1
|
Duan WL, Gu LH, Guo A, Wang XJ, Ding YY, Zhang P, Zhang BG, Li Q, Yang LX. Molecular mechanisms of programmed cell death and potential targeted pharmacotherapy in ischemic stroke (Review). Int J Mol Med 2025; 56:103. [PMID: 40341937 PMCID: PMC12081036 DOI: 10.3892/ijmm.2025.5544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/15/2025] [Indexed: 05/11/2025] Open
Abstract
Stroke poses a threat to the elderly, being the second leading cause of death and the third leading cause of disability worldwide. Ischemic stroke (IS), resulting from arterial occlusion, accounts for ~85% of all strokes. The pathophysiological processes involved in IS are intricate and complex. Currently, tissue plasminogen activator (tPA) is the only Food and Drug Administration‑approved drug for the treatment of IS. However, due to its limited administration window and the risk of symptomatic hemorrhage, tPA is applicable to only ~10% of patients with stroke. Additionally, the reperfusion process associated with thrombolytic therapy can further exacerbate damage to brain tissue. Therefore, a thorough understanding of the molecular mechanisms underlying IS‑induced injury and the identification of potential protective agents is critical for effective IS treatment. Over the past few decades, advances have been made in exploring potential protective drugs for IS. The present review summarizes the specific mechanisms of various forms of programmed cell death (PCD) induced by IS and highlights potential protective drugs targeting different PCD pathways investigated over the last decade. The present review provides a theoretical foundation for basic research and insights for the development of pharmacotherapy for IS.
Collapse
Affiliation(s)
- Wan-Li Duan
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Li-Hui Gu
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Ai Guo
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Xue-Jie Wang
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
- Department of Pathology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Yi-Yue Ding
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Peng Zhang
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Bao-Gang Zhang
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
- Department of Pathology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Qin Li
- Rehabilitation Medicine and Health College, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| | - Li-Xia Yang
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
2
|
Mssillou I, El Abdali Y, Amrati FEZ, Lim A, Khalid A, Makeen HA, Albratty M, Lefrioui Y, Louafi B, Bousta D. Unlocking the therapeutic potential of protocatechualdehyde: pharmacological applications and mechanism insights. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04267-9. [PMID: 40493248 DOI: 10.1007/s00210-025-04267-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Accepted: 05/05/2025] [Indexed: 06/12/2025]
Abstract
Protocatechualdehyde (PCA) is a phenolic aldehyde with many pharmacological features. It is composed of two hydroxyl groups attached to C3 and C4 of the benzene ring. This review aims to discover the source of PCA, discuss its pharmacological efficiency, investigate its implication in related mechanistic pathways, approve safety, and elaborate scientific foundation for future studies. This compound has been detected in various plants, especially Salviae Miltiorrhiza Bumge. which is considered as the main source. PCA has been confirmed to possess strong antioxidant activity by inhibiting free radicals in different in vivo and in vitro models. Interestingly, PCA has an important anticancer potential and is involved in various mechanistic pathways, such as apoptosis in different cancer cells, degradation of cyclin expression, cell cycle arrest, and downregulated regulator proteins. Furthermore, this molecule has a clear effect in reducing inflammation via inhibition of cyclooxygenases-2 (COX-2), tumor necrosis factor-α (TNF-α), and interleukins (IL-6, IL-1β), reduction of NLRP3, GSDMD, and caspase-1 expression, activation of the Nrf2 pathway, and suppression of the ERK/c-Fos/NFATc1 signaling axis. In addition, the antimicrobial proprieties and antibiofilm activity of PCA have been proved on Micrococcus luteus, MRSA, Ralstonia solanacearum, and Yersinia enterocolitica. Also, it is highly reported in neurological illness and cardiovascular treatment. Particularly, PCA exhibits significant anti-atherosclerosis activity, protects skin, and is valuable for endothelial function. Exploring the pharmacokinetic applications, toxicity and recent advances in researches on PCA, we have developed the scientific insights required for future research for the accurate exploitation of PCA in future pre-clinical and clinical trials.
Collapse
Affiliation(s)
- Ibrahim Mssillou
- National Agency of Medicinal and Aromatic Plants, 34025, Taounate, Morocco.
| | - Youness El Abdali
- Laboratory of Biotechnology, Health, Agrofood and Environment (LBEAS), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, 30000, Fez, Morocco
| | - Fatima Ez-Zahra Amrati
- Laboratory of Cell Biology and Molecular Genetics (LBCGM), Department of Biology, Faculty of Sciences, Ibn Zohr University, Agadir, Souss Massa, Morocco
| | - Adrian Lim
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Asaad Khalid
- Health Research Center, Jazan University, P.O. Box 114, 45142, Jazan, Saudi Arabia.
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, Faculty of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, 45142, Jazan, Saudi Arabia
| | - Youssra Lefrioui
- National Agency of Medicinal and Aromatic Plants, 34025, Taounate, Morocco
- Laboratory of Functional Ecology and Environmental Engineering, Faculty of Sciences and Technology, Sidi Mohamed Ben Abdellah University, 30000, Fez, Morocco
| | - Boutaina Louafi
- National Agency of Medicinal and Aromatic Plants, 34025, Taounate, Morocco
- Laboratory of Biotechnology Environment Agrofood and Health (LBEAS), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, 30000, Fez, Morocco
| | - Dalila Bousta
- National Agency of Medicinal and Aromatic Plants, 34025, Taounate, Morocco
| |
Collapse
|
3
|
Dong MQ, Chen Y, Wang Q, Li HQ, Bai LY, Cao X, Xu Y. The novel compound CP-10 suppresses microglia-mediated neuroinflammation and neutrophil chemotaxis and attenuates ischemic brain injury by targeting FPR1. Neuropharmacology 2025; 277:110528. [PMID: 40412758 DOI: 10.1016/j.neuropharm.2025.110528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 05/17/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Ischemic stroke represents a major neurological disorder characterized by significant morbidity, where neuroinflammation plays a central role in exacerbating cerebral injury. Following ischemic events, microglial activation and the subsequent infiltration of peripheral immune cells, particularly neutrophils, contribute to the disruption of the blood-brain barrier and amplify neuronal damage. In the present study, as a part of our ongoing screening experiment to evaluate the anti-inflammatory effects of new compounds, a novel compound, namely N-{2-[(7-chloro-4-oxo-4H-pyrido[1,2-a] pyrimidin-2-yl) methoxy] phenyl}-2,2-dimethylpropanamide (referred to as CP-10 in this study), to test whether it could target microglial activation and neutrophil chemotaxis, both critical contributors to ischemic brain injury. CP-10 exhibited no apparent cytotoxicity to primary microglia or neurons at concentrations up to 30 μM. Transcriptomic analysis revealed that CP-10 modulated a wide range of inflammatory and immune response genes, particularly those associated with neutrophil chemotaxis, such as Cxcl1, Cxcl2, Cxcl3, and Cxcl5. Mechanistically, CP-10 exerted its effects by interacting with formyl peptide receptor 1 (FPR1), a receptor involved in microglial activation and neutrophil recruitment. In vivo, CP-10 significantly reduced infarct volume and neurological deficits in a mouse model of middle cerebral artery occlusion (MCAO), accompanied by reduced neutrophil infiltration and microglial activation in the ischemic penumbra. Furthermore, CP-10 inhibited key inflammatory signaling pathways, including NF-κB and MAPK, downstream of FPR1 activation. These findings position CP-10 as a promising candidate for ischemic stroke therapy, targeting neuroinflammation and immune cell chemotaxis via FPR1 modulation.
Collapse
Affiliation(s)
- Meng-Qi Dong
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
| | - Yan Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Qing Wang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
| | - Hui-Qin Li
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Ling-Yun Bai
- Department of Neurology, Joint Institute of Nanjing Drum Tower Hospital for Life and Health, College of Life Science, Nanjing Normal University, Nanjing, 210008, China
| | - Xiang Cao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China; Department of Neurology, Joint Institute of Nanjing Drum Tower Hospital for Life and Health, College of Life Science, Nanjing Normal University, Nanjing, 210008, China.
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China.
| |
Collapse
|
4
|
Zhang M, Xue SJ, Yang F, Xiao M, Tang YB, Wu Y. LncRNA SNHG7/miR-181b-5p/TLR4 Activates Inflammation And Promotes Pyroptosis Through NF-κB Signaling in Diabetic Nephropathy. Inflammation 2025:10.1007/s10753-025-02295-4. [PMID: 40372612 DOI: 10.1007/s10753-025-02295-4] [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: 10/27/2024] [Revised: 03/06/2025] [Accepted: 03/20/2025] [Indexed: 05/16/2025]
Abstract
MiR-181b-5p plays a critical role in the pyroptosis and injury of kidney tubular cells in diabetic kidney disease (DKD). The long noncoding RNA (lncRNA) small nucleolar RNA hostgene 7 (SNHG7) has been shown to bind to and inhibit the function of miR-181b-5p. However, the precise role of SNHG7 in DKD remains unclear. To address this, the current study measured the expression levels of SNHG7, miR-181b-5p, and Toll-like receptor 4 (TLR4) using RT-qPCR analysis of renal biopsies from both normal individuals and patients with DKD. An in vitro DKD model was subsequently established by exposing HK-2 cells to high glucose (HG). In both DKD tissues and HG-stimulated HK-2 cells, SNHG7 and TLR4 levels were significantly elevated, while miR-181b-5p levels were markedly reduced. Knockdown of SNHG7 resulted in multiple beneficial effects: it effectively attenuated high glucose-induced lactate dehydrogenase (LDH) leakage, restored cell viability, inhibited the production of inflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin 18 (IL-18), and interleukin 1β (IL-1β), and suppressed the activation of the nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 3 (NLRP3)/acysteinyl aspartate-specific proteinase 1 (caspase-1)/gasdermin D (GSDMD) pathway. Mechanistically, SNHG7 functions as a molecular sponge for miR-181b-5p, while miR-181b-5p directly targets TLR4, collectively regulating nuclear factor-kappaB (NF-κB) pathway activation. Moreover, inhibition of miR-181b-5p or up-regulation of TLR4 reversed the protective effects of SNHG7 knockdown. Additionally, co-transfection of a TLR4 over-expression vector with a miR-181b-5p mimic counteracted the effects of miR-181b-5p overexpression on cell viability, LDH leakage, and the expression of inflammatory factors and pyroptosis-related molecules. In summary, SNHG7 acts as a molecular sponge for miR-181b-5p, promoting inflammation and pyroptosis in DKD, which in turn regulates TLR4 expression and the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Min Zhang
- School of Medicine, Xinyang Vocational and Technical College, Xinyang, 464000, China
| | - Sheng-Jiang Xue
- Ladder Molecular Biomedical Research Center, Guangzhou, 510800, China
| | - Feng Yang
- No.2 Department of Pharmacy, Hunan Provincial People'S Hospital, the First Affiliated Hospital of Hunan Normal University), Changsha, 410002, China
| | - Meng Xiao
- School of Medicine, Xinyang Vocational and Technical College, Xinyang, 464000, China
| | - Yong-Bo Tang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Ying Wu
- Department of Nephrology, Hunan Provincial People'S Hospital, the First Affiliated Hospital of Hunan Normal University), No. 61 Jiefangxi Road, Furong District, Changsha, 410002, China.
| |
Collapse
|
5
|
Wang Y, Ma Y, Tuo P, Naeem A, Tang Y, Su Q, Li H, Gao X, Wang X. Forsythoside A Alleviates Acute Alcoholic Liver Injury by Binding to TLR4 to Inhibit the Activation of the NF-κB Pathway. Phytother Res 2025. [PMID: 40342240 DOI: 10.1002/ptr.8485] [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: 12/04/2023] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 05/11/2025]
Abstract
Forsythoside A (FTA) is a key component found in the fruit and leaves of Forsythia suspensa, having anti-inflammatory and antioxidant properties. However, it is unclear whether FTA can have a protective effect against acute alcoholic liver injury (ALI) and how it may exert this effect. This research examined the potential protective effects of FTA against acute ALI using cell and animal models. The protective properties of FTA against acute ALI were attributed to its anti-inflammatory and antioxidant actions by detecting the markers of oxidative stress and inflammation. The underlying mechanism was explored through the utilization of Western blotting, Molecular Docking, and Microscale Thermophoresis techniques. The results showed that pretreatment with high doses of FTA had a significant protective effect on acute ALI in both cell and animal models. The pretreatment with high doses of FTA inhibited alcohol-induced oxidative stress and inflammation, raising antioxidative enzyme activity in both models. Furthermore, FTA has been shown to bind to TLR4, thereby inhibiting alcohol-induced activation of the NF-κB signaling pathway, leading to a decrease in cellular oxidative stress and inflammatory reactions. This interaction also facilitates the ubiquitination-mediated degradation of TLR4, ultimately diminishing its regulatory impact on the NF-κB signaling cascade. FTA has a significant protective effect on acute ALI. It binds to TLR4 to inhibit the activation of the NF-κB signaling pathway by alcohol, thereby reducing oxidative stress and inflammation and exerting a protective effect. The results of our study provide a theoretical basis for the development of FTA for the prevention and treatment of acute ALI.
Collapse
Affiliation(s)
- Yuehua Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, College of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yuying Ma
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
| | - Peng Tuo
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
| | - Abid Naeem
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Yijun Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
| | - Qianying Su
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
| | - Huajian Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
| | - Xiaokang Gao
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, College of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiaoli Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Shiyan, China
| |
Collapse
|
6
|
Kamińska K, Grygier B, Regulska M, Procner M, Leśkiewicz M, Szczęch M, Yang J, Bouzga A, Warszyński P, Lasoń W, Szczepanowicz K, Basta-Kaim A. Multilayered Nanocarriers as a New Strategy for Delivering Drugs with Protective and Anti-inflammatory Potential: Studies in Hippocampal Organotypic Cultures Subjected to Experimental Ischemia. Mol Neurobiol 2025; 62:6333-6351. [PMID: 39786699 PMCID: PMC11953135 DOI: 10.1007/s12035-024-04670-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/16/2024] [Indexed: 01/12/2025]
Abstract
Oxidative stress and neuroinflammation play a pivotal role in pathomechanisms of brain ischemia. Our research aimed to formulate a nanotheranostic system for delivering carnosic acid as a neuroprotective agent with anti-oxidative and anti-inflammatory properties to ischemic brain tissue, mimicked by organotypic hippocampal cultures (OHCs) exposed to oxygen-glucose deprivation (OGD). In the first part of this study, the nanocarriers were formulated by encapsulating two types of nanocores (nanoemulsion (AOT) and polymeric (PCL)) containing CA into multilayer shells using the sequential adsorption of charged nanoobjects method. The newly designed nanoparticles possessed favorable physicochemical characteristics as reflected by zeta potential and other parameters. Next, we demonstrated that the newly designed gadolinium-containing nanoparticles were not toxic to OHCs and did not affect the detrimental effects of OGD on the viability of the hippocampal cells. Importantly, they readily crossed the artificial blood-brain barrier based on the human cerebral microvascular endothelial (hCMEC/D3) cell line. Furthermore, the PCL-Gd carnosic acid-loaded nanoparticles displayed anti-inflammatory potential, expressed as decreased OGD-induced HIF-1α and IL-1β levels. Results of the molecular study revealed a complex mechanism of the nanoformulation on ischemia-related neuroinflammation in OHCs, including anti-inflammatory protein A20 stimulation and moderate attenuation of the NFκB signaling pathway. Summing up, this study points to acceptable biocompatibility of the newly designed CA-containing theranostic nanoformulation and emphasizes their interaction with inflammatory processes commonly associated with the ischemic brain.
Collapse
Affiliation(s)
- Kinga Kamińska
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Beata Grygier
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Magdalena Regulska
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Magdalena Procner
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Monika Leśkiewicz
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Marta Szczęch
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, 30-239, Kraków, Poland
| | - Juan Yang
- SINTEF Material and Chemistry, Forskningsveien 1, NO-0314, Oslo, Norway
| | - Aud Bouzga
- SINTEF Material and Chemistry, Forskningsveien 1, NO-0314, Oslo, Norway
| | - Piotr Warszyński
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, 30-239, Kraków, Poland
| | - Władysław Lasoń
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Krzysztof Szczepanowicz
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, 30-239, Kraków, Poland.
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland.
| |
Collapse
|
7
|
Bu F, Yuan X, Cui X, Guo R. Bibliometric Analysis and Visualized Study of Research on Mesenchymal Stem Cells in Ischemic Stroke. Stem Cell Rev Rep 2025:10.1007/s12015-025-10878-9. [PMID: 40257541 DOI: 10.1007/s12015-025-10878-9] [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] [Accepted: 04/04/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND One of the major global causes of death and disability is ischemic stroke (IS). Mesenchymal stem cells (MSCs) emerge as a cell-based therapy for numerous diseases. Recently, research on the role of MSCs in ischemic stroke has developed rapidly worldwide. Bibliometric analysis of MSCs for IS has not yet been published, though. AIM Through bibliometric analysis, the aim of this study was to assess the current state of research on MSCs in the field of ischemic stroke research worldwide and to identify important results, major research areas, and emerging trends. METHODS Publications related to MSCs in ischemic stroke from January 1, 2002, to December 31, 2022, were obtained from the Web of Science Core Collection (WoSCC). We used HistCite, VOSViewer, CiteSpace, and Bibliometrix for bibliometric analysis and visualization. We employed the Total Global Citation Score (TGCS) to assess the impact of publications. RESULTS The bibliometric analysis included a total of 2,048 publications. The 1,386 papers used in this study were authored by 200 individuals across 200 organizations in 72 countries, published in 202 journals. Cesar V Borlongan published the most documents among high-productivity authors. Michael Chopp was the author with the highest average number of citations per paper, with an average paper citation time of 118.54. We found that research of MSCs in ischemic stroke developed rapidly starting in 2008. Neurosciences were the most productive journals, and Chinese researchers have produced the most research papers in this subject. The most cited article is "Systemic administration of exosomes released from mesenchymal stromal cells promotes functional recovery and neurovascular plasticity after stroke in rats". CONCLUSION This study uses both numbers and descriptions to thoroughly review the research on MSCs related to IS. This information provides valuable experience for researchers to carry out MSCs' work on IS.
Collapse
Affiliation(s)
- Fanwei Bu
- Xinxiang First People's Hospital, Xinxiang, China
| | | | - Xiaocan Cui
- Xinxiang First People's Hospital, Xinxiang, China
| | - Ruyue Guo
- Henan University of Chinese Medicine, Zhengzhou, China.
| |
Collapse
|
8
|
Xu Q, Lu SR, Shi ZH, Yang Y, Yu J, Wang Z, Zhang BS, Hong K. Nutritional status of elderly hypertensive patients and its relation to the occurrence of cognitive impairment. World J Psychiatry 2025; 15:103092. [PMID: 40309587 PMCID: PMC12038662 DOI: 10.5498/wjp.v15.i4.103092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/12/2025] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Hypertension is a common chronic disease in the elderly population, and its association with cognitive impairment has been increasingly recognized. Cognitive impairment, including mild cognitive impairment and dementia, can significantly affect the quality of life and independence of elderly individuals. Therefore, identifying risk factors for cognitive impairment in elderly hypertensive patients is crucial for developing effective interventions and improving health outcomes. Nutritional status is one of the potential factors that may influence cognitive function in elderly hypertensive patients. Malnutrition or inadequate nutrition can lead to various health problems, including weakened immune system, increased susceptibility to infections, and impaired physical and mental function. Furthermore, poor nutritional status has been linked to increased risk of cognitive decline and dementia in various populations. In this observational study, we aimed to investigate the nutritional status of elderly hypertensive patients and its relationship to the occurrence of cognitive impairment. By collecting baseline data on general information, body composition, and clinical indicators, we hope to identify risk factors for cognitive impairment in this patient population. The results of this study are expected to provide more scientific basis for the health management of elderly patients with hypertension, particularly in terms of maintaining good nutritional status and reducing the risk of cognitive impairment. AIM To explore the differences between clinical data and cognitive function of elderly hypertensive patients with different nutritional status, analyze the internal relationship between nutritional statuses and cognitive impairment, and build a nomogram model for predicting nutritional status in elderly hypertensive patients. METHODS The present study retrospectively analyzed 200 elderly patients admitted to our hospital for a hypertension during the period July 1, 2024 to September 30, 2024 as study subjects, and the 200 patients were divided into a modeling cohort (140 patients) and a validation cohort (60 patients) according to the ratio of 7:3. The modeling cohort were divided into a malnutrition group (26 cases), a malnutrition risk group (42 cases), and a normal nutritional status group (72 cases) according to the patients' Mini-Nutritional Assessment Scale (MNA) scores, and the modeling cohort was divided into a hypertension combined with cognitive impairment group (34 cases) and a hypertension cognitively normal group (106 cases) according to the Montreal Cognitive Assessment Scale (MoCA) scores, and the validation cohort was divided into a hypertension combined with cognitive impairment group (14 cases) and hypertension cognitively normal group (46 cases). The study outcome was the occurrence of cognitive impairment in elderly hypertensive patients. Univariate and multivariate logistic regression was used to explore the relationship between the general information of the elderly hypertensive patients and the influence indicators and the occurrence of cognitive impairment, the roadmap prediction model was established and validated, the patient work receiver operating characteristic curve was used to evaluate the predictive efficacy of the model, the calibration curve was used to assess the consistency between the predicted events and the actual events, and the decision curve analysis was used to evaluate the validity of the model. Pearson correlation analysis was used to explore the relationship between nutrition-related indicators and MoCA scores. RESULTS In this research, the modeling cohort comprised 140 cases, while the verification cohort consisted of 60 cases, with no notable discrepancy in the data between the two groups. In the modeling cohort, there were significant differences in body mass index (BMI), albumin (ALB), hemoglobin (Hb) and homocysteine levels among the malnourished group, the malnourished risk group and the normal nutritional status group. The results of univariate and multivariate analysis showed that BMI [odds ratio (OR) = 0.830, P = 0.014], ALB (OR = 0.860, P = 0.028), Hb (OR = 0.939, P = 0.035) and MNA score (OR = 0.640, P = 0.000) were independent protective factors for patients without cognitive impairment, and alkaline phosphatase (ALP) (OR = 1.074, P = 0.000) was an independent risk factor for patients with cognitive impairment. In this study, the prediction nomogram tailored for cognitive deterioration in elderly patients with hypertension demonstrated robust predictive power and a close correspondence between predicted and observed outcomes. This model offers significant potential as a means to forestall cognitive decline in hypertensive elderly patients. ALP was negatively correlated with MoCA score, while BMI, MNA score, Hb and ALB were positively correlated with MoCA score. CONCLUSION BMI, MNA score, Hb and ALB were independent protective factors for cognitive impairment in elderly hypertensive patients and were positively correlated with MoCA score. ALP was an independent risk factor for cognitive impairment in elderly hypertensive patients and was negatively correlated with the MoCA score. The column line graph model established in the study has a good predictive value.
Collapse
Affiliation(s)
- Qiao Xu
- Department of Geriatrics, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi 214023, Jiangsu Province, China
| | - Shou-Rong Lu
- Department of Geriatrics, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi 214023, Jiangsu Province, China
| | - Zi-Hao Shi
- Department of Clinical Nutrition, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi 214023, Jiangsu Province, China
| | - Ying Yang
- Department of Geriatrics, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi 214023, Jiangsu Province, China
| | - Jie Yu
- Department of Geriatrics, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi 214023, Jiangsu Province, China
| | - Zhuo Wang
- Department of Geriatrics, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi 214023, Jiangsu Province, China
| | - Bing-Shan Zhang
- Department of Geriatrics, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi 214023, Jiangsu Province, China
| | - Kan Hong
- Department of Geriatrics, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi People’s Hospital, Wuxi 214023, Jiangsu Province, China
| |
Collapse
|
9
|
Chen Q, Li X, Yang Y, Ni J, Chen J. Combined Analysis of Human and Experimental Rat Samples Identified Biomarkers for Ischemic Stroke. Mol Neurobiol 2025; 62:3794-3812. [PMID: 39325100 PMCID: PMC11790756 DOI: 10.1007/s12035-024-04512-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
The genetic transcription profile and underlying molecular mechanisms of ischemic stroke (IS) remain elusive. To address this issue, four mRNA and one miRNA expression profile of rats with middle cerebral artery occlusion (MCAO) were acquired from the Gene Expression Omnibus (GEO) database. A total of 780 differentially expressed genes (DEGs) and 56 miRNAs (DEMs) were screened. Gene set and functional enrichment analysis revealed that a substantial number of immune-inflammation-related pathways were abnormally activated in IS. Through weighted gene co-expression network analysis, the turquoise module was identified as meaningful. By taking the intersection of the turquoise module genes, DEM-target genes, and all DEGs, 354 genes were subsequently obtained as key IS-related genes. Among them, six characteristic genes were identified using the least absolute shrinkage and selection operator. After validation with three external datasets, transforming growth factor beta 1 (Tgfb1) was selected as the hub gene. This finding was further confirmed by gene expression pattern analysis in both the MCAO model rats and clinical IS patients. Moreover, the expression of the hub genes exhibited a negative correlation with the modified Rankin scale score (P < 0.05). Collectively, these results expand our knowledge of the genetic profile and molecular mechanisms involved in IS and suggest that the Tgfb1 gene is a potential biomarker of this disease.
Collapse
Affiliation(s)
- Qingfa Chen
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Xiaolu Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530022, Guangxi, China
| | - Ye Yang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530022, Guangxi, China
| | - Jun Ni
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, Fujian, China.
| | - Jianmin Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, Fujian, China.
| |
Collapse
|
10
|
Moldoveanu CA, Tomoaia-Cotisel M, Sevastre-Berghian A, Tomoaia G, Mocanu A, Pal-Racz C, Toma VA, Roman I, Ujica MA, Pop LC. A Review on Current Aspects of Curcumin-Based Effects in Relation to Neurodegenerative, Neuroinflammatory and Cerebrovascular Diseases. Molecules 2024; 30:43. [PMID: 39795101 PMCID: PMC11722367 DOI: 10.3390/molecules30010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Curcumin is among the most well-studied natural substances, known for its biological actions within the central nervous system, its antioxidant and anti-inflammatory properties, and human health benefits. However, challenges persist in effectively utilising curcumin, addressing its metabolism and passage through the blood-brain barrier (BBB) in therapies targeting cerebrovascular diseases. Current challenges in curcumin's applications revolve around its effects within neoplastic tissues alongside the development of intelligent formulations to enhance its bioavailability. Formulations have been discovered including curcumin's complexes with brain-derived phospholipids and proteins, or its liposomal encapsulation. These novel strategies aim to improve curcumin's bioavailability and stability, and its capability to cross the BBB, thereby potentially enhancing its efficacy in treating cerebrovascular diseases. In summary, this review provides a comprehensive overview of molecular pathways involved in interactions of curcumin and its metabolites, and brain vascular homeostasis. This review explores cellular and molecular current aspects, of curcumin-based effects with an emphasis on curcumin's metabolism and its impact on pathological conditions, such as neurodegenerative diseases, schizophrenia, and cerebral angiopathy. It also highlights the limitations posed by curcumin's poor bioavailability and discusses ongoing efforts to surpass these impediments to harness the full therapeutic potential of curcumin in neurological disorders.
Collapse
Affiliation(s)
- Claudia-Andreea Moldoveanu
- Department of Molecular Biology and Biotechnology, Babeș-Bolyai University, Clinicilor St., RO-400371 Cluj-Napoca, Romania;
- Department of Experimental Biology and Biochemistry, Institute of Biological Research from Cluj-Napoca, a Branch of NIRDBS Bucharest, 48 Republicii St., RO-400015 Cluj-Napoca, Romania;
| | - Maria Tomoaia-Cotisel
- Research Center of Excellence in Physical Chemistry, Faculty of Chemistry and Chemical Engineering, “Babes-Bolyai University”, 11 Arany Janos St., RO-400028 Cluj-Napoca, Romania or (M.T.-C.); (A.M.); (C.P.-R.); (M.-A.U.)
- Academy of Romanian Scientists, 3 Ilfov St., RO-050044 Bucharest, Romania;
| | - Alexandra Sevastre-Berghian
- Department of Physiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 1 Clinicilor St., RO-400006 Cluj-Napoca, Romania;
| | - Gheorghe Tomoaia
- Academy of Romanian Scientists, 3 Ilfov St., RO-050044 Bucharest, Romania;
- Department of Orthopedics and Traumatology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 47 Gen. Traian Moșoiu St., RO-400132 Cluj-Napoca, Romania
| | - Aurora Mocanu
- Research Center of Excellence in Physical Chemistry, Faculty of Chemistry and Chemical Engineering, “Babes-Bolyai University”, 11 Arany Janos St., RO-400028 Cluj-Napoca, Romania or (M.T.-C.); (A.M.); (C.P.-R.); (M.-A.U.)
| | - Csaba Pal-Racz
- Research Center of Excellence in Physical Chemistry, Faculty of Chemistry and Chemical Engineering, “Babes-Bolyai University”, 11 Arany Janos St., RO-400028 Cluj-Napoca, Romania or (M.T.-C.); (A.M.); (C.P.-R.); (M.-A.U.)
| | - Vlad-Alexandru Toma
- Department of Molecular Biology and Biotechnology, Babeș-Bolyai University, Clinicilor St., RO-400371 Cluj-Napoca, Romania;
- Department of Experimental Biology and Biochemistry, Institute of Biological Research from Cluj-Napoca, a Branch of NIRDBS Bucharest, 48 Republicii St., RO-400015 Cluj-Napoca, Romania;
- Academy of Romanian Scientists, 3 Ilfov St., RO-050044 Bucharest, Romania;
- Centre for Systems Biology, Biodiversity and Bioresources “3B”, Babeș-Bolyai University, 44 Republicii St., RO-400347 Cluj-Napoca, Romania
| | - Ioana Roman
- Department of Experimental Biology and Biochemistry, Institute of Biological Research from Cluj-Napoca, a Branch of NIRDBS Bucharest, 48 Republicii St., RO-400015 Cluj-Napoca, Romania;
| | - Madalina-Anca Ujica
- Research Center of Excellence in Physical Chemistry, Faculty of Chemistry and Chemical Engineering, “Babes-Bolyai University”, 11 Arany Janos St., RO-400028 Cluj-Napoca, Romania or (M.T.-C.); (A.M.); (C.P.-R.); (M.-A.U.)
| | - Lucian-Cristian Pop
- Research Center of Excellence in Physical Chemistry, Faculty of Chemistry and Chemical Engineering, “Babes-Bolyai University”, 11 Arany Janos St., RO-400028 Cluj-Napoca, Romania or (M.T.-C.); (A.M.); (C.P.-R.); (M.-A.U.)
| |
Collapse
|
11
|
Chen L, Wang K, Liu X, Wang L, Zou H, Hu S, Zhou L, Li R, Cao S, Ruan B, Cui Q. Design, synthesis, in vitro and in vivo biological evaluation of pterostilbene derivatives for anti-inflammation therapy. J Enzyme Inhib Med Chem 2024; 39:2315227. [PMID: 38421003 PMCID: PMC10906133 DOI: 10.1080/14756366.2024.2315227] [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: 10/27/2023] [Accepted: 01/07/2024] [Indexed: 03/02/2024] Open
Abstract
Pterostilbene (PST) is a naturally derived stilbene compound in grapes, blueberries, and other fruits. It is also a natural dietary compound with a wide range of biological activities such as antioxidant, anti-inflammatory, antitumor, and so on. Structural modifications based on the chemical scaffold of the pterostilbene skeleton are of great importance for drug discovery. In this study, pterostilbene skeletons were used to design novel anti-inflammatory compounds with high activity and low toxicity. A total of 30 new were found and synthesised, and their anti-inflammatory activity and safety were screened. Among them, compound E2 was the most active (against NO: IC50 = 0.7 μM) than celecoxib. Further studies showed that compound E2 exerted anti-inflammatory activity by blocking LPS-induced NF-κB/MAPK signalling pathway activation. In vivo experiments revealed that compound E2 had a good alleviating effect on acute colitis in mice. In conclusion, compound E2 may be a promising anti-inflammatory lead compound.
Collapse
Affiliation(s)
- Liuzeng Chen
- School of Biology, Food and Environment, Hefei University, Hefei, P. R. China
| | - Ke Wang
- School of Biology, Food and Environment, Hefei University, Hefei, P. R. China
| | - Xiaohan Liu
- School of Biology, Food and Environment, Hefei University, Hefei, P. R. China
| | - Lifan Wang
- School of Biology, Food and Environment, Hefei University, Hefei, P. R. China
| | - Hui Zou
- School of Biology, Food and Environment, Hefei University, Hefei, P. R. China
| | - Shuying Hu
- School of Biology, Food and Environment, Hefei University, Hefei, P. R. China
| | - Lingling Zhou
- School of Biology, Food and Environment, Hefei University, Hefei, P. R. China
| | - Rong Li
- School of Biology, Food and Environment, Hefei University, Hefei, P. R. China
| | - Shiying Cao
- School of Biology, Food and Environment, Hefei University, Hefei, P. R. China
| | - Banfeng Ruan
- School of Biology, Food and Environment, Hefei University, Hefei, P. R. China
| | - Quanren Cui
- Institute of Tobacoo Research, Anhui Academy of Agricultural Sciences, Hefei, P. R. China
| |
Collapse
|
12
|
Zhang W, Guo C, Li Y, Wang H, Wang H, Wang Y, Wu T, Wang H, Cheng G, Man J, Chen S, Fu S, Yang L. Mitophagy mediated by HIF-1α/FUNDC1 signaling in tubular cells protects against renal ischemia/reperfusion injury. Ren Fail 2024; 46:2332492. [PMID: 38584135 PMCID: PMC11000611 DOI: 10.1080/0886022x.2024.2332492] [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: 10/12/2023] [Accepted: 03/14/2024] [Indexed: 04/09/2024] Open
Abstract
Acute kidney injury (AKI) is associated with a high mortality rate. Pathologically, renal ischemia/reperfusion injury (RIRI) is one of the primary causes of AKI, and hypoxia-inducible factor (HIF)-1α may play a defensive role in RIRI. This study assessed the role of hypoxia-inducible factor 1α (HIF-1α)-mediated mitophagy in protection against RIRI in vitro and in vivo. The human tubular cell line HK-2 was used to assess hypoxia/reoxygenation (H/R)-induced mitophagy through different in vitro assays, including western blotting, immunofluorescence staining, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL), and reactive oxygen species (ROS) measurement. Additionally, a rat RIRI model was established for evaluation by renal histopathology, renal Doppler ultrasound, and transmission electron microscopy to confirm the in vitro data. The selective HIF-1α inhibitor LW6 reduced H/R-induced mitophagy but increased H/R-induced apoptosis and ROS production. Moreover, H/R treatment enhanced expression of the FUN14 domain-containing 1 (FUNDC1) protein. Additionally, FUNDC1 overexpression reversed the effects of LW6 on the altered expression of light chain 3 (LC3) BII and voltage-dependent anion channels as well as blocked the effects of HIF-1α inhibition in cells. Pretreatment of the rat RIRI model with roxadustat, a novel oral HIF-1α inhibitor, led to decreased renal injury and apoptosis in vivo. In conclusion, the HIF-1α/FUNDC1 signaling pathway mediates H/R-promoted renal tubular cell mitophagy, whereas inhibition of this signaling pathway protects cells from mitophagy, thus aggravating apoptosis, and ROS production. Accordingly, roxadustat may protect against RIRI-related AKI.
Collapse
Affiliation(s)
- Wenjun Zhang
- Department of Nephrology, Lanzhou University Affiliated Second Hospital, Lanzhou, China
- Gansu Provicne Clinical Research Center for Kidney Diseases, Lanzhou, China
| | - Chao Guo
- Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yi Li
- Department of Anesthesiology, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Hao Wang
- Department of Urology Surgery, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Huabing Wang
- Department of Urology Surgery, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Yingying Wang
- Department of Nephrology, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Tingting Wu
- Department of Functional Examination in Children, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Huinan Wang
- The Second Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Gang Cheng
- The Second Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Jiangwei Man
- Department of Urology Surgery, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Siyu Chen
- Department of Urology Surgery, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Shengjun Fu
- Department of Urology Surgery, Lanzhou University Affiliated Second Hospital, Lanzhou, China
| | - Li Yang
- Department of Urology Surgery, Lanzhou University Affiliated Second Hospital, Lanzhou, China
- Gansu Provicne Clinical Research Center for Urology, Lanzhou, China
| |
Collapse
|
13
|
Li S, Adamu A, Ye Y, Gao F, Mi R, Xue G, Wang Z. (+)-Borneol inhibits neuroinflammation and M1 phenotype polarization of microglia in epileptogenesis through the TLR4-NFκB signaling pathway. Front Neurosci 2024; 18:1497102. [PMID: 39605791 PMCID: PMC11599196 DOI: 10.3389/fnins.2024.1497102] [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: 09/16/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Objective To investigate the effect of (+)-borneol on neuroinflammation and microglia phenotype polarization in epileptogenesis and its possible mechanism. Methods Based on mouse models of status epilepticus (SE) induced by pilocarpine, and treated with 15 mg/kg (+)-borneol, western-blot was used to detect the expressions of NeuN, Iba-1, TLR4, p65 and p-p65 in the hippocampus. Immunofluorescence was used to detect the expression of apoptosis-related proteins Bax and Bcl-2. To explore the effect of (+)-borneol on microglia in vitro, we used the kainic acid-induced microglia model and the concentration of (+)-borneol was 25 μM according to CCK-8 results. The levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-10 (IL-10) in the supernatant of each group was detected by ELISA. The nitric oxide (NO) content in the supernatant was detected by Griess method. The expressions of Iba-1 and TLR4-NFκB signaling pathway-related proteins (TLR4, p65, p-p65) were detected by Western-Blot. Immunofluorescence was used to detect microglia's M1 and M2 phenotype polarization and the expression of Iba-1 and TLR4. Results (+)-borneol reduced hippocampal neuronal injury, apoptosis, and microglia activation by inhibiting the TLR-NFκB signaling pathway in SE mice. TLR4 agonist LPS partially reversed the neuroprotective effect of (+)-borneol. In the KA-induced microglia model, (+)-borneol inhibited microglia activation, M1 phenotype polarization, and secretion of pro-inflammatory cytokines through the TLR4-NFκB signaling pathway. LPS treatment inhibited the therapeutic effects of (+)-borneol. Conclusion (+)-borneol inhibits microglial neuroinflammation and M1 phenotype polarization through TLR4-NFκB signaling pathway and reduces neuronal damage and apoptosis in SE mice. Therefore, (+)-borneol may be a potential drug for epilepsy modification therapy.
Collapse
Affiliation(s)
- Shuo Li
- Second Clinical Medical School, Shanxi Medical University, Taiyuan, China
| | - Alhamdu Adamu
- Second Clinical Medical School, Shanxi Medical University, Taiyuan, China
| | - Yucai Ye
- Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Fankai Gao
- Department of Neurology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Rulin Mi
- Department of Neurology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Guofang Xue
- Department of Neurology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhaojun Wang
- Department of Physiology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
14
|
Jiang H, Zhang C, Lin M, Yin Y, Deng S, Liu W, Zhuo B, Tian G, Du Y, Meng Z. Deciphering the mechanistic impact of acupuncture on the neurovascular unit in acute ischemic stroke: Insights from basic research in a narrative review. Ageing Res Rev 2024; 101:102536. [PMID: 39384155 DOI: 10.1016/j.arr.2024.102536] [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: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Ischemic stroke(IS), a severe acute cerebrovascular disease, not only imposes a heavy economic burden on society but also presents numerous challenges in treatment. During the acute phase, while thrombolysis and thrombectomy serve as primary treatments, these approaches are restricted by a narrow therapeutic window. During rehabilitation, commonly used neuroprotective agents struggle with their low drug delivery efficiency and inadequate preclinical testing, and the long-term pharmacological and toxicity effects of nanomedicines remain undefined. Meanwhile, acupuncture as a therapeutic approach is widely acknowledged for its effectiveness in treating IS and has been recommended by the World Health Organization (WHO) as an alternative and complementary therapy, even though its exact mechanisms remain unclear. This review aims to summarize the known mechanisms of acupuncture and electroacupuncture (EA) in the treatment of IS. Research shows that acupuncture treatment mainly protects the neurovascular unit through five mechanisms: 1) reducing neuronal apoptosis and promoting neuronal repair and proliferation; 2) maintaining the integrity of the blood-brain barrier (BBB); 3) inhibiting the overactivation and polarization imbalance of microglia; 4) regulating the movement of vascular smooth muscle (VSM) cells; 5) promoting the proliferation of oligodendrocyte precursors. Through an in-depth analysis, this review reveals the multi-level, multi-dimensional impact of acupuncture treatment on the neurovascular unit (NVU) following IS, providing stronger evidence and a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Hailun Jiang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Chao Zhang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Mengxuan Lin
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yu Yin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shizhe Deng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Bifang Zhuo
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Guang Tian
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yuzheng Du
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Zhihong Meng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
15
|
Li X, Long J, Yao C, Liu X, Li N, Zhou Y, Li D, Xiong G, Wang K, Hao Y, Chen K, Zhou Z, Ji A, Luo P, Cai T. The role of BTG2/PI3K/AKT pathway-mediated microglial activation in T-2 toxin-induced neurotoxicity. Toxicol Lett 2024; 400:81-92. [PMID: 39147216 DOI: 10.1016/j.toxlet.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 07/07/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
T-2 toxin is one of the mycotoxins widely distributed in human food and animal feed. Our recent work has shown that microglial activation may contribute to T-2 toxin-induced neurotoxicity. However, the molecular mechanisms involved need to be further clarified. To address this, we employed high-throughput transcriptome sequencing and found altered B cell translocation gene 2 (BTG2) expression levels in microglia following T-2 toxin treatment. It has been shown that altered BTG2 expression is involved in a range of neurological pathologies, but whether it's involved in the regulation of microglial activation is unclear. The aim of this study was to investigate the role of BTG2 in T-2 toxin-induced microglial activation. The results of animal experiments showed that T-2 toxin caused neurobehavioral disorders and promoted the expression of microglial BTG2 and pro-inflammatory activation of microglia in hippocampus and cortical, while microglial inhibitor minocycline inhibited these changes. The results of in vitro experiments showed that T-2 toxin enhanced BTG2 expression and pro-inflammatory microglial activation, and inhibited BTG2 expression weakened T-2 toxin-induced microglial activation. Moreover, T-2 toxin activated PI3K/AKT and its downstream NF-κB signaling pathway, which could be reversed after knock-down of BTG2 expression. Meanwhile, the PI3K inhibitor LY294002 also blocked this process. Therefore, BTG2 may be involved in T-2 toxin's ability to cause microglial activation through PI3K/AKT/NF-κB pathway.
Collapse
Affiliation(s)
- Xiukuan Li
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jinyun Long
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Chunyan Yao
- Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiaoling Liu
- Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Na Li
- Chongqing Yongchuan District Center for Disease Control and Prevention, Chongqing 402160, China
| | - Yumeng Zhou
- Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Dawei Li
- Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Guiyuan Xiong
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Kexue Wang
- Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yuhui Hao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ka Chen
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ziyuan Zhou
- Department of Environmental Health, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ailing Ji
- Department of Preventive Medicine, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China.
| | - Peng Luo
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China.
| | - Tongjian Cai
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Department of Epidemiology, College of Preventive Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
16
|
Zheng H, Xiao X, Han Y, Wang P, Zang L, Wang L, Zhao Y, Shi P, Yang P, Guo C, Xue J, Zhao X. Research progress of propofol in alleviating cerebral ischemia/reperfusion injury. Pharmacol Rep 2024; 76:962-980. [PMID: 38954373 DOI: 10.1007/s43440-024-00620-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
Ischemic stroke is a leading cause of adult disability and death worldwide. The primary treatment for cerebral ischemia patients is to restore blood supply to the ischemic region as quickly as possible. However, in most cases, more severe tissue damage occurs, which is known as cerebral ischemia/reperfusion (I/R) injury. The pathological mechanisms of brain I/R injury include mitochondrial dysfunction, oxidative stress, excitotoxicity, calcium overload, neuroinflammation, programmed cell death and others. Propofol (2,6-diisopropylphenol), a short-acting intravenous anesthetic, possesses not only sedative and hypnotic effects but also immunomodulatory and neuroprotective effects. Numerous studies have reported the protective properties of propofol during brain I/R injury. In this review, we summarize the potential protective mechanisms of propofol to provide insights for its better clinical application in alleviating cerebral I/R injury.
Collapse
Affiliation(s)
- Haijing Zheng
- Basic Medical College, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
- Zhengzhou Central Hospital, Zhengzhou, China
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Xian Xiao
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Yiming Han
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Pengwei Wang
- Department of Pharmacy, the First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, Henan, 453100, China
| | - Lili Zang
- Department of Surgery, the First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, China
| | - Lilin Wang
- Department of Pediatric Surgery, the First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, China
| | - Yinuo Zhao
- Basic Medical College, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Peijie Shi
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Pengfei Yang
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| | - Chao Guo
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| | - Jintao Xue
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| | - Xinghua Zhao
- Basic Medical College, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| |
Collapse
|
17
|
Mi R, Cheng H, Chen R, Bai B, Li A, Gao F, Xue G. Effects and mechanisms of long-acting glucagon-like peptide-1 receptor agonist semaglutide on microglia phenotypic transformation and neuroinflammation after cerebral ischemia/reperfusion in rats. Brain Circ 2024; 10:354-365. [PMID: 40012598 PMCID: PMC11850941 DOI: 10.4103/bc.bc_38_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/14/2024] [Accepted: 09/04/2024] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND The optimal method for addressing cerebral ischemic stroke involves promptly restoring blood supply. However, cerebral ischemia-reperfusion injury (CIRI) is an unavoidable consequence of this event. Neuroinflammation is deemed the primary mechanism of CIRI, with various activation phenotypes of microglia playing a pivotal role. Research has demonstrated that long-lasting agonists of the glucagon-like peptide-1 receptor can suppress neuroinflammation and microglial activation. METHODS A transient middle cerebral artery occlusion (tMCAO) rat model was established to investigate the effects of semaglutide. Neurological impairments were evaluated utilizing modified neurological severity score on days 1, 3, and 7 postinterventions. Brains were stained with 2,3,5-Triphenyltetrazolium Chloride to determine infarct volume. To assess the expression of various microglia activation phenotypes and neuroinflammatory biomarkers, we utilized immunohistochemistry and immunoblotting. RESULTS The study demonstrated that semaglutide in the tMCAO model could decrease neurological deficit scores and reduce the size of cerebral infarcts. In addition, we observed low levels of cluster of differentiation 68 (CD68, an indicator of M1 microglial activation) and tumor necrosis factor alpha (a pro-inflammatory mediator). Moreover, the results indicated a rise in the levels of CD206 (an indicator of M2 activation) and transforming growth factor beta (an anti-inflammatory mediator), while simultaneously reducing P65 levels in the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling cascade. CONCLUSION In the CIRI model, semaglutide exhibits notable neuroprotective effects on rats, reducing neuroinflammation through the regulation of microglia phenotype transformation and inhibition of NF-κB activation.
Collapse
Affiliation(s)
- Rulin Mi
- Department of Neurology, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Huifeng Cheng
- Department of Geriatric Medicine, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Rui Chen
- Department of Neurology, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Bo Bai
- Department of Neurology, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - An Li
- Department of Neurology, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Fankai Gao
- Department of Neurology, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Guofang Xue
- Department of Neurology, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
18
|
Huang SC, Huang HC, Liao WL, Kao ST, Cheng CY. Neuroprotective effects of Gastrodia elata Blume on promoting M2 microglial polarization by inhibiting JNK/TLR4/T3JAM/NF-κB signaling after transient ischemic stroke in rats. Front Pharmacol 2024; 15:1469602. [PMID: 39391701 PMCID: PMC11465390 DOI: 10.3389/fphar.2024.1469602] [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: 07/24/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Background Gastrodia elata Blume, also called Tian Ma (TM), has been used to treat stroke for centuries. However, its effects on inflammation in acute cerebral ischemic injury and underlying mechanisms involved in microglial polarization remain unknown. The present study explored the effects of the TM extract on the modulation of microglial M1/M2 polarization 2 days after transient cerebral ischemia. Methods Male Sprague Dawley rats were intracerebroventricularly administered with 1% dimethyl sulfoxide 25 min before cerebral ischemia and subsequently intraperitoneally administered 0.25 g/kg (DO + TM-0.25 g), 0.5 g/kg (DO + TM-0.5 g), or 1 g/kg (DO + TM-1 g) of the TM extract after cerebral ischemia onset. Results DO + TM-0.5 g and DO + TM-1 g treatments downregulated the following: phospho-c-Jun N-terminal kinase (p-JNK)/JNK, tumor necrosis factor (TNF) receptor-associated factor 3 (TRAF3), TRAF3-interacting JNK-activating modulator (T3JAM), p-nuclear factor-kappa B p65 (p-NF-κB p65)/NF-κB p65, ionized calcium-binding adapter molecule 1 (Iba1), CD86, TNF-α, interleukin (IL)-1β, and IL-6 expression and toll-like receptor 4 (TLR4)/Iba1, CD86/Iba1, and p-NF-κB p65/Iba1 coexpression. These treatments also upregulated IL-10, nerve growth factor, and vascular endothelial growth factor A expression and YM-1/2/Iba1 and IL-10/neuronal nuclei coexpression in the cortical ischemic rim. The JNK inhibitor SP600125 exerted similar treatment effects as the DO + TM-0.5 g and DO + TM-1 g treatments. Conclusion DO + TM-0.5 g and DO + TM-1 g/kg treatments attenuate cerebral infarction by inhibiting JNK-mediated signaling. TM likely exerts the neuroprotective effects of promoting M1 to M2 microglial polarization by inhibiting JNK/TLR4/T3JAM/NF-κB-mediated signaling in the cortical ischemic rim 2 days after transient cerebral ischemia.
Collapse
Affiliation(s)
- Shang-Chih Huang
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan
| | - Hui-Chi Huang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Wen-Ling Liao
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Shung-Te Kao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chin-Yi Cheng
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, Hui-Sheng Hospital, Taichung, Taiwan
| |
Collapse
|
19
|
Costet-Mejía A, Trejo-Tapia G, Baca-Ibarra II, Rodríguez-Hernández AA, García-Hernández J, Camacho-Díaz BH, Zamilpa A. An Organic Fraction of Oenothera rosea L'Her Ex. Aiton Prevents Neuroinflammation in a Rat Ischemic Model. Pharmaceuticals (Basel) 2024; 17:1184. [PMID: 39338346 PMCID: PMC11434707 DOI: 10.3390/ph17091184] [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: 08/08/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Oenothera rosea L'Her Ex. Aiton, presenting antioxidant and anti-inflammatory activities, is traditionally used to treat bruises and headaches and as a healing agent. This study aimed to investigate whether its organic fraction (EAOr) has neuroprotective properties against neuroinflammation in the context of ischemia/reperfusion. METHODS The chemical composition of EAOr was determined using HPLC techniques, and its neuroprotective activities were evaluated in a common carotid-artery ligation model for the induction of ischemia/reperfusion (I/R). The animals were supplemented with EAOR for 15 days. On the last day, the animals were rested for one hour, following which the common carotid-artery ligation procedure was performed to induce I/R. The neurological deficit was evaluated at 24 h after I/R using Bederson's scale, and the relative expression of inflammatory genes and structure of hippocampal neurons were analyzed at 48 h. RESULTS The chemical analysis revealed five major compounds in EAOr: gallic acid, rutin, ellagic acid, and glucoside and rhamnoside quercetin. EAOr prevented neurological deficit 24 h after I/R; led to the early activation of the AIF and GFAP genes; reduced Nfkb1, IL-1beta, Il-6 and Casp3 gene expression; and protected hippocampal neurons. CONCLUSIONS Our findings demonstrate that EAOr contains polyphenol-type compounds, which could exert a therapeutic effect through the inhibition of neuroinflammation and neuronal death genes, thus maintaining hippocampal neurons.
Collapse
Affiliation(s)
- Alejandro Costet-Mejía
- Centro de Desarrollo de Productos Bióticos (CEPROBI), Instituto Politécnico Nacional, Yautepec 62739, Morelos, Mexico; (A.C.-M.); (B.H.C.-D.)
- Centro de Investigación Biomédica del Sur (CIBIS), Instituto Mexicano del Seguro Social (IMSS), Xochitepec 62780, Morelos, Mexico
| | - Gabriela Trejo-Tapia
- Centro de Desarrollo de Productos Bióticos (CEPROBI), Instituto Politécnico Nacional, Yautepec 62739, Morelos, Mexico; (A.C.-M.); (B.H.C.-D.)
| | - Itzel Isaura Baca-Ibarra
- Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México 06720, Mexico; (I.I.B.-I.); (J.G.-H.)
| | | | - Julio García-Hernández
- Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México 06720, Mexico; (I.I.B.-I.); (J.G.-H.)
| | - Brenda Hildeliza Camacho-Díaz
- Centro de Desarrollo de Productos Bióticos (CEPROBI), Instituto Politécnico Nacional, Yautepec 62739, Morelos, Mexico; (A.C.-M.); (B.H.C.-D.)
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur (CIBIS), Instituto Mexicano del Seguro Social (IMSS), Xochitepec 62780, Morelos, Mexico
| |
Collapse
|
20
|
Lu W, Wen J. Crosstalk Among Glial Cells in the Blood-Brain Barrier Injury After Ischemic Stroke. Mol Neurobiol 2024; 61:6161-6174. [PMID: 38279077 DOI: 10.1007/s12035-024-03939-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 01/11/2024] [Indexed: 01/28/2024]
Abstract
Blood-brain barrier (BBB) is comprised of brain microvascular endothelial cells (ECs), astrocytes, perivascular microglia, pericytes, neuronal processes, and the basal lamina. As a complex and dynamic interface between the blood and the central nervous system (CNS), BBB is responsible for transporting nutrients essential for the normal metabolism of brain cells and hinders many toxic compounds entering into the CNS. The loss of BBB integrity following stroke induces tissue damage, inflammation, edema, and neural dysfunction. Thus, BBB disruption is an important pathophysiological process of acute ischemic stroke. Understanding the mechanism underlying BBB disruption can uncover more promising biological targets for developing treatments for ischemic stroke. Ischemic stroke-induced activation of microglia and astrocytes leads to increased production of inflammatory mediators, containing chemokines, cytokines, matrix metalloproteinases (MMPs), etc., which are important factors in the pathological process of BBB breakdown. In this review, we discussed the current knowledges about the vital and dual roles of astrocytes and microglia on the BBB breakdown during ischemic stroke. Specifically, we provided an updated overview of phenotypic transformation of microglia and astrocytes, as well as uncovered the crosstalk among astrocyte, microglia, and oligodendrocyte in the BBB disruption following ischemic stroke.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
21
|
Zhang X, Lv D, Li B, Ding Y. Inhaled aerosolized algal polysaccharides: A novel and reliable strategy for treating pneumonia through inflammation and oxidative stress inhibition. Int Immunopharmacol 2024; 137:112532. [PMID: 38908087 DOI: 10.1016/j.intimp.2024.112532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/02/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Sepsis-associated acute lung injury (ALI) poses a significant threat, characterized by inflammation and oxidative damage. Effective drugs targeting these aspects with reliable drug delivery systems are vital for ALI management. This study aimed to evaluate the influence of algal polysaccharides (APs) with aerosolized drug delivery in ALI mice and clarify the underlying mechanism. To induce the sepsis-associated acute lung injury (ALI) model, mice were administered intraperitoneal injections of 10 mg/kg LPS for 48 h in vivo. ALI mice received APs via atomization to arrive at different sites within the lungs. Lung tissue samples and bronchoalveolar lavage fluid (BALF) were collected to access lung injury parameters. Concurrently, western blotting, H&E staining, and immunofluorescence (IF) were applied to investigate the specific impact of APs on ALI. The results showed that APs protect lung tissue against ALI by inhibiting inflammation and mitigating oxidative stress-induced damage. This study highlights promising avenues for ALI intervention using natural compounds with anti-inflammatory and antioxidant properties.
Collapse
Affiliation(s)
- XingXing Zhang
- Department of Neurology, Changshu No. 2 People's Hospital (Affiliated Changshu Hospital of Nantong University), Changshu 215500, Jiangsu, China; School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Demin Lv
- Department of Traumatic Orthopedics, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu, China
| | - Bingxia Li
- Department of Stomatology, The 359th Hospital of the People's Liberation Army of China, Zhenjiang 212001, Jiangsu, China
| | - Yuting Ding
- Department of Neurology, Changshu No. 2 People's Hospital (Affiliated Changshu Hospital of Nantong University), Changshu 215500, Jiangsu, China.
| |
Collapse
|
22
|
Xie L, He M, Ying C, Chu H. Mechanisms of inflammation after ischemic stroke in brain-peripheral crosstalk. Front Mol Neurosci 2024; 17:1400808. [PMID: 38932932 PMCID: PMC11199882 DOI: 10.3389/fnmol.2024.1400808] [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: 03/14/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Stroke is a devastating disease with high morbidity, disability, and mortality, among which ischemic stroke is more common. However, there is still a lack of effective methods to improve the prognosis and reduce the incidence of its complications. At present, there is evidence that peripheral organs are involved in the inflammatory response after stroke. Moreover, the interaction between central and peripheral inflammation includes the activation of resident and peripheral immune cells, as well as the activation of inflammation-related signaling pathways, which all play an important role in the pathophysiology of stroke. In this review, we discuss the mechanisms of inflammatory response after ischemic stroke, as well as the interactions through circulatory pathways between peripheral organs (such as the gut, heart, lung and spleen) and the brain to mediate and regulate inflammation after ischemic stroke. We also propose the potential role of meningeal lymphatic vessels (MLVs)-cervical lymph nodes (CLNs) as a brain-peripheral crosstalk lymphatic pathway in ischemic stroke. In addition, we also summarize the mechanisms of anti-inflammatory drugs in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ling Xie
- Department of Critical Medicine, First People's Hospital of Linping District, Hangzhou, China
| | - Ming He
- Department of Critical Medicine, First People's Hospital of Linping District, Hangzhou, China
| | - Caidi Ying
- Department of Hepatobiliary and Pancreatic Surgery, The Traditional Chinese Medicine Hospital of Ningbo, Ningbo, China
| | - Haifeng Chu
- Department of Neurosurgery, The Traditional Chinese Medicine Hospital of Linping District, Hangzhou, China
| |
Collapse
|
23
|
Li W, Huang X, Wei Y, Yin T, Diao L. Connecting the dots: the role of fatigue in female infertility. Reprod Biol Endocrinol 2024; 22:66. [PMID: 38849828 PMCID: PMC11157719 DOI: 10.1186/s12958-024-01235-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
Fatigue, an increasingly acknowledged symptom in various chronic diseases, has garnered heightened attention, during the medical era of bio-psycho-social model. Its persistence not only significantly compromises an individual's quality of life but also correlates with chronic organ damage. Surprisingly, the intricate relationship between fatigue and female reproductive health, specifically infertility, remains largely unexplored. Our exploration into the existing body of evidence establishes a compelling link between fatigue with uterine and ovarian diseases, as well as conditions associated with infertility, such as rheumatism. This observation suggests a potentially pivotal role of fatigue in influencing overall female fertility. Furthermore, we propose a hypothetical mechanism elucidating the impact of fatigue on infertility from multiple perspectives, postulating that neuroendocrine, neurotransmitter, inflammatory immune, and mitochondrial dysfunction resulting from fatigue and its co-factors may further contribute to endocrine disorders, menstrual irregularities, and sexual dysfunction, ultimately leading to infertility. In addition to providing this comprehensive theoretical framework, we summarize anti-fatigue strategies and accentuate current knowledge gaps. By doing so, our aim is to offer novel insights, stimulate further research, and advance our understanding of the crucial interplay between fatigue and female reproductive health.
Collapse
Grants
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
Collapse
Affiliation(s)
- Wenzhu Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Xiaoyan Huang
- Department of Rheumatology, The University of Hong Kong- Shenzhen Hospital, Shenzhen, 518053, China
| | - Yiqiu Wei
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China.
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518045, China.
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri- implantation, Shenzhen, 518045, China.
| |
Collapse
|
24
|
Yang Y, Fei Y, Xu X, Yao J, Wang J, Liu C, Ding H. Shikonin attenuates cerebral ischemia/reperfusion injury via inhibiting NOD2/RIP2/NF-κB-mediated microglia polarization and neuroinflammation. J Stroke Cerebrovasc Dis 2024; 33:107689. [PMID: 38527567 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107689] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/27/2024] [Accepted: 03/21/2024] [Indexed: 03/27/2024] Open
Abstract
OBJECTIVES Microglia-mediated neuroinflammation plays a crucial role in the pathophysiological process of multiple neurological disorders such as ischemic stroke, which still lacks effective therapeutic agents. Shikonin possesses anti-inflammatory and neuroprotective properties. However, its underlying mechanism remains elusive. This study aimed to investigate whether Shikonin confers protection against cerebral ischemia/reperfusion (I/R) injury by modulating microglial polarization and elucidate the associated mechanisms. METHODS This study employed an oxygen-glucose deprivation and reoxygenation (OGD/R) BV2 microglial cellular model and a middle cerebral artery occlusion/reperfusion (MCAO/R) animal model to investigate the protection and underlying mechanism of Shikonin against ischemic stroke. RESULTS The results demonstrated that Shikonin treatment significantly reduced brain infarction volume and improved neurological function in MCAO/R rats. Simultaneously, Shikonin treatment significantly reduced microglial proinflammatory phenotype and levels of proinflammatory markers (inducible-NO synthase (iNOS), tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and IL-6), increased microglial anti-inflammatory phenotype and levels of anti-inflammatory markers (Arginase-1 (Arg1), transforming growth factor-beta (TGF-β), and IL-10), reversed the expression of Nucleotide-binding oligomerization domain 2 (NOD2) and phosphorylation receptor interacting protein 2 (p-RIP2), and suppressed nuclear factor kappa-B (NF-κB) signaling activation in the ischemic penumbra regions. These effects of Shikonin were further corroborated in OGD/R-treated BV2 cells. Furthermore, overexpression of NOD2 markedly attenuated the neuroprotective effects of Shikonin treatment in MCAO/R rats. NOD2 overexpression also attenuated the regulatory effects of Shikonin on neuroinflammation, microglial polarization, and NF-κB signaling activation. CONCLUSION This study illustrates that Shikonin mitigates inflammation mediated by microglial proinflammatory polarization by inhibiting the NOD2/RIP2/NF-κB signaling pathway, thereby exerting a protective role. The findings uncover a potential molecular mechanism for Shikonin in treating ischemic stroke.
Collapse
Affiliation(s)
- Ya Yang
- College of Pharmacy, Xinjiang Medical University, No. 567 North Shangde Road, Urumqi, Xinjiang 830017, PR China
| | - Yuxiang Fei
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, PR China; School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xuejiao Xu
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jun Yao
- College of Pharmacy, Xinjiang Medical University, No. 567 North Shangde Road, Urumqi, Xinjiang 830017, PR China; Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi, Xinjiang 830017, PR China
| | - Jianing Wang
- Department of Pharmacy, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu 211100, PR China
| | - Chao Liu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, PR China; School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Haiyan Ding
- College of Pharmacy, Xinjiang Medical University, No. 567 North Shangde Road, Urumqi, Xinjiang 830017, PR China; Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi, Xinjiang 830017, PR China.
| |
Collapse
|
25
|
Zhou J, Ye W, Chen L, Li J, Zhou Y, Bai C, Luo L. Triptolide alleviates cerebral ischemia/reperfusion injury via regulating the Fractalkine/CX3CR1 signaling pathway. Brain Res Bull 2024; 211:110939. [PMID: 38574865 DOI: 10.1016/j.brainresbull.2024.110939] [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: 10/21/2023] [Revised: 03/16/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
PURPOSE To evaluate the potential efficacy of Triptolide (TP) on cerebral ischemia/reperfusion injury (CIRI) and to uncover the underlying mechanism through which TP regulates CIRI. METHODS We constructed a middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model to simulate CIRI, and established a lipopolysaccharide (LPS)-stimulated BV-2 cell model to mimic the inflammatory state during CIRI. The neurological deficits score (NS) of mice were measured for assessment of neurologic functions. Both the severity of cerebral infarction and the apoptosis level in mouse brain tissues or cells were respectively evaluated using corresponding techniques. The expression levels of Ionized calcium binding adapter molecule 1 (IBA-1), Inductible Nitric Oxide Synthase (iNOS), Arginase 1 (Arg-1), Tumor necrosis factor-α (TNF-α), Interleukin 1β (IL-1β), Cysteine histoproteinase S (CTSS), Fractalkine, chemokine C-X3-C motif receptor 1 (CX3CR1), BCL-2-associated X protein (BAX), and antiapoptotic proteins (Bcl-2) were detected using immunofluorescence, qRT-PCR as well as Western blot, respectively. RESULTS Relative to the Sham group, treatment with TP attenuated the increased NS, infarct area and apoptosis levels observed in MCAO/R mice. Upregulated expression levels of IBA-1, iNOS, Arg-1, TNF-α and IL-1β were found in MCAO/R mice, while TP suppressed iNOS, TNF-α and IL-1β expression, and enhanced Arg-1 expression in both MCAO/R mice and LPS-stimulated BV-2 cells. Besides, TP inhibited the CTSS/Fractalkine/CX3CR1 pathway activation in both MCAO/R mice and LPS-induced BV-2 cells, while overexpression of CTSS reversed such effect. Co-culturing HT-22 cells with TP+LPS-treated BV-2 cells led to enhanced cell viability and decreased apoptosis levels. However, overexpression of CTSS further aggravated HT-22 cell injury. CONCLUSION TP inhibits not only microglia polarization towards the M1 phenotype by suppressing the CTSS/Fractalkine/CX3CR1 pathway activation, but also HT-22 apoptosis by crosstalk with BV-2 cells, thereby ameliorating CIRI. These findings reveal a novel mechanism of TP in improving CIRI, and offer potential implications for addressing the preventive and therapeutic strategies of CIRI.
Collapse
Affiliation(s)
- Jiajun Zhou
- Department of Neurology, Affiliated Hangzhou Xixi Hospital Zhejiang University of Medicine, Hangzhou, Zhejiang, China
| | - Wei Ye
- Department of Neurology, Affiliated Hangzhou Xixi Hospital Zhejiang University of Medicine, Hangzhou, Zhejiang, China
| | - Ling Chen
- Department of Neurology, Affiliated Hangzhou Xixi Hospital Zhejiang University of Medicine, Hangzhou, Zhejiang, China
| | - Junheng Li
- Department of Neurology, Affiliated Hangzhou Xixi Hospital Zhejiang University of Medicine, Hangzhou, Zhejiang, China
| | - Yijun Zhou
- Department of Liver Diseases, Affiliated Hangzhou Xixi Hospital Zhejiang University of Medicine, Hangzhou, Zhejiang, China
| | - Chunfeng Bai
- Department of Neurology, Affiliated Hangzhou Xixi Hospital Zhejiang University of Medicine, Hangzhou, Zhejiang, China
| | - Lian Luo
- Department of Neurology, Affiliated Hangzhou Xixi Hospital Zhejiang University of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
26
|
Shui X, Chen J, Fu Z, Zhu H, Tao H, Li Z. Microglia in Ischemic Stroke: Pathogenesis Insights and Therapeutic Challenges. J Inflamm Res 2024; 17:3335-3352. [PMID: 38800598 PMCID: PMC11128258 DOI: 10.2147/jir.s461795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
Ischemic stroke is the most common type of stroke, which is the main cause of death and disability on a global scale. As the primary immune cells in the brain that are crucial for preserving homeostasis of the central nervous system microenvironment, microglia have been found to exhibit dual or even multiple effects at different stages of ischemic stroke. The anti-inflammatory polarization of microglia and release of neurotrophic factors may provide benefits by promoting neurological recovery at the lesion in the early phase after ischemic stroke. However, the pro-inflammatory polarization of microglia and secretion of inflammatory factors in the later phase of injury may exacerbate the ischemic lesion, suggesting the therapeutic potential of modulating the balance of microglial polarization to predispose them to anti-inflammatory transformation in ischemic stroke. Microglia-mediated signaling crosstalk with other cells may also be key to improving functional outcomes following ischemic stroke. Thus, this review provides an overview of microglial functions and responses under physiological and ischemic stroke conditions, including microglial activation, polarization, and interactions with other cells. We focus on approaches that promote anti-inflammatory polarization of microglia, inhibit microglial activation, and enhance beneficial cell-to-cell interactions. These targets may hold promise for the creation of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xinyao Shui
- Clinical Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Jingsong Chen
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Ziyue Fu
- Clinical Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Haoyue Zhu
- Clinical Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Hualin Tao
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Zhaoyinqian Li
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| |
Collapse
|
27
|
Tiwari S, Basnet N, Choi JW. Lysophosphatidic Acid Receptor 1 Plays a Pathogenic Role in Permanent Brain Ischemic Stroke by Modulating Neuroinflammatory Responses. Biomol Ther (Seoul) 2024; 32:319-328. [PMID: 38627097 PMCID: PMC11063482 DOI: 10.4062/biomolther.2024.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 05/01/2024] Open
Abstract
Lysophosphatidic acid receptor 1 (LPA1) plays a critical role in brain injury following a transient brain ischemic stroke. However, its role in permanent brain ischemic stroke remains unknown. To address this, we investigated whether LPA1 could contribute to brain injury of mice challenged by permanent middle cerebral artery occlusion (pMCAO). A selective LPA1 antagonist (AM152) was used as a pharmacological tool for this investigation. When AM152 was given to pMCAO-challenged mice one hour after occlusion, pMCAO-induced brain damage such as brain infarction, functional neurological deficits, apoptosis, and blood-brain barrier disruption was significantly attenuated. Histological analyses demonstrated that AM152 administration attenuated microglial activation and proliferation in injured brain after pMCAO challenge. AM152 administration also attenuated abnormal neuroinflammatory responses by decreasing expression levels of pro-inflammatory cytokines while increasing expression levels of anti-inflammatory cytokines in the injured brain. As underlying effector pathways, NF-κB, MAPKs (ERK1/2, p38, and JNKs), and PI3K/Akt were found to be involved in LPA1-dependent pathogenesis. Collectively, these results demonstrate that LPA1 can contribute to brain injury by permanent ischemic stroke, along with relevant pathogenic events in an injured brain.
Collapse
Affiliation(s)
- Supriya Tiwari
- Laboratory of Neuropharmacology, College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Republic of Korea
| | - Nikita Basnet
- Laboratory of Neuropharmacology, College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Republic of Korea
| | - Ji Woong Choi
- Laboratory of Neuropharmacology, College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Republic of Korea
| |
Collapse
|
28
|
Chen P, Wang W, Ban W, Zhang K, Dai Y, Yang Z, You Y. Deciphering Post-Stroke Sleep Disorders: Unveiling Neurological Mechanisms in the Realm of Brain Science. Brain Sci 2024; 14:307. [PMID: 38671959 PMCID: PMC11047862 DOI: 10.3390/brainsci14040307] [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: 02/21/2024] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024] Open
Abstract
Sleep disorders are the most widespread mental disorders after stroke and hurt survivors' functional prognosis, response to restoration, and quality of life. This review will address an overview of the progress of research on the biological mechanisms associated with stroke-complicating sleep disorders. Extensive research has investigated the negative impact of stroke on sleep. However, a bidirectional association between sleep disorders and stroke exists; while stroke elevates the risk of sleep disorders, these disorders also independently contribute as a risk factor for stroke. This review aims to elucidate the mechanisms of stroke-induced sleep disorders. Possible influences were examined, including functional changes in brain regions, cerebrovascular hemodynamics, neurological deficits, sleep ion regulation, neurotransmitters, and inflammation. The results provide valuable insights into the mechanisms of stroke complicating sleep disorders.
Collapse
Affiliation(s)
- Pinqiu Chen
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, Yantai 264005, China; (P.C.)
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Wenyan Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, Yantai 264005, China; (P.C.)
| | - Weikang Ban
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Kecan Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yanan Dai
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Zhihong Yang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yuyang You
- School of Automation, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
29
|
Olas B. New light on changes in the number and function of blood platelets stimulated by cocoa and its products. Front Pharmacol 2024; 15:1366076. [PMID: 38533262 PMCID: PMC10963627 DOI: 10.3389/fphar.2024.1366076] [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: 01/05/2024] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
Hyperactivation of blood platelets, one of the causes of heart attack, and other cardiovascular diseases (CVDs), is influenced by various dietary components, including phenolic compounds from vegetables, fruits, teas, wines, cocoa and its products, including chocolate. The present paper sheds new light on the effect of cocoa and its products, especially dark chocolate, on the number and function of blood platelets, and the anti-platelet activity of their constituent phenolic compounds. A review was performed of papers identified in various electronic databases, including PubMed, Science Direct, Scopus, Web of Knowledge, and Google Scholar, with the aim of determining whether their anti-platelet activity may serve as part of a sweet strategy in countering CVDs. Various studies demonstrate that cocoa consumption, especially in the form of dark chocolate, with a high flavanol concentration, has anti-platelet activity and may play a significant role in cardioprotection; they also note that cocoa consumption may be a good strategy in diminishing cardiovascular risk, including hyperactivation of blood platelets.
Collapse
Affiliation(s)
- Beata Olas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| |
Collapse
|
30
|
Yuan J, Wang Y, Gao J, Zhang X, Xing J. Eicosapentaenoic Acid Alleviates Inflammatory Response and Insulin Resistance in Pregnant Mice With Gestational Diabetes Mellitus. Physiol Res 2024; 73:57-68. [PMID: 38466005 PMCID: PMC11019622 DOI: 10.33549/physiolres.935113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 09/13/2023] [Indexed: 04/26/2024] Open
Abstract
This study investigated the effect of eicosapentaenoic acid (EPA) on insulin resistance in pregnant mice with gestational diabetes mellitus (GDM) and underlying mechanism. C57BL/6 mice fed with a high-fat diet for 4 weeks and the newly gestated were selected and injected with streptozotocin for GDM modeling. We demonstrated that the fasting insulin levels (FINS) and insulin sensitivity index (ISI) in serum and blood glucose level were significantly higher in GDM group than in normal control (NC) group. The low or high dose of EPA intervention reduced these levels, and the effect of high dose intervention was more significant. The area under the curve in GDM group was higher than that of NC group, and then gradually decreased after low or high dose of EPA treatment. The serum levels of TC, TG and LDL were increased in GDM group, while decreased in EPA group. GDM induced down-regulation of HDL level, and the low or high dose of EPA gradually increased this level. The levels of p-AKT2Ser, p-IRS-1Tyr, GLUT4, and ratios of pIRS-1Tyr/IRS-1 and pAKT2Ser/AKT2 in gastrocnemius muscle were reduced in GDM group, while low or high dose of EPA progressively increased these alterations. GDM enhanced TLR4, NF-kappaB p65, IL-1beta, IL-6 and TNF-alpha levels in placental tissues, and these expressions were declined at different dose of EPA, and the decrease was greater at high dose. We concluded that EPA receded the release of inflammatory factors in the placental tissues by inhibiting the activation of TLR4 signaling, thereby alleviating the IR.
Collapse
Affiliation(s)
- J Yuan
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital, Lubei District, Tangshan, Hebei, People's Republic of China.
| | | | | | | | | |
Collapse
|
31
|
Zou Y, Pei J, Wan C, Liu S, Hu B, Li Z, Tang Z. Mechanism of scutellarin inhibition of astrocyte activation to type A1 after ischemic stroke. J Stroke Cerebrovasc Dis 2024; 33:107534. [PMID: 38219378 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 01/16/2024] Open
Abstract
OBJECTIVE The aim of this study was to investigate the effects of scutellarin on the activation of astrocytes into the A1 type following cerebral ischemia and to explore the underlying mechanism. METHODS In vivo, a mouse middle cerebral artery wire embolism model was established to observe the regulation of astrocyte activation to A1 type by scutellarin, and the effects on neurological function and brain infarct volume. In vitro, primary astrocytes were cultured to establish an oxygen-glucose deprivation model, and the mRNA and protein expression of C3, a specific marker of A1-type astrocytes pretreated with scutellarin, were examined. The neurons were cultured in vitro to detect the toxic effects of ischemia-hypoxia-activated A1 astrocyte secretion products on neurons, and to observe whether scutellarin could reduce the neurotoxicity of A1 astrocytes. To validate the signaling pathway-related proteins regulated by scutellarin on C3 expression in astrocytes. RESULTS The results showed that scutellarin treatment reduced the volume of cerebral infarcts and attenuated neurological deficits in mice caused by middle cerebral artery embolism. Immunofluorescence and Western blot showed that treatment with scutellarin down-regulated middle cerebral artery embolism and OGD/R up-regulated A1-type astrocyte marker C3. The secretory products of ischemia-hypoxia-activated A1-type astrocytes were toxic to neurons and induced an increase in neuronal apoptosis, and astrocytes treated with scutellarin reduced the toxic effects on neurons. Further study revealed that scutellarin inhibited the activation of NF-κB signaling pathway and thus inhibited the activation of astrocytes to A1 type.
Collapse
Affiliation(s)
- Yongwei Zou
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wuhua District, Kunming, Yunnan Province, China
| | - Jingchun Pei
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wuhua District, Kunming, Yunnan Province, China
| | - Cheng Wan
- Department of Medical Imaging, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shuangshuang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wuhua District, Kunming, Yunnan Province, China
| | - Bin Hu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wuhua District, Kunming, Yunnan Province, China
| | - Zhigao Li
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wuhua District, Kunming, Yunnan Province, China
| | - Zhiwei Tang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wuhua District, Kunming, Yunnan Province, China.
| |
Collapse
|
32
|
Briones-Valdivieso C, Briones F, Orellana-Urzúa S, Chichiarelli S, Saso L, Rodrigo R. Novel Multi-Antioxidant Approach for Ischemic Stroke Therapy Targeting the Role of Oxidative Stress. Biomedicines 2024; 12:501. [PMID: 38540114 PMCID: PMC10968576 DOI: 10.3390/biomedicines12030501] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/03/2024] [Accepted: 02/10/2024] [Indexed: 01/03/2025] Open
Abstract
Stroke is a major contributor to global mortality and disability. While reperfusion is essential for preventing neuronal death in the penumbra, it also triggers cerebral ischemia-reperfusion injury, a paradoxical injury primarily caused by oxidative stress, inflammation, and blood-brain barrier disruption. An oxidative burst inflicts marked cellular damage, ranging from alterations in mitochondrial function to lipid peroxidation and the activation of intricate signalling pathways that can even lead to cell death. Thus, given the pivotal role of oxidative stress in the mechanisms of cerebral ischemia-reperfusion injury, the reinforcement of the antioxidant defence system has been proposed as a protective approach. Although this strategy has proven to be successful in experimental models, its translation into clinical practice has yielded inconsistent results. However, it should be considered that the availability of numerous antioxidant molecules with a wide range of chemical properties can affect the extent of injury; several groups of antioxidant molecules, including polyphenols, carotenoids, and vitamins, among other antioxidant compounds, can mitigate this damage by intervening in multiple signalling pathways at various stages. Multiple clinical trials have previously been conducted to evaluate these properties using melatonin, acetyl-L-carnitine, chrysanthemum extract, edaravone dexborneol, saffron, coenzyme Q10, and oleoylethanolamide, among other treatments. Therefore, multi-antioxidant therapy emerges as a promising novel therapeutic option due to the potential synergistic effect provided by the simultaneous roles of the individual compounds.
Collapse
Affiliation(s)
| | - Felipe Briones
- Institute for Public Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany;
| | - Sofía Orellana-Urzúa
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile;
| | - Silvia Chichiarelli
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine Sapienza University, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile;
| |
Collapse
|
33
|
Sun R, Wang YF, Yang X. Knockdown of IFIT3 ameliorates multiple sclerosis via selectively regulating M1 polarization of microglia in an experimental autoimmune encephalomyelitis model. Int Immunopharmacol 2024; 128:111501. [PMID: 38232539 DOI: 10.1016/j.intimp.2024.111501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/19/2024]
Abstract
The key to the treatment of multiple sclerosis (MS) is to promote the transition from inflammation-induced demyelination to remyelination. Polarization of microglia towards M1 or M2 phenotype is critical in this transition. Interferon induced protein with tetratricopeptide repeats 3 (IFIT3) is involved in inflammatory reaction and up-regulated in M1-polarized macrophages. However, its effect on microglia during MS has not been reported. In this paper, we demonstrated the important role of IFIT3 in selectively regulating microglia polarization. The expression of IFIT3 was increased when microglia were polarized towards M1, but did not change under M2 polarization. The knockdown of IFIT3 selectively inhibited M1 polarization, while M2 polarization was not affected by IFIT3 silencing. Furthermore, the activation of signal transducer and activator of transcription 1 (STAT1) and nuclear factor kappa-B (NF-ĸB) signaling in M1 polarized microglia was suppressed by downregulating IFIT3. In experimental autoimmune encephalitis (EAE) mice, an animal model of MS, IFIT3 expression was upregulated. The disease progression, inflammatory infiltration and demyelination in the EAE mice were alleviated by silencing IFIT3. The inhibitory effects of IFIT3 knockdown on M1 polarization and STAT1 and NF-ĸB pathways were also confirmed in the spinal cord of EAE mice. In summary, our findings suggest that IFIT3 selectively intensified microglia polarization towards the pro-inflammatory M1 phenotype, and may contribute to the progression of MS.
Collapse
Affiliation(s)
- Ran Sun
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, China
| | - Yan-Fang Wang
- Department of Orthopedics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, China
| | - Xue Yang
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, China.
| |
Collapse
|
34
|
Min XL, Jia WJ, Guo L, Jing R, Zhao XH, Hu JY, Li XH, Liu W, Wang T, Dou XK. Brain microvascular endothelial cell-derived exosomes transmitting circ_0000495 promote microglial M1-polarization and endothelial cell injury under hypoxia condition. FASEB J 2024; 38:e23387. [PMID: 38193649 DOI: 10.1096/fj.202301637r] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/15/2023] [Accepted: 12/11/2023] [Indexed: 01/10/2024]
Abstract
Human brain microvascular endothelial cells (HBMVECs) and microglia play critical roles in regulating cerebral homeostasis during ischemic stroke. However, the role of HBMVECs-derived exosomes in microglia polarization after stroke remains unknown. We isolated exosomes (Exos) from oxygen glucose deprivation (OGD)-exposed HBMVECs, before added them into microglia. Microglia polarization markers were tested using RT-qPCR or flow cytometry. Inflammatory cytokines were measured with ELISA. Endothelial cell damage was assessed by cell viability, apoptosis, apoptosis-related proteins, oxidative stress, and angiogenic activity using CCK-8, flow cytometry, western blot, ELISA, and endothelial tube formation assay, respectively. We also established middle cerebral artery occlusion (MCAO) mice model to examine the function of circ_0000495 on stroke in vivo. Our study found that HBMVECs-Exos reduced M2 markers (IL-10, CD163, and CD206), increased M1 markers (TNF-α, IL-1β, and IL-12), CD86-positive cells, and inflammatory cytokines (TNF-α and IL-1β), indicating the promotion of microglial M1-polarization. Microglial M1-polarization induced by HBMVECs-Exos reduced viability and promoted apoptosis and oxidative stress, revealing the aggravation of endothelial cell damage. However, circ_0000495 silencing inhibited HBMVECs-Exos-induced alterations. Mechanistically, circ_0000495 adsorbed miR-579-3p to upregulate toll-like receptor 4 (TLR4) in microglia; miR-579-3p suppressed HBMVECs-Exos-induced alterations via declining TLR4; furthermore, Yin Yang 1 (YY1) transcriptionally activated circ_0000495 in HBMVECs. Importantly, circ_0000495 aggravated ischemic brain injury in vivo via activating TLR4/nuclear factor-κB (NF-κB) pathway. Collectively, OGD-treated HBMVECs-Exos transmitted circ_0000495 to regulate miR-579-3p/TLR4/NF-κB axis in microglia, thereby facilitating microglial M1-polarization and endothelial cell damage.
Collapse
Affiliation(s)
- Xiao-Li Min
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wen-Ji Jia
- Department of Neurology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li Guo
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Rui Jing
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiao-Hong Zhao
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jia-Yi Hu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xu-Hui Li
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wei Liu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xing-Kui Dou
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
35
|
Li R, Song M, Zheng Y, Zhang J, Zhang S, Fan X. Naoxueshu oral liquid promotes hematoma absorption by targeting CD36 in M2 microglia via TLR4/MyD88/NF-κB signaling pathway in rats with intracerebral hemorrhage. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117116. [PMID: 37659762 DOI: 10.1016/j.jep.2023.117116] [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: 06/10/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Intracerebral hemorrhage (ICH) is a major public health issue that leads to elevated rates of death and disability and has few proven treatments. Naoxueshu oral liquid (NXS), a TCM patent drug, is widely used in patients with ICH. Although a series of clinical studies have confirmed the efficacy and safety of NXS, the underlying mechanism of hematoma absorption is unclear. AIM OF THE STUDY Our work aimed to elucidate the effect and mechanism of NXS on hematoma absorption in rats with ICH. MATERIALS AND METHODS Induction of ICH model in the rats with intracerebral injection of collagenase VII, followed by treatment with NXS and Edaravone as a control neuroprotection medication. Neural functional recovery was assessed using mNSS, foot fault test, corner test, forelimb grip-traction test, and adhesive removal test. Hematoma absorption was assessed by the spectrophotometric hemoglobin assay with Drabkin's reagent. The protein expression of CD36, M2 microglia marker (CD206 and YM-1) and TLR4/MyD88/NF-κB pathway related proteins were determined by Western blot and immunofluorescence. RESULTS NXS could significantly ameliorate the ICH recovery of neural and locomotor function as well as reduce hemorrhage volume. NXS could increase the expression of CD36 expressed in M2 microglia and promote M2 microglia polarization. Simultaneously, NXS significantly suppressed protein expressions of TLR4, MyD88, and NF-κB following ICH in rats. The results indicated that lipopolysaccharide (LPS), TLR4 specific agonist, could partially reverse the change in ICH rats administrated with NXS. CONCLUSIONS NXS promotes hematoma absorption by targeting CD36 expression in M2 microglia via TLR4/MyD88/NF-κB signaling pathway in rats with ICH. Collectively, current research provides a novel theoretical basis for the clinical application of NXS.
Collapse
Affiliation(s)
- Ruoqi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Meiying Song
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yingyi Zheng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Jiaxue Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Shanshan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| |
Collapse
|
36
|
Zhang BW, Sun KH, Liu T, Zou W. The Crosstalk Between Immune Cells After Intracerebral Hemorrhage. Neuroscience 2024; 537:93-104. [PMID: 38056621 DOI: 10.1016/j.neuroscience.2023.11.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/28/2023] [Accepted: 11/15/2023] [Indexed: 12/08/2023]
Abstract
The inflammatory mechanism of intracerebral hemorrhage (ICH) has been widely studied, and it is believed that the regulation of this mechanism is of great significance to the prognosis. In the early stage of the acute phase of ICH, the release of a large number of inflammatory factors around the hematoma can recruit more inflammatory cells to infiltrate the area, further release inflammatory factors, cause an inflammatory cascade reaction, aggravate the volume of cerebral hematoma and edema and further destroy the blood-brain barrier (BBB), according to this, the crosstalk between cells may be of great significance in secondary brain injury (SBI). Because most of the cells recruited are inflammatory immune cells, this paper mainly discusses the cells based on the inflammatory mechanism to discuss their functions after ICH, we found that among the main cells inherent in the brain, glial cells account for the majority, of which microglia are the most widely studied and it can interact with a variety of cells, which is reflected in the literature researches on its pathogenesis and treatment. We believe that exploring multi-mechanism and multi-cell regulated drugs may be the future development trend, and the existing research, the comparison and unification of modeling methods, and the observation of long-term efficacy may be the first problem that researchers need to solve.
Collapse
Affiliation(s)
- Bai-Wen Zhang
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Ke-Han Sun
- Rehabilitation Department, Maternal and Child Health Hospital of Xing-an League, Ulanhot City, Inner Mongolia 137400, China
| | - Ting Liu
- Rehabilitation Department, Pengzhou Traditional Chinese Medicine Hospital, Chengdu 611930, China
| | - Wei Zou
- The Third Acupuncture Department, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
37
|
Xu W, Wang X, Hou X, Yang Y, Ma R, Lv R, Yin Q. The role of microglia in the pathogenesis of diabetic-associated cognitive dysfunction. Front Endocrinol (Lausanne) 2024; 14:1246979. [PMID: 38274227 PMCID: PMC10808430 DOI: 10.3389/fendo.2023.1246979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 12/29/2023] [Indexed: 01/27/2024] Open
Affiliation(s)
- Wenwen Xu
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xinyu Wang
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xunyao Hou
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yan Yang
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Rongrong Ma
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Renjun Lv
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qingqing Yin
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
38
|
Hasan GM, Anwar S, Shamsi A, Sohal SS, Hassan MI. The neuroprotective potential of phytochemicals in traumatic brain injury: mechanistic insights and pharmacological implications. Front Pharmacol 2024; 14:1330098. [PMID: 38239205 PMCID: PMC10794744 DOI: 10.3389/fphar.2023.1330098] [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: 10/30/2023] [Accepted: 12/15/2023] [Indexed: 01/22/2024] Open
Abstract
Traumatic brain injury (TBI) leads to brain damage, comprising both immediate primary damage and a subsequent cascade of secondary injury mechanisms. The primary injury results in localized brain damage, while the secondary damage initiates inflammatory responses, followed by the disruption of the blood-brain barrier, infiltration of peripheral blood cells, brain edema, and the release of various immune mediators, including chemotactic factors and interleukins. TBI disrupts molecular signaling, cell structures, and functions. In addition to physical tissue damage, such as axonal injuries, contusions, and haemorrhages, TBI interferes with brain functioning, impacting cognition, decision-making, memory, attention, and speech capabilities. Despite a deep understanding of the pathophysiology of TBI, an intensive effort to evaluate the underlying mechanisms with effective therapeutic interventions is imperative to manage the repercussions of TBI. Studies have commenced to explore the potential of employing natural compounds as therapeutic interventions for TBI. These compounds are characterized by their low toxicity and limited interactions with conventional drugs. Moreover, many natural compounds demonstrate the capacity to target various aspects of the secondary injury process. While our understanding of the pathophysiology of TBI, there is an urgent need for effective therapeutic interventions to mitigate its consequences. Here, we aimed to summarize the mechanism of action and the role of phytochemicals against TBI progression. This review discusses the therapeutic implications of various phytonutrients and addresses primary and secondary consequences of TBI. In addition, we highlighted the roles of emerging phytochemicals as promising candidates for therapeutic intervention of TBI. The review highlights the neuroprotective roles of phytochemicals against TBI and the mechanistic approach. Furthermore, our efforts focused on the underlying mechanisms, providing a better understanding of the therapeutic potential of phytochemicals in TBI therapeutics.
Collapse
Affiliation(s)
- Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
39
|
Yuhan L, Khaleghi Ghadiri M, Gorji A. Impact of NQO1 dysregulation in CNS disorders. J Transl Med 2024; 22:4. [PMID: 38167027 PMCID: PMC10762857 DOI: 10.1186/s12967-023-04802-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
NAD(P)H Quinone Dehydrogenase 1 (NQO1) plays a pivotal role in the regulation of neuronal function and synaptic plasticity, cellular adaptation to oxidative stress, neuroinflammatory and degenerative processes, and tumorigenesis in the central nervous system (CNS). Impairment of the NQO1 activity in the CNS can result in abnormal neurotransmitter release and clearance, increased oxidative stress, and aggravated cellular injury/death. Furthermore, it can cause disturbances in neural circuit function and synaptic neurotransmission. The abnormalities of NQO1 enzyme activity have been linked to the pathophysiological mechanisms of multiple neurological disorders, including Parkinson's disease, Alzheimer's disease, epilepsy, multiple sclerosis, cerebrovascular disease, traumatic brain injury, and brain malignancy. NQO1 contributes to various dimensions of tumorigenesis and treatment response in various brain tumors. The precise mechanisms through which abnormalities in NQO1 function contribute to these neurological disorders continue to be a subject of ongoing research. Building upon the existing knowledge, the present study reviews current investigations describing the role of NQO1 dysregulations in various neurological disorders. This study emphasizes the potential of NQO1 as a biomarker in diagnostic and prognostic approaches, as well as its suitability as a target for drug development strategies in neurological disorders.
Collapse
Affiliation(s)
- Li Yuhan
- Epilepsy Research Center, Münster University, Münster, Germany
- Department of Breast Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Ali Gorji
- Epilepsy Research Center, Münster University, Münster, Germany.
- Department of Neurosurgery, Münster University, Münster, Germany.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
40
|
Lekhak N, Bhattarai HK. Phytochemicals in Cancer Chemoprevention: Preclinical and Clinical Studies. Cancer Control 2024; 31:10732748241302902. [PMID: 39629692 PMCID: PMC11615997 DOI: 10.1177/10732748241302902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/11/2024] [Accepted: 11/11/2024] [Indexed: 12/08/2024] Open
Abstract
Phytochemicals, chemicals from plants, have garnered huge attention for their potential ability to prevent cancer. In vivo and preclinical models show that they do so often by affecting the hallmarks of cancer. Phytochemicals affect key pathways involved in the survival, genome maintenance, proliferation, senescence, and transendothelial migration of cancer cells. Some phytochemicals, namely antioxidants, can scavenge and quench reactive oxygen species (ROS) to prevent lipid peroxidation and DNA damage. They also trigger apoptosis by stopping the cell cycle at checkpoints to initiate the DNA damage response. Numerous in vitro and in vivo studies suggest that phytochemicals hinder cancer onset and progression by modifying major cell signaling pathways such as JAK/STAT, PI3K/Akt, Wnt, NF-kB, TGF-β, and MAPK. It is a well-known fact that the occurrence of cancer is in itself a very intricate process involving multiple mechanisms concurrently. Cancer prevention using phytochemicals is also an equally complex process that requires investigation and understanding of a myriad of processes going on in the cells and tissues. While many in vitro and preclinical studies have established that phytochemicals may be potential chemopreventive agents of cancer, their role in clinical randomized control trials needs to be established. This paper aims to shed light on the dynamics of chemoprevention using phytochemicals.
Collapse
Affiliation(s)
- Nitish Lekhak
- Department of Biotechnology, Kathmandu University, Dhulikhel, Nepal
| | | |
Collapse
|
41
|
Long Y, Li XQ, Deng J, Ye QB, Li D, Ma Y, Wu YY, Hu Y, He XF, Wen J, Shi A, Yu S, Shen L, Ye Z, Zheng C, Li N. Modulating the polarization phenotype of microglia - A valuable strategy for central nervous system diseases. Ageing Res Rev 2024; 93:102160. [PMID: 38065225 DOI: 10.1016/j.arr.2023.102160] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
Central nervous system (CNS) diseases have become one of the leading causes of death in the global population. The pathogenesis of CNS diseases is complicated, so it is important to find the patterns of the disease to improve the treatment strategy. Microglia are considered to be a double-edged sword, playing both harmful and beneficial roles in CNS diseases. Therefore, it is crucial to understand the progression of the disease and the changes in the polar phenotype of microglia to provide guidance in the treatment of CNS diseases. Microglia activation may evolve into different phenotypes: M1 and M2 types. We focused on the roles that M1 and M2 microglia play in regulating intercellular dialogues, pathological reactions and specific diseases in CNS diseases. Importantly, we summarized the strategies used to modulate the polarization phenotype of microglia, including traditional pharmacological modulation, biological therapies, and physical strategies. This review will contribute to the development of potential strategies to modulate microglia polarization phenotypes and provide new alternative therapies for CNS diseases.
Collapse
Affiliation(s)
- Yu Long
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Xiao-Qiu Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jie Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Qiao-Bo Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yin Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yuan-Yuan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yue Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Xiao-Fang He
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jing Wen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Ai Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Shuang Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Lin Shen
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medine, Tianjin, China.
| | - Zhen Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Chuan Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Nan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
42
|
Dassamiour S, Bensaad MS, Ghebache W. Utility of phenolic acids in neurological disorders. ADVANCEMENT OF PHENOLIC ACIDS IN DRUG DISCOVERY 2024:295-344. [DOI: 10.1016/b978-0-443-18538-0.00015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
43
|
Sun C, Rahman MSU, Enkhjargal B, Peng J, Zhou K, Xie Z, Wu L, Zhang T, Zhu Q, Tang J, Zeng Y, Zhang JH, Xu S. Osteopontin modulates microglial activation states and attenuates inflammatory responses after subarachnoid hemorrhage in rats. Exp Neurol 2024; 371:114585. [PMID: 37884185 DOI: 10.1016/j.expneurol.2023.114585] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/10/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
AIMS Osteopontin (OPN) has demonstrated neuroprotective effects in various stroke models. Its role in neuroinflammation after brain injury remains to be elucidated. This study aims to clarify the effect of OPN on neuroinflammation, particularly on the functional states of microglia after subarachnoid hemorrhage (SAH). METHODS 77 rats were randomly divided into the following groups: Sham, SAH 24 h, SAH + rOPN, SAH + Vehicle (PBS), SAH + OPN siRNA, and SAH + Scr siRNA, SAH + rOPN+Fib-14 and SAH + rOPN+DMSO. Modified Garcia and beam balance tests were used to evaluate neurobehavioral outcomes. Semi-quantitative immunofluorescence staining was performed to measure expression of myeloperoxidase (MPO) and microglia activation state markers CD16, CD206 after SAH and recombinant OPN treatment. The quantification of microglia activation and functional markers CD16, CD206, TNF-α and IL-10 were further evaluated using Western-blotting. RESULTS Nasal administration of rOPN improved neurological dysfunction, attenuated neutrophil infiltration, and decreased expression of phenotypic and functional markers of pro-inflammatory microglia CD16 and TNF-α. It also promoted an anti-inflammatory microglial state, as evidenced by increased expression of CD206 and IL-10. Furthermore, after blocking the phosphorylation of FAK signaling, the effects of rOPN on microglial activation states were partially reversed. The downstream pathways of STAT3 and NF-κB also exhibited consistent changes, suggesting the involvement of the STAT3 and NF-κB pathways in OPN's modulation of microglial activation via integrin-FAK signaling. CONCLUSION OPN attenuates inflammatory responses after SAH by promoting an anti-inflammatory microglial state, potentially mediated through the integrin-FAK-STAT3 and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Chengmei Sun
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Muhammad Saif Ur Rahman
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Budbazar Enkhjargal
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jianhua Peng
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Keren Zhou
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Zhiyi Xie
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Lingyun Wu
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Tongyu Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Qiquan Zhu
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Yujia Zeng
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA.
| | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
44
|
Nie H, Ge J, Yang K, Peng Z, Wu H, Yang T, Mei Z. Naotaifang III Protects Against Cerebral Ischemia Injury Through LPS/TLR4 Signaling Pathway in the Microbiota-Gut-Brain Axis. Drug Des Devel Ther 2023; 17:3571-3588. [PMID: 38058793 PMCID: PMC10697094 DOI: 10.2147/dddt.s421658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023] Open
Abstract
Background Ischemic stroke (IS) is a leading cause of mortality worldwide. Naotaifang III is a new Chinese herbal formula to treat IS. Previous studies have shown that Astragali Radix, Puerariae Lobatae Radix, Chuanxiong Rhizoma, and Rhei Radix Et Rhizoma in Naotaifang III were able to regulate the imbalance of intestinal microbiota during cerebral ischemia injury. Methods Rats were randomly divided into sham operation group, normal control group, middle cerebral artery occlusion (MCAO) group, intestinal microbiota imbalance MCAO group, Naotaifang III group, and normal bacteria transplantation group, with 15 rats in each group. Then, neurological function scores and cerebral infarction volume were detected; haematoxylin and eosin staining and Golgi silver staining were used to observe morphological changes in brain tissue. Meanwhile, the lipopolysaccharide (LPS) and cerebral cortex interleukin (IL)-1β were detected by enzyme-linked immunosorbent assay (ELISA); the expressions of Toll-like receptor (TLR)-4 and nuclear factor kappa-B (NF-κB) proteins were detected by immunofluorescence and Western blot. The cecal flora was detected by 16S rDNA. The results showed that gut dysbiosis aggravated cerebral ischemic injury and significantly increased the expression of LPS, TLR4, NF-κB, and IL-1β, which could be significantly reversed by Naotaifang III or normal bacterial transplantation. Naotaifang III may exert a protective effect on neuroinflammatory injury after MCAO through the LPS/TLR4 signaling pathway in the microbe-gut-brain axis. In summary, Naotaifang III may induce anti-neuroinflammatory molecular mechanisms and signaling pathways through the microbe-gut-brain axis. Results The results showed that gut dysbiosis aggravated cerebral ischemic injury and significantly increased the expression of LPS, TLR4, NF-κB, and IL-1β, which could be significantly reversed by Naotaifang III or normal bacterial transplantation. Naotaifang III may exert a protective effect on neuroinflammatory injury after MCAO through the LPS/TLR4 signaling pathway in the microbe-gut-brain axis. Conclusion Naotaifang III may induce anti-neuroinflammatory molecular mechanisms and signaling pathways through the microbe-gut-brain axis.
Collapse
Affiliation(s)
- Huifang Nie
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
- Hunan Academy of Chinese Medicine, Changsha, Hunan, 410006, People’s Republic of China
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Zhuli Peng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Haihui Wu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Tong Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| |
Collapse
|
45
|
Du X, Amin N, Xu L, Botchway BOA, Zhang B, Fang M. Pharmacological intervention of curcumin via the NLRP3 inflammasome in ischemic stroke. Front Pharmacol 2023; 14:1249644. [PMID: 37915409 PMCID: PMC10616488 DOI: 10.3389/fphar.2023.1249644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023] Open
Abstract
Ischemic-induced neuronal injury arises due to low oxygen/nutrient levels and an inflammatory response that exacerbates neuronal loss. NOD-like receptor family pyrin domain-containing 3 (NLRP3) is an important regulator of inflammation after ischemic stroke, with its inhibition being involved in nerve regeneration. Curcumin, a main active ingredient in Chinese herbs, plays a positive role in neuronal repair and neuroprotection by regulating the NLRP3 signaling pathway. Nevertheless, the signaling mechanisms relating to how curcumin regulates NLRP3 inflammasome in inflammation and neural restoration following ischemic stroke are unknown. In this report, we summarize the main biological functions of the NLRP3 inflammasome along with the neuroprotective effects and underlying mechanisms of curcumin via impairment of the NLRP3 pathway in ischemic brain injury. We also discuss the role of medicinal interventions that target the NLRP3 and potential pathways, as well as possible directions for curcumin therapy to penetrate the blood-brain barrier (BBB) and hinder inflammation in ischemic stroke. This report conclusively demonstrates that curcumin has neuroprotective properties that inhibit inflammation and prevent nerve cell loss, thereby delaying the progression of ischemic brain damage.
Collapse
Affiliation(s)
- Xiaoxue Du
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Nashwa Amin
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt
| | - Linhao Xu
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Benson O. A. Botchway
- Department of Neurology, Children’s Hospital of Zhejiang University School of Medicine, National Clinical Research Centre for Child Health, Hangzhou, China
- Pharmacy Department, Bupa Cromwell Hospital, London, United Kingdom
| | - Bo Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Marong Fang
- Department of Neurology, Children’s Hospital of Zhejiang University School of Medicine, National Clinical Research Centre for Child Health, Hangzhou, China
| |
Collapse
|
46
|
Wang Y, Liu W, Geng P, Du W, Guo C, Wang Q, Zheng GQ, Jin X. Role of Crosstalk between Glial Cells and Immune Cells in Blood-Brain Barrier Damage and Protection after Acute Ischemic Stroke. Aging Dis 2023; 15:2507-2525. [PMID: 37962453 PMCID: PMC11567273 DOI: 10.14336/ad.2023.1010] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/10/2023] [Indexed: 11/15/2023] Open
Abstract
Blood-brain barrier (BBB) damage is the main pathological basis for acute ischemic stroke (AIS)-induced cerebral vasogenic edema and hemorrhagic transformation (HT). Glial cells, including microglia, astrocytes, and oligodendrocyte precursor cells (OPCs)/oligodendrocytes (OLs) play critical roles in BBB damage and protection. Recent evidence indicates that immune cells also have an important role in BBB damage, vasogenic edema and HT. Therefore, regulating the crosstalk between glial cells and immune cells would hold the promise to alleviate AIS-induced BBB damage. In this review, we first introduce the roles of glia cells, pericytes, and crosstalk between glial cells in the damage and protection of BBB after AIS, emphasizing the polarization, inflammatory response and crosstalk between microglia, astrocytes, and other glia cells. We then describe the role of glial cell-derived exosomes in the damage and protection of BBB after AIS. Next, we specifically discuss the crosstalk between glial cells and immune cells after AIS. Finally, we propose that glial cells could be a potential target for alleviating BBB damage after AIS and we discuss some molecular targets and potential strategies to alleviate BBB damage by regulating glial cells after AIS.
Collapse
Affiliation(s)
- Yihui Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Wencao Liu
- Shanxi Provincial People's Hospital, Taiyuan 030001, China.
| | - Panpan Geng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK.
| | - Qian Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Guo-qing Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
47
|
Rahman Z, Ghuge S, Dandekar MP. Partial blood replacement ameliorates middle cerebral artery occlusion generated neurological aberrations by intervening TLR4 and NLRP3 cascades in rats. Metab Brain Dis 2023; 38:2339-2354. [PMID: 37402080 DOI: 10.1007/s11011-023-01259-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 06/26/2023] [Indexed: 07/05/2023]
Abstract
Acute ischemic stroke is a catastrophic medical condition that causes severe disability and mortality if the sufferer escapes treatment within a stipulated timeframe. While timely intervention with clot-bursting agents like tissue-plasminogen activators abrogates some post-stroke neurologic deficits, no neuroprotective therapy is yet promisingly addresses the post-recanalization neuroinflammation in post-stroke survivors. Herein, we investigated the effect of partial blood replacement therapy (BRT), obtained from healthy and treadmill-trained donor rats, on neurological deficits, and peripheral and central inflammatory cascades using the ischemia-reperfusion animal paradigm. The cerebral ischemia-reperfusion was induced in rats by occlusion of the middle cerebral artery (MCAO) for 90 min, followed by reperfusion. Rats underwent MCAO surgery displayed remarkable sensorimotor and motor deficits in rotarod, foot fault, adhesive removal, and paw whisker tests till 5 days post-surgery. These behavior abnormalities were ameliorated in the BRT-recipient MCAO rats. BRT also reduced the infarct volume and neuronal death in the ipsilateral hemisphere revealed by TTC and cresyl violet staining compared to the MCAO group. Rats received BRT infusion exhibited the reduced expression of glial fibrillary acidic protein, ionized calcium-binding adaptor molecule-1 (Iba-1), and MyD88 on day 5 post-MCAO in immunohistochemistry and immunofluorescent assays. Moreover, elevated levels of toll-like receptor 4 (TLR4) and mRNA expression of IL-1β, TNF-α, matrix metalloproteinase-9 and NLRP3, and decreased levels of zonula occludens-1 in MCAO rats, were reversed following BRT. These findings suggest that the partial BRT may rescind MCAO-induced neurological dysfunctions and cerebral injury by intervening in the TLR4 and NLRP3 pathways in rats.
Collapse
Affiliation(s)
- Ziaur Rahman
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Shubham Ghuge
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India.
| |
Collapse
|
48
|
Litke R, Vicari J, Huang BT, Shapiro L, Roh KH, Silver A, Talreja P, Palacios N, Yoon Y, Kellner C, Kaniskan H, Vangeti S, Jin J, Ramos-Lopez I, Mobbs C. Novel small molecules inhibit proteotoxicity and inflammation: Mechanistic and therapeutic implications for Alzheimer's Disease, healthspan and lifespan- Aging as a consequence of glycolysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544352. [PMID: 37398396 PMCID: PMC10312632 DOI: 10.1101/2023.06.12.544352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Inflammation drives many age-related, especially neurological, diseases, and likely mediates age-related proteotoxicity. For example, dementia due to Alzheimer's Disease (AD), cerebral vascular disease, many other neurodegenerative conditions is increasingly among the most devastating burdens on the American (and world) health system and threatens to bankrupt the American health system as the population ages unless effective treatments are developed. Dementia due to either AD or cerebral vascular disease, and plausibly many other neurodegenerative and even psychiatric conditions, is driven by increased age-related inflammation, which in turn appears to mediate Abeta and related proteotoxic processes. The functional significance of inflammation during aging is also supported by the fact that Humira, which is simply an antibody to the pro-inflammatory cytokine TNF-a, is the best-selling drug in the world by revenue. These observations led us to develop parallel high-throughput screens to discover small molecules which inhibit age-related Abeta proteotoxicity in a C. elegans model of AD AND LPS-induced microglial TNF-a. In the initial screen of 2560 compounds (Microsource Spectrum library) to delay Abeta proteotoxicity, the most protective compounds were, in order, phenylbutyrate, methicillin, and quetiapine, which belong to drug classes (HDAC inhibitors, beta lactam antibiotics, and tricyclic antipsychotics, respectably) already robustly implicated as promising to protect in neurodegenerative diseases, especially AD. RNAi and chemical screens indicated that the protective effects of HDAC inhibitors to reduce Abeta proteotoxicity are mediated by inhibition of HDAC2, also implicated in human AD, dependent on the HAT Creb binding protein (Cbp), which is also required for the protective effects of both dietary restriction and the daf-2 mutation (inactivation of IGF-1 signaling) during aging. In addition to methicillin, several other beta lactam antibiotics also delayed Abeta proteotoxicity and reduced microglial TNF-a. In addition to quetiapine, several other tricyclic antipsychotic drugs also delayed age-related Abeta proteotoxicity and increased microglial TNF-a, leading to the synthesis of a novel congener, GM310, which delays Abeta as well as Huntingtin proteotoxicity, inhibits LPS-induced mouse and human microglial and monocyte TNF-a, is highly concentrated in brain after oral delivery with no apparent toxicity, increases lifespan, and produces molecular responses highly similar to those produced by dietary restriction, including induction of Cbp inhibition of inhibitors of Cbp, and genes promoting a shift away from glycolysis and toward metabolism of alternate (e.g., lipid) substrates. GM310, as well as FDA-approved tricyclic congeners, prevented functional impairments and associated increase in TNF-a in a mouse model of stroke. Robust reduction of glycolysis by GM310 was functionally corroborated by flux analysis, and the glycolytic inhibitor 2-DG inhibited microglial TNF-a and other markers of inflammation, delayed Abeta proteotoxicity, and increased lifespan. These results support the value of phenotypic screens to discover drugs to treat age-related, especially neurological and even psychiatric diseases, including AD and stroke, and to clarify novel mechanisms driving neurodegeneration (e.g., increased microglial glycolysis drives neuroinflammation and subsequent neurotoxicity) suggesting novel treatments (selective inhibitors of microglial glycolysis).
Collapse
|
49
|
Kong AHY, Wu AJ, Ho OKY, Leung MMK, Huang AS, Yu Y, Zhang G, Lyu A, Li M, Cheung KH. Exploring the Potential of Aptamers in Targeting Neuroinflammation and Neurodegenerative Disorders: Opportunities and Challenges. Int J Mol Sci 2023; 24:11780. [PMID: 37511539 PMCID: PMC10380291 DOI: 10.3390/ijms241411780] [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: 06/30/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Neuroinflammation is the precursor for several neurodegenerative diseases (NDDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Targeting neuroinflammation has emerged as a promising strategy to address a wide range of CNS pathologies. These NDDs still present significant challenges in terms of limited and ineffective diagnosis and treatment options, driving the need to explore innovative and novel therapeutic alternatives. Aptamers are single-stranded nucleic acids that offer the potential for addressing these challenges through diagnostic and therapeutic applications. In this review, we summarize diagnostic and therapeutic aptamers for inflammatory biomolecules, as well as the inflammatory cells in NDDs. We also discussed the potential of short nucleotides for Aptamer-Based Targeted Brain Delivery through their unique features and modifications, as well as their ability to penetrate the blood-brain barrier. Moreover, the unprecedented opportunities and substantial challenges of using aptamers as therapeutic agents, such as drug efficacy, safety considerations, and pharmacokinetics, are also discussed. Taken together, this review assesses the potential of aptamers as a pioneering approach for target delivery to the CNS and the treatment of neuroinflammation and NDDs.
Collapse
Affiliation(s)
- Anna Hau-Yee Kong
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Aston Jiaxi Wu
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Olivia Ka-Yi Ho
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Maggie Ming-Ki Leung
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Alexis Shiying Huang
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong SAR, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong SAR, China
| | - Aiping Lyu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong SAR, China
| | - Min Li
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - King-Ho Cheung
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
50
|
Zhao D, Chen X, Wang R, Pang H, Wang J, Liu L. Determining the chemical profile of Caragana jubata (Pall.) Poir. by UPLC-QTOF-MS analysis and evaluating its anti-ischemic stroke effects. JOURNAL OF ETHNOPHARMACOLOGY 2023; 309:116275. [PMID: 36806344 DOI: 10.1016/j.jep.2023.116275] [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: 12/21/2022] [Revised: 02/04/2023] [Accepted: 02/12/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Caragana jubata, belonging to the Leguminosae family, is a shrubby medicinal plant distributed in high-altitude areas of China. The red heartwood of C. jubata is the original source of 'zuomuxing', a Tibetan medicine that promotes blood circulation and removes blood stasis to treat different diseases associated with the blood. AIM OF THE STUDY To date, research on the chemical constituents of C. jubata remains very limited. The anti-ischemic stroke (anti-IS) effects of this plant have not been studied. The aim of the present study was to analyze the chemical profile of C. jubata, establish various anti-IS models to comprehensively evaluate the anti-IS effects of C. jubata, and explore the mechanism of action. MATERIALS AND METHODS Ultraperformance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS) was chosen to analyze the chemical profile. A middle cerebral artery occlusion reperfusion (MCAO/R) model, zebrafish cerebral thrombosis model, and oxygen-glucose deprivation/reperfusion (OGD/R) model in PC12/BV2 cells were used to thoroughly evaluate the anti-IS effects of C. jubata. Additionally, western blotting and immunofluorescence staining were used to detect the mechanism of action. RESULTS Fifty-three compounds were identified from a 95% ethanol extract of C. jubata (ECJ) by UPLC-QTOF-MS analysis. 17 and 7 compounds were identified from C. jubata and the genus Caragana for the first time. ECJ was found to attenuate infarct size and reduce brain edema and neurological scores in MCAO/R rats. ECJ notably reduced the zebrafish cerebral thrombosis incidence in a dose-dependent manner compared with that in the model group. Surprisingly, compared to the positive control drug aspirin, 50 μg/ml ECJ exhibited a better therapeutic effect than aspirin at 30 μg/ml. Additionally, ECJ significantly increased the viability of PC12/BV2 cells injured by OGD/R. Moreover, ECJ inhibited the protein expression of M1 markers (TNF-α, iNOS, and IL-1β) and increased that of M2 markers (Arg-1 and CD206) in OGD/R-injured BV2 cells. ECJ significantly decreased the immunofluorescence intensity of CD16 and increased that of CD206. CONCLUSIONS The results from UPLC-QTOF-MS analysis showed that ECJ was rich in flavonoids. The results from pharmacological experiments verified the anti-IS effects of C. jubata in vivo and in vitro for the first time. In addition, ECJ could regulate the polarization of microglia. The present study highlights the medicinal value of C. jubata, thus providing a theoretical basis for the further development of new drugs from C. jubata to treat IS.
Collapse
Affiliation(s)
- Dingping Zhao
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China.
| | - Xiqing Chen
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China.
| | - Ruiqi Wang
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China.
| | - Hanqing Pang
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
| | - Jianbin Wang
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
| | - Liang Liu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|