1
|
Demir M, Elbe H, Cetinavci D, Saruhan E. Effects of Troxerutin on Oxidative Stress, Inflammation and Galectin- 3 Expression in Intracerebroventricular Kainic Acid-Induced Neurotoxicity. Inflammation 2025:10.1007/s10753-025-02301-9. [PMID: 40237932 DOI: 10.1007/s10753-025-02301-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
Excitotoxicity caused by excessive concentration of the excitatory neurotransmitter glutamate causes neuronal cell death and promotes neurodegenerative disorders. The neuroexcitant neurotoxin kainic acid (KA) induces excitotoxicity, leading to neuronal death via oxidative stress and inflammation, and its experimental use is widespread. This study was designed to determine the protective effect of Troxerutin (TXR) and its relationship with Galectin-3 (Gal-3) in experimental excitotoxicity with neuroinflammation and oxidative stress. Fifty male Wistar rats were divided into five groups (n = 10): Control group rats received intraperitoneal (ip) normal saline for 6 days. Sham group rats received a single dose of intracerebroventricular (icv) normal saline on the first day. KA group rats were treated with a single dose of KA; icv-0.5 μg/μl). TXR group rats treated with TXR for 6 days: ip-100 mg/kg) and KA + TXR group rats treated with KA (single dose) and TXR (6 days). It was observed that malondialdehyde (MDA) and interleukin-1β (IL-1β) levels increased and reduced glutathione (GSH) levels decreased in the cerebral cortex of rats with KA neurotoxicity. TXR treatment caused a significant improvement in MDA and GSH levels and a significant decrease in IL-1β levels in rats with the excitotoxicity model. Gal-3 expressions in the hippocampus and cerebellum increased in KA-treated rats, whereas TXR treatment decreased Gal-3 expressions. In addition, histopathological changes caused by KA administration showed improvement in TXR-treated groups. In conclusion, the findings showed that TXR treatment attenuated KA-induced neurotoxicity by reducing oxidative tissue damage, inflammatory response and Gal-3 expression.
Collapse
Affiliation(s)
- Mehmet Demir
- Department of Physiology, Faculty of Medicine, Karabuk University, Karabuk, Turkey.
| | - Hulya Elbe
- Department of Histology and Embryology, Faculty of Medicine, Mugla Sıtkı Kocman University, Mugla, Turkey
| | - Dilan Cetinavci
- Department of Histology and Embryology, Mugla Training and Research Hospital, Mugla, Turkey
| | - Ercan Saruhan
- Department of Medical Biochemistry, Faculty of Medicine, Mugla Sıtkı Kocman University, Mugla, Turkey
| |
Collapse
|
2
|
Alatawi FS, Omran AME, Rashad E, Abdel-Rahman ON, Soliman AF. Rutin attenuates bleomycin-induced acute lung injury via miR-9-5p mediated NF-κB signaling inhibition: network pharmacology analysis and experimental evidence. Front Pharmacol 2025; 16:1522690. [PMID: 40110126 PMCID: PMC11920148 DOI: 10.3389/fphar.2025.1522690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Although successfully used as a chemotherapeutic agent in various malignant diseases, acute lung injury (ALI) is one of the major limitations of bleomycin (BLM). Seeking reliable natural remedies, this study aimed to explore the potential effect of rutin on BLM-induced ALI. Methods Targets of rutin and ALI were collected using various databases. Enrichment analyses of common targets were conducted, a protein-protein interaction (PPI) network was constructed, the hub genes were identified, and the upstream miRNA interacting with the top hub gene was later predicted. A BLM-induced ALI rat model was established to verify rutin potential effects, and the selected hub gene expression with its upstream regulatory miRNA and a downstream set of targets were examined to elucidate the action mechanism. Results A total of 147 genes have been identified as potential therapeutic targets of rutin to treat BLM-induced ALI. Data from the enrichment and PPI analyses and the prediction of the upstream miRNAs indicated that the most worthwhile pair to study was miR-9a-5p/Nfkb1. In vivo findings showed that rutin administration significantly ameliorated pulmonary vascular permeability, inflammatory cells alveolar infiltration, induction of proinflammatory cytokines in the bronchoalveolar lavage fluid, and lung histology. Mechanistically, rutin downregulated the gene expression level of Nfkb1, Ptgs2, Il18, and Ifng, alongside their protein products, NF-κB p50, COX-2, IL-18, and IFN-γ, accompanied by an upregulation of rno-miR-9a-5p, Il10, and IL-10 expression in lung tissues. Conclusion Combining network pharmacology and an in vivo study revealed that miR-9-5p/Nfkb1 axis could mediate the meliorative effect of rutin against BLM-induced ALI.
Collapse
Affiliation(s)
- Fatema S Alatawi
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Awatif M E Omran
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Eman Rashad
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Omnia N Abdel-Rahman
- Biological and Geological Sciences Department, Faculty of Education, Ain Shams University, Cairo, Egypt
| | - Ahmed F Soliman
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
3
|
Yang B, Lin Y, Huang Y, Shen YQ, Chen Q. Thioredoxin (Trx): A redox target and modulator of cellular senescence and aging-related diseases. Redox Biol 2024; 70:103032. [PMID: 38232457 PMCID: PMC10827563 DOI: 10.1016/j.redox.2024.103032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/03/2023] [Accepted: 01/04/2024] [Indexed: 01/19/2024] Open
Abstract
Thioredoxin (Trx) is a compact redox-regulatory protein that modulates cellular redox state by reducing oxidized proteins. Trx exhibits dual functionality as an antioxidant and a cofactor for diverse enzymes and transcription factors, thereby exerting influence over their activity and function. Trx has emerged as a pivotal biomarker for various diseases, particularly those associated with oxidative stress, inflammation, and aging. Recent clinical investigations have underscored the significance of Trx in disease diagnosis, treatment, and mechanistic elucidation. Despite its paramount importance, the intricate interplay between Trx and cellular senescence-a condition characterized by irreversible growth arrest induced by multiple aging stimuli-remains inadequately understood. In this review, our objective is to present a comprehensive and up-to-date overview of the structure and function of Trx, its involvement in redox signaling pathways and cellular senescence, its association with aging and age-related diseases, as well as its potential as a therapeutic target. Our review aims to elucidate the novel and extensive role of Trx in senescence while highlighting its implications for aging and age-related diseases.
Collapse
Affiliation(s)
- Bowen Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yumeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yibo Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
4
|
Park GT, Lim JK, Choi EB, Lim MJ, Yun BY, Kim DK, Yoon JW, Hong YG, Chang JH, Bae SH, Ahn JY, Kim JH. Transplantation of adipose tissue-derived microvascular fragments promotes therapy of critical limb ischemia. Biomater Res 2023; 27:70. [PMID: 37455318 DOI: 10.1186/s40824-023-00395-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/15/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Adipose tissue-derived microvascular fragments are functional vessel segments derived from arterioles, capillaries, and veins. Microvascular fragments can be used as vascularization units in regenerative medicine and tissue engineering containing microvascular networks. However, the in vivo therapeutic and vascularization properties of human microvascular fragments have not been investigated. METHODS In this study, we isolated microvascular fragments, stromal vascular fractions, and mesenchymal stem cells from human lipoaspirate and studied their therapeutic efficacy and in vivo vasculogenic activity in a murine model of hindlimb ischemia. In addition, in vivo angiogenic activity and engraftment of microvascular fragments into blood vessels were measured using Matrigel plug assay. RESULTS Both microvascular fragments and stromal vascular fractions contain not only mesenchymal stem cells but also endothelial progenitor cells. In a Matrigel plug assay, microvascular fragments increased the number of blood vessels containing red blood cells more than mesenchymal stem cells and stromal vascular fractions did. The engraftment of the microvascular fragments transplanted in blood vessels within the Matrigel plug significantly increased compared to the engraftment of mesenchymal stem cells and stromal vascular fractions. Moreover, intramuscular injection of microvascular fragments markedly increased blood flow in the ischemic hindlimbs and alleviated tissue necrosis compared to that of mesenchymal stem cells or stromal vascular fractions. Furthermore, transplanted microvascular fragments formed new blood vessels in ischemic limbs. CONCLUSIONS These results suggest that microvascular fragments show improved engraftment efficiency and vasculogenic activity in vivo and are highly useful for treating ischemic diseases and in tissue engineering. Adipose tissue-derived microvascular fragments are vascularization units in regenerative medicine and tissue engineering containing microvascular networks. Intramuscular injection of microvascular fragments markedly increased blood flow in the ischemic hindlimbs and alleviated tissue necrosis. The present study suggests that microvascular fragments show improved engraftment efficiency and vasculogenic activity in vivo and are highly useful for treating ischemic diseases and in tissue engineering.
Collapse
Affiliation(s)
- Gyu Tae Park
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea
| | - Jae Kyung Lim
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea
| | - Eun-Bae Choi
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea
| | - Mi-Ju Lim
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea
| | - Bo-Young Yun
- UVA Surgery Clinic, Busan, 47537, Republic of Korea
| | - Dae Kyoung Kim
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea
| | - Jung Won Yoon
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea
| | - Yoon Gi Hong
- BS The Body Aesthetic Plastic Surgery Clinic, Busan, 47287, Republic of Korea
| | - Jae Hoon Chang
- BS The Body Aesthetic Plastic Surgery Clinic, Busan, 47287, Republic of Korea
| | - Seong Hwan Bae
- Department of Plastic and Reconstructive Surgery, College of Medicine, Pusan National University, Busan, Gyeongsangnam-do, 49241, Republic of Korea
| | - Jung Yong Ahn
- UVA Surgery Clinic, Busan, 47537, Republic of Korea.
| | - Jae Ho Kim
- Department of Physiology, College of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea.
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do, 50612, Republic of Korea.
| |
Collapse
|
5
|
Zhou J, Li L, Qu M, Tan J, Sun G, Luo F, Zhong P, He C. Electroacupuncture pretreatment protects septic rats from acute lung injury by relieving inflammation and regulating macrophage polarization. Acupunct Med 2022:9645284221118588. [PMID: 36039902 DOI: 10.1177/09645284221118588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Macrophage polarization toward the M2 phenotype may attenuate inflammation and have a therapeutic effect in acute lung injury (ALI). OBJECTIVE To investigate the role of electroacupuncture (EA) pretreatment on the inflammatory response and macrophage polarization in a septic rat model of lipopolysaccharide (LPS)-induced ALI. METHODS Male Sprague Dawley rats (n = 24) were randomly divided into three groups (n = 8 each): control (Ctrl), ALI (LPS) and pre-EA (LPS + EA pretreatment). ALI and pre-EA rats were injected with LPS via the caudal vein. Pulmonary edema was assessed by left upper pulmonary lobe wet-to-dry (W/D) ratios. Lung injury scores were obtained from paraffin-embedded and hematoxylin and eosin-stained sections of the left lower pulmonary lobe. Inflammatory activation was quantified using serum tumor necrosis factor (TNF)-α, interleukin (IL)-1β, transforming growth factor (TGF)-β and IL-10 levels measured by enzyme linked immunosorbent assay (ELISA). Macrophage phenotype was determined by real-time quantitative polymerase chain reaction (RT-qPCR) and Western blotting. RESULTS Mean lung W/D ratio was significantly lower and serum IL-1β levels were decreased in pre-EA rats compared to ALI rats (P < 0.05). TNF-α mRNA expression was decreased and mannose receptor (MR) and Arg1 mRNA expression was increased in the lung tissues of pre-EA rats compared to ALI rats (P < 0.01). Arg1 protein expression was similarly increased in the lung tissues of pre-EA rats compared to ALI rats (P < 0.05). CONCLUSION EA pretreatment may play a protective role by promoting macrophage polarization to the M2 phenotype in a septic rat model of LPS-induced ALI.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Li
- Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Mengjian Qu
- Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Jinqu Tan
- Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Guanghua Sun
- Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Fu Luo
- Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Peirui Zhong
- Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Chengqi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Vidhya R, Anbumani VI, Dinakara Rao A, Anuradha CV. Identification of novel human neutrophil elastase inhibitors from dietary phytochemicals using in silico and in vitro studies. J Biomol Struct Dyn 2022; 40:3451-3461. [PMID: 33222615 DOI: 10.1080/07391102.2020.1847685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Ramachandran Vidhya
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar, Tamil Nadu, India
| | | | - Ampasala Dinakara Rao
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry, India
| | | |
Collapse
|
7
|
Sahu D, Bishwal SC, Malik MZ, Sahu S, Kaushik SR, Sharma S, Saini E, Arya R, Rastogi A, Sharma S, Sen S, Singh RKB, Liu CJ, Nanda RK, Panda AK. Troxerutin-Mediated Complement Pathway Inhibition is a Disease-Modifying Treatment for Inflammatory Arthritis. Front Cell Dev Biol 2022; 10:845457. [PMID: 35433699 PMCID: PMC9009527 DOI: 10.3389/fcell.2022.845457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/03/2022] [Indexed: 12/01/2022] Open
Abstract
Troxerutin (TXR) is a phytochemical reported to possess anti-inflammatory and hepatoprotective effects. In this study, we aimed to exploit the antiarthritic properties of TXR using an adjuvant-induced arthritic (AIA) rat model. AIA-induced rats showed the highest arthritis score at the disease onset and by oral administration of TXR (50, 100, and 200 mg/kg body weight), reduced to basal level in a dose-dependent manner. Isobaric tags for relative and absolute quantitative (iTRAQ) proteomics tool were employed to identify deregulated joint homogenate proteins in AIA and TXR-treated rats to decipher the probable mechanism of TXR action in arthritis. iTRAQ analysis identified a set of 434 proteins with 65 deregulated proteins (log2 case/control≥1.5) in AIA. Expressions of a set of important proteins (AAT, T-kininogen, vimentin, desmin, and nucleophosmin) that could classify AIA from the healthy ones were validated using Western blot analysis. The Western blot data corroborated proteomics findings. In silico protein–protein interaction study of tissue-proteome revealed that complement component 9 (C9), the major building blocks of the membrane attack complex (MAC) responsible for sterile inflammation, get perturbed in AIA. Our dosimetry study suggests that a TXR dose of 200 mg/kg body weight for 15 days is sufficient to bring the arthritis score to basal levels in AIA rats. We have shown the importance of TXR as an antiarthritic agent in the AIA model and after additional investigation, its arthritic ameliorating properties could be exploited for clinical usability.
Collapse
Affiliation(s)
- Debasis Sahu
- Product Development Cell, National Institute of Immunology, New Delhi, India
- Department of Orthopedics Surgery, New York University School of Medicine, New York, NY, United States
- *Correspondence: Debasis Sahu, ; Ranjan Kumar Nanda, ; Amulya Kumar Panda,
| | - Subasa Chandra Bishwal
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Md. Zubbair Malik
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Sukanya Sahu
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Sandeep Rai Kaushik
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Shikha Sharma
- Amity Institute of Forensic Sciences, Amity University, Noida, India
| | - Ekta Saini
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Rakesh Arya
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Archana Rastogi
- Department of Pathology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Sandeep Sharma
- Department of Medical Laboratory Sciences, Lovely Professional University, Phagwara, India
| | - Shanta Sen
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - R. K. Brojen Singh
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Chuan-Ju Liu
- Department of Orthopedics Surgery, New York University School of Medicine, New York, NY, United States
| | - Ranjan Kumar Nanda
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
- *Correspondence: Debasis Sahu, ; Ranjan Kumar Nanda, ; Amulya Kumar Panda,
| | - Amulya Kumar Panda
- Product Development Cell, National Institute of Immunology, New Delhi, India
- *Correspondence: Debasis Sahu, ; Ranjan Kumar Nanda, ; Amulya Kumar Panda,
| |
Collapse
|
8
|
Herbal Active Ingredients: Potential for the Prevention and Treatment of Acute Lung Injury. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5543185. [PMID: 34258266 PMCID: PMC8245226 DOI: 10.1155/2021/5543185] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023]
Abstract
Acute lung injury (ALI) is a life-threatening clinical syndrome with high morbidity and mortality. The main pathological features of ALI are increased alveolar-capillary membrane permeability, edema, uncontrolled migration of neutrophils to the lungs, and diffuse alveolar damage, resulting in acute hypoxemic respiratory failure. Glucocorticoids, aspirin, and other anti-inflammatory drugs are commonly used to treat ALI. Respiratory supports, such as a ventilator, are used to alleviate hypoxemia. Many treatment methods are available, but they cannot significantly ameliorate the quality of life of patients with ALI and reduce mortality rates. Herbal active ingredients, such as flavonoids, terpenoids, saponins, alkaloids, and quinonoids, exhibit advantages for ALI prevention and treatment, but the underlying mechanism needs further study. This paper summarizes the role of herbal active ingredients in anti-ALI therapy and progresses in the understanding of their mechanisms. The work also provides some references and insights for the discovery and development of novel drugs for ALI prevention and treatment.
Collapse
|
9
|
He YQ, Zhou CC, Yu LY, Wang L, Deng JL, Tao YL, Zhang F, Chen WS. Natural product derived phytochemicals in managing acute lung injury by multiple mechanisms. Pharmacol Res 2021; 163:105224. [PMID: 33007416 PMCID: PMC7522693 DOI: 10.1016/j.phrs.2020.105224] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 12/14/2022]
Abstract
Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS) as common life-threatening lung diseases with high mortality rates are mostly associated with acute and severe inflammation in lungs. With increasing in-depth studies of ALI/ARDS, significant breakthroughs have been made, however, there are still no effective pharmacological therapies for treatment of ALI/ARDS. Especially, the novel coronavirus pneumonia (COVID-19) is ravaging the globe, and causes severe respiratory distress syndrome. Therefore, developing new drugs for therapy of ALI/ARDS is in great demand, which might also be helpful for treatment of COVID-19. Natural compounds have always inspired drug development, and numerous natural products have shown potential therapeutic effects on ALI/ARDS. Therefore, this review focuses on the potential therapeutic effects of natural compounds on ALI and the underlying mechanisms. Overall, the review discusses 159 compounds and summarizes more than 400 references to present the protective effects of natural compounds against ALI and the underlying mechanism.
Collapse
Affiliation(s)
- Yu-Qiong He
- Institute of Chinese Materia Madica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Can-Can Zhou
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Lu-Yao Yu
- Institute of Chinese Materia Madica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Liang Wang
- Institute of Chinese Materia Madica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiu-Ling Deng
- Institute of Chinese Materia Madica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yu-Long Tao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Feng Zhang
- Institute of Chinese Materia Madica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.
| | - Wan-Sheng Chen
- Institute of Chinese Materia Madica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.
| |
Collapse
|
10
|
Jafari-Khataylou Y, Emami SJ, Mirzakhani N. Troxerutin attenuates inflammatory response in lipopolysaccharide-induced sepsis in mice. Res Vet Sci 2020; 135:469-478. [PMID: 33261826 DOI: 10.1016/j.rvsc.2020.11.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 11/17/2020] [Accepted: 11/22/2020] [Indexed: 12/16/2022]
Abstract
Troxerutin (Tx), known as vitamin P4 is a derivative of natural bioflavonoid rutin. Tx possesses different biological activities such as antioxidant, anticancer, and anti-inflammatory. The current study was conducted to determine potential therapeutic effect of Tx in lipopolysaccharides (LPS)-induced sepsis in mice. In LPS-induced sepsis, the mice were treated intraperitoneally (ip) with Tx twice daily. Therapeutic effect was assessed by measuring serum level of cytokines, alanine aminotransferase (ALT) and lactate dehydrogenase (LDH). Level of nitric oxide (NO), superoxide dismutase (SOD), catalase (CAT), Myeloperoxidase (MPO) and Malondialdehyde (MDA) was measured. Expression of CD40 receptor on leucocytes was measured using flowcytometry. Splenocyte proliferation was evaluated using MTT assay. The effect of Tx on survival rate during administration of lethal dose of LPS was investigated. The results showed that Tx inhibited LPS induced NO production. Inflammatory pathways were suppressed by reduction of inflammatory cytokines production. Further, elevated CD40 expression of leucocytes and proliferation of splenocytes markedly reduced in Tx treated group. Antioxidant defense system was enhanced by increased activity of SOD and CAT and decreased level of MDA. MPO, ALT and LDH activity. Additionally, treatment with Tx significantly increased the mean survival time of mice compared with the LPS treated group. Histologically, Tx treatment decreased inflammatory cells infiltration and histopathologicl changes in the liver. Our findings showed that reduced inflammatory parameters, improved antioxidant activity, reduced histological lesions and increased survival rate. These findings suggest that Tx is an effective anti-inflammatory agent for the treatment of LPS-induced sepsis.
Collapse
Affiliation(s)
- Yaser Jafari-Khataylou
- Assistant Professor of Immunology, Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Seyyed Jamal Emami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Navideh Mirzakhani
- Assistant Professor of Pathology, Department of Pathobiology, Faculty of Veterinary Medicine, Amol University of Special Modern Technologies, Amol, Iran
| |
Collapse
|
11
|
Vidhya R, Anuradha CV. Anti-inflammatory effects of troxerutin are mediated through elastase inhibition. Immunopharmacol Immunotoxicol 2020; 42:423-435. [PMID: 32762381 DOI: 10.1080/08923973.2020.1806870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
CONTEXT Obesity is a chronic low-grade inflammatory state associated with immune cell infiltration into the adipose tissue (AT). We hypothesize that the anti-obesity and anti-inflammatory effects of troxerutin (TX) are mediated through inhibition of elastase. OBJECTIVE To determine the inhibitory effect of TX on elastase in vitro and in tumor necrosis factor alpha (TNFα) induced 3T3-L1 adipocytes and the molecular interaction of TX with human neutrophil elastase (HNE). MATERIALS AND METHODS Differentiated 3T3-L1 adipocytes were pretreated with TX, elastatinal (ELAS) or sodium salicylate (SAL) before exposure to TNFα. Lipid accumulation, reactive oxygen species (ROS) generation and oxidant-antioxidant balance were examined. The mRNA and protein expression of TNFα, interleukin-6, monocyte chemoattractant protein-1, adiponectin, leptin, resistin, chemerin, and elastase were analyzed. Elastase inhibition by TX and ELAS in a cell free system and docking studies for HNE with TX and ELAS were performed. RESULTS TX, ELAS or SAL pretreatment had lowered lipid droplets formation and TG content. TX suppressed ROS generation, oxidative stress and improved antioxidant status. The expression of inflammatory cytokines and elastase was downregulated while that of adiponectin was upregulated by TX. The concentration required to produce 50% inhibition in vitro (IC50) was 11.5 μM for TX and 16.9 μM for ELAS. TX showed hydrogen bonding and hydrophobic interactions with elastase. DISCUSSION TNFα induces inflammation of 3T3-L1 cells through elastase activation. TX inhibits elastase activity, downregulates expression and binds with elastase. CONCLUSION The antioxidant and anti-inflammatory activities of TX in AT could be of relevance in the management of obesity.
Collapse
Affiliation(s)
- Ramachandran Vidhya
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar, Tamil Nadu, India
| | | |
Collapse
|