1
|
Asadi Y, Moundounga RK, Chakroborty A, Pokokiri A, Wang H. FOXOs and their roles in acute and chronic neurological disorders. Front Mol Biosci 2025; 12:1538472. [PMID: 40260403 PMCID: PMC12010098 DOI: 10.3389/fmolb.2025.1538472] [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: 12/02/2024] [Accepted: 02/10/2025] [Indexed: 04/23/2025] Open
Abstract
The forkhead family of transcription factors of class O (FOXOs) consisting of four functionally related proteins, FOXO1, FOXO3, FOXO4, and FOXO6, are mammalian homologs of daf-16 in Caenorhabditis elegans and were previously identified as tumor suppressors, oxidative stress sensors, and cell survival modulators. Under normal physiological conditions, FOXO protein activities are negatively regulated by phosphorylation via the phosphoinositide 3-kinase (PI3K)-Akt pathway, a well-known cell survival pathway: Akt phosphorylates FOXOs to inactivate their transcriptional activity by relocalizing FOXOs from the nucleus to the cytoplasm for degradation. However, under oxidative stress or absent the cellular survival drive of growth factors, FOXO proteins translocate to the nucleus and upregulate a series of target genes, thereby promoting cell growth arrest and cell death and altering mitochondrial homeostasis. FOXO gene expression is also regulated by other transcriptional factors such as p53 or autoregulation by their activities and end products. Here we summarize the structure, posttranslational modifications, and translocation of FOXOs linking to their transcriptional control of cellular functions, survival, and death, emphasizing their role in regulating the cellular response to some acute insults and chronic neurological disorders. This review will conclude with a brief section on potential therapeutic interventions that can be used to modulate FOXOs' activities when treating acute and chronic neurological disorders.
Collapse
Affiliation(s)
- Yasin Asadi
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Rozenn K. Moundounga
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Anand Chakroborty
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Augustina Pokokiri
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Hongmin Wang
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
2
|
Cong S, Wang N, Pei H, Li Z, Meng Y, Maimaitituersun S, Zhao X, Wan R, Wan Q, Luo L, Bian Y, Wen W, Cui H. Syringin inhibits the crosstalk between macrophages and fibroblast-like synoviocytes to treat rheumatoid arthritis via PDE4. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156401. [PMID: 39842374 DOI: 10.1016/j.phymed.2025.156401] [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: 11/06/2024] [Revised: 12/28/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Syringin (SRG) is well-known for its anti-inflammatory effects. However, its pharmacological mechanisms against rheumatoid arthritis (RA) are not fully understood. MATERIALS AND METHODS We assessed the anti-RA effects of SRG using a collagen-induced arthritis (CIA) rat model. And, we employed single-cell RNA sequencing (scRNA-seq) to analyze the changes in cell types and gene expression in the synovial tissues. Building on these observations, we investigated the effects of SRG on M1 macrophage polarization and RA-fibroblast-like synoviocytes (FLS) proliferation. RESULTS Our findings highlighted the anti-RA effects of SRG on CIA rat. scRNA-seq of rat synovial tissues revealed significant changes in M1 and RA-FLS. Specifically, SRG decreased the levels of inflammatory factors in the supernatants of LPS and IFN-γ induced THP-1 cells and downregulated M1-polarized markers in these cells. Further analysis indicated that SRG's regulation of phosphodiesterase 4 (PDE4) and its associated factors was crucial for its anti-M1 polarization effects. Besides, we found that SRG inhibited the activation of FLS in vivo but showed no direct effects on RA-FLS in vitro. However, in RA-FLS, co-cultured with supernatant from SRG-treated M1-polarized THP-1 cells exhibited lower ability of cell proliferation and activation as compared to co-cultured with supernatant from M1-polarized THP-1 cells. CONCLUSION By integrating scRNA-seq analysis with in vivo and in vitro validations, our study revealed that SRG achieved its anti-RA effects by blocking the interaction between macrophages and RA-FLS, with PDE4 playing a central role. This study may provide a novel research paradigm in studying the multi-cell regulatory mechanisms of natural compounds.
Collapse
Affiliation(s)
- Shan Cong
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi 830054, China
| | - Ning Wang
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Huan Pei
- School of Basic Medical, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Zixuan Li
- Medical School, Tibet University, Lhasa 850000, China
| | - Yan Meng
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi 830054, China
| | - Saimire Maimaitituersun
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi 830054, China
| | - Xue Zhao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi 830054, China
| | - Rong Wan
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi 830054, China
| | - Qianqian Wan
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Li Luo
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi 830054, China.
| | - Yuhong Bian
- School of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Weibo Wen
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China.
| | - Huantian Cui
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China.
| |
Collapse
|
3
|
Zhang L, Hu Z, Li Z, Lin Y. Crosstalk among mitophagy, pyroptosis, ferroptosis, and necroptosis in central nervous system injuries. Neural Regen Res 2024; 19:1660-1670. [PMID: 38103229 PMCID: PMC10960298 DOI: 10.4103/1673-5374.389361] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/28/2023] [Accepted: 09/24/2023] [Indexed: 12/18/2023] Open
Abstract
Central nervous system injuries have a high rate of resulting in disability and mortality; however, at present, effective treatments are lacking. Programmed cell death, which is a genetically determined form of active and ordered cell death with many types, has recently attracted increasing attention due to its functions in determining the fate of cell survival. A growing number of studies have suggested that programmed cell death is involved in central nervous system injuries and plays an important role in the progression of brain damage. In this review, we provide an overview of the role of programmed cell death in central nervous system injuries, including the pathways involved in mitophagy, pyroptosis, ferroptosis, and necroptosis, and the underlying mechanisms by which mitophagy regulates pyroptosis, ferroptosis, and necroptosis. We also discuss the new direction of therapeutic strategies targeting mitophagy for the treatment of central nervous system injuries, with the aim to determine the connection between programmed cell death and central nervous system injuries and to identify new therapies to modulate programmed cell death following central nervous system injury. In conclusion, based on these properties and effects, interventions targeting programmed cell death could be developed as potential therapeutic agents for central nervous system injury patients.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zhigang Hu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zhenxing Li
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yixing Lin
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| |
Collapse
|
4
|
Qian Q, Pan J, Yang J, Wang R, Luo K, Wu Z, Ma S, Wang Y, Li M, Gao Y. Syringin: a naturally occurring compound with medicinal properties. Front Pharmacol 2024; 15:1435524. [PMID: 39104400 PMCID: PMC11298447 DOI: 10.3389/fphar.2024.1435524] [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: 05/20/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Syringin, a phenylpropanoid glycoside, is widely distributed in various plants, such as Acanthopanax senticosus (Rupr. et Maxim.) Harms, Syringa reticulata (BL) Hara var. mandshurica (Maxim.) Hara, and Ilex rotunda Thumb. It serves as the main ingredient in numerous listed medicines, health products, and foods with immunomodulatory, anti-tumor, antihyperglycemic, and antihyperlipidemic effects. This review aims to systematically summarize syringin, including its physicochemical properties, plant sources, extraction and separation methods, total synthesis approaches, pharmacological activities, drug safety profiles, and preparations and applications. It will also cover the pharmacokinetics of syringin, followed by suggestions for future application prospects. The information on syringin was obtained from internationally recognized scientific databases through the Internet (PubMed, CNKI, Google Scholar, Baidu Scholar, Web of Science, Medline Plus, ACS Elsevier, and Flora of China) and libraries. Syringin, extraction and separation, pharmacological activities, preparations and applications, and pharmacokinetics were chosen as the keywords. According to statistics, syringin can be found in 23 families more than 60 genera, and over 100 species of plants. As a key component in many Chinese herbal medicines, syringin holds significant research value due to its unique sinapyl alcohol structure. Its diverse pharmacological effects include immunomodulatory activity, tumor suppression, hypoglycemic action, and hypolipidemic effects. Additionally, it has been shown to provide neuroprotection, liver protection, radiation protection, cardioprotection, and bone protection. Related preparations such as Aidi injection, compound cantharidin capsule, and Tanreqing injection have been widely used in clinical settings. Other studies on syringin such as extraction and isolation, total synthesis, safety profile assessment, and pharmacokinetics have also made progress. It is crucial for medical research to deeply explore its mechanism of action, especially regarding immunity and tumor therapy. Meanwhile, more robust support is needed to improve the utilization of plant resources and to develop extraction means adapted to the needs of industrial biochemistry to further promote economic development while protecting people's health.
Collapse
Affiliation(s)
- Qingyuan Qian
- College of Pharmacy, Lanzhou University, Lanzhou, China
- Institute of Radiation Medicine Sciences, Beijing, China
| | - Jinchao Pan
- Institute of Radiation Medicine Sciences, Beijing, China
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Jun Yang
- Institute of Radiation Medicine Sciences, Beijing, China
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
| | - Renjie Wang
- Institute of Radiation Medicine Sciences, Beijing, China
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
| | - Kai Luo
- Institute of Radiation Medicine Sciences, Beijing, China
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhenhui Wu
- Institute of Radiation Medicine Sciences, Beijing, China
| | - Shuhe Ma
- Institute of Radiation Medicine Sciences, Beijing, China
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yuguang Wang
- Institute of Radiation Medicine Sciences, Beijing, China
| | - Maoxing Li
- College of Pharmacy, Lanzhou University, Lanzhou, China
- Institute of Radiation Medicine Sciences, Beijing, China
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yue Gao
- Institute of Radiation Medicine Sciences, Beijing, China
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| |
Collapse
|
5
|
Liu QQ, Wu GH, Wang XC, Xiong XW, Rui-Wang, Yao BL. The role of Foxo3a in neuron-mediated cognitive impairment. Front Mol Neurosci 2024; 17:1424561. [PMID: 38962803 PMCID: PMC11220205 DOI: 10.3389/fnmol.2024.1424561] [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: 04/28/2024] [Accepted: 06/06/2024] [Indexed: 07/05/2024] Open
Abstract
Cognitive impairment (COI) is a prevalent complication across a spectrum of brain disorders, underpinned by intricate mechanisms yet to be fully elucidated. Neurons, the principal cell population of the nervous system, orchestrate cognitive processes and govern cognitive balance. Extensive inquiry has spotlighted the involvement of Foxo3a in COI. The regulatory cascade of Foxo3a transactivation implicates multiple downstream signaling pathways encompassing mitochondrial function, oxidative stress, autophagy, and apoptosis, collectively affecting neuronal activity. Notably, the expression and activity profile of neuronal Foxo3a are subject to modulation via various modalities, including methylation of promoter, phosphorylation and acetylation of protein. Furthermore, upstream pathways such as PI3K/AKT, the SIRT family, and diverse micro-RNAs intricately interface with Foxo3a, engendering alterations in neuronal function. Through several downstream routes, Foxo3a regulates neuronal dynamics, thereby modulating the onset or amelioration of COI in Alzheimer's disease, stroke, ischemic brain injury, Parkinson's disease, and traumatic brain injury. Foxo3a is a potential therapeutic cognitive target, and clinical drugs or multiple small molecules have been preliminarily shown to have cognitive-enhancing effects that indirectly affect Foxo3a. Particularly noteworthy are multiple randomized, controlled, placebo clinical trials illustrating the significant cognitive enhancement achievable through autophagy modulation. Here, we discussed the role of Foxo3a in neuron-mediated COI and common cognitively impaired diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Bao-Le Yao
- Department of Rehabilitation Medicine, Ganzhou People’s Hospital, Ganzhou, China
| |
Collapse
|
6
|
Xie M, Long H, Tian S, Zhu Z, Meng P, Du K, Wang Y, Guo D, Wang H, Peng Q. Saikosaponin F ameliorates depression-associated dry eye disease by inhibiting TRIM8-induced TAK1 ubiquitination. Int Immunopharmacol 2024; 130:111749. [PMID: 38430804 DOI: 10.1016/j.intimp.2024.111749] [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: 10/19/2023] [Revised: 02/15/2024] [Accepted: 02/21/2024] [Indexed: 03/05/2024]
Abstract
AIMS Saikosaponin F (SsF) is one of the major active ingredients of Radix Bupleuri, an herb widely used in the treatment of depression. Studies have shown that dry eye disease often occurs together with depression. The aim of this study is to investigate whether SsF can improve depression-associated dry eye disease and explore the underlying mechanism. METHODS Behavioral test was used to verify the effect of SsF on CUMS-induced depression-like behaviors in mice. Corneal fluorescein staining, phenol red cotton thread test and periodic acid-Schiff (PAS) staining were used to observe the effect of SsF on depression-associated dry eye disease. Western blot (WB) was performed to observe the expression of TAK1 protein and key proteins of NF-κB and MAPK (P38) inflammatory pathways in the hippocampus and cornea. Immunohistochemical staining was used to observe the expression of microglia, and immunoprecipitation was used to observe K63-linked TAK1 ubiquitination. Subsequently, we constructed a viral vector sh-TAK1 to silence TAK1 protein to verify whether SsF exerted its therapeutic effect based on TAK1. The expression of inflammatory factors such as IL-1β, TNF-α and IL-18 in hippocampus and cornea were detected by ELISA. Overexpression of TRIM8 (OE-TRIM8) by viral vector was used to verify whether SsF improved depression-associated dry eye disease based on TRIM8. RESULTS SsF treatment significantly improved the depression-like behavior, increased tear production and restored corneal injury in depression-related dry eye model mice. SsF treatment downregulated TAK1 expression and TRIM8-induced K63-linked TAK1 polyubiquitination, while inhibiting the activation of NF-κB and MAPK (P38) inflammatory pathways and microglial expression. In addition, selective inhibition of TAK1 expression ameliorated depression-associated dry eye disease, while overexpression of TRIM8 attenuated the therapeutic effect of SsF on depression-associated dry eye disease. CONCLUSION SsF inhibited the polyubiquitination of TAK1 by acting on TRIM8, resulting in the downregulation of TAK1 expression, inhibition of inflammatory response, and improvement of CUMS-induced depression-associated dry eye disease.
Collapse
Affiliation(s)
- Mingxia Xie
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Hongping Long
- Center for Medical Research and Innovation, The First Hospital of Hunan University of Chinese Medicine, Changsha 410002, China
| | - Sainan Tian
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhengqing Zhu
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Pan Meng
- Center for Medical Research and Innovation, The First Hospital of Hunan University of Chinese Medicine, Changsha 410002, China; College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ke Du
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yajing Wang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Dongwei Guo
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Hanqing Wang
- College of Pharmacy, Ningxia Medical University, Yinchuan 750003, China.
| | - Qinghua Peng
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; Center for Medical Research and Innovation, The First Hospital of Hunan University of Chinese Medicine, Changsha 410002, China.
| |
Collapse
|
7
|
Huang X, Tan J, Ji Y, Luo J, Zhao Y, Zhao J. BRCC3 mediates inflammation and pyroptosis in cerebral ischemia/reperfusion injury by activating the NLRP6 inflammasome. CNS Neurosci Ther 2024; 30:e14697. [PMID: 38544474 PMCID: PMC10973773 DOI: 10.1111/cns.14697] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 04/04/2024] Open
Abstract
AIMS Neuroinflammation and pyroptosis are key mediators of cerebral ischemia/reperfusion (I/R) injury-induced pathogenic cascades. BRCC3, the human homolog of BRCC36, is implicated in neurological disorders and plays a crucial role in neuroinflammation and pyroptosis. However, its effects and potential mechanisms in cerebral I/R injury in mice are unclear. METHODS Cellular localization of BRCC3 and the interaction between BRCC3 and NLRP6 were assessed. Middle cerebral artery occlusion/reperfusion (MCAO) and oxygen-glucose deprivation/reoxygenation (OGD/R) models were established in mice and HT22 cells, respectively, to simulate cerebral I/R injury in vivo and in vitro. RESULTS BRCC3 protein expression peaked 24 h after MCAO and OGD/R. BRCC3 knockdown reduced the inflammation and pyroptosis caused by cerebral I/R injury and ameliorated neurological deficits in mice after MCAO. The effects of BRCC3 on inflammation and pyroptosis may be mediated by NLRP6 inflammasome activation. Moreover, both BRCC3 and its N- and C-terminals interacted with NLRP6, and both BRCC3 and its terminals reduced NLRP6 ubiquitination. Additionally, BRCC3 affected the interaction between NLRP6 and ASC, which may be related to inflammasome activation. CONCLUSION BRCC3 shows promise as a novel target to enhance neurological recovery and attenuate the inflammatory responses and pyroptosis caused by NLRP6 activation in cerebral I/R injury.
Collapse
Affiliation(s)
- Xiaohuan Huang
- Department of PathologyChongqing Medical UniversityChongqingChina
- Department of PathologyChongqing Three Gorges Medical CollegeWanzhouChina
| | - Junyi Tan
- Department of PathophysiologyChongqing Medical UniversityChongqingChina
| | - Yanyan Ji
- Department of PathologyChongqing Medical UniversityChongqingChina
| | - Jing Luo
- Department of PathologyChongqing Medical UniversityChongqingChina
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yong Zhao
- Department of PathologyChongqing Medical UniversityChongqingChina
| | - Jing Zhao
- Department of PathophysiologyChongqing Medical UniversityChongqingChina
| |
Collapse
|
8
|
Wang J, He W, Yue H, Zhao P, Li J. Effective-components combination alleviates PM2.5-induced inflammation by evoking macrophage autophagy in COPD. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117537. [PMID: 38043756 DOI: 10.1016/j.jep.2023.117537] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bufei Yishen formula (BYF) is clinically used to treat chronic obstructive pulmonary disease (COPD). Effective-component compatibility (ECC) is a combination of five active components derived from BYF, which has an equal effect on COPD to BYF. Our previous study has also demonstrated that ECC can protect COPD rats against PM2.5 exposure. However, the precise mechanisms remain to be elucidated. AIM OF THE STUDY To explore the mechanism underlying the anti-inflammatory effects of ECC-BYF against PM2.5-accelerated COPD. MATERIALS AND METHODS MH-S macrophages were stimulated by PM2.5 suspension to establish an in vitro model. Western blotting and immunofluorescent staining were used to measure the protein levels of autophagy markers. ELISA and quantitative PCR were used to detect the levels of inflammatory cytokines. In vivo, an established PM2.5-accelerated COPD rat model was used to determine the protective effect of ECC-BYF. Lung function, pathology, autophagy, and inflammatory mediators were detected. RESULTS Firstly, we observed a significantly increased number of macrophages in the lungs upon PM2.5 exposure. Then, decreased autophagy flux while elevated inflammation was detected in PM2.5-exposed rats and MH-S cells. In MH-S cells, ECC-BYF significantly suppressed the PM2.5-increased inflammatory cytokines production, which was accompanied by the enhancement of autophagy flux. An autophagy inhibitor counteracted the anti-inflammatory effect elicited by ECC-BYF. In addition, ECC-BYF stimulated Foxo3 nuclear translocation and upregulated Foxo3 expression, whereas Foxo3 knockdown abrogated the inhibitory effect of ECC-BYF on inflammation. In PM2.5-accelerated COPD rats, ECC-BYF also attenuated the autophagy disruption and increased Foxo3 in the lungs, finally resulting in a suppression of pulmonary inflammation and an enhancement of lung function. CONCLUSION ECC-BYF can ameliorate PM2.5-aggravated inflammation in COPD, which might be associated with the enhancement of autophagy flux in alveolar macrophages through the activation of Foxo3 signals.
Collapse
Affiliation(s)
- Jing Wang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, Henan Province, 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China.
| | - Weijing He
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, Henan Province, 450046, China
| | - Huiyu Yue
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, Henan Province, 450046, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, Henan Province, 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, Henan Province, 450046, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China.
| |
Collapse
|
9
|
Xu LL, Xie JQ, Shen JJ, Ying MD, Chen XZ. Neuron-derived exosomes mediate sevoflurane-induced neurotoxicity in neonatal mice via transferring lncRNA Gas5 and promoting M1 polarization of microglia. Acta Pharmacol Sin 2024; 45:298-311. [PMID: 37803140 PMCID: PMC10789735 DOI: 10.1038/s41401-023-01173-9] [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: 05/16/2023] [Accepted: 09/17/2023] [Indexed: 10/08/2023]
Abstract
Sevoflurane exposure during rapid brain development induces neuronal apoptosis and causes memory and cognitive deficits in neonatal mice. Exosomes that transfer genetic materials including long non-coding RNAs (lncRNAs) between cells play a critical role in intercellular communication. However, the lncRNAs found in exosomes derived from neurons treated with sevoflurane and their potential role in promoting neurotoxicity remain unknown. In this study, we investigated the role of cross-talk of newborn mouse neurons with microglial cells in sevoflurane-induced neurotoxicity. Mouse hippocampal neuronal HT22 cells were exposed to sevoflurane, and then co-cultured with BV2 microglial cells. We showed that sevoflurane treatment markedly increased the expression of the lncRNA growth arrest-specific 5 (Gas5) in neuron-derived extracellular vesicles, which inhibited neuronal proliferation and induced neuronal apoptosis by promoting M1 polarization of microglia and the release of inflammatory cytokines. We further revealed that the exosomal lncRNA Gas5 significantly upregulated Foxo3 as a competitive endogenous RNA of miR-212-3p in BV2 cells, and activated the NF-κB pathway to promote M1 microglial polarization and the secretion of inflammatory cytokines, thereby exacerbating neuronal damage. In neonatal mice, intracranial injection of the exosomes derived from sevoflurane-treated neurons into the bilateral hippocampi significantly increased the proportion of M1 microglia, inhibited neuronal proliferation and promoted apoptosis, ultimately leading to neurotoxicity. Similar results were observed in vitro in BV2 cells treated with the CM from HT22 cells after sevoflurane exposure. We conclude that sevoflurane induces the transfer of lncRNA Gas5-containing exosomes from neurons, which in turn regulates the M1 polarization of microglia and contributes to neurotoxicity. Thus, modulating the expression of lncRNA Gas5 or the secretion of exosomes could be a strategy for addressing sevoflurane-induced neurotoxicity.
Collapse
Affiliation(s)
- Li-Li Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Jia-Qian Xie
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jian-Jun Shen
- Department of Anesthesia, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Mei-Dan Ying
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China.
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Xin-Zhong Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
10
|
R N, Kh M, Hegde SN, Begum N, Kukkupuni SK, Gowda M, Narendran P. De novo genome assembly and annotation of the medicinal plant Tinospora cordifolia (Willd.) Miers ex Hook. f. & Thom's. Funct Integr Genomics 2023; 23:330. [PMID: 37935874 DOI: 10.1007/s10142-023-01262-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 11/09/2023]
Abstract
Indian natural climbing shrub Tinospora cordifolia, often known as "Guduchi" and "Amrita," is a highly esteemed medicinal plant in the Indian system of medicine (ISM). It is a member of the Menispermaceae family which consists of a rich source of protein, micronutrients, and rich source of bioactive components which are used in treating various systemic diseases. The current study was designed to know the biological characterization of the plant genome and biosynthesis of plant metabolites essential for its medicinal applications. Tinospora cordifolia's complete genome was sequenced using Illumina HiSeq2500 sequencing technology. The draft genome was assembled through a de novo method. An integrative genome annotation approach was used to perform functional gene prediction. The pathway analysis was carried out using the KEGG database. The total genome size obtained after genome assembly was 894 Mb with an N50 of 9148 bp. The integrative annotation approach resulted in 35,111 protein-coding genes. In addition, genes responsible for the synthesis of syringin, a secondary metabolite found in plants, were identified. In comparison to the standard drug (dopamine, rasagiline, and selegiline), syringin's molecular docking exhibited a greater binding affinity from the range of - 4.3 to - 6.6 kcal/mol for all the targets of Parkinson's disease and for Alzheimer's targets; it has shown the maximum potency from the range of - 6.5 to - 7.4 kcal/mol with respect to the standard drug (donepezil, galantamine, and rivastigmine). This study provides the genomic information of Tinospora cordifolia which is helpful in understanding genomic insights and metabolic pathways connected to the corresponding plant genome and predicts the possible useful effect for the molecular characterization of therapeutic drugs.
Collapse
Affiliation(s)
- Namitha R
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), 74/2, Post Attur via Yelahanka, Jarakabande Kaval, Bengaluru, 560 064, India
| | - Manasa Kh
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), 74/2, Post Attur via Yelahanka, Jarakabande Kaval, Bengaluru, 560 064, India
| | - Santhosh N Hegde
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), 74/2, Post Attur via Yelahanka, Jarakabande Kaval, Bengaluru, 560 064, India
| | - Noorunnisa Begum
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), 74/2, Post Attur via Yelahanka, Jarakabande Kaval, Bengaluru, 560 064, India
| | - Subrahmanya Kumar Kukkupuni
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), 74/2, Post Attur via Yelahanka, Jarakabande Kaval, Bengaluru, 560 064, India
| | - Malali Gowda
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), 74/2, Post Attur via Yelahanka, Jarakabande Kaval, Bengaluru, 560 064, India.
| | - Pavithra Narendran
- The University of Trans-Disciplinary Health Sciences and Technology (TDU), 74/2, Post Attur via Yelahanka, Jarakabande Kaval, Bengaluru, 560 064, India.
| |
Collapse
|
11
|
Singh VK, Thakur DC, Rajak N, Mishra A, Kumar A, Giri R, Garg N. The multi-protein targeting potential of bioactive syringin in inflammatory diseases: using molecular modelling and in-silico analysis of regulatory elements. J Biomol Struct Dyn 2023; 42:12851-12862. [PMID: 37882327 DOI: 10.1080/07391102.2023.2273440] [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: 08/02/2023] [Accepted: 10/14/2023] [Indexed: 10/27/2023]
Abstract
Inflammation plays a crucial role in the onset or progression of a variety of acute and chronic diseases. Non-steroidal anti-inflammatory drugs (NSAIDs) are the only available FDA-approved therapy. The therapeutic outcome of NSAIDs is still finite due to off-target effects and extreme side effects on other vital organs. Bioactive syringin has been manifested to hold anti-osteoporosis, cardiac hypertrophy, alter autophagy, anti-cancer, neuro-preventive effects, etc. However, its multi-protein targeting potential in inflammation mostly remains unexplored. In the present work, we have checked the multi-protein targeting potential of bioactive glycoside syringin in inflammatory diseases. Based on the binding score of protein-ligand complexes, glycoside syringin scored greater than -7 kcal/mol against 12 inflammatory proteins. Our molecular dynamic simulation study (200 ns) confirmed that bioactive syringin remained inside the binding cavity of inflammatory proteins (JAK1, TYK2, and COX1) in a stable conformation. Further, our co-expression analysis suggests that these genes play an essential role in multiple pathways and are regulated by multiple miRNAs. Our study demonstrates that bioactive glycoside syringin might be a multi-protein targeting potential against inflammatory diseases and could be further investigated utilizing different preclinical approaches.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Vipendra Kumar Singh
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, India
| | - D C Thakur
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, India
| | - Naina Rajak
- Faculty of Ayurveda, Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Anand Mishra
- Molecular Plant Pathology Laboratory, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
| | - Ankur Kumar
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, India
| | - Rajanish Giri
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, India
| | - Neha Garg
- Faculty of Ayurveda, Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
12
|
Jeong M, Ju Y, Kwon H, Kim Y, Hyun KY, Choi GE. Protocatechuic Acid and Syringin from Saussurea neoserrata Nakai Attenuate Prostaglandin Production in Human Keratinocytes Exposed to Airborne Particulate Matter. Curr Issues Mol Biol 2023; 45:5950-5966. [PMID: 37504292 PMCID: PMC10378452 DOI: 10.3390/cimb45070376] [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: 06/09/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/29/2023] Open
Abstract
Saussurea neoserrata Nakai offers a reliable and efficient source of antioxidants that can help alleviate adverse skin reactions triggered by air pollutants. Air pollutants, such as particulate matter (PM), have the ability to infiltrate the skin and contribute to the higher occurrence of cardiovascular, cerebrovascular, and respiratory ailments. Individuals with compromised skin barriers are particularly susceptible to the impact of PM since it can be absorbed more readily through the skin. This study investigated the impact of protocatechuic acid and syringin, obtained from the n-BuOH extract of S. neoserrata Nakai, on the release of PGE2 and PGD2 induced by PM10. Additionally, it examined the gene expression of the synthesis of PGE2 and PGD2 in human keratinocytes. The findings of this research highlight the potential of utilizing safe and efficient plant-derived antioxidants in dermatological and cosmetic applications to mitigate the negative skin reactions caused by exposure to air pollution.
Collapse
Affiliation(s)
- Myeongguk Jeong
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| | - Yeongdon Ju
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
- Medical Science Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hyeokjin Kwon
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| | - Yeeun Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| | - Kyung-Yae Hyun
- Department of Clinical Laboratory Science, Dong-Eui University, Busan 47340, Republic of Korea
| | - Go-Eun Choi
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| |
Collapse
|
13
|
ERGON EY, ÇELİK A, DİNİZ G, ÇOLAK R, ÖZDEMİR SA, ÇALKAVUR Ş, YILMAZ O. Evaluation of syringin's neuroprotective effect in a model of neonatal hypoxic-ischemic brain injury. Turk J Med Sci 2023; 53:1312-1320. [PMID: 38813032 PMCID: PMC10763795 DOI: 10.55730/1300-0144.5697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 10/26/2023] [Accepted: 06/21/2023] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND/AIM A significant cause of mortality and morbidity in the neonatal era is hypoxic-ischemic encephalopathy (HIE). This study examined the histopathological analysis and neuroprotective impact of syringin (SYR) in an experimental HIE rat model. MATERIAL AND METHODS On the 7th postnatal day, 24 Wistar albino rats were evaluated in 3 groups using the HIE model under gas anesthesia. In the experiment, Group A received 10 mg/kg SYR plus dimethyl sulfoxide (DMSO), Group B received DMSO only, and Group C served as a sham group. Immunohistochemical techniques were used to assess apoptotic cell measurement and proinflammatory cytokines (TNF-α and IL-1β primary antibodies). RESULTS Rats suffering from hypoxic-ischemic brain damage had their apoptosis assessed. The SYR and sham groups had statistically fewer cells undergoing apoptosis (p < 0.001). There was no difference between the groups in terms of IL-1β and TNF-α during immunohistochemical staining. Neuronal degeneration was significantly lower in the histological evaluation of the hippocampus in the SYR group (p = 0.01). A statistically significant difference (p = 0.01) was observed between the SYR and the control groups regarding pericellular and perivascular edema. CONCLUSION SYR reduced apoptosis, perivascular and pericellular edema, and neuronal degeneration in rat cerebral tissue. These results raise the possibility that SYR may have a neuroprotective effect on the harm brought on by HIE. This is the first investigation of SYR's function within the HIE paradigm.
Collapse
Affiliation(s)
- Ezgi Yangın ERGON
- Neonatal Intensive Care Unit, Pediatric Division, Dr Behçet Uz Children’s Education and Research Hospital, İzmir,
Turkiye
| | - Aslı ÇELİK
- Department of Laboratory Animal Science, Faculty of Health Sciences, Dokuz Eylül University, İzmir,
Turkiye
| | - Gülden DİNİZ
- Department of Pathology, Medical Faculty, İzmir Democracy University, İzmir,
Turkiye
| | - Rüya ÇOLAK
- Neonatal Intensive Care Unit, Pediatric Division, Medikal Park Florya Hospital, Aydın University Medical Faculty, İstanbul,
Turkiye
| | - Senem Alkan ÖZDEMİR
- Neonatal Intensive Care Unit, Pediatric Division, Dr Behçet Uz Children’s Education and Research Hospital, İzmir,
Turkiye
| | - Şebnem ÇALKAVUR
- Neonatal Intensive Care Unit, Pediatric Division, Dr Behçet Uz Children’s Education and Research Hospital, İzmir,
Turkiye
| | - Osman YILMAZ
- Department of Laboratory Animal Science, Faculty of Health Sciences, Dokuz Eylül University, İzmir,
Turkiye
| |
Collapse
|
14
|
Chen B, Jin W. A comprehensive review of stroke-related signaling pathways and treatment in western medicine and traditional Chinese medicine. Front Neurosci 2023; 17:1200061. [PMID: 37351420 PMCID: PMC10282194 DOI: 10.3389/fnins.2023.1200061] [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: 04/04/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
This review provides insight into the complex network of signaling pathways and mechanisms involved in stroke pathophysiology. It summarizes the historical progress of stroke-related signaling pathways, identifying potential interactions between them and emphasizing that stroke is a complex network disease. Of particular interest are the Hippo signaling pathway and ferroptosis signaling pathway, which remain understudied areas of research, and are therefore a focus of the review. The involvement of multiple signaling pathways, including Sonic Hedgehog (SHH), nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE), hypoxia-inducible factor-1α (HIF-1α), PI3K/AKT, JAK/STAT, and AMPK in pathophysiological mechanisms such as oxidative stress and apoptosis, highlights the complexity of stroke. The review also delves into the details of traditional Chinese medicine (TCM) therapies such as Rehmanniae and Astragalus, providing an analysis of the recent status of western medicine in the treatment of stroke and the advantages and disadvantages of TCM and western medicine in stroke treatment. The review proposes that since stroke is a network disease, TCM has the potential and advantages of a multi-target and multi-pathway mechanism of action in the treatment of stroke. Therefore, it is suggested that future research should explore more treasures of TCM and develop new therapies from the perspective of stroke as a network disease.
Collapse
Affiliation(s)
- Binhao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Jin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
15
|
Omorou M, Huang Y, Gao M, Mu C, Xu W, Han Y, Xu H. The forkhead box O3 (FOXO3): a key player in the regulation of ischemia and reperfusion injury. Cell Mol Life Sci 2023; 80:102. [PMID: 36939886 PMCID: PMC11072419 DOI: 10.1007/s00018-023-04755-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 02/10/2023] [Accepted: 03/09/2023] [Indexed: 03/21/2023]
Abstract
Forkhead box O3 is a protein encoded by the FOXO3 gene expressed throughout the body. FOXO3 could play a crucial role in longevity and many other pathologies, such as Alzheimer's disease, glioblastoma, and stroke. This study is a comprehensive review of the expression of FOXO3 under ischemia and reperfusion (IR) and the molecular mechanisms of its regulation and function. We found that the expression level of FOXO3 under ischemia and IR is tissue-specific. Specifically, the expression level of FOXO3 is increased in the lung and intestinal epithelial cells after IR. However, FOXO3 is downregulated in the kidney after IR and in the skeletal muscles following ischemia. Interestingly, both increased and decreased FOXO3 expression have been reported in the brain, liver, and heart following IR. Nevertheless, these contribute to stimulating ischemia and reperfusion injury via the induction of inflammatory response, apoptosis, autophagy, mitophagy, pyroptosis, and oxidative damage. These results suggest that FOXO3 could play protective effects in some organs and detrimental effects in others against IR injury. Most importantly, these findings indicate that controlling FOXO3 expression, genetically or pharmacologically, could contribute to preventing or treating ischemia and reperfusion damage.
Collapse
Affiliation(s)
- Moussa Omorou
- Department of Biochemistry and Molecular Biology, Jiamusi University School of Basic Medical Sciences, Jiamusi, 154000, Heilongjiang, People's Republic of China
| | - Yiwei Huang
- Department of Biochemistry and Molecular Biology, Jiamusi University School of Basic Medical Sciences, Jiamusi, 154000, Heilongjiang, People's Republic of China
| | - Meng Gao
- Department of Biochemistry and Molecular Biology, Jiamusi University School of Basic Medical Sciences, Jiamusi, 154000, Heilongjiang, People's Republic of China
| | - Chenxi Mu
- Department of Biochemistry and Molecular Biology, Jiamusi University School of Basic Medical Sciences, Jiamusi, 154000, Heilongjiang, People's Republic of China
| | - Weijing Xu
- Department Epidemiology and Health Statistics, Jiamusi University School of Public Health, Jiamusi, 154000, Heilongjiang, People's Republic of China
| | - Yuchun Han
- Department of Biochemistry and Molecular Biology, Jiamusi University School of Basic Medical Sciences, Jiamusi, 154000, Heilongjiang, People's Republic of China
| | - Hui Xu
- Department of Biochemistry and Molecular Biology, Jiamusi University School of Basic Medical Sciences, Jiamusi, 154000, Heilongjiang, People's Republic of China.
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, Jiamusi, 154000, Heilongjiang, People's Republic of China.
| |
Collapse
|
16
|
Vasquez RD, Aventurado CA, Castillo AL. Syringin as TGF-βR1, HER2, EGFR, FGFR4 Kinase, and MMP-2
Inhibitor and Potential Cytotoxic Agent against ER+ Breast Cancer Cells. CURRENT ENZYME INHIBITION 2023; 19:55-64. [DOI: 10.2174/1573408019666221107145705] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/27/2022] [Accepted: 09/14/2022] [Indexed: 01/03/2025]
Abstract
Background:
Breast cancer is currently the most diagnosed cancer worldwide. Neoplastic
cells and components of the tumor microenvironment trigger enzymes and receptors to facilitate
cancer advancement. Syringin, a natural phenylpropanoid glycoside, has been reported to
possess anti-cancer activity and affinity with numerous druggable targets of breast carcinoma.
Objectives:
This work aims to evaluate the effects of syringin on the growth of breast cancer cells
(MCF-7) and normal dermal fibroblast cells (HDFn) and its ability to inhibit the protein targets of
breast cancer.
Methods:
Syringin was investigated on cell lines in vitro via MTT assay. Using non-cell-based activity
assay kits, its influence on the activity of transforming growth factor-beta receptor type 1
(TGF-βR1), human epidermal growth factor receptor (HER2), epidermal growth factor receptor
(EGFR), fibroblast growth factor receptor 4 (FGFR4), and matrix metalloproteinase-2 (MMP-2)
was evaluated.
Results:
Syringin exhibited significant cytotoxicity against MCF-7 cells (IC50: 32.11 μM for 24
hours and 21.35 μM for 48 hours) and was non-toxic on healthy HDFn cells (IC50: >100 μM for 24
and 48 hours). It significantly suppressed the activity of cancer and angiogenesis regulating enzymes
in vitro with commendable IC50 values on TGF-βR1 kinase (IC50: 6.48 μM), HER2 kinase
(IC50: 7.18 μM), EGFR kinase (IC50: 12.38 μM), FGFR4 kinase (IC50: 16.03 μM), and MMP-2
(IC50: 16.07 μM).
Conclusion:
Findings showed the selective toxicity of syringin on breast cancer cells and its potential
against pro-angiogenic enzymes. These discoveries strongly indicate the significance and
therapeutic potential of syringin in targeted cancer therapy.
Collapse
Affiliation(s)
- Ross D. Vasquez
- The Graduate School, University of Santo Tomas, Manila, 1015, Philippines
- Faculty of Pharmacy, University of
Santo Tomas, Manila, 1015, Philippines
- Research Center for the Natural and Applied Sciences, University of
Santo Tomas, Manila 1015, Philippines
| | | | - Agnes L. Castillo
- The Graduate School, University of Santo Tomas, Manila, 1015, Philippines
- Faculty of Pharmacy, University of
Santo Tomas, Manila, 1015, Philippines
- Research Center for the Natural and Applied Sciences, University of
Santo Tomas, Manila 1015, Philippines
| |
Collapse
|
17
|
Zhao D, Liu K, Wang J, Shao H. Syringin exerts anti-inflammatory and antioxidant effects by regulating SIRT1 signaling in rat and cell models of acute myocardial infarction. Immun Inflamm Dis 2023; 11:e775. [PMID: 36840496 PMCID: PMC9950876 DOI: 10.1002/iid3.775] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/29/2022] [Accepted: 01/13/2023] [Indexed: 02/26/2023] Open
Abstract
INTRODUCTION This study aimed to investigate the role of syringin in improving heart function during myocardial ischemia/reperfusion (I/R) and to determine whether the sirtuin 1/peroxisome proliferator-activated receptor gamma coactivator 1 alpha (SIRT1/PGC-1α) pathway contributes to this cardioprotective effect in vivo and in vitro. METHODS H9c2 cells were incubated with H2 O2 for 12 h. The effect of syringin was assessed by measuring cell viability; the apoptotic rate; Keap1/NRF2/HO-1 activation; and the levels of proinflammatory cytokines, oxidative products, and antioxidative enzymes. In addition, SIRT1 was silenced via short hairpin RNA (shRNA)-SIRT1 transfection to evaluate its involvement in syringin-mediated protection. Syringin rescued cells from H2 O2 -induced reductions in viability, antioxidative enzyme levels, and NRF2/HO-1 activation; likewise, syringin inhibited apoptosis, inflammation, and oxidative stress. We also created a rat model of I/R by ligating the left anterior descending coronary artery for 30 min, followed by reperfusion for 12 min. Syringin was then intraperitoneally injected, and the effect on infarct size and cardiac function was examined after 7 days. NRF2/HO-1 activity and the levels of myocardial proinflammatory cytokines, oxidative products, and antioxidative enzymes were measured. RESULTS In comparison to the untreated I/R group, the syringin treatment group exhibited improved cardiac function and reduced cardiac lesion and infarct size. Syringin administration also markedly reduced the levels of proinflammatory cytokines and reactive oxygen species and promoted antioxidative enzyme expression and NRF2/HO-1 pathway activation. CONCLUSIONS Syringin may serve a protective role in animal and cell models of I/R by improving cardiac function, inhibiting the inflammatory response, and activating the antioxidative response.
Collapse
Affiliation(s)
- Di Zhao
- Department of Cardiology IThe Third Affiliated Hospital of Qiqihar Medical UniversityQiqiharHeilongjiangChina
| | - Ketong Liu
- Department of CardiologyThe Fifth Affiliated Hospital (Zhuhai) of Zunyi Medical UniversityZhuhaiGuangdongChina
| | - Jian Wang
- Department of Cardiology IThe Third Affiliated Hospital of Qiqihar Medical UniversityQiqiharHeilongjiangChina
| | - Haifeng Shao
- Department of Cardiology IThe Third Affiliated Hospital of Qiqihar Medical UniversityQiqiharHeilongjiangChina
| |
Collapse
|
18
|
Meng C, Chen S, He Q, Tan J, Wu J, Zhao J. IKZF3 modulates cerebral ischemia/reperfusion injury by inhibiting neuroinflammation. Int Immunopharmacol 2023; 114:109480. [PMID: 36525791 DOI: 10.1016/j.intimp.2022.109480] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022]
Abstract
Neuroinflammation is a key mediator to the pathogenic cascades induced by cerebral ischemia-reperfusion (I/R) injury. IKZF3, a key zinc finger transcription factor in the Ikaros family, has already been shown to modulate a wide range of cell functions and the production of inflammatory mediators. However, the effects of IKZF3 on inflammation and the potential mechanism after cerebral I/R injury remain unclear. In this study, we evaluated the effect of IKZF3 on HT-22 cells under oxygen-glucose deprivation and reoxygenation (OGD/R) in vitro and in mice with MACO in vivo. We found that IKZF3 expression peaked at 12 h after MCAO and OGD/R, and there was high expression of IKZF3 in brain tissues and HT-22 cells. IKZF3 knockdown exacerbated the damage by OGD-induced HT-22 cells injury and MCAO-induced brain injury in mice by regulating the production of inflammatory factors, which promoted the phosphorylation and nuclear transfer of NF-ĸB and may bind with NF-ĸB-p65 in vivo and in vitro. Our results suggested that IKZF3 may provide a new target in improve neurological recovery and reducing neuroinflammation after cerebral I/R injury.
Collapse
Affiliation(s)
- Changchang Meng
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China
| | - Shiyu Chen
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China
| | - Qi He
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China
| | - Junyi Tan
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China
| | - Jingxian Wu
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China.
| | - Jing Zhao
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
19
|
Du Y, Li C, Xu S, Yang J, Wan H, He Y. LC-MS/MS combined with blood-brain dual channel microdialysis for simultaneous determination of active components of astragali radix-safflower combination and neurotransmitters in rats with cerebral ischemia reperfusion injury: Application in pharmacokinetic and pharmacodynamic study. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154432. [PMID: 36113188 DOI: 10.1016/j.phymed.2022.154432] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/23/2022] [Accepted: 09/02/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Astragali Radix-Safflower combination (ARSC) is widely utilized in clinic to treat cerebral ischemia/reperfusion injury (CI/RI). Whereas, there is no in-depth research of the pharmacokinetics (PK) and pharmacodynamics (PD) analysis of ARSC after intragastric administration in rats with CI/RI. PURPOSE The purpose of this research is to investigate the PK characteristics of eight active ingredients (astragaloside IV, calycosin, calycosin-7-O-β-glucoside, formononetin, ononin, hydroxysafflor yellow A, syringin and vernine) of ARSC, and the regulation of neurotransmitters disorders, revealing the pharmacodynamic substance basis and the mechanism of ARSC in treating CI/RI from the molecular level. METHODS We established a new method which based on blood-brain dual channel microdialysis (MD) coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS) to continuously gather, and determine the components of ARSC and neurotransmitters related to CI/RI in vivo. The collected data were analyzed by sigmoid-Emax function. The neurotransmitters primarily regulated in CI/RI rat were discussed by principal component analysis and the compound most associated with total pharmacodynamics was chosen by partial least squares regression. RESULTS The validated LC-MS/MS method had specificity and selectivity to simultaneously analyze the concentration of eight active components of ARSC extract and five neurotransmitters of CI/RI rats. The recovery rates of brain MD probe and blood MD probe were stable within six hours. The MD probes recovery rates decreased with the increase of flow rates, but the solution concentration had little effect on the probes recovery rates. It was feasible to correct the recovery rates of probes in vivo by using reverse dialysis method. All eight active ingredients of ARSC could pass across the blood brain barrier after CI/RI. ARSC regulated the release of glutamate (Glu), γ-aminobutyric acid (GABA), dopamine (DA), 5-hydroxytryptamine (5-HT) and aspartic acid (Asp). Notably, astragaloside IV and hydroxysafflor yellow A might have better regulatory effect on neurotransmitters in comparison with other six measured components of ARSC, and Glu was the neurotransmitter mainly regulated in CI/RI rats. CONCLUSION The ARSC was able to treat CI/RI through ameliorating neurotransmitters disorders. There was a hysteresis between the peaked drug concentration and maximum therapeutic effect of ARSC. The drug effective concentrations range of ASIV, calycosin, calycosin-7-O-β-glucoside, syringin and vernine in blood microdialysate and calycosin, syringin, vernine in brain microdialysate were narrow, which need be paid attention in clinical use.
Collapse
Affiliation(s)
- Yu Du
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chang Li
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shouchao Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiehong Yang
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Haitong Wan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
20
|
Bocharova OA, Shevchenko VE, Kazeev IV, Sheichenko OP, Ionov NS, Bocharov EV, Karpova RV, Aksenov AA, Poroikov VV, Kucheryanu VG, Kosorukov VS. Analysis of Eleutherosides by Tandem Mass Spectrometry: Possibilities of Standardizing a Multi-Phytoadaptogen Formulation for Preventive Oncology. Pharm Chem J 2022. [DOI: 10.1007/s11094-022-02712-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
21
|
Li XT, Zhou JC, Zhou Y, Ren YS, Huang YH, Wang SM, Tan L, Yang ZY, Ge YW. Pharmacological effects of Eleutherococcus senticosus on the neurological disorders. Phytother Res 2022; 36:3490-3504. [PMID: 35844057 DOI: 10.1002/ptr.7555] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/02/2022] [Accepted: 06/23/2022] [Indexed: 11/07/2022]
Abstract
Eleutherococcus senticosus is a medicinal plant widely used in traditional medicine and edible remedies with effects on anti-fatigue, sleep improvement, and memory enhancement. Recently, the application of E. senticosus to neurological disorders has been a focus. However, its overall pharmacological effect on neural diseases and relevant mechanisms are needed in an in-depth summary. In this review, the traditional uses and the therapeutic effect of E. senticosus on the treatment of fatigue, depression, Alzheimer's disease, Parkinson's disease, and cerebral ischemia were summarized. In addition, the underlying mechanisms involved in the anti-oxidative damage, anti-inflammation, neurotransmitter modulation, improvement of neuronal growth, and anti-apoptosis were discussed. This review will accelerate the understanding of the neuroprotective effects brought from the E. senticosus, and impetus its development as a phytotherapy agent against neurological disorders.
Collapse
Affiliation(s)
- Xi-Tao Li
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jie-Chun Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yu Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ying-Shan Ren
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yu-Hong Huang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shu-Mei Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou, China
| | - Long Tan
- State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Zhi-You Yang
- College of Food Science and Technology, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Institute of Nutrition and Marine Drugs, Guangdong Ocean University, Zhanjiang, China
| | - Yue-Wei Ge
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
22
|
Wang F, Yuan C, Liu B, Yang YF, Wu HZ. Syringin exerts anti-breast cancer effects through PI3K-AKT and EGFR-RAS-RAF pathways. J Transl Med 2022; 20:310. [PMID: 35794555 PMCID: PMC9258109 DOI: 10.1186/s12967-022-03504-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/24/2022] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
Breast cancer (BC) is one of the most common malignant tumors with the highest mortality in the world. Modern pharmacological studies have shown that Syringin has an inhibitory effect on many tumors, but its anti-BC efficacy and mechanism are still unclear.
Methods
First, Syringin was isolated from Acanthopanax senticosus (Rupr. & Maxim.) Harms (ASH) by systematic solvent extraction and silica gel chromatography column. The plant name is composed of genus epithet, species additive words and the persons’ name who give its name. Then, the hub targets of Syringin against BC were revealed by bioinformatics. To provide a more experimental basis for later research, the hub genes which could be candidate biomarkers of BC and a ceRNA network related to them were obtained. And the potential mechanism of Syringin against BC was proved in vitro experiments.
Results
Syringin was obtained by liquid chromatography-mass spectrometry (LC–MS), nuclear magnetic resonance (NMR), and high-performance liquid chromatography (HPLC). Bioinformatics results showed that MAP2K1, PIK3CA, HRAS, EGFR, Caspase3, and PTGS2 were the hub targets of Syringin against BC. And PIK3CA and HRAS were related to the survival and prognosis of BC patients, the PIK3CA-hsa-mir-139-5p-LINC01278 and PIK3CA-hsa-mir-375 pathways might be closely related to the mechanism of Syringin against BC. In vitro experiments confirmed that Syringin inhibited the proliferation and migration and promoted apoptosis of BC cells through the above hub targets.
Conclusions
Syringin against BC via PI3K-AKT-PTGS2 and EGFR-RAS-RAF-MEK-ERK pathways, and PIK3CA and HRAS are hub genes for adjuvant treatment of BC.
Graphical Abstract
Collapse
|
23
|
The Signaling Pathways and Targets of Natural Compounds from Traditional Chinese Medicine in Treating Ischemic Stroke. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103099. [PMID: 35630576 PMCID: PMC9148018 DOI: 10.3390/molecules27103099] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 11/17/2022]
Abstract
Ischemic stroke (IS) is a common neurological disorder associated with high disability rates and mortality rates. At present, recombinant tissue plasminogen activator (r-tPA) is the only US(FDA)-approved drug for IS. However, due to the narrow therapeutic window and risk of intracerebral hemorrhage, r-tPA is currently used in less than 5% of stroke patients. Natural compounds have been widely used in the treatment of IS in China and have a wide range of therapeutic effects on IS by regulating multiple targets and signaling pathways. The keywords "ischemia stroke, traditional Chinese Medicine, Chinese herbal medicine, natural compounds" were used to search the relevant literature in PubMed and other databases over the past five years. The results showed that JAK/STAT, NF-κB, MAPK, Notch, Nrf2, and PI3K/Akt are the key pathways, and SIRT1, MMP9, TLR4, HIF-α are the key targets for the natural compounds from traditional Chinese medicine in treating IS. This study aims to update and summarize the signaling pathways and targets of natural compounds in the treatment of IS, and provide a base of information for the future development of effective treatments for IS.
Collapse
|
24
|
Wang Y, Wu H, Han Z, Sheng H, Wu Y, Wang Y, Guo X, Zhu Y, Li X, Wang Y. Guhong injection promotes post-stroke functional recovery via attenuating cortical inflammation and apoptosis in subacute stage of ischemic stroke. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:154034. [PMID: 35276592 DOI: 10.1016/j.phymed.2022.154034] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND As a leading cause of death and disability, alternative therapies for stroke are still limited by its complicated pathophysiological manifestations. Guhong injection (GHI), consisting of safflower aqueous extract and aceglutamide, has been widely applied for the clinical treatment of cerebrovascular diseases, especially ischemic stroke and post-stroke recovery, in China. Recently, a series of studies have reported the positive effect of GHI against cerebral ischemia/reperfusion injury via targeting various molecular mechanisms. However, questions remain on whether treatment with GHI contributes to better functional recovery after stroke and if so, the potential mechanisms and active substances. PURPOSE The aim of this work was to explore the potential therapeutic possibilities of GHI for the neurological and behavioral recovery after stroke and to investigate the underlying molecular mechanisms as well as active substances. METHODS The neural and motor deficits as well as cortical lesions after GHI treatment were investigated in a mouse model of transient ischemic stroke. Based on the substance identification of GHI, network pharmacology combined with an experimental verification method was used to systematically decipher the biological processes and signaling pathways closely related to GHI intervention in response to post-stroke functional outcomes. Subsequently, ingenuity pathway analysis (IPA) analysis was performed to determine the anti-stroke active substances targeting to the hub targets involved in the significant molecular pathways regulated by GHI treatment. RESULTS Therapeutically, administration of GHI observably ameliorated the post-stroke recovery of neural and locomotor function as well as reduced infarct volume and histopathological damage to the cerebral cortex in subacute stroke mice. According to 26 identified or tentatively characterized substances in GHI, the compound-target-pathway network was built. Bioinformatics analysis suggested that inflammatory and apoptotic pathways were tightly associated with the anti-stroke effect of GHI. Based on protein-protein interaction network analysis, the hub targets (such as NF-κB p65, TNF-α, IL-6, IL-1β, Bax, Bcl-2, and Caspase-3) involved in inflammation and apoptosis were selected. On the one hand, immunofluorescence and ELISA results showed that GHI (10 ml/kg) treatment obviously reduced NF-κB p65 nuclear translocation as well as decreased the abnormally elevated concentrations of proinflammatory cytokines (TNF-α, IL-6, and IL-1β) in damaged cortex tissues. On the other hand, GHI (10 ml/kg) treatment significantly downregulated the number of TUNEL-positive apoptotic cells in ischemic cortex and effectively restored the abnormal expression of Bax, Bcl-2, and Caspase-3. Based on the results of IPA, hydroxysafflor yellow A, baicalin, scutellarin, gallic acid, syringin, chlorogenic acid, kaempferol, kaempferol-3-O-β-rutinoside, and rutin acted synergistically on core targets, which could be considered as the active substances of GHI. CONCLUSION Overall, the current findings showed that the beneficial action of GHI on improving post-stroke functional recovery of subacute stroke mice partly via the modulation of cortical inflammation and apoptosis. These findings not only provide a reliable reference for the clinical application of GHI, but also shed light on a promising alternative therapeutic strategy for ischemic stroke patients.
Collapse
Affiliation(s)
- Yule Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Huimin Wu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Zhu Han
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Hongda Sheng
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Yuhan Wu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Yingchao Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, 291 Fucheng Road, Qiantang District, Hangzhou 310020, China
| | - Xinran Guo
- School of Humanities and Management, Wannan Medical College, 34 Yinhunan Road, Jinghu District, Wuhu 241001, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, Jinghai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Xuecai Li
- Tonghua Guhong Pharmaceutical Co., Ltd., 5099 Jianguo Road, Meihekou 135099, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, Jinghai District, Tianjin 301617, China.
| |
Collapse
|
25
|
Platonin protects against cerebral ischemia/reperfusion injury in rats by inhibiting NLRP3 inflammasomes via BNIP3/LC3 signaling mediated autophagy. Brain Res Bull 2022; 180:12-23. [DOI: 10.1016/j.brainresbull.2021.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/04/2021] [Accepted: 12/20/2021] [Indexed: 12/26/2022]
|
26
|
Lee SY, Lee DY, Kang JH, Jeong JW, Kim JH, Kim HW, Oh DH, Kim JM, Rhim SJ, Kim GD, Kim HS, Jang YD, Park Y, Hur SJ. Alternative experimental approaches to reduce animal use in biomedical studies. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
27
|
Todorova V, Ivanov K, Ivanova S. Comparison between the Biological Active Compounds in Plants with Adaptogenic Properties ( Rhaponticum carthamoides, Lepidium meyenii, Eleutherococcus senticosus and Panax ginseng). PLANTS (BASEL, SWITZERLAND) 2021; 11:64. [PMID: 35009068 PMCID: PMC8747685 DOI: 10.3390/plants11010064] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 06/01/2023]
Abstract
BACKGROUND In the 1960s, research into plant adaptogens began. Plants with adaptogenic properties have rich phytochemical compositions and have been used by humanity since ancient times. However, it is not still clear whether the adaptogenic properties are because of specific compounds or because of the whole plant extracts. The aim of this review is to compare the bioactive compounds in the different parts of these plants. METHODS The search strategy was based on studies related to the isolation of bioactive compounds from Rhaponticum carthamoides, Lepidium meyenii, Eleutherococcus senticosus, and Panax ginseng. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed. RESULTS This review includes data from 259 articles. The phytochemicals isolated from Rhaponticum carthamoides, Lepidium meyenii, Eleutherococcus senticosus, and Panax ginseng were described and classified in several categories. CONCLUSIONS Plant species have always played an important role in drug discovery because their effectiveness is based on the hundreds of years of experience with folk medicine in different nations. In our view, there is great potential in the near future for some of the phytochemicals found in these plants species to become pharmaceutical agents.
Collapse
Affiliation(s)
- Velislava Todorova
- Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria; (K.I.); (S.I.)
| | | | | |
Collapse
|
28
|
Medicarpin Protects Cerebral Microvascular Endothelial Cells Against Oxygen-Glucose Deprivation/Reoxygenation-Induced Injury via the PI3K/Akt/FoxO Pathway: A Study of Network Pharmacology Analysis and Experimental Validation. Neurochem Res 2021; 47:347-357. [PMID: 34523056 DOI: 10.1007/s11064-021-03449-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/14/2021] [Accepted: 09/04/2021] [Indexed: 10/20/2022]
Abstract
Medicarpin, a pterocarpan class of naturally occurring phytoestrogen possesses various biological functions. However, the effect of medicarpin on oxygen-glucose deprivation-reoxygenation (OGD/R)-induced injury in human cerebral microvascular endothelial cells (HCMECs) remains largely unknown. Target genes of medicarpin were predicted from PharmMapper. Target genes of ischemic stroke were predicted from public databases GeneCards and DisGeNET. Kyoto Encyclopedia of Genes and Genomes pathway enrichment of the intersecting targets was analyzed via DAVID 6.8. Cell viability was evaluated using CCK-8 assay. Malondialdehyde content, superoxide dismutase activity, and glutathione level were detected using corresponding commercially available kits. Cell death was assessed by TUNEL assays. Expression of protein kinase B (Akt), phosphorylated-Akt, forkhead box protein O1, phosphorylated-FoxO1, FoxO3a, and phosphorylated-FoxO3a (p-FoxO3a) was detected by western blot analysis. The intersecting targets of medicarpin and ischemic stroke were significantly enriched in phosphatidylinositol 3-kinase (PI3K)/Akt and FoxO pathways. Medicarpina attenuated OGD/R-evoked viability inhibition, oxidative stress, and cell death in HCMECs. Additionally, medicarpin activated the PI3K/Akt and FoxO pathways in OGD/R-induced HCMECs. Inhibition of PI3K/Akt pathway abrogated the neuroprotective effect of medicarpin on OGD/R-induced injury and activation of FoxO pathway in HCMECs. In conclusion, medicarpin suppressed OGD/R-induced injury in HCMECs by activating PI3K/Akt/FoxO pathway.
Collapse
|
29
|
Yan N, Xu Z, Qu C, Zhang J. Dimethyl fumarate improves cognitive deficits in chronic cerebral hypoperfusion rats by alleviating inflammation, oxidative stress, and ferroptosis via NRF2/ARE/NF-κB signal pathway. Int Immunopharmacol 2021; 98:107844. [PMID: 34153667 DOI: 10.1016/j.intimp.2021.107844] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022]
Abstract
Cerebrovascular disease and its risk factors cause persistent decrease of cerebral blood flow, chronic cerebral hypoperfusion (CCH) is the major foundation of vascular cognitive impairment (VCI). The hippocampus is extremely vulnerable to cerebral ischemia and hypoxia. Oxidative stress and neuroinflammation injury are important pathophysiological mechanisms of this process, which is closely related to hippocampal neurons damage and loss. Dimethyl fumarate (DMF), an FDA-approved therapeutic for multiple sclerosis (MS), plays a protective role in multiple neurological disorders. Studies have shown that DMF exerts anti-inflammatory and antioxidant effects via the NRF2/ARE/NF-κB signaling pathway. Thus, this study aimed to evaluate the neuroprotective effect of DMF in the CCH rat model. Ferroptosis, a novel defined iron-dependent cell death form, were found to be strongly associated with the pathophysiology of CCH. Emerging evidences have shown that inhibition of ferroptosis by targeting NRF2 exerted neuroprotective effect in neurodegeneration diseases. We also investigated whether DMF can alleviate cognitive deficits through inhibition of ferroptosis by the NRF2 signaling pathway in this study. DMF was intragastric for consecutive five weeks (100 mg/kg/day). Then behavior test and histological, molecular, and biochemical analysis were performed. We found that DMF treatment significantly improved cognitive deficits and partially reversed hippocampus neuronal damage and loss caused by CCH. And DMF treatment decreased hippocampus IL-1β, TNF-α, and IL-6 pro-inflammatory cytokines concentration, and mediated the NF-κB signaling pathway. And DMF also alleviated hippocampus oxidative stress through reducing MDA, and increasing GSH and SOD levels, which are also closely associated with ferroptosis. Besides, DMF treatment reduced the expression of PTGS2, and increased the expression of FTH1 and xCT, and the iron content is also reduced, which were the important features related to ferroptosis. Furthermore, DMF activated the NRF2/ARE signaling pathway and upregulated the expression of HO-1, NQO1 and GPX4. These outcomes indicated that DMF can improve cognitive impairment in rats with CCH, possibly through alleviating neuroinflammation, oxidative stress damage and inhibiting ferroptosis of hippocampal neurons. Overall, our results provide new evidence for the neuroprotective role of DMF.
Collapse
Affiliation(s)
- Nao Yan
- Department of Neurology, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China; Hubei Clinical Research Center for Dementias and Cognitive Impairments, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China
| | - Zhipeng Xu
- Department of Neurology, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China; Hubei Clinical Research Center for Dementias and Cognitive Impairments, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China
| | - Changhua Qu
- Department of Neurology, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China; Hubei Clinical Research Center for Dementias and Cognitive Impairments, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China
| | - JunJian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China; Hubei Clinical Research Center for Dementias and Cognitive Impairments, Zhongnan Hospital of Wuhan University, No. 169, Donghu Road, Wuhan 430071, Hubei, China.
| |
Collapse
|