped PI3K/AKT pathway activation. miR-373-3p negatively modulated TFAP4, and TFAP4 overexpression inverted miR-373-3p-mediated anti-tumor effects. Additionally, TFAP4 enhanced IGF1 expression, and promoted IGF1R-PI3K/AKT pathway activation. Collectively, miR-373-3p functions as an anti-tumor gene in HCC by inhibiting TFAP4/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Hongbin Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Nan Wang
- Emergency Internal Medicine, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuntian Xu
- Emergency Internal Medicine, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiao Chang
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing Ke
- Department of Infectious Diseases, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jun Yin
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
27
|
Qu Q, Li Y, Dong Q, Li S, Du H, Wang Z, Gong X, Zhang W, Lv W, Chao L, Liu M, Tang X, Guo S. Comparative Evaluation of Forsythiae Fructus From Different Harvest Seasons and Regions by HPLC/NIR Analysis and Anti-inflammatory and Antioxidant Assays. Front Pharmacol 2021; 12:737576. [PMID: 34899295 PMCID: PMC8652199 DOI: 10.3389/fphar.2021.737576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022] Open
Abstract
Forsythiae Fructus (FF), the dry fruit of Forsythia suspensa (Thunb.) Vahl, has a long history of use in traditional Chinese Medicine for its heat-clearing and detoxifying properties. It possesses clinical therapeutic effects and biological functions showing efficacy in handling different diseases. To investigate the FF differences in Henan, Shanxi, and Shaanxi in August and October, the surface morphology, mid-infrared and near-infrared spectrums, and HPLC were analyzed. Concurrently, the anti-inflammatory and antioxidant effects on LPS-induced J774A.1 cells were evaluated by western blot and RT-qPCR. The results showed that FF from different Harvest Seasons and Regions are provided with different microstructures and mid-infrared and near-infrared spectrums, and the levels of forsythiaside A and phillyrin of FF from Shanxi in August and phillygenin of FF from Shaanxi in August were the highest. Meanwhile, FF from Shanxi and Shaanxi in August markedly reduced the levels of inflammatory cytokines and mediators (TNF-α, IL-1β, NF-κB, and iNOS) and the protein expression levels of phosphorylated total IKKα/β and nuclear NF-κB. In August, SXFF and SAXFF also promoted the mRNA expression levels of HO-1 and NQO1 and the protein expression levels of HO-1 and nuclear Nrf2 and suppressed the protein expression levels of KEAP1. Spearman correlation analysis showed that phillygenin had a strong correlation with the protein expression on LPS-induced J774A.1 cells. In summary, our results showed that FF from harvest seasons and regions contributed to the distinct differences in microstructure, the mid-infrared and near-infrared spectrums, and compound content. More importantly, FF from Shanxi and Shaanxi in August showed marked anti-inflammatory and antioxidant activities, but with some differences, which may be because of different contents of phillygenin and phillyrin of lignans in FF.
Collapse
Affiliation(s)
- Qian Qu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuefei Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qi Dong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shupeng Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hongliang Du
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhihua Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaopei Gong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wenchang Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Weijie Lv
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Limin Chao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Mengjie Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xinggang Tang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Shining Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Guangdong Research Center for Veterinary Traditional Chinese Medicine and Natural Medicine Engineering Technology, Guangzhou, China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
28
|
Hu Y, Tao W. Microenvironmental Variations After Blood-Brain Barrier Breakdown in Traumatic Brain Injury. Front Mol Neurosci 2021; 14:750810. [PMID: 34899180 PMCID: PMC8662751 DOI: 10.3389/fnmol.2021.750810] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is linked to several pathologies. The blood-brain barrier (BBB) breakdown is considered to be one of the initial changes. Further, the microenvironmental alteration following TBI-induced BBB breakdown can be multi-scaled, constant, and dramatic. The microenvironmental variations after disruption of BBB includes several pathological changes, such as cerebral blood flow (CBF) alteration, brain edema, cerebral metabolism imbalances, and accumulation of inflammatory molecules. The modulation of the microenvironment presents attractive targets for TBI recovery, such as reducing toxic substances, inhibiting inflammation, and promoting neurogenesis. Herein, we briefly review the pathological alterations of the microenvironmental changes following BBB breakdown and outline potential interventions for TBI recovery based on microenvironmental modulation.
Collapse
Affiliation(s)
- Yue Hu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiwei Tao
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
29
|
Jiang Q, Wei D, He X, Gan C, Long X, Zhang H. Phillyrin Prevents Neuroinflammation-Induced Blood-Brain Barrier Damage Following Traumatic Brain Injury via Altering Microglial Polarization. Front Pharmacol 2021; 12:719823. [PMID: 34744713 PMCID: PMC8565465 DOI: 10.3389/fphar.2021.719823] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Phillyrin (Phi) is the main polyphenolic compound found in Forsythia suspensa. Recent studies have revealed that Phi has potent antioxidative and anti-inflammatory effects. However, whether Phi could relieve blood-brain barrier (BBB) damage following traumatic brain injury (TBI) remains unknown. Materials and Methods: Lipopolysaccharide (LPS) was used to activate primary microglia, which were then treated with different doses of Phi or the peroxisome proliferator-activated receptor-gamma (PPARγ) antagonist (GW9662). CCK-8 assay was used for evaluating cell viability, and the cytokines (including IL-1β, IL-6, TNFα, IL-4, IL-10, and TGFβ), microglial phenotypic markers (iNOS, COX2, and CD86 for "M1" polarization; Arg1, Ym1, and CD206 for "M2" polarization), PPARγ, and NF-κB were determined by RT-PCR, Western blot, or cellular immunofluorescence. Primary cultured mouse brain microvascular endothelial cells (BMECs) were stimulated by the condition medium (CM) from microglia. The cell viability, angiogenesis, and tight junction of BMECs were determined via CCK-8 assay, tube formation assay, and Western blot (for detecting MMP3, MMP9, ZO1, claudin-5, and occludin). Furthermore, the mouse TBI model was constructed and treated with Phi and/or GW9662. The BBB integrity was evaluated by H&E staining, Evans blue staining, and tissue immunofluorescence. Results: Phi markedly restrained the pro-inflammatory ("M1" state) cytokines and promoted anti-inflammatory ("M2" polarization) cytokines in LPS-mediated microglia. Phi mitigated "M1" polarization and promoted "M2" polarization of microglia via enhancing PPARγ and inhibiting the NF-κB pathway. The PPARγ antagonist GW9662 significantly repressed Phi-mediated anti-inflammatory effects. Meanwhile, Phi enhanced the viability, tube formation ability, and cell junction of BMECs. In the TBI mouse model, Phi promoted "M2" polarization, whereas it repressed the "M1" polarization of microglia. In addition, Phi reduced TBI-mediated BBB damage. However, the protective effects of Phi were reversed mainly by GW9662 treatment. Conclusion: Phi prevents BBB damage via inhibiting the neuroinflammation of microglia through the PPARγ/NF-κB pathway, which provides a potential therapeutic drug against TBI.
Collapse
Affiliation(s)
- Qian Jiang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Wei
- Department of Neurosurgery, Tianyou Hospital Affiliated to Wuhan University of Science & Technology, Wuhan, China
| | - Xuejun He
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Gan
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobing Long
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Yang Y, Zhao F, Yuan Z, Wang C, Chen K, Xiao W. Inhibition of miR-218-5p reduces myocardial ischemia-reperfusion injury in a Sprague-Dawley rat model by reducing oxidative stress and inflammation through MEF2C/NF-κB pathway. Int Immunopharmacol 2021; 101:108299. [PMID: 34749249 DOI: 10.1016/j.intimp.2021.108299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/14/2021] [Accepted: 10/20/2021] [Indexed: 12/01/2022]
Abstract
Following myocardial ischemia, myocardial reperfusion injury causes oxidative stress (OS) and inflammation, leading to myocardial cell apoptosis and necrosis. Recently, emerging studies have shown that microRNAs (miRNAs) contribute to the pathophysiology associated with myocardial ischemia-reperfusion (I/R). In this study, we conducted both in-vitro and in-vivo experiments to explore the role of miR-218-5p in ischemia-reperfusion (I/R)- or oxygen and glucose deprivation/reperfusion (OGD/R)-mediated cardiomyocyte injury. A total 44 Sprague-Dawley (SD) rats were used, and randomly divided into four groups, control group (n = 11), miR-218-5p-in group (n = 11), I/R group (n = 11), I/R + miR-218-5p-in group (n = 11). Our data showed that miR-218-5p was overexpressed in H9C2 cardiomyocytes under OGD/R treatment. miR-218-5p inhibition reduced the lactate dehydrogenase (LDH) activity and the levels of reactive oxygen species (ROS), malondialdehyde (MDA) and superoxide dismutase (SOD), as well as the expression of tumor necrosis factor alpha (TNF-α), interleukin (IL-1β), and IL-6. Oppositely, miR-218-5p overexpression aggravated OGD/R-mediated damage on H9C2 cells, whereas nuclear factor kappa B (NF-κB) pathway inhibition or myocyte enhancer factor 2C (MEF2C) upregulation reversed miR-218-5p mimics-mediated effects. Bioinformatics analysis predicted that miR-218-5p targeted and dampened its expression, which was testified by the dual-luciferase reporter assay and RNA pull-down assay. In vivo, inhibiting miR-218-5p declined LDH activities and ROS, MDA and SOD levels in rat myocardial tissues under I/R injury, alleviated myocardial fibrosis and inflammatory reactions, and reduced myocardial infarction area. Overall, inhibition of miR-218-5p choked oxidative stress and inflammation in myocardial I/R injury via targeting MEF2C/NF-κB axis, thus relieving the disease progression.
Collapse
Affiliation(s)
- Yang Yang
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, China
| | - Fenglong Zhao
- Department of Cardiology & Nephrology, Wuyi People's Hospital, Wuyi, Hebei 053400, China
| | - Zhe Yuan
- Department of Emergency, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, China
| | - Chuanqiang Wang
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, China
| | - Ke Chen
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, China
| | - Wenliang Xiao
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, China.
| |
Collapse
|
31
|
Wang T, Wen X, Zhang Z, Xie M, Zhou J. Phillyrin ameliorates diabetic nephropathy through the PI3K/Akt/GSK-3β signalling pathway in streptozotocin-induced diabetic mice. Hum Exp Toxicol 2021; 40:S487-S496. [PMID: 34649470 DOI: 10.1177/09603271211051598] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Diabetic nephropathy is a progressive kidney disease resulting from long-term hyperglycaemia in diabetic patients, and the underlying mechanism is complex and lacks effective treatments. Various active ingredients in Chinese herbs have been shown to alleviate renal injury and improve DN in recent years. Phillyrin, a natural medicinal active compound extracted from the Oleaceae family, has various pharmacological effects, including antioxidative, antiapoptotic and antiobesity effects. However, the role of phillyrin and its underlying mechanism in DN have not yet been explored. To investigate the effects of phillyrin on DN and its potential mechanisms of action, we performed experiments using streptozotocin (STZ)-induced DN mice as models. Phillyrin significantly reduced the levels of fasting blood glucose (FBG) and glycosylated haemoglobin A1c (HbA1c), downregulated the levels of serum blood urea nitrogen (BUN), serum creatinine (Scr), serum and urine β2-microglobulins (β2-MG) and improved the pathological changes of the kidney in a DN mouse model. Phillyrin also increased the level of antioxidants and attenuated oxidative damage in DN model mice. In addition, phillyrin inhibited Glycogen synthase kinase-3β (GSK-3β) activity by activating the PI3K/Akt signalling pathway, increased the Bcl-2/Bax ratio, reduced the release of cytochrome c from the mitochondria to the cytoplasm, subsequently inhibited the activation of caspase-3 and ultimately suppressed renal cell apoptosis. These findings suggested that phillyrin could be a new promising therapeutic strategy for DN, and this protective effect might be related to suppressing oxidative stress and apoptosis via the PI3K/Akt/GSK-3β pathway.
Collapse
Affiliation(s)
- Tianyang Wang
- School of Medicine, 117771Yichun University, Yichun, P.R. China
| | - Xuejiao Wen
- School of Medicine, 117771Yichun University, Yichun, P.R. China
| | - Ziwen Zhang
- School of Medicine, 117771Yichun University, Yichun, P.R. China
| | - Minjuan Xie
- School of Medicine, 117771Yichun University, Yichun, P.R. China
| | - Jie Zhou
- School of Medicine, 117771Yichun University, Yichun, P.R. China
| |
Collapse
|
32
|
The neuroprotective effect of phillyrin in intracerebral hemorrhagic mice is produced by activation of the Nrf2 signaling pathway. Eur J Pharmacol 2021; 909:174439. [PMID: 34425100 DOI: 10.1016/j.ejphar.2021.174439] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022]
Abstract
Phillyrin, a natural plant extract, has significant antioxidant and anti-apoptotic effects. However, its effect on intracerebral hemorrhage (ICH) remains unclear. In this study, we investigated a potential role for phillyrin in the regulation of the oxidative stress and apoptosis induced by ICH. A model of ICH was induced by collagenase IV (0.2 U in 1 μl sterile normal saline) in male C57BL/6J (B6) mice and different doses of phillyrin (5, 15, or 30 mg/kg) were intraperitoneally (i.p.) injected at 30 min, 6 h, and 22 h after modeling. We found that phillyrin significantly reduced neural function and lesion volume, improved injury of white and grey matter around the lesion, decreased apoptosis and oxidative stress, increased the expression of nuclear factor-erythroid 2-related factor 2 (Nrf2), heme oxygenase 1(HO-1), NADPH quinone oxidoreductase 1 (NQO1) and Superoxide Dismutase-1(SOD-1) in vitro and in vivo, and protected neurons from the stimulation of hemin by promoting Nrf2 nuclear translocation. Treatment with ML385 (Nrf2 inhibitor) completely reversed the protective effects of phillyrin in vivo after ICH injury. Based on our findings, we conclude that phillyrin treatment alleviates ICH injury-induced apoptosis and oxidative stress via activation of the Nrf2 signaling pathway, highlighting a potential role for phillyrin as an ICH therapeutic.
Collapse
|
33
|
Xie H, Chai H, Du X, Cui R, Dong Y. Overexpressing long non-coding RNA OIP5-AS1 ameliorates sepsis-induced lung injury in a rat model via regulating the miR-128-3p/Sirtuin-1 pathway. Bioengineered 2021; 12:9723-9738. [PMID: 34592882 PMCID: PMC8809967 DOI: 10.1080/21655979.2021.1987132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Sepsis, resulting from infections, is a systemic inflammatory response syndrome with a high fatality rate. The present study revolves around probing into the function and molecular mechanism of long non-coding RNA OIP5 antisense RNA 1 (lncRNA OIP5-AS1) in modulating acute lung injury (ALI) mediated by sepsis. Here, a sepsis model was constructed using cecal ligation and puncture (CLP) surgery in vivo. The alveolar macrophage cell line NR8383 and the alveolar type II cell line RLE-6TN were dealt with lipopolysaccharide (LPS) for in-vitro experiments. We discovered that OIP5-AS1 and Sirtuin1 (SIRT1) were markedly down-regulated in sepsis models elicited by CLP or LPS, while miR-128-3p experienced a dramatic up-regulation. OIP5-AS1 overexpression attenuated NR8383 and RLE-6TN cell apoptosis triggered by LPS and suppressed the expressions of nuclear factor kappa B (NF-κB), inducible nitric oxide synthase (iNOS), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in NR8383 and RLE-6TN cells, whereas miR-128-3p overexpression resulted in the opposite phenomenon. Moreover, OIP5-AS1 overexpression relieved lung edema, lung epithelial cell apoptosis, infiltration of myeloperoxidase (MPO)-labeled polymorphonuclear neutrophils (PMN), inflammatory responses triggered by CLP in vivo. Mechanistically, miR-128-3p, which targeted SIRT1, was hobbled by OIP5-AS1. All in all, OIP5-AS1 overexpression enhanced sepsis-induced ALI by modulating the miR-128-3p/SIRT1 pathway, which helps create new insights into sepsis treatment.
Collapse
Affiliation(s)
- Haibo Xie
- Department of Critical Care Medicine, Zhoushan Maternal and Child Health Hospital, Zhoushan, Zhejiang,316000, China
| | - Hanfei Chai
- Department of Critical Care Medicine, Zhoushan Maternal and Child Health Hospital, Zhoushan, Zhejiang,316000, China
| | - Xiaohong Du
- Department of Critical Care Medicine, Zhoushan Maternal and Child Health Hospital, Zhoushan, Zhejiang,316000, China
| | - Rongna Cui
- Department of Critical Care Medicine, Zhoushan Maternal and Child Health Hospital, Zhoushan, Zhejiang,316000, China
| | - Yinan Dong
- Department of Critical Care Medicine, Zhoushan Maternal and Child Health Hospital, Zhoushan, Zhejiang,316000, China
| |
Collapse
|
34
|
Cai L, Ge B, Xu S, Chen X, Yang H. Up-regulation of circARF3 reduces blood-brain barrier damage in rat subarachnoid hemorrhage model via miR-31-5p/MyD88/NF-κB axis. Aging (Albany NY) 2021; 13:21345-21363. [PMID: 34511434 PMCID: PMC8457610 DOI: 10.18632/aging.203468] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022]
Abstract
Inflammation events have been found to aggravate brain injury and blood-brain barrier (BBB) damage following subarachnoid hemorrhage (SAH). This study probed the role and mechanism of a novel circRNA, circARF3, in regulating the BBB injury in SAH rats and hypoxia-induced vascular endothelial cell (VEC) injury in vitro. Levels of circARF3 and miR-31-5p were monitored by RT-PCR. The expression of inflammatory factors IL-1β and TNF-α was verified by ELISA. In vivo SAH model was constructed in Sprague Dawley (SD) rats. The BBB integrity and cerebral edema, as well as the neurological functions of the rats were evaluated. The apoptotic neurons and microglia in brain lesions were examined by immunohistochemistry (IHC). The MyD88/NF-κB pathway was tested by Western blot. Furthermore, gain-of functional assay were constructed to explore the effects of circARF3 and miR-31-5p in primary cultured brain microvascular endothelial cell (BMEC) injury and microglial inflammation induced by oxygen and glucose deprivation (OGD). circARF3 was significantly down-regulated in plasma and CSF in SAH patients with higher Fisher stages. In the SAH rat model, overexpressing circARF3 improved BBB integrity and neurological score, decreased neuronal apoptosis and microglial activation in ipsilateral basal cortex, with declined miR-31-5p expression and MyD88-NF-κB activation. In vitro, overexpressing circARF3 attenuated OGD-mediated integrity destruction of BMECs and microglial induced neuroinflammation, while overexpressing miR-31-5p had opposite effects. Mechanistically, circARF3 sponged miR-31-5p as an endogenous competitive RNA and dampens its expression, thus inactivating MyD88-NF-κB pathway. CircARF3 attenuates BBB destruction in SAH rats by regulating the miR-31-5p-activated MyD88-NF-κB pathway.
Collapse
Affiliation(s)
- Li Cai
- Department of Neurosurgery, Guangxi International Zhuang Medicine Hospital, Nanning 530221, Guangxi, China
| | - Beihai Ge
- Department of Neurology, Guangxi Zhuang Autonomous Region Brain Hospital, Liuzhou 545005, Guangxi, China
| | - Shengbo Xu
- Department of Neurosurgery, Guangxi International Zhuang Medicine Hospital, Nanning 530221, Guangxi, China
| | - Xiangwen Chen
- Department of Neurosurgery, Guangxi International Zhuang Medicine Hospital, Nanning 530221, Guangxi, China
| | - Hong Yang
- Department of Neurosurgery, Guangxi International Zhuang Medicine Hospital, Nanning 530221, Guangxi, China
| |
Collapse
|
35
|
Du X, Shi L, Cao W, Zuo B, Zhou A. Add-on effect of Chinese herbal medicine in the treatment of mild to moderate COVID-19: A systematic review and meta-analysis. PLoS One 2021; 16:e0256429. [PMID: 34415962 PMCID: PMC8378756 DOI: 10.1371/journal.pone.0256429] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 08/06/2021] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Coronavirus disease 2019 (COVID-19) has emerged as a global pandemic since its outbreak in Wuhan, China. It is an urgent task to prevent and treat COVID-19 effectively early. In China's experience combating the COVID-19 pandemic, Chinese herbal medicine (CHM) has played an indispensable role. A large number of epidemiological investigations have shown that mild to moderate COVID-19 accounts for the largest proportion of cases. It is of great importance to treat such COVID-19 cases, which can help control epidemic progression. Many trials have shown that CHM combined with conventional therapy in the treatment of mild to moderate COVID-19 was superior to conventional therapy alone. This review was designed to evaluate the add-on effect of CHM in the treatment of mild to moderate COVID-19. METHODS Eight electronic databases including PubMed, EMBASE, Cochrane Central Register of Controlled Trials, the Clinical Trials.gov website, China National Knowledge Infrastructure (CNKI), China Science and Technology Journal Database (VIP), Wanfang Database and China Biology Medicine (CBM) were searched from December 2019 to March 2021 without language restrictions. Two reviewers searched and selected studies, and extracted data according to inclusion and exclusion criteria independently. Cochrane Risk of Bias (ROB) tool was used to assess the methodological quality of the included RCTs. Review Manager 5.3.0 software was used for statistical analysis. RESULTS Twelve eligible RCTs including 1393 participants were included in this meta-analysis. Our meta-analyses found that lung CT parameters [RR = 1.26, 95% CI (1.15, 1.38), P<0.00001] and the clinical cure rate [RR = 1.26, 95%CI (1.16, 1.38), P<0.00001] of CHM combined with conventional therapy in the treatment of mild to moderate COVID-19 were better than those of conventional therapy. The rate of conversion to severe cases [RR = 0.48, 95%CI (0.32, 0.73), P = 0.0005], TCM symptom score of fever [MD = -0.62, 95%CI (-0.79, -0.45), P<0.00001], cough cases [RR = 1.43, 95%CI (1.16, 1.75), P = 0.0006], TCM symptom score of cough[MD = -1.07, 95%CI (-1.29, -0.85), P<0.00001], TCM symptom score of fatigue[MD = -0.66, 95%CI (-1.05, -0.28), P = 0.0007], and CRP[MD = -5.46, 95%CI (-8.19, -2.72), P<0.0001] of combination therapy was significantly lower than that of conventional therapy. The WBC count was significantly higher than that of conventional therapy[MD = 0.38, 95%CI (0.31, 0.44), P<0.00001]. Our meta-analysis results were robust through sensitivity analysis. CONCLUSION Chinese herbal medicine combined with conventional therapy may be effective and safe in the treatment of mild to moderate COVID-19. More high-quality RCTs are needed in the future.
Collapse
Affiliation(s)
- Xuqin Du
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, People's Republic of China
| | - Lipeng Shi
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, People's Republic of China
| | - Wenfu Cao
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, People's Republic of China.,Department of Chinese Traditional Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Biao Zuo
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, People's Republic of China
| | - Aimin Zhou
- Department of Cardiovascular U nit, Traditional Chinese medicine hospital Dianjiang Chongqing, Chongqing, People's Republic of China
| |
Collapse
|
36
|
Zhao F, Guo Z, Hou F, Fan W, Wu B, Qian Z. Magnoflorine Alleviates "M1" Polarized Macrophage-Induced Intervertebral Disc Degeneration Through Repressing the HMGB1/Myd88/NF-κB Pathway and NLRP3 Inflammasome. Front Pharmacol 2021; 12:701087. [PMID: 34366853 PMCID: PMC8343137 DOI: 10.3389/fphar.2021.701087] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/12/2021] [Indexed: 12/16/2022] Open
Abstract
Intervertebral disc degeneration (IDD) is related to the deterioration of nucleus pulposus (NP) cells due to hypertrophic differentiation and calcification. The imbalance of pro-inflammatory (M1 type) and anti-inflammatory (M2 type) macrophages contributes to maintaining tissue integrity. Here, we aimed to probe the effect of Magnoflorine (MAG) on NP cell apoptosis mediated by “M1” polarized macrophages. THP-1 cells were treated with lipopolysaccharide (LPS) to induce “M1” polarized macrophages. Under the treatment with increasing concentrations of MAG, the expression of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, IL-18), high mobility group box protein 1 (HMGB1), as well as myeloid differentiation factor 88 (MyD88), nuclear factor kappa B (NF-κB) and NOD-like receptor 3 (NLRP3) inflammasomes in THP-1 cells were determined. What’s more, human NP cells were treated with the conditioned medium (CM) from THP-1 cells. The NP cell viability and apoptosis were evaluated. Western blot (WB) was adopted to monitor the expression of apoptosis-related proteins (Bax, Caspase3, and Caspase9), catabolic enzymes (MMP-3, MMP-13, ADAMTS-4, and ADAMTS-5), and extracellular matrix (ECM) compositions (collagen II and aggrecan) in NP cells. As a result, LPS evidently promoted the expression of pro-inflammatory cytokines and HMGB1, the MyD88-NF-κB activation, and the NLRP3 inflammasome profile in THP-1 cells, while MAG obviously inhibited the "M1″ polarization of THP-1 cells. After treatment with “M1” polarized THP-1 cell CM, NP cell viability was decreased, while cell apoptosis, the pro-inflammatory cytokines, apoptosis-related proteins, and catabolic enzymes were distinctly up-regulated, and ECM compositions were reduced. After treatment with MAG, NP cell damages were dramatically eased. Furthermore, MAG dampened the HMGB1 expression and inactivated the MyD88/NF-κB pathway and NLRP3 inflammasome in NP cells. In conclusion, this study confirmed that MAG alleviates “M1” polarized macrophage-mediated NP cell damage by inactivating the HMGB1-MyD88-NF-κB pathway and NLRP3 inflammasome, which provides a new reference for IDD treatment.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Zhenye Guo
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Fushan Hou
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Wei Fan
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Binqiang Wu
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Zhonglai Qian
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
37
|
Fan Y, Bi Y, Chen H. Salidroside Improves Chronic Stress Induced Depressive Symptoms Through Microglial Activation Suppression. Front Pharmacol 2021; 12:635762. [PMID: 34168556 PMCID: PMC8217647 DOI: 10.3389/fphar.2021.635762] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 05/25/2021] [Indexed: 11/29/2022] Open
Abstract
Depression is a severe neurological disorder highly associated with chronic mental stress stimulation, which involves chronic inflammation and microglial activation in the central nervous system (CNS). Salidroside (SLDS) has been reported to exhibit anti-neuroinflammatory and protective properties on neurological diseases. However, the mechanism underlying the effect of SLDS on depressive symptoms has not been well elaborated. In the present study, the effects of SLDS on depressive behaviors and microglia activation in mice CNS were investigated. Behavioral tests, including Forced swimming test (FST), Open field test (OFT) and Morris water maze (MWM) revealed that SLDS treatment attenuated the depressive behaviors in stress mice. SLDS treatment significantly reduced the microglial immunoreactivity for both Iba-1 and CD68, characteristic of deleterious M1 phenotype in hippocampus of stress mice. Additionally, SLDS inhibited microglial activation involving the suppression of ERK1/2, P38 MAPK and p65 NF-κB activation and thus reduced the expression and release of neuroinflammatory cytokines in stress mice as well as in lipopolysaccharide (LPS)-induced primary microglia. Also, SLDS changed microglial morphology, attachment and reduced the phagocytic ability in LPS-induced primary microglia. The results demonstrated that SLDS treatment could improve the depressive symptoms caused by unpredictable chronic stress, indicating a potential therapeutic application of SLDS in depression treatment by interfering microglia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Yang Fan
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Yajuan Bi
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Haixia Chen
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
38
|
Wang Z, Yang L. Chinese herbal medicine: Fighting SARS-CoV-2 infection on all fronts. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113869. [PMID: 33485973 PMCID: PMC7825841 DOI: 10.1016/j.jep.2021.113869] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/07/2021] [Accepted: 01/18/2021] [Indexed: 05/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes coronavirus disease 2019 (COVID-19), a highly pathogenic virus that has spread rapidly across the entire world. There is a critical need to develop safe and effective drugs, especially broad-spectrum antiviral and organ protection agents in order to treat and prevent this dangerous disease. It is possible that Chinese herbal medicine may play an essential role in the treatment of patients with SARS-CoV-2 infection. AIM OF THE REVIEW We aim to review the use of Chinese herbal medicine in the treatment of COVID-19 both in vitro and in clinical practice. Our goal was to provide a better understanding of the potential therapeutic effects of Chinese herbal medicine and to establish a "Chinese protocol" for the treatment of COVID-19. MATERIALS AND METHODS We systematically reviewed published research relating to traditional Chinese herbal medicines and the treatment of SARS-CoV-2 from inception to the 6th January 2021 by screening a range of digital databases (Web of Science, bioRxiv, medRxiv, China National Knowledge Infrastructure, X-MOL, Wanfang Data, Google Scholar, PubMed, Elsevier, and other resources) and public platforms relating to the management of clinical trials. We included the active ingredients of Chinese herbal medicines, monomer preparations, crude extracts, and formulas for the treatment of COVID-19. RESULTS In mainland China, a range of Chinese herbal medicines have been recognized as very promising anti-SARS-CoV-2 agents, including active ingredients (quercetagetin, osajin, tetrandrine, proscillaridin A, and dihydromyricetin), monomer preparations (xiyanping injection, matrine-sodium chloride injection, diammonium glycyrrhizinate enteric-coated capsules, and sodium aescinate injection), crude extracts (Scutellariae Radix extract and garlic essential oil), and formulas (Qingfei Paidu decoction, Lianhuaqingwen capsules, and Pudilan Xiaoyan oral liquid). All these agents have potential activity against SARS-CoV-2 and have attracted significant attention due to their activities both in vitro and in clinical practice. CONCLUSIONS As a key component of the COVID-19 treatment regimen, Chinese herbal medicines have played an irreplaceable role in the treatment of SARS-CoV-2 infection. The "Chinese protocol" has already demonstrated clear clinical importance. The use of Chinese herbal medicines that are capable of inhibiting SARS-Cov-2 infection may help to address this immediate unmet clinical need and may be attractive to other countries that are also seeking new options for effective COVID-19 treatment. Our analyses suggest that countries outside of China should also consider protocols involving Chinese herbal medicines combat this fast-spreading viral infection.
Collapse
Affiliation(s)
- Zhonglei Wang
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, PR China; School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, PR China.
| | - Liyan Yang
- School of Physics and Physical Engineering, Qufu Normal University, Qufu, 273165, PR China.
| |
Collapse
|
39
|
Xin Y, Tang L, Chen J, Chen D, Wen W, Han F. Inhibition of miR‑101‑3p protects against sepsis‑induced myocardial injury by inhibiting MAPK and NF‑κB pathway activation via the upregulation of DUSP1. Int J Mol Med 2021; 47:20. [PMID: 33448324 PMCID: PMC7849984 DOI: 10.3892/ijmm.2021.4853] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023] Open
Abstract
Numerous studies have found that microRNAs (miRNAs or miRs) are aberrantly expressed when sepsis occurs. The present study aimed to investigate the role of miR-101-3p in sepsis-induced myocardial injury and to elucidate the underlying mechanisms. Models of myocardial injury were established both in vivo and in vitro. The results revealed that miR-101-3p was upregulated in the serum of patients with sepsis-induced cardiomyopathy (SIC) and positively correlated with the levels of pro-inflammatory cytokines (including IL-1β, IL-6 and TNF-α). Subsequently, rats were treated with miR-101-3p inhibitor to suppress miR-101-3p and were then exposed to lipopolysaccharide (LPS). The results revealed that LPS induced marked cardiac dysfunction, apoptosis and inflammation. The inhibition of miR-101-3p markedly attenuated sepsis-induced myocardial injury by attenuating apoptosis and the expression of pro-inflammatory cytokines. Mechanistically, dual specificity phosphatase-1 (DUSP1) was found to be a functional target of miR-101-3p. The downregulation of miR-101-3p led to the overexpression of DUSP1, and the inactivation of the MAPK p38 and NF-κB pathways. Moreover, blocking DUSP1 by short hairpin RNA against DUSP1 (sh-DUSP1) significantly reduced the myocardial protective effects mediated by the inhibition of miR-101-3p. Collectively, the findings of the present study demonstrate that the inhibition of miR-101-3p exerts cardioprotective effects by suppressing MAPK p38 and NF-κB pathway activation, and thus attenuating inflammation and apoptosis dependently by enhancing DUSP1 expression.
Collapse
Affiliation(s)
- Ye Xin
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Li Tang
- Department of Gastroenterology, Xinqiao Hospital of Army Medical University, Chongqing 400037, P.R. China
| | - Jing Chen
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Dong Chen
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wen Wen
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Fugang Han
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
40
|
Xu J, Pei Y, Lu J, Liang X, Li Y, Wang J, Zhang Y. LncRNA SNHG7 alleviates IL-1β-induced osteoarthritis by inhibiting miR-214-5p-mediated PPARGC1B signaling pathways. Int Immunopharmacol 2021; 90:107150. [PMID: 33296783 DOI: 10.1016/j.intimp.2020.107150] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND As a common joint disease, osteoarthritis (OA) is the main cause of limited joint mobility and disability. The role of lncRNAs in the regulation of OA is increasingly discovered. Therefore, further exploring the function of SNHG7 in OA is of great significance for understanding its occurrence and development. METHODS We used interleukin-1β (IL-1β) to treat to establish an OA model primary on chondrocytes in vitro, and gain- and loss of function assays of SNHG7 and miR-214-5p were conducted. The cell viability and apoptosis of chondrocytes were detected by CCK8 assay, BrdU assay and flow cytometry. The inflammatory cytokines (IL-1β, IL-6 and TNF-α), NLRP3 inflammasome, protein level of PPARGC1B, PPARγ, P38 and NF-κB were determined by RT-PCR and/or western blot. RESULTS The results showed that SNHG7 was distinctly downregulated, while miR-214-5p was significantly upregulated in OA patients and primary chondrocytes treated with IL-1β. In addition, SNHG7 enhanced cell viability, inhibited apoptosis and inflammation of IL-1β-mediated chondrocytes. In contrast, miR-214-5p upregulation reduced viability, promoted apoptosis and inflammation of chondrocytes. Mechanistically, SNHG7 served as a competitive endogenous RNA by sponging miR-214-5p, which targeted PPARGC1B. Besides, the results of the compensation experiment affirmed that miR-214-5p attenuates SNHG7-mediated protective effects on IL-1β-mediated chondrocytes against apoptosis and inflammation, and activating PPARγ pathway markedly dampened the cytotoxic effects of miR-214-5p. CONCLUSIONS Collectively, The above results confirmed that SNHG7 prevents IL-1β induced OA by inhibiting NLRP3 inflammasome and apoptosis through miR-214-5p/PPARGC1B axis.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cartilage, Articular/drug effects
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Case-Control Studies
- Cells, Cultured
- Chondrocytes/drug effects
- Chondrocytes/metabolism
- Chondrocytes/pathology
- Humans
- Inflammasomes/metabolism
- Inflammation Mediators/metabolism
- Interleukin-1beta/toxicity
- Knee Joint/drug effects
- Knee Joint/metabolism
- Knee Joint/pathology
- Mice, Inbred C57BL
- MicroRNAs/genetics
- MicroRNAs/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Osteoarthritis, Knee/genetics
- Osteoarthritis, Knee/metabolism
- Osteoarthritis, Knee/pathology
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Signal Transduction
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Mice
Collapse
Affiliation(s)
- Junkui Xu
- Foot Ankle Center, The Xi'an Honghui Hospital, Xi'an 710054, Shaanxi, China
| | - Yanjiang Pei
- Department of Urology Surgery, The Xi'an Honghui Hospital, Xi'an 710054, Shaanxi, China
| | - Jun Lu
- Foot Ankle Center, The Xi'an Honghui Hospital, Xi'an 710054, Shaanxi, China
| | - Xiaojun Liang
- Foot Ankle Center, The Xi'an Honghui Hospital, Xi'an 710054, Shaanxi, China
| | - Yi Li
- Foot Ankle Center, The Xi'an Honghui Hospital, Xi'an 710054, Shaanxi, China
| | - Junhu Wang
- Foot Ankle Center, The Xi'an Honghui Hospital, Xi'an 710054, Shaanxi, China
| | - Yingang Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| |
Collapse
|
41
|
Chu X, Wang C, Wu Z, Fan L, Tao C, Lin J, Chen S, Lin Y, Ge Y. JNK/c-Jun-driven NLRP3 inflammasome activation in microglia contributed to retinal ganglion cells degeneration induced by indirect traumatic optic neuropathy. Exp Eye Res 2020; 202:108335. [PMID: 33141050 DOI: 10.1016/j.exer.2020.108335] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Indirect traumatic optic neuropathy (ITON) is a major cause of permanent loss of vision after blunt head trauma. Neuroinflammation plays a crucial role in neurodegenerative diseases. The present study concentrated on JNK/c-Jun-driven NLRP3 inflammasome activation in microglia during the degeneration of retinal ganglion cells (RGCs) in ITON. METHODS An impact acceleration (IA) model was employed to induce ITON, which could produce significant neurodegeneration in the visual system. Pharmacological approaches were employed to disrupt JNK and to explore whether JNK and the microglial response contribute to RGC death and axonal degeneration. RESULTS Our results indicated that the ITON model induced significant RGC death and axonal degeneration and activated JNK/c-Jun signaling, which could further induce the microglial response and NLRP3 inflammasome activation. Moreover, JNK disruption is sufficient to suppress NLRP3 inflammasome activation in microglia and to prevent RGC death and axonal degeneration. CONCLUSIONS ITON could promote JNK/c-Jun signaling, which further activates the NLRP3 inflammasome in microglia and contributes to the degeneration of axons and death of RGCs. JNK inhibition is able to suppress the inflammatory reaction and improve RGC survival. Although further work is needed to determine whether pharmacological inhibition of the NLRP3 inflammasome can prevent ITON, our findings indicated that such intervention could be promising for translational work.
Collapse
Affiliation(s)
- Xiaoqi Chu
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Chun Wang
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Zheng Wu
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Liting Fan
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Chunmei Tao
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Jiaqi Lin
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Shuang Chen
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Yongzhong Lin
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China.
| | - Yusong Ge
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China.
| |
Collapse
|
42
|
Liu KL, Yu XJ, Sun TZ, Wang YC, Chen MX, Su YW, Zhang HC, Chen YM, Gao HL, Shi XL, Qi J, Li Y, Li HB, Dong WJ, He JK, Kang YM. Effects of seawater immersion on open traumatic brain injury in rabbit model. Brain Res 2020; 1743:146903. [PMID: 32445716 DOI: 10.1016/j.brainres.2020.146903] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 05/10/2020] [Accepted: 05/19/2020] [Indexed: 11/19/2022]
Abstract
We emulated instances of open traumatic brain injuries (TBI) in a maritime disaster. New Zealand rabbit animal models were used to evaluate the pathophysiological changes in open TBI with and without the influence of artificial seawater. New Zealand rabbits were randomly divided into 3 groups. Control group consisted of only normal animals. Animals in TBI and TBI + Seawater groups underwent craniotomy with dura mater incised and brain tissue exposed to free-fall impact. Afterward, only TBI + Seawater group received on-site artificial seawater infusion. Brain water content (BWC) and permeability of blood-brain barrier (BBB) were assessed. Reactive oxygen species levels were measured. Western blotting and immunofluorescence were employed to detect: apoptosis-related factors Caspase-3, Bax and Bcl-2; angiogenesis-related factors CD31 and CD34; astrogliosis-related factor glial fibrillary acidic protein (GFAP); potential neuron injury indicator neuron-specific enolase (NSE). Hematoxylin & eosin, Masson-trichrome and Nissl stainings were performed for pathological observations. Comparing to Control group, TBI group manifested abnormal neuronal morphology; increased BWC; compromised BBB integrity; increased ROS, Bax, CD31, CD34, Caspase-3 and GFAP expressions; decreased Bcl-2 and NSE expression. Seawater immersion caused all changes, except BWC, to become more significant. Seawater immersion worsens the damage inflicted to brain tissue by open TBI. It aggravates hypoxia in brain tissue, upregulates ROS expression, increases neuron sensitivity to apoptosis-inducing factors, and promotes angiogenesis as well as astrogliosis.
Collapse
Affiliation(s)
- Kai-Li Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Xiao-Jing Yu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Tian-Ze Sun
- Department of Human Anatomy and Histology and Embryology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi an 710061, China
| | - Yi-Chang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Meng-Xuan Chen
- College of Stomatology, Xi'an Jiaotong University, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, 98 XiWu Road, Xi'an, Shaanxi 710004, People's Republic of China
| | - Yan-Wen Su
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Hao-Chen Zhang
- School of Clinical Medicine, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yan-Mei Chen
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Hong-Li Gao
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Xiao-Lian Shi
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Jie Qi
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Ying Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Hong-Bao Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Wei-Jiang Dong
- Department of Human Anatomy and Histology and Embryology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi an 710061, China.
| | - Jian-Kang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China.
| |
Collapse
|