1
|
Shen P, Zhang L, Jiang X, Yu B, Zhang J. Targeting HMGB1 and Its Interaction with Receptors: Challenges and Future Directions. J Med Chem 2024; 67:21671-21694. [PMID: 39648929 DOI: 10.1021/acs.jmedchem.4c01912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
High mobility group box 1 (HMGB1) is a nonhistone chromatin protein predominantly located in the nucleus. However, under pathological conditions, HMGB1 can translocate from the nucleus to the cytoplasm and subsequently be released into the extracellular space through both active secretion and passive release mechanisms. The distinct cellular locations of HMGB1 facilitate its interaction with various endogenous and exogenous factors, allowing it to perform diverse functions across a range of diseases. This Perspective provides a comprehensive overview of the structure, release mechanisms, and multifaceted roles of HMGB1 in disease contexts. Furthermore, it introduces the development of both small molecule and macromolecule inhibitors targeting HMGB1 and its interaction with receptors. A detailed analysis of the predicted pockets is also presented, aiming to establish a foundation for the future design and development of HMGB1 inhibitors.
Collapse
Affiliation(s)
- Pingping Shen
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Libang Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xuewa Jiang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jian Zhang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
2
|
Daneshpour A, Shaka Z, Rezaei N. Interplay of cell death pathways and immune responses in ischemic stroke: insights into novel biomarkers. Rev Neurosci 2024:revneuro-2024-0128. [PMID: 39681004 DOI: 10.1515/revneuro-2024-0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024]
Abstract
Stroke is a severe neurological disease and a major worldwide issue, mostly manifesting as ischemic stroke (IS). In order to create effective treatments for IS, it is imperative to fully understand the underlying pathologies, as the existing therapeutic choices are inadequate. Recent investigations have shown the complex relationships between several programmed cell death (PCD) pathways, including necroptosis, ferroptosis, and pyroptosis, and their correlation with immune responses during IS. However, this relationship is still unclear. To address this gap, this review study explored the cellular interactions in the immune microenvironment of IS. Then, to validate prior findings and uncover biomarkers, the study investigated bioinformatics studies. Several pathways, including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), Toll-like receptor 4 (TLR4), and receptor-interacting protein kinase (RIPK), were involved in PCD-immune interactions. The bioinformatics studies reported key biomarkers such as glutathione peroxidase 4 (GPX4), NOD-like receptor family pyrin domain containing 3 (NLRP3), gasdermin D (GSDMD), and TLR4, which have important implications in ferroptosis, cuproptosis, pyroptosis, and necroptosis respectively. These biomarkers were associated with PCD mechanisms such as oxidative stress and inflammatory reactions. The immune infiltration analysis consistently revealed a significant correlation between PCD pathways and detrimental immune cells, such as neutrophils and γδ T cells. Conversely, M2 macrophages and T helper cells showed protective effects. In conclusion, considering the intricate network of interactions between immune responses and PCD pathways, this study emphasized the necessity of a paradigm shift in therapeutic approaches to address the injuries that are related to this complex network.
Collapse
Affiliation(s)
- Arian Daneshpour
- Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| | - Zoha Shaka
- Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793 Iran
- Department of Immunology, School of Medicine, 48439 Tehran University of Medical Sciences , Tehran, 1416634793 Iran
| |
Collapse
|
3
|
Wang C, Li Y, Zhang Y, Smerin D, Gu L, Jiang S, Xiong X. Triolein alleviates ischemic stroke brain injury by regulating autophagy and inflammation through the AKT/mTOR signaling pathway. Mol Med 2024; 30:242. [PMID: 39639187 PMCID: PMC11622655 DOI: 10.1186/s10020-024-00995-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Triolein, a symmetric triglyceride exhibiting anti-inflammatory and antioxidant properties, has demonstrated potential in mitigating cellular damage. However, its therapeutic efficacy in ischemic stroke (IS) and underlying molecular mechanisms remain elusive. Given the critical roles of inflammation and autophagy in IS pathogenesis, this study aimed to elucidate the effects of triolein in IS and investigate its mechanism of action. METHODS We evaluated the impact of triolein using both in vitro oxygen-glucose deprivation/reoxygenation (OGD/R) and in vivo middle cerebral artery occlusion (MCAO/R) models. Neurological function and cerebral infarct volume were assessed 72 h post-reperfusion. Autophagy was quantified through monodansyl cadaverine (MDC) labeling of autophagic vesicles and Western blot analysis of autophagy-related proteins. Microglial activation was visualized via immunofluorescence, while inflammatory cytokine expression was quantified using RT-qPCR. The cytoprotective effect of triolein on OGD/R-induced HT22 cells was evaluated using Cell Counting Kit-8 and lactate dehydrogenase release assays. The involvement of the Protein kinase B/Mechanistic target of rapamycin kinase (AKT/mTOR) pathway was assessed through Western blot analysis. RESULTS Triolein administration significantly reduced infarct volume, enhanced neurological recovery, and attenuated M1 microglial activation and inflammation in MCAO/R-induced mice. Western blot analysis and MDC labeling revealed that triolein exerted an inhibitory effect on post-IS autophagy. Notably, in the BV2-induced OGD/R model, triolein demonstrated an autophagy-dependent suppression of the inflammatory response. Furthermore, triolein inhibited the activation of the AKT/mTOR signaling pathway, consequently attenuating autophagy and mitigating the post-IS inflammatory response. CONCLUSIONS This study provides novel evidence that triolein exerts neuroprotective effects by inhibiting post-stroke inflammation through an autophagy-dependent mechanism. Moreover, the modulation of the AKT/mTOR signaling pathway appears to be integral to the neuroprotective efficacy of triolein. These findings elucidate potential therapeutic strategies for IS management and warrant further investigation.
Collapse
Affiliation(s)
- Chaoqun Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuntao Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yonggang Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | | | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shuting Jiang
- Department of Breast Surgery, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China.
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
4
|
Hu W, Kuang X, Zhang Y, Luo Y, Zhang L. Neuroprotective effects of phenylacetylglycine via β2AR on cerebral ischemia/reperfusion injury in rats. Saudi Pharm J 2024; 32:102210. [PMID: 39697474 PMCID: PMC11653535 DOI: 10.1016/j.jsps.2024.102210] [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: 07/04/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
Phenylacetylglycine (PAGly) is a small molecule derived from phenylalanine in the gut via glycine degradation and conjugation. It has been associated with both the progression of atherosclerosis and protective effects on the myocardium. This study evaluated the function and the underlying mechanisms of PAGly in a rat cerebral ischemia/reperfusion (I/R) injury model. The results indicated that PAGly markedly alleviated cerebral infarct volume (P = 0.0024) and improved the neurobehavioral outcomes (P = 0.0149) after I/R injury. PAGly is structurally analogous to catecholamines and binds to β2-adrenergic receptors (β2AR) on microglia without altering the expression of these receptors (P = 0.9137), but instead inhibiting their activity. It was also observed that when β2AR was engaged in microglia, PAGly suppressed the release of TNF-α (P = 0.0018), IL-1β (P = 0.0310), and IL-6 (P = 0.0017), thereby reducing neuronal apoptosis (P = 0.000003). Furthermore, the protective effect of PAGly diminished after the administration of β2AR-specific agonist fenoterol (P = 0.0055). These data indicate that PAGly mitigates cerebral I/R injury by inhibiting microglial inflammation via β2AR, highlighting its potential as a therapeutic agent. These findings position PAGly as a promising candidate for therapeutic intervention in cerebrovascular injuries, warranting further exploration in clinical settings.
Collapse
Affiliation(s)
- Wenjie Hu
- School of Biological Science, Jining Medical University, Rizhao 276826, Shandong Province, PR China
| | - Xueyan Kuang
- Qingdao West Coast New Area Center for Disease Control and Prevention, Qingdao 266427, Shandong Province, PR China
| | - Yao Zhang
- School of Biological Science, Jining Medical University, Rizhao 276826, Shandong Province, PR China
| | - Yimin Luo
- School of Biological Science, Jining Medical University, Rizhao 276826, Shandong Province, PR China
| | - Litao Zhang
- School of Biological Science, Jining Medical University, Rizhao 276826, Shandong Province, PR China
| |
Collapse
|
5
|
Xie P, Xia M, Long T, Guo D, Cao W, Sun P, Yu W. GIV/Girdin Modulation of Microglial Activation in Ischemic Stroke: Impact of FTO-Mediated m6A Modification. Mol Neurobiol 2024:10.1007/s12035-024-04604-8. [PMID: 39560901 DOI: 10.1007/s12035-024-04604-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/12/2024] [Accepted: 10/28/2024] [Indexed: 11/20/2024]
Abstract
Ischemic stroke (IS) is one of the most common causes of death in the world. The lack of effective pharmacological treatments for IS was primarily due to a lack of understanding of its pathogenesis. Gα-Interacting vesicle-associated protein (GIV/Girdin) is a multi-modular signal transducer and guanine nucleotide exchange factor that controls important signaling downstream of multiple receptors. The purpose of this study was to investigate the role of GIV in IS. In the present study, we found that GIV is highly expressed in the central nervous system (CNS). GIV protein level was decreased, while GIV transcript level was increased in the middle cerebral artery occlusion reperfusion (MCAO/R) mice model. Additionally, GIV was insensitive lipopolysaccharide (LPS) exposure. Interestingly, we found that GIV overexpression dramatically restrained microglial activation, inflammatory response, and M1 polarization in BV-2 microglia induced by oxygen-glucose deprivation and reoxygenation (OGD/R). On the contrary, GIV knockdown had the opposite impact. Mechanistically, we found that GIV activated the Wnt/β-catenin signaling pathway by interacting with DVL2 (disheveled segment polarity protein 2). Notably, m6A demethylase fat mass and obesity-associated protein (FTO) decreased the N6-methyladenosine (m6A) modification-mediated increase of GIV expression and attenuated the inflammatory response in BV-2 stimulated by OGD/R. Taken together, our results demonstrate that GIV inhibited the inflammatory response via activating the Wnt/β-catenin signaling pathway which expression regulated in an FTO-mediated m6A modification in IS. These results broaden our understanding of the role of the FTO-GIV axis in IS development.
Collapse
Affiliation(s)
- Peng Xie
- Key Laboratory of Molecular Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China.
- Department of Human AnatomySchool of Basic Medical ScienceGuian New District, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Mingyan Xia
- Key Laboratory of Molecular Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Human AnatomySchool of Basic Medical ScienceGuian New District, Guizhou Medical University, Guiyang, Guizhou, China
| | - Tingting Long
- Department of Human AnatomySchool of Basic Medical ScienceGuian New District, Guizhou Medical University, Guiyang, Guizhou, China
| | - Dongfen Guo
- Key Laboratory of Molecular Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Human AnatomySchool of Basic Medical ScienceGuian New District, Guizhou Medical University, Guiyang, Guizhou, China
| | - Wenpeng Cao
- Department of Human AnatomySchool of Basic Medical ScienceGuian New District, Guizhou Medical University, Guiyang, Guizhou, China
| | - Ping Sun
- Department of Neurology, The Second People's Hospital of Guiyang, Guiyang, Guiyang, China
| | - Wenfeng Yu
- Key Laboratory of Molecular Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China.
- Department of Human AnatomySchool of Basic Medical ScienceGuian New District, Guizhou Medical University, Guiyang, Guizhou, China.
- Key Laboratory of Human Brain Bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
6
|
Han D, Wang F, Jiang Q, Qiao Z, Zhuang Y, An Q, Li Y, Tang Y, Li C, Shen D. Enhancing Cardioprotection Through Neutrophil-Mediated Delivery of 18β-Glycyrrhetinic Acid in Myocardial Ischemia/Reperfusion Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406124. [PMID: 39264272 PMCID: PMC11558124 DOI: 10.1002/advs.202406124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/08/2024] [Indexed: 09/13/2024]
Abstract
Myocardial ischemia/reperfusion injury (MI/RI) generates reactive oxygen species (ROS) and initiates inflammatory responses. Traditional therapies targeting specific cytokines or ROS often prove inadequate. An innovative drug delivery system (DDS) is developed using neutrophil decoys (NDs) that encapsulate 18β-glycyrrhetinic acid (GA) within a hydrolyzable oxalate polymer (HOP) and neutrophil membrane vesicles (NMVs). These NDs are responsive to hydrogen peroxide (H2O2), enabling controlled GA release. Additionally, NDs adsorb inflammatory factors, thereby reducing inflammation. They exhibit enhanced adhesion to inflamed endothelial cells (ECs) and improved penetration. Once internalized by cardiomyocytes through clathrin-mediated endocytosis, NDs protect against ROS-induced damage and inhibit HMGB1 translocation. In vivo studies show that NDs preferentially accumulate in injured myocardium, reducing infarct size, mitigating adverse remodeling, and enhancing cardiac function, all while maintaining favorable biosafety profiles. This neutrophil-based system offers a promising targeted therapy for MI/RI by addressing both inflammation and ROS, holding potential for future clinical applications.
Collapse
Affiliation(s)
- Dongjian Han
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Fuhang Wang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Qingjiao Jiang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Zhentao Qiao
- Department of Vascular and Endovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Yuansong Zhuang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Quanxu An
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Yuhang Li
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Yazhe Tang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Chenyao Li
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Deliang Shen
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| |
Collapse
|
7
|
Chen Q, Wang J, Xiong X, Chen J, Wang B, Yang H, Zhou J, Deng H, Gu L, Tian J. Blood-Brain Barrier-Penetrating Metal-Organic Framework Antioxidant Nanozymes for Targeted Ischemic Stroke Therapy. Adv Healthc Mater 2024:e2402376. [PMID: 39373278 DOI: 10.1002/adhm.202402376] [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: 06/28/2024] [Revised: 08/20/2024] [Indexed: 10/08/2024]
Abstract
Overproduction of reactive oxygen species (ROS) during reperfusion in ischemic stroke (IS) severely impedes neuronal survival and results in high rates of morbidity and disability. The effective blood-brain barrier (BBB) penetration and brain delivery of antioxidative agents remain the biggest challenge in treating ischemic reperfusion-induced cerebrovascular and neural injury. In this study, a metal-organic framework (MOF) nanozyme (MIL-101-NH2(Fe/Cu)) with ROS scavenging activities to encapsulate neuroprotective agent rapamycin is fabricated and decorating the exterior with BBB-targeting protein ligands (transferrin), thereby realizing enhanced drug retention and controlled release within ischemic lesions for the synergistic treatment of IS. Through the receptor-mediated transcellular pathway, the transferrin-coated MOF nanoparticles achieved efficient transport across the BBB and targeted accumulation at the cerebral ischemic injury site of mice with middle cerebral artery occlusion/reperfusion (MCAO/R), wherein the nanocarrier exhibited catalytic activities of ROS decomposition into O2 and H2O2-responsive rapamycin release. By its BBB-targeting, antioxidative, anti-inflammatory, and antiapoptotic properties, the MOF nanosystem addressed multiple pathological factors of IS and realized remarkable neuroprotective effects, leading to the substantial reduction of cerebral infarction volume and accelerated recovery of nerve functions in the MCAO/R mouse model. This MOF-based nanomedicine provides valuable design principles for effective IS therapy with multi-mechanism synergies.
Collapse
Affiliation(s)
- Qing Chen
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jin Wang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaoxing Xiong
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Junyang Chen
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Bo Wang
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Haixia Yang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jianliang Zhou
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Hongping Deng
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jian Tian
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
8
|
Shen L, Yang J, Zhu Z, Li W, Cui J, Gu L. Elevated Serum HMGB1 Levels and Their Association with Recurrence of Acute Ischaemic Stroke. J Inflamm Res 2024; 17:6887-6894. [PMID: 39372585 PMCID: PMC11451516 DOI: 10.2147/jir.s477415] [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: 05/08/2024] [Accepted: 09/21/2024] [Indexed: 10/08/2024] Open
Abstract
Purpose The study aimed to investigate the correlation between baseline serum levels of high mobility group box 1 (HMGB1) and the recurrence of acute ischemic stroke (AIS). Patients and Methods A total of 544 AIS patients were enrolled and followed up monthly. Serum HMGB1 levels were measured using enzyme-linked immunosorbent assay (ELISA). The primary endpoint was the first recurrence of AIS. Results During a median follow-up period of 43 months, 62 of the 544 AIS patients experienced a recurrence. Both HMGB1 levels and national institute of health stroke scale (NIHSS) scores were significantly higher in the recurrence group compared to the no-recurrence group (p<0.05). According to the receiver operating characteristic curve analysis, the combination (0.855, 95% CI: 0.800-0.911) of HMGB1 (0.745, 95% CI: 0.663-0.826) and NIHSS (0.822, 95% CI: 0.758-0.886) had a higher value for predicting AIS recurrence than either of them (p<0.05). Kaplan-Meier analyses demonstrated that the cumulative survival without AIS recurrence was significantly lower in patients in the high HMGB1 level group than in the low HMGB1 level group (p<0.05). The multifactorial Cox analyses indicated that elevated baseline serum HMGB1 levels (HR: 7.489, 95% CI:4.383-12.795) were a highly effective predictor of recurrence in AIS. Conclusion Elevated baseline serum HMGB1 levels were found to be a highly effective predictor of recurrence in AIS.
Collapse
Affiliation(s)
- Liping Shen
- Department of Neurology, Jiangyin Hospital Affiliated to Nantong University, Jiangyin, Jiangsu, People’s Republic of China
| | - Jiangsheng Yang
- Department of Neurology, Jiangyin Hospital Affiliated to Nantong University, Jiangyin, Jiangsu, People’s Republic of China
| | - Zufu Zhu
- Department of Neurology, Jiangyin Hospital Affiliated to Nantong University, Jiangyin, Jiangsu, People’s Republic of China
| | - Weizhang Li
- Department of Neurology, Jiangyin Hospital Affiliated to Nantong University, Jiangyin, Jiangsu, People’s Republic of China
| | - Junyou Cui
- Department of Neurology, Jiangyin Hospital Affiliated to Nantong University, Jiangyin, Jiangsu, People’s Republic of China
| | - Lingyun Gu
- Department of Neurology, Jiangyin Hospital Affiliated to Nantong University, Jiangyin, Jiangsu, People’s Republic of China
| |
Collapse
|
9
|
Wu F, Zhang Z, Ma S, He Y, He Y, Ma L, Lei N, Deng W, Wang F. Microenvironment-responsive nanosystems for ischemic stroke therapy. Theranostics 2024; 14:5571-5595. [PMID: 39310102 PMCID: PMC11413776 DOI: 10.7150/thno.99822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Ischemic stroke, a common neurological disorder caused by impaired blood supply to the brain, presents a therapeutic challenge. Conventional treatments like thrombolysis and neuroprotection drugs lack ideal drug delivery systems, limiting their effectiveness. Selectively delivering therapies to the ischemic cerebral tissue holds great potential for preventing and/or treating ischemia-related pathological symptoms. The unique pathological microenvironment of the brain after ischemic stroke, characterized by hypoxia, acidity, and inflammation, offers new possibilities for targeted drug delivery. Pathological microenvironment-responsive nanosystems, extensively investigated in tumors with hypoxia-responsive systems as an example, could also respond to the ischemic cerebral microenvironment and achieve brain-targeted drug delivery and release. These emerging nanosystems are gaining traction for ischemic stroke treatment. In this review, we expound on the cerebral pathological microenvironment and clinical treatment strategies of ischemic stroke, highlight various stimulus-responsive materials employed in constructing ischemic stroke microenvironment-responsive nano delivery systems, and discuss the application of these microenvironment-responsive nanosystems in microenvironment regulation for ischemic stroke treatment.
Collapse
Affiliation(s)
- Fang Wu
- Department of Neuro-Intensive Care Unit, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Medical Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhijian Zhang
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Shengnan Ma
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Zhengzhou, 450052, Henan, China
| | - Yanyan He
- Department of Neuro-Intensive Care Unit, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuxi He
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Lixia Ma
- Department of Neuro-Intensive Care Unit, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ningjing Lei
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenjing Deng
- Department of Neuro-Intensive Care Unit, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Fazhan Wang
- Medical Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| |
Collapse
|
10
|
Liu W, Liu L, Li H, Xie Y, Bai J, Guan J, Qi H, Sun J. Targeted pathophysiological treatment of ischemic stroke using nanoparticle-based drug delivery system. J Nanobiotechnology 2024; 22:499. [PMID: 39164747 PMCID: PMC11337765 DOI: 10.1186/s12951-024-02772-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
Ischemic stroke poses significant challenges in terms of mortality and disability rates globally. A key obstacle to the successful treatment of ischemic stroke lies in the limited efficacy of administering therapeutic agents. Leveraging the unique properties of nanoparticles for brain targeting and crossing the blood-brain barrier, researchers have engineered diverse nanoparticle-based drug delivery systems to improve the therapeutic outcomes of ischemic stroke. This review provides a concise overview of the pathophysiological mechanisms implicated in ischemic stroke, encompassing oxidative stress, glutamate excitotoxicity, neuroinflammation, and cell death, to elucidate potential targets for nanoparticle-based drug delivery systems. Furthermore, the review outlines the classification of nanoparticle-based drug delivery systems according to these distinct physiological processes. This categorization aids in identifying the attributes and commonalities of nanoparticles that target specific pathophysiological pathways in ischemic stroke, thereby facilitating the advancement of nanomedicine development. The review discusses the potential benefits and existing challenges associated with employing nanoparticles in the treatment of ischemic stroke, offering new perspectives on designing efficacious nanoparticles to enhance ischemic stroke treatment outcomes.
Collapse
Affiliation(s)
- Wei Liu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Lubin Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Hong Li
- Clinical Laboratory, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, 266033, China
| | - Yutong Xie
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Ju Bai
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Jialiang Guan
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Hongzhao Qi
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Jinping Sun
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
11
|
Zhu L, Zhong W, Meng X, Yang X, Zhang W, Tian Y, Li Y. Polymeric nanocarriers delivery systems in ischemic stroke for targeted therapeutic strategies. J Nanobiotechnology 2024; 22:424. [PMID: 39026255 PMCID: PMC11256638 DOI: 10.1186/s12951-024-02673-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024] Open
Abstract
Ischemic stroke is a complex, high-mortality disease with multifactorial etiology and pathogenesis. Currently, drug therapy is mainly used treat ischemic stroke in clinic, but there are still some limitations, such as limited blood-brain barrier (BBB) penetration efficiency, a narrow treatment time window and drug side effects. Recent studies have pointed out that drug delivery systems based on polymeric nanocarriers can effectively improve the insufficient treatment for ischemic stroke. They can provide neuronal protection by extending the plasma half-life of drugs, enhancing the drug's permeability to penetrate the BBB, and targeting specific structures and cells. In this review, we classified polymeric nanocarriers used for delivering ischemic stroke drugs and introduced their preparation methods. We also evaluated the feasibility and effectiveness and discussed the existing limitations and prospects of polymeric nanocarriers for ischemic stroke treatment. We hoped that this review could provide a theoretical basis for the future development of nanomedicine delivery systems for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Weijie Zhong
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Xuchen Meng
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Xiaosheng Yang
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Wenchuan Zhang
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China
| | - Yayuan Tian
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China.
| | - Yi Li
- Department of Neurosurgery, Ninth People Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
12
|
Zhang K, Du L, Li Z, Huo Z, Shen L, Gao S, Jia Y, Zhu M, Xu B. M2 Macrophage-Derived Small Extracellular Vesicles Ameliorate Pyroptosis and Intervertebral Disc Degeneration. Biomater Res 2024; 28:0047. [PMID: 38952714 PMCID: PMC11214826 DOI: 10.34133/bmr.0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/13/2024] [Indexed: 07/03/2024] Open
Abstract
Intervertebral discs (IVDs) have a limited self-regenerative capacity and current strategies for IVD regeneration are unsatisfactory. Recent studies showed that small extracellular vesicles derived from M2 macrophage cells (M2-sEVs) inhibited inflammation by delivery of various bioactive molecules to recipient cells, which indicated that M2-sEVs may offer a therapeutic strategy for the repair of IVDs. Herein, we investigated the roles and mechanisms of M2-sEVs on IVD regeneration. The in vitro results demonstrated that M2-sEVs inhibited pyroptosis, preserved cellular viability, and promoted migration of nucleus pulposus cells (NPCs). Bioinformatics analysis and verification experiments of microRNA (miR) expression showed that miR-221-3p was highly expressed in M2-sEVs. The mechanism of action was explored and indicated that M2-sEVs inhibited pyroptosis of NPCs through transfer of miR-221-3p, which suppressed the expression levels of phosphatase and tensin homolog and NOD-, LRR-, and pyrin domain-containing protein 3. Moreover, we fabricated decellularized ECM-hydrogel (dECM) for sustained release of M2-sEVs, which exhibited biocompatibility and controlled release properties. The in vivo results revealed that dECM-hydrogel containing M2-sEVs (dECM/M2-sEVs) delayed the degeneration of intervertebral disc degeneration (IDD) models. In addition to demonstrating a promising therapeutic for IDD, this study provided valuable data for furthering the understanding of the roles and mechanisms of M2-sEVs in IVD regeneration.
Collapse
Affiliation(s)
- Kaihui Zhang
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital,
Tianjin University, Tianjin 300211, China
| | - Lilong Du
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital,
Tianjin University, Tianjin 300211, China
| | - Zhenhua Li
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital,
Tianjin University, Tianjin 300211, China
| | - Zhenxin Huo
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital,
Tianjin University, Tianjin 300211, China
| | - Li Shen
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital,
Tianjin University, Tianjin 300211, China
| | - Shan Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yiming Jia
- Department of Stomatology, Chifeng Municipal Hospital, Chifeng, Inner Mongolia 024000, China
| | - Meifeng Zhu
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education),
Nankai University, Tianjin 300071, China
| | - Baoshan Xu
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital,
Tianjin University, Tianjin 300211, China
| |
Collapse
|
13
|
Li Y, Cao Y, Ma K, Ma R, Zhang M, Guo Y, Song H, Sun N, Zhang Z, Yang W. A Triple-Responsive Polymeric Prodrug Nanoplatform with Extracellular ROS Consumption and Intracellular H 2O 2 Self-Generation for Imaging-Guided Tumor Chemo-Ferroptosis-Immunotherapy. Adv Healthc Mater 2024; 13:e2303568. [PMID: 38319010 DOI: 10.1002/adhm.202303568] [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: 10/17/2023] [Revised: 01/10/2024] [Indexed: 02/07/2024]
Abstract
High reactive oxygen species (ROS) levels in tumor microenvironment (TME) impair both immunogenic cell death (ICD) efficacy and T cell activity. Furthermore, tumor escapes immunosurveillance via programmed death-1/programmed death ligand-1 (PD-L1) signal, and the insufficient intracellular hydrogen peroxide weakens ferroptosis efficacy. To tackle the above issues, a glutathione (GSH)/ROS/pH triple-responsive prodrug nanomedicine that encapsulates Fe2O3 nanoparticle via electrostatic interaction is constructed for magnetic resonance imaging (MRI)-guided multi-mode theranostics with chemotherapy/ferroptosis/immunotherapy. The diselenide bond consumes ROS in TME to increase T cells and ICD efficacy, the cleavage of which facilitates PD-L1 antagonist D peptide release to block immune checkpoint. After intracellular internalization, Fe2O3 nanoparticle is released in the acidic endosome for MRI simultaneously with lipid peroxides generation for tumor ferroptosis. Doxorubicin is cleaved from polymers in the condition of high intracellular GSH level accompanied by tumor ICD, which simultaneously potentiates ferroptosis by NADPH oxidase mediated H2O2 self-generation. In vivo results indicate that the nanoplatform strengthens tumor ICD, induces cytotoxic T lymphocytes proliferation, inhibits 4T1 tumor regression and metastasis, and prolongs survival median. In all, a new strategy is proposed in strengthening ICD and T cells activity cascade with ferroptosis as well as immune checkpoint blockade for effective tumor immunotherapy.
Collapse
Affiliation(s)
- Yongjuan Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yongjian Cao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Kunru Ma
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Rong Ma
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Mengzhe Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yichen Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Haiwei Song
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, 138673
| | - Nannan Sun
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Zhengzhou University, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, Henan, 450001, China
| | - Weijing Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Zhengzhou University, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, Henan, 450001, China
| |
Collapse
|
14
|
Huang H, Zheng S, Wu J, Liang X, Li S, Mao P, He Z, Chen Y, Sun L, Zhao X, Cai A, Wang L, Sheng H, Yao Q, Chen R, Zhao Y, Kou L. Opsonization Inveigles Macrophages Engulfing Carrier-Free Bilirubin/JPH203 Nanoparticles to Suppress Inflammation for Osteoarthritis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400713. [PMID: 38593402 PMCID: PMC11165524 DOI: 10.1002/advs.202400713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/21/2024] [Indexed: 04/11/2024]
Abstract
Osteoarthritis (OA) is a chronic inflammatory disease characterized by cartilage destruction, synovitis, and osteophyte formation. Disease-modifying treatments for OA are currently lacking. Because inflammation mediated by an imbalance of M1/M2 macrophages in the synovial cavities contributes to OA progression, regulating the M1 to M2 polarization of macrophages can be a potential therapeutic strategy. Basing on the inherent immune mechanism and pathological environment of OA, an immunoglobulin G-conjugated bilirubin/JPH203 self-assembled nanoparticle (IgG/BRJ) is developed, and its therapeutic potential for OA is evaluated. After intra-articular administration, IgG conjugation facilitates the recognition and engulfment of nanoparticles by the M1 macrophages. The internalized nanoparticles disassemble in response to the increased oxidative stress, and the released bilirubin (BR) and JPH203 scavenge reactive oxygen species (ROS), inhibit the nuclear factor kappa-B pathway, and suppress the activated mammalian target of rapamycin pathway, result in the repolarization of macrophages and enhance M2/M1 ratios. Suppression of the inflammatory environment by IgG/BRJ promotes cartilage protection and repair in an OA rat model, thereby improving therapeutic outcomes. This strategy of opsonization involving M1 macrophages to engulf carrier-free BR/JPH203 nanoparticles to suppress inflammation for OA therapy holds great potential for OA intervention and treatment.
Collapse
Affiliation(s)
- Huirong Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Shimin Zheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| | - Jianing Wu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| | - Xindan Liang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Shengjie Li
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Pengfei Mao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| | - Zhinan He
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Yahui Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Lining Sun
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Xinyu Zhao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| | - Aimin Cai
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| | - Luhui Wang
- Department of UltrasonographyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325015China
| | - Huixiang Sheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| | - Ying‐Zheng Zhao
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhou325035China
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of PharmacyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhou325027China
| |
Collapse
|
15
|
Li J, Wang Z, Li J, Zhao H, Ma Q. HMGB1: A New Target for Ischemic Stroke and Hemorrhagic Transformation. Transl Stroke Res 2024:10.1007/s12975-024-01258-5. [PMID: 38740617 DOI: 10.1007/s12975-024-01258-5] [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: 02/29/2024] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 05/16/2024]
Abstract
Stroke in China is distinguished by its high rates of morbidity, recurrence, disability, and mortality. The ultra-early administration of rtPA is essential for restoring perfusion in acute ischemic stroke, though it concurrently elevates the risk of hemorrhagic transformation. High-mobility group box 1 (HMGB1) emerges as a pivotal player in neuroinflammation after brain ischemia and ischemia-reperfusion. Released passively by necrotic cells and actively secreted, including direct secretion of HMGB1 into the extracellular space and packaging of HMGB1 into intracellular vesicles by immune cells, glial cells, platelets, and endothelial cells, HMGB1 represents a prototypical damage-associated molecular pattern (DAMP). It is intricately involved in the pathogenesis of atherosclerosis, thromboembolism, and detrimental inflammation during the early phases of ischemic stroke. Moreover, HMGB1 significantly contributes to neurovascular remodeling and functional recovery in later stages. Significantly, HMGB1 mediates hemorrhagic transformation by facilitating neuroinflammation, directly compromising the integrity of the blood-brain barrier, and enhancing MMP9 secretion through its interaction with rtPA. As a systemic inflammatory factor, HMGB1 is also implicated in post-stroke depression and an elevated risk of stroke-associated pneumonia. The role of HMGB1 extends to influencing the pathogenesis of ischemia by polarizing various subtypes of immune and glial cells. This includes mediating excitotoxicity due to excitatory amino acids, autophagy, MMP9 release, NET formation, and autocrine trophic pathways. Given its multifaceted role, HMGB1 is recognized as a crucial therapeutic target and prognostic marker for ischemic stroke and hemorrhagic transformation. In this review, we summarize the structure and redox properties, secretion and pathways, regulation of immune cell activity, the role of pathophysiological mechanisms in stroke, and hemorrhage transformation for HMGB1, which will pave the way for developing new neuroprotective drugs, reduction of post-stroke neuroinflammation, and expansion of thrombolysis time window.
Collapse
Affiliation(s)
- Jiamin Li
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Zixin Wang
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Jiameng Li
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Haiping Zhao
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China.
| | - Qingfeng Ma
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China.
| |
Collapse
|
16
|
Yin N, Wang W, Pei F, Zhao Y, Liu C, Guo M, Zhang K, Zhang Z, Shi J, Zhang Y, Wang Z, Liu J. A Neutrophil Hijacking Nanoplatform Reprograming NETosis for Targeted Microglia Polarizing Mediated Ischemic Stroke Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305877. [PMID: 38444306 PMCID: PMC11077645 DOI: 10.1002/advs.202305877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 01/02/2024] [Indexed: 03/07/2024]
Abstract
Precise and efficient regulation of microglia is vital for ischemic stroke therapy and prognosis. The infiltration of neutrophils into the brain provides opportunities for regulatory drugs across the blood-brain barrier, while hindered by neutrophil extracellular traps (NETs) and targeted delivery of intracerebral drugs to microglia. This study reports an efficient neutrophil hijacking nanoplatform (referred to as APTS) for targeted A151 (a telomerase repeat sequence) delivery to microglia without the generation of NETs. In the middle cerebral artery occlusion (MCAO) mouse model, the delivery efficiency to ischemic stroke tissues increases by fourfold. APTS dramatically reduces the formation of NETs by 2.2-fold via reprogramming NETosis to apoptosis in neutrophils via a reactive oxygen species scavenging-mediated citrullinated histone 3 inhibition pathway. Noteworthy, A151 within neutrophils is repackaged into apoptotic bodies following the death pattern reprogramming, which, when engulfed by microglia, polarizes microglia to an anti-inflammatory M2 phenotype. After four times treatment, the cerebral infarction area in the APTS group decreases by 5.1-fold. Thus, APTS provides a feasible, efficient, and practical drug delivery approach for reshaping the immune microenvironment and treating brain disorders in the central nervous system.
Collapse
Affiliation(s)
- Na Yin
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
- Collaborative Innovation Center of New Drug Research and Safety EvaluationZhengzhou UniversityZhengzhou450001China
| | - Wenya Wang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
- Collaborative Innovation Center of New Drug Research and Safety EvaluationZhengzhou UniversityZhengzhou450001China
| | - Fei Pei
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
- Collaborative Innovation Center of New Drug Research and Safety EvaluationZhengzhou UniversityZhengzhou450001China
| | - Yuzhen Zhao
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
- Collaborative Innovation Center of New Drug Research and Safety EvaluationZhengzhou UniversityZhengzhou450001China
| | - Changhua Liu
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
- Collaborative Innovation Center of New Drug Research and Safety EvaluationZhengzhou UniversityZhengzhou450001China
| | - Mingming Guo
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
- Collaborative Innovation Center of New Drug Research and Safety EvaluationZhengzhou UniversityZhengzhou450001China
| | - Kaixiang Zhang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
- Collaborative Innovation Center of New Drug Research and Safety EvaluationZhengzhou UniversityZhengzhou450001China
| | - Zhenzhong Zhang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
- Collaborative Innovation Center of New Drug Research and Safety EvaluationZhengzhou UniversityZhengzhou450001China
| | - Jinjin Shi
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
- Collaborative Innovation Center of New Drug Research and Safety EvaluationZhengzhou UniversityZhengzhou450001China
| | - Yun Zhang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
- Collaborative Innovation Center of New Drug Research and Safety EvaluationZhengzhou UniversityZhengzhou450001China
| | - Zhi‐Hao Wang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
- Collaborative Innovation Center of New Drug Research and Safety EvaluationZhengzhou UniversityZhengzhou450001China
| | - Junjie Liu
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou UniversityZhengzhou450001China
- Collaborative Innovation Center of New Drug Research and Safety EvaluationZhengzhou UniversityZhengzhou450001China
| |
Collapse
|
17
|
Battaglini M, Marino A, Montorsi M, Carmignani A, Ceccarelli MC, Ciofani G. Nanomaterials as Microglia Modulators in the Treatment of Central Nervous System Disorders. Adv Healthc Mater 2024; 13:e2304180. [PMID: 38112345 DOI: 10.1002/adhm.202304180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Indexed: 12/21/2023]
Abstract
Microglia play a pivotal role in the central nervous system (CNS) homeostasis, acting as housekeepers and defenders of the surrounding environment. These cells can elicit their functions by shifting into two main phenotypes: pro-inflammatory classical phenotype, M1, and anti-inflammatory alternative phenotype, M2. Despite their pivotal role in CNS homeostasis, microglia phenotypes can influence the development and progression of several CNS disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, ischemic stroke, traumatic brain injuries, and even brain cancer. It is thus clear that the possibility of modulating microglia activation has gained attention as a therapeutic tool against many CNS pathologies. Nanomaterials are an unprecedented tool for manipulating microglia responses, in particular, to specifically target microglia and elicit an in situ immunomodulation activity. This review focuses the discussion on two main aspects: analyzing the possibility of using nanomaterials to stimulate a pro-inflammatory response of microglia against brain cancer and introducing nanostructures able to foster an anti-inflammatory response for treating neurodegenerative disorders. The final aim is to stimulate the analysis of the development of new microglia nano-immunomodulators, paving the way for innovative and effective therapeutic approaches for the treatment of CNS disorders.
Collapse
Affiliation(s)
- Matteo Battaglini
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Attilio Marino
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Margherita Montorsi
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Alessio Carmignani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Maria Cristina Ceccarelli
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| |
Collapse
|
18
|
Cao Y, Yu Y, Pan L, Han W, Zeng F, Wang J, Mei Q, Liu C. Sulfated Polysaccharide-Based Nanocarrier Drives Microenvironment-Mediated Cerebral Neurovascular Remodeling for Ischemic Stroke Treatment. NANO LETTERS 2024; 24:5214-5223. [PMID: 38649327 DOI: 10.1021/acs.nanolett.4c00650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Stroke is a leading cause of global mortality and severe disability. However, current strategies used for treating ischemic stroke lack specific targeting capabilities, exhibit poor immune escape ability, and have limited drug release control. Herein, we developed an ROS-responsive nanocarrier for targeted delivery of the neuroprotective agent rapamycin (RAPA) to mitigate ischemic brain damage. The nanocarrier consisted of a sulfated chitosan (SCS) polymer core modified with a ROS-responsive boronic ester enveloped by a red blood cell membrane shell incorporating a stroke homing peptide. When encountering high levels of intracellular ROS in ischemic brain tissues, the release of SCS combined with RAPA from nanoparticle disintegration facilitates effective microglia polarization and, in turn, maintains blood-brain barrier integrity, reduces cerebral infarction, and promotes cerebral neurovascular remodeling in a mouse stroke model involving transient middle cerebral artery occlusion (tMCAO). This work offers a promising strategy to treat ischemic stroke therapy.
Collapse
Affiliation(s)
- Yinli Cao
- School of Medicine, Shanghai University, Shanghai 200444, People's Republic of China
| | - Yuanman Yu
- The State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Lina Pan
- The State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Weili Han
- School of Medicine, Shanghai University, Shanghai 200444, People's Republic of China
| | - Feng Zeng
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, People's Republic of China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Qiyong Mei
- School of Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, People's Republic of China
| | - Changsheng Liu
- The State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| |
Collapse
|
19
|
Wu Y, Sun J, Lin Q, Wang D, Hai J. Sustained release of vascular endothelial growth factor A and basic fibroblast growth factor from nanofiber membranes reduces oxygen/glucose deprivation-induced injury to neurovascular units. Neural Regen Res 2024; 19:887-894. [PMID: 37843225 PMCID: PMC10664103 DOI: 10.4103/1673-5374.382252] [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: 11/16/2022] [Revised: 04/12/2023] [Accepted: 06/16/2023] [Indexed: 10/17/2023] Open
Abstract
Upregulation of vascular endothelial growth factor A/basic fibroblast growth factor (VEGFA/bFGF) expression in the penumbra of cerebral ischemia can increase vascular volume, reduce lesion volume, and enhance neural cell proliferation and differentiation, thereby exerting neuroprotective effects. However, the beneficial effects of endogenous VEGFA/bFGF are limited as their expression is only transiently increased. In this study, we generated multilayered nanofiber membranes loaded with VEGFA/bFGF using layer-by-layer self-assembly and electrospinning techniques. We found that a membrane containing 10 layers had an ideal ultrastructure and could efficiently and stably release growth factors for more than 1 month. This 10-layered nanofiber membrane promoted brain microvascular endothelial cell tube formation and proliferation, inhibited neuronal apoptosis, upregulated the expression of tight junction proteins, and improved the viability of various cellular components of neurovascular units under conditions of oxygen/glucose deprivation. Furthermore, this nanofiber membrane decreased the expression of Janus kinase-2/signal transducer and activator of transcription-3 (JAK2/STAT3), Bax/Bcl-2, and cleaved caspase-3. Therefore, this nanofiber membrane exhibits a neuroprotective effect on oxygen/glucose-deprived neurovascular units by inhibiting the JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Yifang Wu
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jun Sun
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qi Lin
- Department of Pharmacy, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dapeng Wang
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian Hai
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
20
|
Lan Z, Qu L, Liang Y, Chen L, Xu S, Ge J, Xue Z, Bao X, Xia S, Yang H, Huang J, Xu Y, Zhu X. AZD1390, an ataxia telangiectasia mutated inhibitor, attenuates microglia-mediated neuroinflammation and ischemic brain injury. CNS Neurosci Ther 2024; 30:e14696. [PMID: 38668740 PMCID: PMC11048048 DOI: 10.1111/cns.14696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 02/09/2024] [Accepted: 03/06/2024] [Indexed: 04/28/2024] Open
Abstract
AIMS Excessive neuroinflammation mediated mainly by microglia plays a crucial role in ischemic stroke. AZD1390, an ataxia telangiectasia mutated (ATM) specific inhibitor, has been shown to promote radio-sensitization and survival in central nervous system malignancies, while the role of AZD1390 in ischemic stroke remains unknown. METHODS Real-time PCR, western blot, immunofluorescence staining, flow cytometry and enzyme-linked immunosorbent assays were used to assess the activation of microglia and the release of inflammatory cytokines. Behavioral tests were performed to measure neurological deficits. 2,3,5-Triphenyltetrazolium chloride staining was conducted to assess the infarct volume. The activation of NF-κB signaling pathway was explored through immunofluorescence staining, western blot, co-immunoprecipitation and proximity ligation assay. RESULTS The level of pro-inflammation cytokines and activation of NF-κB signaling pathway was suppressed by AZD1390 in vitro and in vivo. The behavior deficits and infarct size were partially restored with AZD1390 treatment in experimental stroke. AZD1390 restrict ubiquitylation and sumoylation of the essential regulatory subunit of NF-κB (NEMO) in an ATM-dependent and ATM-independent way respectively, which reduced the activation of the NF-κB pathway. CONCLUSION AZD1390 suppressed NF-κB signaling pathway to alleviate ischemic brain injury in experimental stroke, and attenuated microglia activation and neuroinflammation, which indicated that AZD1390 might be an attractive agent for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Zhen Lan
- Department of NeurologyNanjing Drum Tower Hospital, Clinical College of Nanjing Medical UniversityNanjingJiangsuChina
| | - Long‐jie Qu
- Department of NeurologyNanjing Drum Tower Hospital, Clinical College of Nanjing Medical UniversityNanjingJiangsuChina
| | - Ying Liang
- Department of NeurologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Li‐qiu Chen
- Department of NeurologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Shuai Xu
- Department of NeurologyNanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Jian‐wei Ge
- Department of NeurologyNanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Zhi‐wei Xue
- Department of NeurologyNanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Xin‐yu Bao
- Department of NeurologyNanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingJiangsuChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingJiangsuChina
- Nanjing Neuropsychiatry Clinic Medical CenterNanjingJiangsuChina
| | - Sheng‐nan Xia
- Department of NeurologyNanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingJiangsuChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingJiangsuChina
- Nanjing Neuropsychiatry Clinic Medical CenterNanjingJiangsuChina
| | - Hai‐yan Yang
- Department of NeurologyNanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingJiangsuChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingJiangsuChina
- Nanjing Neuropsychiatry Clinic Medical CenterNanjingJiangsuChina
| | - Jing Huang
- Department of NeurologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Yun Xu
- Department of NeurologyNanjing Drum Tower Hospital, Clinical College of Nanjing Medical UniversityNanjingJiangsuChina
- Department of NeurologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjingJiangsuChina
- Department of NeurologyNanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingJiangsuChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingJiangsuChina
- Nanjing Neuropsychiatry Clinic Medical CenterNanjingJiangsuChina
| | - Xiao‐lei Zhu
- Department of NeurologyNanjing Drum Tower Hospital, Clinical College of Nanjing Medical UniversityNanjingJiangsuChina
- Department of NeurologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjingJiangsuChina
- Department of NeurologyNanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingJiangsuChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingJiangsuChina
- Nanjing Neuropsychiatry Clinic Medical CenterNanjingJiangsuChina
| |
Collapse
|
21
|
Zhan Y, Dai Y, Ding Z, Lu M, He Z, Chen Z, Liu Y, Li Z, Cheng G, Peng S, Liu Y. Application of stimuli-responsive nanomedicines for the treatment of ischemic stroke. Front Bioeng Biotechnol 2024; 11:1329959. [PMID: 38370870 PMCID: PMC10869484 DOI: 10.3389/fbioe.2023.1329959] [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: 11/27/2023] [Indexed: 02/20/2024] Open
Abstract
Ischemic stroke (IS) refers to local brain tissue necrosis which is caused by impaired blood supply to the carotid artery or vertebrobasilar artery system. As the second leading cause of death in the world, IS has a high incidence and brings a heavy economic burden to all countries and regions because of its high disability rate. In order to effectively treat IS, a large number of drugs have been designed and developed. However, most drugs with good therapeutic effects confirmed in preclinical experiments have not been successfully applied to clinical treatment due to the low accumulation efficiency of drugs in IS areas after systematic administration. As an emerging strategy for the treatment of IS, stimuli-responsive nanomedicines have made great progress by precisely delivering drugs to the local site of IS. By response to the specific signals, stimuli-responsive nanomedicines change their particle size, shape, surface charge or structural integrity, which enables the enhanced drug delivery and controlled drug release within the IS tissue. This breakthrough approach not only enhances therapeutic efficiency but also mitigates the side effects commonly associated with thrombolytic and neuroprotective drugs. This review aims to comprehensively summarize the recent progress of stimuli-responsive nanomedicines for the treatment of IS. Furthermore, prospect is provided to look forward for the better development of this field.
Collapse
Affiliation(s)
- Yongyi Zhan
- Zhuhai Interventional Medical Center, Cerebrovascular Diseases Department, Zhuhai Clinical Medical College of Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Yue Dai
- Zhuhai Interventional Medical Center, Cerebrovascular Diseases Department, Zhuhai Clinical Medical College of Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Zhejing Ding
- Zhuhai Interventional Medical Center, Cerebrovascular Diseases Department, Zhuhai Clinical Medical College of Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Mingtian Lu
- Zhuhai Interventional Medical Center, Cerebrovascular Diseases Department, Zhuhai Clinical Medical College of Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Zehua He
- Zhuhai Interventional Medical Center, Cerebrovascular Diseases Department, Zhuhai Clinical Medical College of Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Zhengwei Chen
- Zhuhai Interventional Medical Center, Cerebrovascular Diseases Department, Zhuhai Clinical Medical College of Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Yongkang Liu
- Zhuhai Interventional Medical Center, Cerebrovascular Diseases Department, Zhuhai Clinical Medical College of Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Zhongliang Li
- Zhuhai Interventional Medical Center, Cerebrovascular Diseases Department, Zhuhai Clinical Medical College of Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Guangsen Cheng
- Zhuhai Interventional Medical Center, Cerebrovascular Diseases Department, Zhuhai Clinical Medical College of Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Shaojun Peng
- Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Yu Liu
- Zhuhai Interventional Medical Center, Cerebrovascular Diseases Department, Zhuhai Clinical Medical College of Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| |
Collapse
|
22
|
Fan L, Jin L, Tang T, Zheng Y, Chen Z, Lin H, Ding C, Wang T, Chen H, Guo Y, Xu C, Zhou H, Wu X, Fu X, Yan F, Mao Z, Chen G. Neutrophil-like pH-responsive pro-efferocytic nanoparticles improve neurological recovery by promoting erythrophagocytosis after intracerebral hemorrhage. Theranostics 2024; 14:283-303. [PMID: 38164152 PMCID: PMC10750197 DOI: 10.7150/thno.90370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/06/2023] [Indexed: 01/03/2024] Open
Abstract
Rationale: Intracerebral hemorrhage (ICH) is a devastating cerebrovascular disease resulting from blood extravasating into the brain parenchyma. Escalation of erythrophagocytosis (a form of efferocytosis), avoiding the consequent release of the detrimental erythrocyte lysates, may be a promising target of ICH management. The ADAM17 inhibitor and liver X receptor (LXR) agonist could promote efficient efferocytosis and injury repair. Nevertheless, the poor bioavailability and restriction of the blood-brain barrier (BBB) hinder their application. Therefore, it is needed that biocompatible and smart nanoplatforms were designed and synthesized to realize effective therapy targeting erythrophagocytosis. Methods: We first assessed the synergistic effect of therapeutic GW280264X (an ADAM17 inhibitor) and desmosterol (an LXR agonist) on erythrophagocytosis in vitro. Then a pH-responsive neutrophil membrane-based nanoplatform (NPEOz) served as a carrier to accurately deliver therapeutic GW280264X and desmosterol to the damaged brain was prepared via co-extrusion. Afterwards, their pH-responsive performance was valued in vitro and targeting ability was assessed through fluorescence image in vivo. Finally, the pro-erythrophagocytic and anti-neuroinflammatory ability of the nanomedicine and related mechanisms were investigated. Results: After the synergistical effect of the above two drugs on erythrophagocytosis was confirmed, we successfully developed neutrophil-disguised pH-responsive nanoparticles to efficiently co-deliver them. The nanoparticles could responsively release therapeutic agents under acidic environments, and elicit favorable biocompatibility and ability of targeting injury sites. D&G@NPEOz nanoparticles enhanced erythrophagocytosis through inhibiting shedding of the efferocytotic receptors MERTK/AXL mediated by ADAM17 and accelerating ABCA-1/ABCG-1-mediated cholesterol efflux regulated by LXR respectively. In addition, the nano-formulation was able to modulate the inflammatory microenvironment by transforming efferocytes towards a therapeutic phenotype with reducing the release of proinflammatory cytokines while increasing the secretion of anti-inflammatory factors, and improve neurological function. Conclusions: This biomimetic nanomedicine is envisaged to offer an encouraging strategy to effectively promote hematoma and inflammation resolution, consequently alleviate ICH progression.
Collapse
Affiliation(s)
- Linfeng Fan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Tianchi Tang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Yonghe Zheng
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Zihang Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Haopu Lin
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Chao Ding
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Tingting Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Huaijun Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Yinghan Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Chaoran Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Hang Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Xinyan Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Xiongjie Fu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Feng Yan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Gao Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| |
Collapse
|
23
|
Deng X, Hu Z, Zhou S, Wu Y, Fu M, Zhou C, Sun J, Gao X, Huang Y. Perspective from single-cell sequencing: Is inflammation in acute ischemic stroke beneficial or detrimental? CNS Neurosci Ther 2024; 30:e14510. [PMID: 37905592 PMCID: PMC10805403 DOI: 10.1111/cns.14510] [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: 07/05/2023] [Revised: 09/24/2023] [Accepted: 10/08/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Acute ischemic stroke (AIS) is a common cerebrovascular event associated with high incidence, disability, and poor prognosis. Studies have shown that various cell types, including microglia, astrocytes, oligodendrocytes, neurons, and neutrophils, play complex roles in the early stages of AIS and significantly affect its prognosis. Thus, a comprehensive understanding of the mechanisms of action of these cells will be beneficial for improving stroke prognosis. With the rapid development of single-cell sequencing technology, researchers have explored the pathophysiological mechanisms underlying AIS at the single-cell level. METHOD We systematically summarize the latest research on single-cell sequencing in AIS. RESULT In this review, we summarize the phenotypes and functions of microglia, astrocytes, oligodendrocytes, neurons, neutrophils, monocytes, and lymphocytes, as well as their respective subtypes, at different time points following AIS. In particular, we focused on the crosstalk between microglia and astrocytes, oligodendrocytes, and neurons. Our findings reveal diverse and sometimes opposing roles within the same cell type, with the possibility of interconversion between different subclusters. CONCLUSION This review offers a pioneering exploration of the functions of various glial cells and cell subclusters after AIS, shedding light on their regulatory mechanisms that facilitate the transformation of detrimental cell subclusters towards those that are beneficial for improving the prognosis of AIS. This approach has the potential to advance the discovery of new specific targets and the development of drugs, thus representing a significant breakthrough in addressing the challenges in AIS treatment.
Collapse
Affiliation(s)
- Xinpeng Deng
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Ziliang Hu
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Shengjun Zhou
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Yiwen Wu
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Menglin Fu
- School of Economics and ManagementChina University of GeosciencesWuhanChina
| | - Chenhui Zhou
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Jie Sun
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Xiang Gao
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Yi Huang
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| |
Collapse
|
24
|
Sri Kanaka Durga Vijayalakshmi G, Puvvada N. Recent Advances in Chemically Engineered Nanostructures Impact on Ischemic Stroke Treatment. ACS OMEGA 2023; 8:45188-45207. [PMID: 38075770 PMCID: PMC10701887 DOI: 10.1021/acsomega.3c06228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 02/12/2024]
Abstract
Stroke is a serious public health problem that raises expenses for society and causes long-term impairment and death. However, due to restricted blood-brain barrier (BBB) penetration, there are few treatment alternatives for treating stroke. Recanalization techniques, neuroprotective medications, and recovery techniques are all forms of treatment. The ischemic stroke treatment window is too narrow for logical and efficient therapy, and detection is possible only in advanced stages. BBB integrity disruption, neurotoxicity, and the brief half-life of therapeutic thrombolytics are the key molecular pathogenic causes of ischemic stroke. Existing neuroprotective drugs' inability to promote the recovery of ischemic brain tissue after a stroke is another factor that contributes to the disease's progression, chronic nature, and severity. A possible approach to getting around these medication restrictions and boosting the effectiveness of therapies is nanotechnology. In order to get around these drug-related restrictions and boost the effectiveness of therapies for neurological conditions such as stroke, nanotechnology has emerged as a viable option. These problems might be avoided by using nanoparticle-based methods to create a thrombolytic medication that is safe to use after the tissue plasminogen activator (tPA) treatment window has passed. The idea of using biomimetic nanoparticles in the future for the treatment of ischemic stroke through immunotherapy and stem cell therapy is highlighted, along with recent advancements in the study of nanomaterials for ischemic stroke diagnostics and treatment.
Collapse
Affiliation(s)
| | - Nagaprasad Puvvada
- Department of Chemistry,
School of Advanced Sciences, VIT-AP University, Amaravathi, Andhra Pradesh 522237, India
| |
Collapse
|
25
|
Kang X, Cao Y, Sun G, Fei D, Kang K, Meng X, Zhao M. CircPTP4A2 Promotes Microglia Polarization in Cerebral Ischemic Stroke via miR-20b-5p/YTHDF1/TIMP2 Axis. Neuromolecular Med 2023; 25:501-515. [PMID: 37704831 DOI: 10.1007/s12017-023-08751-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/12/2023] [Indexed: 09/15/2023]
Abstract
Activated microglia play dual roles in ischemic stroke (IS) according to its polarization states. Herein, we investigated the function of circPTP4A2 in regulating microglia polarization in IS. IS models were established by MACO/R and OGD/R treatment. TTC staining was employed to detect cerebral infarct size. Cell vitality was measured using CCK-8 assay. CD16 and CD206 levels were examined using flow cytometry. The interactions between circPTP4A2, miR-20b-5p, and YTHDF1 were analyzed by dual-luciferase reporter gene, RIP, or RNA pull-down assays. circPTP4A2 was upregulated in IS patients. circPTP4A2 knockdown alleviated MCAO/R-induced cerebral injury in mice. circPTP4A2 knockdown promoted microglia M2 polarization after OGD/R. circPTP4A2 promoted YTHDF1 expression by sponging miR-20b-5p. The promoting effect of circPTP4A2 knockdown on microglia M2 polarization was abrogated by miR-20b-5p inhibition. YTHDF1 activated the NF-κB pathway by increasing TIMP2 mRNA stability and expression. circPTP4A2 downregulation promoted microglia M2 polarization to inhibit IS development by regulating the miR-20b-5p/YTHDF1/TIMP2/NF-κB axis.
Collapse
Affiliation(s)
- Xianxin Kang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Nangang District, Harbin, 150000, Heilongjiang Province, People's Republic of China
| | - Yanhui Cao
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Nangang District, Harbin, 150000, Heilongjiang Province, People's Republic of China
| | - Guodong Sun
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Nangang District, Harbin, 150000, Heilongjiang Province, People's Republic of China
| | - Dongsheng Fei
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Nangang District, Harbin, 150000, Heilongjiang Province, People's Republic of China
| | - Kai Kang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Nangang District, Harbin, 150000, Heilongjiang Province, People's Republic of China
| | - Xianglin Meng
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Nangang District, Harbin, 150000, Heilongjiang Province, People's Republic of China
| | - Mingyan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, Postal Street, Nangang District, Harbin, 150000, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
26
|
Wang X, Liu Y, Li M, Ju Y, Tang J, Chen T, Lin X, Gu N, Yang F. Neuroinflammation catching nanobubbles for microglia-neuron unit modulation against epilepsy. Biomaterials 2023; 302:122302. [PMID: 37666103 DOI: 10.1016/j.biomaterials.2023.122302] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023]
Abstract
Epilepsy is a common neurological disease caused by synchronous firing of hyperexcitable neurons. Currently, patients with epilepsy are typically treated with antiseizure medicines that work by interrupting the hyperexcitability or hypersynchrony of localized neurons or by inhibiting excitatory neurotransmission. However, these drugs do not treat the underlying causes of epilepsy, and nearly one-third of patients have seizures that cannot be controlled by these medications. Animal and clinical evidence suggests that inflammation caused by neuronal and non-neuronal cells within the epilepsy lesion could play a central role in seizure disorders. Here we report a gas-filled nanobubble (NB) conjugated with diammonium glycyrrhizinate (DG) drugs and sphingosine-1-phosphate (S1P) molecules (S1P@DG-NBs) on the lipid shell for targeted therapy and real-time ultrasound visualization applications against neuroinflammatory injury. Affinity of S1P@DG-NBs for the S1P receptor endows these NBs with enhanced targeting capability to the neuroinflammatory microenvironment of epilepsy, where the DG drugs modulate endothelium-microglia-neuron inflammation by inhibiting high-mobility group box 1 molecules and downregulating the Toll-like receptor 4 signaling pathway, resulting in anti-inflammatory M2 microglia that exert anti-epilepsy effects. Our results show that this technology can enhance visualization of epileptic brain and deliver drugs with anti-inflammatory and immunomodulatory properties to ameliorate seizures symptoms.
Collapse
Affiliation(s)
- Xiao Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Yang Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Mingxi Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Yongxu Ju
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Jian Tang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Tiandong Chen
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Xubo Lin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, 100191, PR China
| | - Ning Gu
- Medical School, Nanjing University, Nanjing, 210093, PR China
| | - Fang Yang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, PR China.
| |
Collapse
|
27
|
Zhu Z, Lu H, Jin L, Gao Y, Qian Z, Lu P, Tong W, Lo PK, Mao Z, Shi H. C-176 loaded Ce DNase nanoparticles synergistically inhibit the cGAS-STING pathway for ischemic stroke treatment. Bioact Mater 2023; 29:230-240. [PMID: 37502677 PMCID: PMC10371767 DOI: 10.1016/j.bioactmat.2023.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/15/2023] [Accepted: 07/04/2023] [Indexed: 07/29/2023] Open
Abstract
The neuroinflammatory responses following ischemic stroke cause irreversible nerve cell death. Cell free-double strand DNA (dsDNA) segments from ischemic tissue debris are engulfed by microglia and sensed by their cyclic GMP-AMP synthase (cGAS), which triggers robust activation of the innate immune stimulator of interferon genes (STING) pathway and initiate the chronic inflammatory cascade. The decomposition of immunogenic dsDNA and inhibition of the innate immune STING are synergistic immunologic targets for ameliorating neuroinflammation. To combine the anti-inflammatory strategies of STING inhibition and dsDNA elimination, we constructed a DNase-mimetic artificial enzyme loaded with C-176. Nanoparticles are self-assembled by amphiphilic copolymers (P[CL35-b-(OEGMA20.7-co-NTAMA14.3)]), C-176, and Ce4+ which is coordinated with nitrilotriacetic acid (NTA) group to form corresponding catalytic structures. Our work developed a new nano-drug that balances the cGAS-STING axis to enhance the therapeutic impact of stroke by combining the DNase-memetic Ce4+ enzyme and STING inhibitor synergistically. In conclusion, it is a novel approach to modulating central nervus system (CNS) inflammatory signaling pathways and improving stroke prognosis.
Collapse
Affiliation(s)
- Zhixin Zhu
- Department of Orthopedics, 1st Affiliated Hospital of Zhejiang University School of Medicine, Qingchun Road 79, Hangzhou, 31000, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haipeng Lu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yong Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhefeng Qian
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Pan Lu
- Department of Orthopedics, 1st Affiliated Hospital of Zhejiang University School of Medicine, Qingchun Road 79, Hangzhou, 31000, China
| | - Weijun Tong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Pik Kwan Lo
- Department of Chemistry and State Key Laboratory of Marine Pollution, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haifei Shi
- Department of Orthopedics, 1st Affiliated Hospital of Zhejiang University School of Medicine, Qingchun Road 79, Hangzhou, 31000, China
| |
Collapse
|
28
|
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: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [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
|
29
|
Nyandoro VO, Omolo CA, Ismail EA, Yong L, Govender T. Inflammation-responsive drug delivery nanosystems for treatment of bacterial-induced sepsis. Int J Pharm 2023; 644:123346. [PMID: 37633537 DOI: 10.1016/j.ijpharm.2023.123346] [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: 08/15/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Sepsis, a complication of dysregulated host immune systemic response to an infection, is life threatening and causes multiple organ injuries. Sepsis is recognized by WHO as a big contributor to global morbidity and mortality. The heterogeneity in sepsis pathophysiology, antimicrobial resistance threat, the slowdown in the development of antimicrobials, and limitations of conventional dosage forms jeopardize the treatment of sepsis. Drug delivery nanosystems are promising tools to overcome some of these challenges. Among the drug delivery nanosystems, inflammation-responsive nanosystems have attracted considerable interest in sepsis treatment due to their ability to respond to specific stimuli in the sepsis microenvironment to release their payload in a precise, targeted, controlled, and rapid manner compared to non-responsive nanosystems. These nanosystems posit superior therapeutic potential to enhance sepsis treatment. This review critically evaluates the recent advances in the design of drug delivery nanosystems that are inflammation responsive and their potential in enhancing sepsis treatment. The sepsis microenvironment's unique features, such as acidic pH, upregulated receptors, overexpressed enzymes, and enhanced oxidative stress, that form the basis for their design have been adequately discussed. These inflammation-responsive nanosystems have been organized into five classes namely: Receptor-targeted nanosystems, pH-responsive nanosystems, redox-responsive nanosystems, enzyme-responsive nanosystems, and multi-responsive nanosystems. Studies under each class have been thematically grouped and discussed with an emphasis on the polymers used in their design, nanocarriers, key characterization, loaded actives, and key findings on drug release and therapeutic efficacy. Further, this information is concisely summarized into tables and supplemented by inserted figures. Additionally, this review adeptly points out the strengths and limitations of the studies and identifies research avenues that need to be explored. Finally, the challenges and future perspectives on these nanosystems have been thoughtfully highlighted.
Collapse
Affiliation(s)
- Vincent O Nyandoro
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa; Department of Pharmaceutical Chemistry and Pharmaceutics, School of Pharmacy, Kabarak University, Nakuru, Kenya
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa; Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, Nairobi, Kenya.
| | - Eman A Ismail
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Liu Yong
- Wenzhou Institute, University of Chinese Academy of Sciences (WIUCAS), China
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
30
|
Quan W, Kong S, Li S, Ouyang Q, Lu S, Guo J, Wu K, Zhao W, Luo H. Anti-Photoaging Effects of Nanocomposites of Amphiphilic Chitosan/18β-Glycyrrhetinic Acid. Molecules 2023; 28:molecules28114362. [PMID: 37298838 DOI: 10.3390/molecules28114362] [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: 03/28/2023] [Revised: 05/11/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Improving the transdermal absorption of weakly soluble drugs for topical use can help to prevent and treat skin photoaging. Nanocrystals of 18β-glycyrrhetinic acid (i.e., NGAs) prepared by high-pressure homogenization and amphiphilic chitosan (ACS) were used to form ANGA composites by electrostatic adsorption, and the optimal ratio of NGA to ACS was 10:1. Dynamic light scattering analysis and zeta potential analysis were used to evaluate the nanocomposites' suspension, and the results showed that mean particle size was 318.8 ± 5.4 nm and the zeta potential was 30.88 ± 1.4 mV after autoclaving (121 °C, 30 min). The results of CCK-8 showed that the half-maximal inhibitory concentration (IC50) of ANGAs (71.9 μg/mL) was higher than that of NGAs (51.6 μg/mL), indicating that the cytotoxicity of ANGAs was weaker than that of NGAs at 24 h. After the composite had been prepared as a hydrogel, the vertical diffusion (Franz) cells were used to investigate skin permeability in vitro, and it was shown that the cumulative permeability of the ANGA hydrogel increased from 56.5 ± 1.4% to 75.3 ± 1.8%. The efficacy of the ANGA hydrogel against skin photoaging was studied by constructing a photoaging animal model under ultraviolet (UV) irradiation and staining. The ANGA hydrogel improved the photoaging characteristics of UV-induced mouse skin significantly, improved structural changes (e.g., breakage and clumping of collagen and elastic fibers in the dermis) significantly, and improved skin elasticity, while it inhibited the abnormal expression of matrix metalloproteinase (MMP)-1 and MMP-3 significantly, thereby reducing the damage caused by UV irradiation to the collagen-fiber structure. These results indicated that the NGAs could enhance the local penetration of GA into the skin and significantly improve the photoaging of mouse skin. The ANGA hydrogel could be used to counteract skin photoaging.
Collapse
Affiliation(s)
- Weiyan Quan
- Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
| | - Songzhi Kong
- Department of Applied Chemistry, School of Chemistry and Environmental Science, Guangdong Ocean University, Zhanjiang 524088, China
| | - Sidong Li
- Department of Applied Chemistry, School of Chemistry and Environmental Science, Guangdong Ocean University, Zhanjiang 524088, China
| | - Qianqian Ouyang
- Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
| | - Sitong Lu
- Songshan Lake Materials Laboratory, Dongguan 523808, China
| | - Jiaqi Guo
- Department of Applied Chemistry, School of Chemistry and Environmental Science, Guangdong Ocean University, Zhanjiang 524088, China
| | - Kefeng Wu
- Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
| | - Wei Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Hui Luo
- Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
| |
Collapse
|
31
|
Mo J, Hu J, Cheng X. The role of high mobility group box 1 in neuroinflammatory related diseases. Biomed Pharmacother 2023; 161:114541. [PMID: 36963363 DOI: 10.1016/j.biopha.2023.114541] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 03/26/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a ubiquitous and highly conserved non-histone DNA-binding protein with different biological functions according to its subcellular localization. It is widely believed that HMGB1, which is released into the extracellular space, plays a key role in the inflammatory response. In recent years, numerous studies have shown that the development of various neurological diseases such as epilepsy, Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), cerebrovascular disease and traumatic brain injury (TBI) are inextricably linked to inflammation. We will review the mechanisms of HMGB1 and its receptors in nervous system inflammation to provide a basis for further development of new HMGB1-based therapies.
Collapse
Affiliation(s)
- Jialu Mo
- The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei, China
| | - Jiao Hu
- The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei, China
| | - Xianglin Cheng
- The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei, China.
| |
Collapse
|
32
|
Regulated extravascular microenvironment via reversible thermosensitive hydrogel for inhibiting calcium influx and vasospasm. Bioact Mater 2023; 21:422-435. [PMID: 36185746 PMCID: PMC9483581 DOI: 10.1016/j.bioactmat.2022.08.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/18/2022] [Accepted: 08/13/2022] [Indexed: 11/22/2022] Open
Abstract
Arterial vasospasm after microsurgery can cause severe obstruction of blood flow manifested as low tissue temperature, leading to tissue necrosis. The timely discovery and synchronized treatment become pivotal. In this study, a reversible, intelligent, responsive thermosensitive hydrogel system is constructed employing both the gel–sol transition and the sol–gel transition. The “reversible thermosensitive (RTS)” hydrogel loaded with verapamil hydrochloride is designed to dynamically and continuously regulate the extravascular microenvironment by inhibiting extracellular calcium influx. After accurate implantation and following in situ gelation, the RTS hydrogel reverses to the sol state causing massive drug release to inhibit vasospasm when the tissue temperature drops to the predetermined transition temperature. Subsequent restoration of the blood supply alleviates further tissue injury. Before the temperature drops, the RTS hydrogel maintains the gel state as a sustained-release reservoir to prevent vasospasm. The inhibition of calcium influx and vasospasm in vitro and in vivo is demonstrated using vascular smooth muscle cells, mice mesenteric arterial rings, and vascular ultrasonic Doppler detection. Subsequent animal experiments demonstrate that RTS hydrogel can promote tissue survival and alleviate tissue injury responding to temperature change. Therefore, this RTS hydrogel holds therapeutic potential for diseases requiring timely detection of temperature change. Proposing a new strategy for the discovery and treatment for diseases requiring timely detection of temperature change. Applying both the gel–sol transition and the sol–gel transition of PEG/PLGA triblock polymers. Realizing the dynamical and continuous regulation of the extravascular microenvironment.
Collapse
|
33
|
Zhai L, Pei H, Shen H, Guan Q, Sheng J. Mechanism of neocryptotanshinone in protecting against cerebral ischemic injury: By suppressing M1 polarization of microglial cells and promoting cerebral angiogenesis. Int Immunopharmacol 2023; 116:109815. [PMID: 36773571 DOI: 10.1016/j.intimp.2023.109815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/16/2023] [Accepted: 01/28/2023] [Indexed: 02/11/2023]
Abstract
AIM This study explored the protective function and mechanism of neocryptotanshinone (NEO) on cerebral ischemia. METHODS Lipopolysaccharide/γ-interferon(LPS/IFN-γ)was employed to mimic the polarization of mouse microglial cells BV2. After NEO treatment, the M1 polarization level of BV2 cells was identified using flow cytometry (FCM), fluorescent cell staining and enzyme linked immunosorbent assay(ELISA). Moreover, the mouse endothelial cells bEnd.3 were applied to be the study objects, which were intervened with NEO under the hypoxic condition. Thereafter, based on in-vitro tubule formation assay and fluorescence staining, the in-vitro tubule formation ability of bEnd.3 cells was detected. By adopting middle cerebral artery occlusion(MCAO) method, we constructed the mouse model of cerebral ischemia. After NEO intervention, the pathological changes of brain tissues were identified, while CD34 expression was measured by immunohistochemical (IHC) staining, nerve injury was detected by Nissl staining, and the changes in neurological behaviors of mice were also detected. RESULTS Our results showed that NEO suppressed M1 polarization of BV2 cells, which exerted its effect through suppressing NF-κB and STAT3 signals, thereby decreasing the levels of iNOS, CD11b and inflammatory factors. NEO stimulated tubule formation in bEnd.3 cells based on the hypoxic situation, which exerted its effect through activating the Vascularendothelial growth factor-Vascular Endothelial Growth Factor Receptor 2-Notch homolog 1(VFGF-VEGFR2-Notch1) signal. Furthermore, NEO suppressed cerebral ischemia in mice and lowered the ischemic penumbra. NEO also improved the neurological behaviors of mice, increased the CD34 levels and decreased the expression of inflammatory factors. CONCLUSION NEO has well protective effect against cerebral ischemia, and its mechanisms are related to suppressing M1 polarization of microglial cells and promoting cerebral angiogenesis, which are the mechanisms of NEO in treating ischemic encephalopathy.
Collapse
Affiliation(s)
- Liping Zhai
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, China
| | - Hongyan Pei
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China.
| | - Heping Shen
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, China.
| | - Qiaobing Guan
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, China.
| | - Jian Sheng
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, China.
| |
Collapse
|
34
|
18β-Glycyrrhetinic Acid Ameliorates Neuroinflammation Linked Depressive Behavior Instigated by Chronic Unpredictable Mild Stress via Triggering BDNF/TrkB Signaling Pathway in Rats. Neurochem Res 2023; 48:551-569. [PMID: 36307572 PMCID: PMC9616426 DOI: 10.1007/s11064-022-03779-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/06/2022] [Accepted: 09/30/2022] [Indexed: 02/04/2023]
Abstract
Evidence shows that inflammatory responses may encompass the onset of severe depressive illness. Traditionally used licorice contains 18β-glycyrrhetinic acid (18βGA), which has been demonstrated to reduce inflammation and oxidative stress. This study investigates the antidepressant effects of 18βGA and the underlying mechanism in rats exposed to chronic unpredictable mild stress (CUMS). Wistar rats were exposed to CUMS for 36 consecutive days to establish depression. 18βGA (10, 20, and 50 mg/kg) or fluoxetine was given once daily (from day 30 to day 36). Thereafter, behavior parameters (sucrose preference test, forced-swimming test, open-field test, body weight), pro-inflammatory cytokines, neurotransmitters, adrenocorticotropic hormone (ACTH), corticosterone (CORT), and liver biomarkers were studied. Immunohistochemistry and western blot analyses were conducted to investigate the protein's expression. 18βGA (20 and 50 mg/kg) treatment increased sucrose intake, locomotion in the open-field test, decreased immobility time in the forced swim test, and improved body weight in CUMS-exposed rats. The therapy of 18βGA dramatically declined cytokines, ACTH and CORT and improved 5HT and norepinephrine in CUMS rats. Furthermore, BDNF and TrkB proteins were down-regulated in CUMS group, which was increased to varying degrees by 18βGA at doses of 20 and 50 mg/kg. Therefore, 18βGA ameliorates depressive-like behavior persuaded by chronic unpredictable mild stress, decreases neuroinflammation, liver biomarkers, stress hormones, and improves body weight, brain neurotransmitter concentration via activating on BDNF/TrkB signaling pathway in both PFC and hippocampus in rats.
Collapse
|
35
|
Hoseinzadeh A, Ghoddusi Johari H, Anbardar MH, Tayebi L, Vafa E, Abbasi M, Vaez A, Golchin A, Amani AM, Jangjou A. Effective treatment of intractable diseases using nanoparticles to interfere with vascular supply and angiogenic process. Eur J Med Res 2022; 27:232. [PMID: 36333816 PMCID: PMC9636835 DOI: 10.1186/s40001-022-00833-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022] Open
Abstract
Angiogenesis is a vital biological process involving blood vessels forming from pre-existing vascular systems. This process contributes to various physiological activities, including embryonic development, hair growth, ovulation, menstruation, and the repair and regeneration of damaged tissue. On the other hand, it is essential in treating a wide range of pathological diseases, such as cardiovascular and ischemic diseases, rheumatoid arthritis, malignancies, ophthalmic and retinal diseases, and other chronic conditions. These diseases and disorders are frequently treated by regulating angiogenesis by utilizing a variety of pro-angiogenic or anti-angiogenic agents or molecules by stimulating or suppressing this complicated process, respectively. Nevertheless, many traditional angiogenic therapy techniques suffer from a lack of ability to achieve the intended therapeutic impact because of various constraints. These disadvantages include limited bioavailability, drug resistance, fast elimination, increased price, nonspecificity, and adverse effects. As a result, it is an excellent time for developing various pro- and anti-angiogenic substances that might circumvent the abovementioned restrictions, followed by their efficient use in treating disorders associated with angiogenesis. In recent years, significant progress has been made in different fields of medicine and biology, including therapeutic angiogenesis. Around the world, a multitude of research groups investigated several inorganic or organic nanoparticles (NPs) that had the potential to effectively modify the angiogenesis processes by either enhancing or suppressing the process. Many studies into the processes behind NP-mediated angiogenesis are well described. In this article, we also cover the application of NPs to encourage tissue vascularization as well as their angiogenic and anti-angiogenic effects in the treatment of several disorders, including bone regeneration, peripheral vascular disease, diabetic retinopathy, ischemic stroke, rheumatoid arthritis, post-ischemic cardiovascular injury, age-related macular degeneration, diabetic retinopathy, gene delivery-based angiogenic therapy, protein delivery-based angiogenic therapy, stem cell angiogenic therapy, and diabetic retinopathy, cancer that may benefit from the behavior of the nanostructures in the vascular system throughout the body. In addition, the accompanying difficulties and potential future applications of NPs in treating angiogenesis-related diseases and antiangiogenic therapies are discussed.
Collapse
Affiliation(s)
- Ahmad Hoseinzadeh
- Thoracic and Vascular Surgery Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Surgery, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Ghoddusi Johari
- Thoracic and Vascular Surgery Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Surgery, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA
| | - Ehsan Vafa
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Abbasi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Golchin
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Jangjou
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
36
|
Scavenging of reactive oxygen species can adjust the differentiation of tendon stem cells and progenitor cells and prevent ectopic calcification in tendinopathy. Acta Biomater 2022; 152:440-452. [PMID: 36108965 DOI: 10.1016/j.actbio.2022.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022]
Abstract
Tendinopathy is a common disorder that leads to pain and impaired quality of life. Recent studies revealed that osteogenic differentiation of tendon stem/progenitor cells (TSPCs) played an important role in the pathogenesis of tendon calcification and tendinopathy. In this study, we found that the growth hormone-releasing hormone agonist (GA) can prevent matrix degradation and osteogenic differentiation in TSPCs. As oxidative stress is a key factor in the osteogenic differentiation of TSPCs, we used bovine serum albumin/heparin nanoparticles (BHNPs), which have biocompatibility and drug loading capacity, to scavenge reactive oxygen species (ROS) and achieve sustained release of GA at the site of inflammation. The newly developed BHNPs@GA had a synergetic effect on reducing ROS production in TSPCs. In addition, BHNPs@GA effectively inhibited tendon calcification and promoted collagen formation in a rat model of tendinopathy. Focusing on the ROS underlying the differentiation and dedifferentiation of TSPCs, this work demonstrated that sustained release of GA targeting ROS and ectopic ossification is a practical therapeutic strategy for treating tendinopathy. STATEMENT OF SIGNIFICANCE: Osteogenic differentiation of tendon stem/progenitor cells (TSPCs) plays an important role in the pathogenesis of ectopic calcification in tendinopathy. In this study, we found that growth hormone-releasing hormone agonist (GA) can reduce reactive oxygen species (ROS) production and adjust TSPCs differentiation. Bovine serum albumin/heparin nanoparticles (BHNPs) were developed to encapsulate GA and achieve sustained release of GA at the site of inflammation. The developed compound, BHNPs@GA, with a synergistic effect of inhibiting ROS and thus, can effectively adjust TSPCs differentiation, inhibit tendon calcification, and promote collagen formation in tendinopathy. This study highlighted the role of ROS underlying the differentiation and dedifferentiation of TSPCs in tendinopathy, and findings may help to identify new therapeutic targets and develop novel strategy for treating tendinopathy.
Collapse
|
37
|
Li YX, Wang HB, Jin JB, Yang CL, Hu JB, Li J. Advances in the research of nano delivery systems in ischemic stroke. Front Bioeng Biotechnol 2022; 10:984424. [PMID: 36338131 PMCID: PMC9634573 DOI: 10.3389/fbioe.2022.984424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/07/2022] [Indexed: 11/29/2022] Open
Abstract
Ischemic stroke is the most common type of cerebrovascular disease with high disability rate and mortality. The blood-brain barrier (BBB) protects the homeostasis of the brain's microenvironment and impedes the penetration of 98% of drugs. Therefore, effective treatment requires the better drug transport across membranes and increased drug distribution. Nanoparticles are a good choice for drugs to cross BBB. The main pathways of nano delivery systems through BBB include passive diffusion, adsorption-mediated endocytosis, receptor-mediated transport, carrier-mediated transport, etc. At present, the materials used in brain-targeted delivery can be divided into natural polymer, synthetic polymers, inorganic materials and phospholipid. In this review, we first introduced several ways of nano delivery systems crossing the BBB, and then summarized their applications in ischemic stroke. Based on their potential and challenges in the treatment of ischemic stroke, new ideas and prospects are proposed for designing feasible and effective nano delivery systems.
Collapse
Affiliation(s)
- Yi-Xuan Li
- Faculty of Materials Science and Chemical Engineering, Ningbo University, Ningbo, China
| | - Hong-Bo Wang
- Department of Pharmacy, Ningbo University Affiliated Yangming Hospital, Yuyao, China
| | - Jian-Bo Jin
- Department of Pharmacy, Ningbo University Affiliated Yangming Hospital, Yuyao, China
| | - Chun-Lin Yang
- Department of Pharmacy, Ningbo University Affiliated Yangming Hospital, Yuyao, China
| | - Jing-Bo Hu
- Faculty of Materials Science and Chemical Engineering, Ningbo University, Ningbo, China
| | - Jing Li
- Department of Pharmacy, Ningbo University Affiliated Yangming Hospital, Yuyao, China
- *Correspondence: Jing Li,
| |
Collapse
|