1
|
Lan Y, Zou C, Nong F, Huang Q, Zeng J, Song W, Liang G, Wei Q, Pan M, Zou D, Long Y. Decoding immune cell dynamics in ischemic stroke: insights from single-cell RNA sequencing analysis. Front Aging Neurosci 2025; 17:1549518. [PMID: 40303468 PMCID: PMC12037566 DOI: 10.3389/fnagi.2025.1549518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Background Ischemic stroke (IS) is a leading cause of adult disability worldwide. The inflammatory processes involved are complex, making it challenging to fully understand the pathological mechanisms of IS. Phagocytosis plays an important role in eliminating neurotoxic or damaged neurons resulting from inflammatory responses. This study employed bioinformatics methods to analyze single-cell RNA sequencing (scRNA-seq) data to investigate the cell types and molecular biological processes involved in IS. Methods scRNA-seq data for IS were obtained from the Gene Expression Omnibus (GEO). Following sample screening and reprocessing, 5,582 single cells were identified from healthy controls and patients with IS. Uniform manifold approximation and projection (UMAP) was utilized to further explore the cellular composition in IS. Functional enrichment analysis of differentially expressed genes was conducted to identify transcriptional regulators, whereas cell developmental trajectories were predicted to uncover potential cell fate decisions. iTALK was employed to identify potential ligand-receptor axes within the cell-type immune microenvironment of IS. Results Based on scRNA-seq data analysis, we identified four cell types and their associated subclusters, along with genes exhibiting significant differential expression within these subclusters. Phagocytosis was significantly enriched in cell types linked to IS, while the differentiation trajectories of subpopulations in IS was different. Additionally, multiple receptor-ligand axes were identified, indicating diverse interactions within the immune microenvironment of IS. Conclusion This study demonstrated that phagocytosis in IS cell types critically influences disease progression. It also predicted the trajectories of infarct cells. These findings provide valuable insights into the molecular and cellular mechanisms underlying IS and highlight potential pathways for therapeutic intervention.
Collapse
Affiliation(s)
- Yating Lan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Chun Zou
- Department of Rehabilitation, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Feiyu Nong
- Department of Rehabilitation, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qi Huang
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jingyi Zeng
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wenyi Song
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Guining Liang
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qingyan Wei
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Mika Pan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Donghua Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yaobin Long
- Department of Rehabilitation, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
2
|
Torres VO, Turchan-Cholewo J, Colson MK, Yanev P, Britsch DRS, Cotter KM, McAtee AM, Ujas TA, Mercurio D, Kong X, Plautz EJ, Joshi CR, Matsui TK, Mori E, Cajigas-Hernandez A, Zuurbier K, Estus S, Goldberg MP, Monson NL, Stowe AM. B cells upregulate NMDARs, respond to extracellular glutamate, and express mature BDNF to protect the brain from ischemic injury. Neurobiol Dis 2025; 207:106819. [PMID: 39900302 PMCID: PMC11948303 DOI: 10.1016/j.nbd.2025.106819] [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: 10/16/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/05/2025] Open
Abstract
Following stroke, B cells enter brain regions outside of the ischemic injury to mediate functional recovery. Although B cells produce neurotrophins that support remote plasticity, including brain-derived neurotrophic factor (BDNF), it remains unclear which signal(s) activate B cells in the absence of infarct-localized pro-inflammatory cues. Activation of N-methyl-d-aspartate (NMDA)-type receptor (NMDAR) subunits on neurons can upregulate mature BDNF (mBDNF) production from a pro-BDNF precursor, but whether this occurs in B cells is unknown. We identified GluN2A and GluN2B NMDAR subunits on B cells that respond to glutamate and mediate nearly half of the glutamate-induced Ca2+ responses in activated B cell subsets. Ischemic stroke recruits GluN2A+ B cells into the ipsilesional hemisphere and both stroke and neurophysiologic levels of glutamate regulate gene and surface expression. Regardless of injury, pro-BDNF+ B cells localize to spleen/circulation whereas mBDNF+ B cells localize to the brain, including in aged male and female mice. We confirmed B cell-derived BDNF was required for in vitro and in vivo B cell-mediated neuroprotection. Lastly, GluN2A, GluN2B, glutamate-induced Ca2+ responses, and BDNF expression were all clinically confirmed in B cells from healthy donors, with BDNF+ B cells present in post-stroke human parenchyma. These data suggest that B cells express functional NMDARs that respond to glutamate, enhance NMDAR signaling with activation, and upregulate mature BDNF expression within the brain. This study identifies potential glutamate-induced neurotrophic roles for B cells in the brain; an immune response to neurotransmitters unique from established pro-inflammatory stimuli and relevant to any CNS-localized injury or disease.
Collapse
Affiliation(s)
- Vanessa O Torres
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA; Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Jadwiga Turchan-Cholewo
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Mary K Colson
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Pavel Yanev
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Daimen R S Britsch
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Katherine M Cotter
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Annabel M McAtee
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Thomas A Ujas
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Domenico Mercurio
- Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA
| | - Xiangmei Kong
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA
| | - Erik J Plautz
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA
| | - Chaitanya R Joshi
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA
| | - Takeshi K Matsui
- Department of Neural and Muscular Physiology, Shimane University School of Medicine, 89-1 Enya-cho, Izumo-shi 693-8501, Shimane, Japan; Department of Future Basic Medicine, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8521 Nara, Japan
| | - Eiichiro Mori
- Department of Future Basic Medicine, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8521 Nara, Japan
| | - Ambar Cajigas-Hernandez
- Department of Neuroscience, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9111, USA
| | - Kielen Zuurbier
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9148, USA
| | - Steven Estus
- Department of Physiology, University of Kentucky, 741 S. Limestone, BBSRB B243, Lexington, KY 40536, USA
| | - Mark P Goldberg
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA; Department of Neurology, Institute for Integration of Medicine and Science, UT Health San Antonio, 7703 Floyd Curl Drive, MSC 7883, San Antonio, TX 78229, USA
| | - Nancy L Monson
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA; Department of Immunology, University of Texas Southwestern Medical Center, 6124 Harry Hines Blvd., Dallas, TX 75390-9093, USA
| | - Ann M Stowe
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Suite NL9.114, Dallas, TX 75390-8813, USA; Department of Neurology, University of Kentucky, 740 S. Limestone, Kentucky Clinic J-455, Lexington, KY 40536, USA.
| |
Collapse
|
3
|
Yang G, Su R, Bu J, Li Y, Lin X, Jin J, Zhang Y, Zhuang P, Guo H, Yin Q. Emerging role of adaptive immunity in diabetes-induced cognitive impairment: from the periphery to the brain. Metab Brain Dis 2025; 40:102. [PMID: 39821703 DOI: 10.1007/s11011-025-01532-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/09/2025] [Indexed: 01/19/2025]
Abstract
Diabetic cognitive impairment (DCI) is a central nervous system complication induced by peripheral metabolic dysfunction of diabetes mellitus. Cumulative studies have shown that neuro-immune crosstalk is involved in the pathological progression of DCI. However, current studies mostly focus on the interaction between innate immunity cells and neurons, while ignoring the role of adaptive immunity cells in DCI. Notably, recent studies have revealed adaptive immune cells are involved in cognitive development and the progression of neurodegenerative diseases. Equally important, accumulated past studies have also shown that diabetic patients experience imbalanced peripheral adaptive immune homeostasis and disrupted transmission of adaptive immune cells to the central system. Therefore, this review first updated the cognitive mechanism of adaptive immune regulation, and then summarized the contribution of adaptive immunity to DCI from the aspects of peripheral adaptive immune homeostasis, transmission pathways, and brain tissue infiltration. Furthermore, we also summarized the potential of anti-diabetic drugs to regulate adaptive immunity, and looked forward to the potential value of regulatory adaptive immunity in the prevention and treatment of DCI, to provide a new strategy for the prevention and treatment of DCI.
Collapse
Affiliation(s)
- Genhui Yang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Runtao Su
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jie Bu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ying Li
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xueling Lin
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jiahui Jin
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yanjun Zhang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Pengwei Zhuang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| | - Hong Guo
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Qingsheng Yin
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
4
|
Iadecola C, Anrather J. The immunology of stroke and dementia. Immunity 2025; 58:18-39. [PMID: 39813992 PMCID: PMC11736048 DOI: 10.1016/j.immuni.2024.12.008] [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: 09/03/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
Ischemic stroke and vascular cognitive impairment, caused by a sudden arterial occlusion or more subtle but protracted vascular insufficiency, respectively, are leading causes of morbidity and mortality worldwide with limited therapeutic options. Innate and adaptive immunity have long been implicated in neurovascular injury, but recent advances in methodology and new experimental approaches have shed new light on their contributions. A previously unappreciated dynamic interplay of brain-resident, meningeal, and systemic immune cells with the ischemic brain and its vasculature has emerged, and new insights into the frequent overlap between vascular and Alzheimer pathology have been provided. Here, we critically review these recent findings, place them in the context of current concepts on neurovascular pathologies and Alzheimer's disease, and highlight their impact on recent stroke and Alzheimer therapies.
Collapse
Affiliation(s)
- Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
5
|
Sang Q, Kang C, Liu D, Wang L, Liu X, Li J. Polyphyllin VII ameliorates neuroinflammation and brain injury via modulating Treg/Th17 balance in a mouse model of cerebral ischemia-reperfusion injury. Int Immunopharmacol 2024; 143:113423. [PMID: 39447415 DOI: 10.1016/j.intimp.2024.113423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 09/08/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
Dysregulation of Th17 and Treg cells contributes to the pathophysiology of cerebral ischemia. Metabolic changes of peripheral CD4+ T cells lead to the imbalance of Treg/Th17 polarization, which represents a promising strategy for post-stroke therapy. Polyphyllin VII (PVII), a steroidal saponin extracted from traditional Chinese herb Rhizoma Paridis, has multiple bioactivities, but the potential function of PVII in cerebral ischemia-reperfusion injury is not elucidated yet. In our study, a mouse transient middle cerebral artery occlusion (MCAO) model was constructed. TTC staining, H&E staining, TUNEL staining, ELISA assay, flow cytometry, western blot, RT-qPCR, Open-field test, Morris water maze test, hanging wire test, rotarod test and foot-fault test were performed to evaluate the potential function of PVII in MCAO mice. We found that PVII showed protective effects on cerebral ischemia-reperfusion injury by reducing infarct volume, ameliorating brain injury and neuroinflammation, and improving long-term functional recovery of MCAO mice. PVII promoted Treg infiltration and suppressed infiltration of Th1/Th17 cells in ischemic brain in vivo. Moreover, PVII impaired peripheral CD4+ T cell activation and modulated Treg/Th17 differentiation in vitro. Mechanistically, PVII suppressed mTORC1 activation to influence glycolytic metabolism and ROS generation of T cells, thus leads to the imbalance of Treg/Th17 polarization towards Treg skewed. Furthermore, reactivation of mTORC1 by MHY1485 abolished the influence of PVII on brain injury and neuroinflammation in MCAO mice. Our data provided a novel role of PVII in cerebral ischemia-reperfusion injury via manipulating Treg/Th17 imbalance.
Collapse
Affiliation(s)
- Qiuling Sang
- Department of Neuroelectrophysiology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Chunyang Kang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Dingxi Liu
- Department of Clinical Medicine, Zunyi Medical University, Zhuhai 519041, China
| | - Libo Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Xiaoyang Liu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130000, China.
| | - Jia Li
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
6
|
Chen Y, Zhu Y, Huang C, Qu Y, Zhu Y. Identification and Validation of Ferroptosis Related Genes in Ischemic Stroke and Its Effect on the Peripheral Immune Landscape. Int J Gen Med 2024; 17:6377-6392. [PMID: 39720573 PMCID: PMC11668250 DOI: 10.2147/ijgm.s485612] [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: 08/15/2024] [Accepted: 12/05/2024] [Indexed: 12/26/2024] Open
Abstract
Background This research utilized a combination of gene databases associated with ferroptosis and online gene expression data from ischemic stroke samples to pinpoint ferroptosis-related genes (FRGs) in ischemic stroke cases. Methods By employing Random Forest (RF) and Support Vector Machine (SVM) models based on these genes, an overlap of genes from both models was identified as "Hub" genes. Through consensus clustering analysis using Hub genes, two distinct clusters of FRGs were revealed in ischemic stroke samples. Examination of the correlation between these molecular subtypes and the immune microenvironment highlighted a close link between gene expression levels and immune cell infiltration. Significantly different gene expression and functions within the FRG clusters underscored the pivotal role of Hub genes in the immune microenvironment. A gene diagnostic model related to ferroptosis was developed and validated to elucidate the significance of the identified genes. Results The results demonstrated that the Hub gene-based classification model effectively differentiated between ischemic stroke patients and normal samples, achieving an AUC of 0.900, signifying clinical relevance. Conclusion This study successfully identified ferroptosis-related genes in ischemic stroke, offering insights that could contribute to the formulation of future comprehensive treatment approaches.
Collapse
Affiliation(s)
- Yan Chen
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, People’s Republic of China
| | - Yanmei Zhu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, People’s Republic of China
| | - Cong Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, People’s Republic of China
| | - Youyang Qu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, People’s Republic of China
| | - Yulan Zhu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, People’s Republic of China
| |
Collapse
|
7
|
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
|
8
|
Yang C, Cai YX, Wang ZF, Tian SF, Li ZQ. Tertiary lymphoid structures in the central nervous system. Trends Mol Med 2024:S1471-4914(24)00281-8. [PMID: 39578120 DOI: 10.1016/j.molmed.2024.10.014] [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: 08/01/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
Tertiary lymphoid structures (TLSs) frequently occur at sites of chronic inflammation. A more advanced stage of multiple sclerosis (MS) has been associated with certain TLSs. However, tumor-associated TLSs have been shown to correlate with a greater treatment response rate and a better prognosis in glioma mouse models. In this review, we evaluate the clinical significances of TLSs in prognosis and treatment response, as well as the status of TLS-directed therapies targeting alternative biochemical pathways in various central nervous system (CNS) disorders. Potential molecular mechanisms underlying the development of TLSs are also discussed. Exploring these areas may provide an essential understanding of the processes behind disease advancement, uncover new therapeutic objectives, and detect biomarkers that forecast disease progression and treatment efficacy.
Collapse
Affiliation(s)
- Chao Yang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yu-Xiang Cai
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ze-Fen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Su-Fang Tian
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
9
|
Wang C, Gu L, Zhang Y, Gao Y, Jian Z, Xiong X. Bibliometric insights into the inflammation and mitochondrial stress in ischemic stroke. Exp Neurol 2024; 378:114845. [PMID: 38838802 DOI: 10.1016/j.expneurol.2024.114845] [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: 03/22/2024] [Revised: 05/19/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Research in the areas of inflammation and mitochondrial stress in ischemic stroke is rapidly expanding, but a comprehensive overview that integrates bibliometric trends with an in-depth review of molecular mechanisms is lacking. OBJECTIVE To map the evolving landscape of research using bibliometric analysis and to detail the molecular mechanisms that underpin these trends, emphasizing their implications in ischemic stroke. METHODS We conducted a bibliometric analysis to identify key trends, top contributors, and focal research themes. In addition, we review recent research advances in mitochondrial stress and inflammation in ischemic stroke to gain a detailed understanding of the pathophysiological processes involved. CONCLUSION Our integrative approach not only highlights the growing research interest and collaborations but also provides a detailed exploration of the molecular mechanisms that are central to the pathology of ischemic stroke. This synthesis offers valuable insights for researchers and paves the way for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Chaoqun Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, 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
| | - Yonggang Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yikun Gao
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China; Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
10
|
Zhong L, Xie Z, Han Z, Fan J, Wang R, Tao Z, Ma Q, Luo Y. Long Non-Coding H19 in Lymphocytes: Prognostic Value in Acute Ischemic Stroke Patients. Pharmaceuticals (Basel) 2024; 17:1008. [PMID: 39204113 PMCID: PMC11357374 DOI: 10.3390/ph17081008] [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: 06/21/2024] [Revised: 07/19/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
Acute ischemic stroke (AIS) is a cerebrovascular disease that seriously affects the physical and mental health and quality of life of patients. However, there is a lack of reliable prognostic prediction methods. The main objective of this study was to investigate the prognostic value of long non-coding RNA (lncRNA) H19 in lymphocytes of patients with AIS, and to construct a prognostic prediction model for AIS including lncRNA H19 in lymphocytes, which would provide new ideas for the prognostic evaluation of AIS. Poor prognosis was defined when the patient's modified Rankin scale (mRS) score at 3 months after AIS onset was greater than 2. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to measure the level of lncRNA H19 in lymphocytes. Spearman correlation analysis revealed a positive correlation between lncRNA H19 and mRS score at 3 months after AIS onset (r = 0.1977, p = 0.0032), while lncRNA H19 was negatively correlated with white blood cells counts, lymphocytes counts, and neutrophils counts. Logistic regression analysis identified lncRNA H19 as an independent predictor of poor prognosis (OR = 3.062 [1.69-5.548], p < 0.001). Moreover, a nomogram prediction model incorporating lncRNA H19 in lymphocytes demonstrated effective discrimination, calibration, and clinical applicability in predicting AIS outcomes. The findings suggest that lncRNA H19 in lymphocytes could be a valuable prognostic indicator and a potential pharmacological target for AIS patients, and might be a novel pathway for enhanced prognostic evaluation and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Liyuan Zhong
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; (L.Z.); (Z.X.); (Z.H.); (J.F.); (R.W.); (Z.T.)
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Zixian Xie
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; (L.Z.); (Z.X.); (Z.H.); (J.F.); (R.W.); (Z.T.)
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Ziping Han
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; (L.Z.); (Z.X.); (Z.H.); (J.F.); (R.W.); (Z.T.)
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Junfen Fan
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; (L.Z.); (Z.X.); (Z.H.); (J.F.); (R.W.); (Z.T.)
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Rongliang Wang
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; (L.Z.); (Z.X.); (Z.H.); (J.F.); (R.W.); (Z.T.)
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Zhen Tao
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; (L.Z.); (Z.X.); (Z.H.); (J.F.); (R.W.); (Z.T.)
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Qingfeng Ma
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yumin Luo
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; (L.Z.); (Z.X.); (Z.H.); (J.F.); (R.W.); (Z.T.)
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
- Beijing Geriatric Medical Research Center, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| |
Collapse
|
11
|
Zhao P, Zhang G, Wang Y, Wei C, Wang Z, Zhai W, Shen Y, Shi L, Sun L. Peripheral immunity is associated with cognitive impairment after acute minor ischemic stroke and transient ischemic attack. Sci Rep 2024; 14:16201. [PMID: 39003356 PMCID: PMC11246473 DOI: 10.1038/s41598-024-67172-w] [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: 09/07/2023] [Accepted: 07/09/2024] [Indexed: 07/15/2024] Open
Abstract
Immunoinflammation is associated with the development of post-stroke cognitive impairment (PSCI), however, peripheral immunity has not been fully explored. We aimed to investigate the association between PSCI and peripheral immune indicators, including neutrophil, lymphocyte, and mononuclear percentages and counts; the systemic immune inflammation index; platelet-to-lymphocyte ratio; neutrophil-to-lymphocyte ratio (NLR); and lymphocyte-to-monocyte ratio. A total of 224 patients with acute minor ischemic stroke or transient ischemic attack with 6-12 months of follow-up were included. PSCI was defined as a Montreal Cognitive Assessment score < 22 during the follow-up period. We performed logistic regression, subgroup analyses based on age and sex, and further established predictive models. We found that increased innate immunity indicators (neutrophils, neutrophil percentage) increased the risk of PSCI, whereas increased adaptive immunity indicator (lymphocytes) were protective against PSCI, especially in patients aged 50-65 years. Neutrophil percentage and NLR improved the predictive efficacy of the models that included demographic, clinical, and imaging information, with the area under the curve increased from 0.765 to 0.804 and 0.803 (P = 0.042 and 0.049, respectively). We conducted a comprehensive analysis of peripheral immunity in PSCI, providing a novel perspective on the early detection, etiology, and treatment of PSCI.
Collapse
Affiliation(s)
- PanPan Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - GuiMei Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - YongChun Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - ChunXiao Wei
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - ZiCheng Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - WeiJie Zhai
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - YanXin Shen
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Lin Shi
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 71#, Changchun, 130021, China.
| |
Collapse
|
12
|
Feng J, Li Y, Wang C, Wang Y, Wan Y, Zheng M, Chen T, Xiao X. Peripheral blood transcriptomic analysis identifies potential inflammation and immune signatures for central retinal artery occlusion. Sci Rep 2024; 14:7398. [PMID: 38548806 PMCID: PMC10978867 DOI: 10.1038/s41598-024-57052-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/13/2024] [Indexed: 04/01/2024] Open
Abstract
Central retinal artery occlusion (CRAO) is an acute retinal ischaemic disease, but early diagnosis is challenging due to a lack of biomarkers. Blood samples were collected from CRAO patients and cataract patients. Gene expression profiles were distinct between arterial/venous CRAO blood (A-V group) and venous CRAO/control blood (V-C group) samples. Differentially expressed genes (DEGs) were subjected to GO and KEGG enrichment analyses. Hub genes were identified by Cytoscape and used to predict gene interactions via GeneMANIA. Immune cell infiltration was analysed by CIBERSORT. More than 1400 DEGs were identified in the A-V group and 112 DEGs in the V-C group compared to controls. The DEGs in both groups were enriched in the ribosome pathway, and those in the V-C group were also enriched in antigen processing/MHC pathways. Network analysis identified ribosomal proteins (RPS2 and RPS5) as the core genes of the A-V group and MHC genes (HLA-F) as the core genes of the V-C group. Coexpression networks showed ribosomal involvement in both groups, with additional immune responses in the V-C group. Immune cell analysis indicated increased numbers of neutrophils and T cells. Ribosomal and MHC-related genes were identified as potential CRAO biomarkers, providing research directions for prevention, diagnosis, treatment and prognosis.
Collapse
Affiliation(s)
- Jiaqing Feng
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China
| | - Ying Li
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China
| | - Chuansen Wang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China
| | - Yuedan Wang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China
| | - Yuwei Wan
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China
| | - Mengxue Zheng
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China
| | - Ting Chen
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China.
| | - Xuan Xiao
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China.
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
13
|
Mergenthaler P, Balami JS, Neuhaus AA, Mottahedin A, Albers GW, Rothwell PM, Saver JL, Young ME, Buchan AM. Stroke in the Time of Circadian Medicine. Circ Res 2024; 134:770-790. [PMID: 38484031 DOI: 10.1161/circresaha.124.323508] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/15/2024] [Indexed: 03/19/2024]
Abstract
Time-of-day significantly influences the severity and incidence of stroke. Evidence has emerged not only for circadian governance over stroke risk factors, but also for important determinants of clinical outcome. In this review, we provide a comprehensive overview of the interplay between chronobiology and cerebrovascular disease. We discuss circadian regulation of pathophysiological mechanisms underlying stroke onset or tolerance as well as in vascular dementia. This includes cell death mechanisms, metabolism, mitochondrial function, and inflammation/immunity. Furthermore, we present clinical evidence supporting the link between disrupted circadian rhythms and increased susceptibility to stroke and dementia. We propose that circadian regulation of biochemical and physiological pathways in the brain increase susceptibility to damage after stroke in sleep and attenuate treatment effectiveness during the active phase. This review underscores the importance of considering circadian biology for understanding the pathology and treatment choice for stroke and vascular dementia and speculates that considering a patient's chronotype may be an important factor in developing precision treatment following stroke.
Collapse
Affiliation(s)
- Philipp Mergenthaler
- Center for Stroke Research Berlin (P.M., A.M.B.), Charité - Universitätsmedizin Berlin, Germany
- Department of Neurology with Experimental Neurology (P.M.), Charité - Universitätsmedizin Berlin, Germany
- Stroke Research, Radcliffe Department of Medicine (P.M., J.S.B., A.A.N., A.M., A.M.B.), University of Oxford, United Kingdom
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Joyce S Balami
- Stroke Research, Radcliffe Department of Medicine (P.M., J.S.B., A.A.N., A.M., A.M.B.), University of Oxford, United Kingdom
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Ain A Neuhaus
- Stroke Research, Radcliffe Department of Medicine (P.M., J.S.B., A.A.N., A.M., A.M.B.), University of Oxford, United Kingdom
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, United Kingdom (A.A.N.)
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Amin Mottahedin
- Stroke Research, Radcliffe Department of Medicine (P.M., J.S.B., A.A.N., A.M., A.M.B.), University of Oxford, United Kingdom
- Nuffield Department of Clinical Neurosciences (A.M., P.M.R.), University of Oxford, United Kingdom
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Gregory W Albers
- Department of Neurology, Stanford Hospital, Palo Alto, CA (G.W.A.)
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Peter M Rothwell
- Nuffield Department of Clinical Neurosciences (A.M., P.M.R.), University of Oxford, United Kingdom
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences (P.M.R.), University of Oxford, United Kingdom
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Jeffrey L Saver
- Department of Neurology and Comprehensive Stroke Center, Geffen School of Medicine, University of Los Angeles, CA (J.L.S.)
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (M.E.Y.)
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Alastair M Buchan
- Center for Stroke Research Berlin (P.M., A.M.B.), Charité - Universitätsmedizin Berlin, Germany
- Stroke Research, Radcliffe Department of Medicine (P.M., J.S.B., A.A.N., A.M., A.M.B.), University of Oxford, United Kingdom
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| |
Collapse
|
14
|
Wu S, Tabassum S, Payne CT, Hu H, Gusdon AM, Choi HA, Ren XS. Updates of the role of B-cells in ischemic stroke. Front Cell Neurosci 2024; 18:1340756. [PMID: 38550918 PMCID: PMC10972894 DOI: 10.3389/fncel.2024.1340756] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/27/2024] [Indexed: 10/11/2024] Open
Abstract
Ischemic stroke is a major disease causing death and disability in the elderly and is one of the major diseases that seriously threaten human health and cause a great economic burden. In the early stage of ischemic stroke, neuronal structure is destroyed, resulting in death or damage, and the release of a variety of damage-associated pattern molecules induces an increase in neuroglial activation, peripheral immune response, and secretion of inflammatory mediators, which further exacerbates the damage to the blood-brain barrier, exacerbates cerebral edema, and microcirculatory impairment, triggering secondary brain injuries. After the acute phase of stroke, various immune cells initiate a protective effect, which is released step by step and contributes to the repair of neuronal cells through phenotypic changes. In addition, ischemic stroke induces Central Nervous System (CNS) immunosuppression, and the interaction between the two influences the outcome of stroke. Therefore, modulating the immune response of the CNS to reduce the inflammatory response and immune damage during stroke is important for the protection of brain function and long-term recovery after stroke, and modulating the immune function of the CNS is expected to be a novel therapeutic strategy. However, there are fewer studies on B-cells in brain function protection, which may play a dual role in the stroke process, and the understanding of this cell is still incomplete. We review the existing studies on the mechanisms of the role of B-cells, inflammatory response, and immune response in the development of ischemic stroke and provide a reference for the development of adjuvant therapeutic drugs for ischemic stroke targeting inflammatory injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xuefang S. Ren
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, United States
| |
Collapse
|
15
|
Ma X, Zuo Y, Hu X, Chen S, Zhong K, Xue R, Gui S, Liu K, Li S, Zhu X, Yang J, Deng Z, Liu X, Xu Y, Liu S, Shi Z, Zhou M, Tang Y. Terminally differentiated cytotoxic CD4 + T cells were clonally expanded in the brain lesion of radiation-induced brain injury. CNS Neurosci Ther 2024; 30:e14682. [PMID: 38499993 PMCID: PMC10948588 DOI: 10.1111/cns.14682] [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: 12/11/2023] [Revised: 02/04/2024] [Accepted: 02/25/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Accumulating evidence supports the involvement of adaptive immunity in the development of radiation-induced brain injury (RIBI). Our previous work has emphasized the cytotoxic function of CD8+ T cells in RIBI. In this study, we aimed to investigate the presence and potential roles of cytotoxic CD4+ T cells (CD4+ CTLs) in RIBI to gain a more comprehensive understanding of adaptive immunity in this context. MAIN TEXT Utilizing single-cell RNA sequencing (scRNA-seq), we analyzed 3934 CD4+ T cells from the brain lesions of four RIBI patients and identified six subclusters within this population. A notable subset, the cytotoxic CD4+ T cells (CD4+ CTLs), was marked with high expression of cytotoxicity-related genes (NKG7, GZMH, GNLY, FGFBP2, and GZMB) and several chemokine and chemokine receptors (CCL5, CX3CR1, and CCL4L2). Through in-depth pseudotime analysis, which simulates the development of CD4+ T cells, we observed that the CD4+ CTLs exhibited signatures of terminal differentiation. Their functions were enriched in protein serine/threonine kinase activity, GTPase regulator activity, phosphoprotein phosphatase activity, and cysteine-type endopeptidase activity involved in the apoptotic signaling pathway. Correspondingly, mice subjected to gamma knife irradiation on the brain showed a time-dependent infiltration of CD4+ T cells, an increase of MHCII+ cells, and the existence of CD4+ CTLs in lesions, along with an elevation of apoptotic-related proteins. Finally, and most crucially, single-cell T-cell receptor sequencing (scTCR-seq) analysis at the patient level determined a large clonal expansion of CD4+ CTLs in lesion tissues of RIBI. Transcriptional factor-encoding genes TBX21, RORB, and EOMES showed positive correlations with the cytotoxic functions of CD4+ T cells, suggesting their potential to distinguish RIBI-related CD4+ CTLs from other subsets. CONCLUSION The present study enriches the understanding of the transcriptional landscape of adaptive immune cells in RIBI patients. It provides the first description of a clonally expanded CD4+ CTL subset in RIBI lesions, which may illuminate new mechanisms in the development of RIBI and offer potential biomarkers or therapeutic targets for the disease.
Collapse
Affiliation(s)
- Xueying Ma
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - You Zuo
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Xia Hu
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhouChina
- Jiangmen Central HospitalAffiliated Jiangmen Hospital of Sun Yat‐sen UniversityJiangmenChina
| | - Sitai Chen
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Ke Zhong
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of Pharmacy, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Ruiqi Xue
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shushu Gui
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Kejia Liu
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shaojian Li
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Xiaoqiu Zhu
- Department of Anesthesiology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Jingwen Yang
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhenhong Deng
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Xiaolu Liu
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yongteng Xu
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceSun Yat‐sen UniversityGuangzhouChina
| | - Zhongshan Shi
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhouChina
- Jiangmen Central HospitalAffiliated Jiangmen Hospital of Sun Yat‐sen UniversityJiangmenChina
| | - Yamei Tang
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Brain Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
16
|
Fu R, Zhao L, Guo Y, Qin X, Xu W, Cheng X, Zhang Y, Xu S. AIM2 inflammasome: A potential therapeutic target in ischemic stroke. Clin Immunol 2024; 259:109881. [PMID: 38142900 DOI: 10.1016/j.clim.2023.109881] [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: 09/28/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Ischemic stroke (IS) is a significant global public health issue with a high incidence, disability, and mortality rate. A robust inflammatory cascade with complex and wide-ranging mechanisms occurs following ischemic brain injury. Inflammasomes are multiprotein complexes in the cytoplasm that modulate the inflammatory response by releasing pro-inflammatory cytokines and inducing cellular pyroptosis. Among these inflammasomes, the Absent in Melanoma 2 (AIM2) inflammasome shows the ability to detect a wide range of pathogen DNAs, thereby triggering an inflammatory response. Recent studies have indicated that the aberrant expression of AIM2 inflammasome in various cells is closely associated with the pathological processes of ischemic brain injury. This paper summarizes the expression and regulatory role of AIM2 in CNS and peripheral immune cells and discusses current therapeutic approaches targeting AIM2 inflammasome. These findings aim to serve as a reference for future research in this field.
Collapse
Affiliation(s)
- Rong Fu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Linna Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Yuying Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Xiaoli Qin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenzhe Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xueqi Cheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunsha Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shixin Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
17
|
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
|
18
|
Zbesko JC, Stokes J, Becktel DA, Doyle KP. Targeting foam cell formation to improve recovery from ischemic stroke. Neurobiol Dis 2023; 181:106130. [PMID: 37068641 PMCID: PMC10993857 DOI: 10.1016/j.nbd.2023.106130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/31/2023] [Accepted: 04/14/2023] [Indexed: 04/19/2023] Open
Abstract
Inflammation is a crucial part of the healing process after an ischemic stroke and is required to restore tissue homeostasis. However, the inflammatory response to stroke also worsens neurodegeneration and creates a tissue environment that is unfavorable to regeneration for several months, thereby postponing recovery. In animal models, inflammation can also contribute to the development of delayed cognitive deficits. Myeloid cells that take on a foamy appearance are one of the most prominent immune cell types within chronic stroke infarcts. Emerging evidence indicates that they form as a result of mechanisms of myelin lipid clearance becoming overwhelmed, and that they are a key driver of the chronic inflammatory response to stroke. Therefore, targeting lipid accumulation in foam cells may be a promising strategy for improving recovery. The aim of this review is to provide an overview of current knowledge regarding inflammation and foam cell formation in the brain in the weeks and months following ischemic stroke and identify targets that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Jacob C Zbesko
- Department of Immunobiology, University of Arizona, United States
| | - Jessica Stokes
- Department of Pediatrics, University of Arizona, United States
| | | | - Kristian P Doyle
- Department of Immunobiology, University of Arizona, United States; Departments of Neurology, Neurosurgery, Psychology, Arizona Center on Aging, and the BIO5 Institute, University of Arizona, United States.
| |
Collapse
|
19
|
Liu Y, Chen S, Liu S, Wallace KL, Zille M, Zhang J, Wang J, Jiang C. T-cell receptor signaling modulated by the co-receptors: Potential targets for stroke treatment. Pharmacol Res 2023; 192:106797. [PMID: 37211238 DOI: 10.1016/j.phrs.2023.106797] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/02/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Stroke is a severe and life-threatening disease, necessitating more research on new treatment strategies. Infiltrated T lymphocytes, an essential adaptive immune cell with extensive effector function, are crucially involved in post-stroke inflammation. Immediately after the initiation of the innate immune response triggered by microglia/macrophages, the adaptive immune response associated with T lymphocytes also participates in the complex pathophysiology of stroke and partially informs the outcome of stroke. Preclinical and clinical studies have revealed the conflicting roles of T cells in post-stroke inflammation and as potential therapeutic targets. Therefore, exploring the mechanisms that underlie the adaptive immune response associated with T lymphocytes in stroke is essential. The T-cell receptor (TCR) and its downstream signaling regulate T lymphocyte differentiation and activation. This review comprehensively summarizes the various molecules that regulate TCR signaling and the T-cell response. It covers both the co-stimulatory and co-inhibitory molecules and their roles in stroke. Because immunoregulatory therapies targeting TCR and its mediators have achieved great success in some proliferative diseases, this article also summarizes the advances in therapeutic strategies related to TCR signaling in lymphocytes after stroke, which can facilitate translation. DATA AVAILABILITY: No data was used for the research described in the article.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Shuai Chen
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Simon Liu
- Medical Genomics Unit, National Human Genome Research Institute, Bethesda, MD, 20814, USA
| | - Kevin L Wallace
- College of Mathematical and Natural Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, A-1090 Vienna, Austria
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, 450000, Zhengzhou, P. R. China.
| | - Jian Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China; Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, P. R. China.
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China.
| |
Collapse
|
20
|
Maheshwari S, Dwyer LJ, Sîrbulescu RF. Inflammation and immunomodulation in central nervous system injury - B cells as a novel therapeutic opportunity. Neurobiol Dis 2023; 180:106077. [PMID: 36914074 PMCID: PMC10758988 DOI: 10.1016/j.nbd.2023.106077] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 03/13/2023] Open
Abstract
Acute injury to the central nervous system (CNS) remains a complex and challenging clinical need. CNS injury initiates a dynamic neuroinflammatory response, mediated by both resident and infiltrating immune cells. Following the primary injury, dysregulated inflammatory cascades have been implicated in sustaining a pro-inflammatory microenvironment, driving secondary neurodegeneration and the development of lasting neurological dysfunction. Due to the multifaceted nature of CNS injury, clinically effective therapies for conditions such as traumatic brain injury (TBI), spinal cord injury (SCI), and stroke have proven challenging to develop. No therapeutics that adequately address the chronic inflammatory component of secondary CNS injury are currently available. Recently, B lymphocytes have gained increasing appreciation for their role in maintaining immune homeostasis and regulating inflammatory responses in the context of tissue injury. Here we review the neuroinflammatory response to CNS injury with particular focus on the underexplored role of B cells and summarize recent results on the use of purified B lymphocytes as a novel immunomodulatory therapeutic for tissue injury, particularly in the CNS.
Collapse
Affiliation(s)
- Saumya Maheshwari
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Liam J Dwyer
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruxandra F Sîrbulescu
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Wang YR, Cui WQ, Wu HY, Xu XD, Xu XQ. The role of T cells in acute ischemic stroke. Brain Res Bull 2023; 196:20-33. [PMID: 36906042 DOI: 10.1016/j.brainresbull.2023.03.005] [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: 12/29/2022] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
Acute ischemic stroke (AIS) is associated with high rates of disability and mortality, exerting a substantial impact on overall survival and health-related quality of life. Treatment of AIS remains challenging given that the underlying pathologic mechanisms remain unclear. However, recent research has demonstrated that the immune system plays a key role in the development of AIS. Numerous studies have reported infiltration of T cells into ischemic brain tissue. While some types of T cells can promote the development of inflammatory responses and aggravate ischemic damage in patients with AIS, other T cells appear to exert neuroprotective effects via immunosuppression and other mechanisms. In this review, we discuss the recent findings regarding the infiltration of T cells into ischemic brain tissue, and the mechanisms governing how T cells can facilitate tissue injury or neuroprotection in AIS. Factors influencing the function of T cells, such as intestinal microflora and sex differences, are also discussed. We also explore the recent research on the effect of non-coding RNA on T cells after stroke, as well as the potential for specifically targeting T cells in the treatment of stroke patients.
Collapse
Affiliation(s)
- Yi-Ran Wang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wen-Qiang Cui
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China; First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong-Yun Wu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China; First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiang-Dong Xu
- Experimental Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiang-Qing Xu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China; First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
22
|
Wang Y, Zhi H, Zhang X. Effect of Huangdisan grain on improving cognitive impairment in VD rats and its mechanism in immune inflammatory response. J Neuroimmunol 2023; 377:578058. [PMID: 36871311 DOI: 10.1016/j.jneuroim.2023.578058] [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: 09/09/2022] [Revised: 02/16/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND Vascular dementia (VD) is the second most common type of dementia after Alzheimer's disease (AD). Although the incidence rate is very high, there is no definitive treatment for VD. And it has serious impact on the quality of life of VD patients. In recent years, more and more studies about the clinical efficacy and pharmacological effects of traditional Chinese medicine (TCM) in the treatment of VD have been conducted. And Huangdisan grain has been used to treat VD patients with a good curative effect in clinic. OBJECTIVE This study was designed to investigate the effect of Huangdisan grain on the inflammatory response and cognitive function of VD rats modeled by bilateral common carotid artery occlusion (BCCAO), that aimed to improve the treatment methods for VD. METHODS 8-week-old healthy SPF male Wistar rats (280 ± 20 g) were randomly divided into the normal group (Gn, n = 10), sham operated group (Gs, n = 10), and operated group (Go, n = 35). The VD rat models in Go group were established by BCCAO. 8 weeks after surgery, the operated rats were screened by the hidden platform trail of Morris Water Maze (MWM), and the rats with cognitive dysfunction were further randomly divided into the impaired group (Gi, n = 10) and TCM group (Gm, n = 10). The VD rats in Gm group were given the intragastric administration of Huangdisan grain decoction once a day for 8 weeks, and the other groups were given intragastric administration of normal saline. Then the cognitive ability of rats in each group was detected by the MWM Test. The lymphocyte subsets in peripheral blood and hippocampus of rats were measured by flow cytometry. The levels of cytokines (IL-1β, IL-2, IL-4, IL-10, TNF-α, INF-γ, MIP-2, COX-2, iNOS) in peripheral blood and hippocampus were measured by ELISA (enzyme linked immunosorbent assay). The number of Iba-1+ CD68+ co-positive cells in the CA1 region of hippocampus was measured by immunofluorescence. RESULTS Compared with the Gn group, the escape latencies of the Gi group were prolonged (P < 0.01), the time spent in the former platform quadrant was shortened (P < 0.01), and the number of times of crossing over the former platform location was reduced (P < 0.05). But compared with the Gi group, the escape latencies of Gm group were shortened (P < 0.01), the time spent in the former platform quadrant was prolonged (P < 0.05), and the number of times of crossing over the former platform location was increased (P < 0.05). The number of Iba-1+ CD68+ co-positive cells in the CA1 region of hippocampus of VD rats in Gi group was increased (P < 0.01) compared with the Gn group. And the proportions of T Cells, CD4+ T Cells, CD8+ T Cells in the hippocampus were increased (P < 0.01). The level of pro-inflammatory cytokines in the hippocampus was increased significantly, such as IL-1β (P < 0.01), IL-2 (P < 0.01), TNF-α (P < 0.05), IFN-γ (P < 0.01), COX-2 (P < 0.01), MIP-2 (P < 0.01) and iNOS (P < 0.05). And the level of IL-10 (P < 0.01), a kind of anti-inflammatory cytokine, was decreased. The proportions of T Cells (P < 0.05), CD4+ T Cells (P < 0.01) and NK Cells (P < 0.05) in the peripheral blood of the VD rats in Gi group were decreased, and the level of IL-1β, IL-2, TNF-α, IFN-γ, COX-2, MIP-2 and iNOS was increased significantly (P < 0.01) compared with the Gn group. Meanwhile, the level of IL-4 and IL-10 was decreased (P < 0.01). Huangdisan grain could reduce the number of Iba-1+ CD68+ co-positive cells in the CA1 region of hippocampus (P < 0.01), decrease the proportions of T Cells, CD4+ T Cells, CD8+ T Cells and the level of IL-1β, MIP-2 in hippocampus (P < 0.01) of VD rats. Moreover, it could rise the proportion of NK Cells (P < 0.01) and the level of IL-4 (P < 0.05), IL-10 (P < 0.05), and decrease the level of IL-1β (P < 0.01), IL-2 (P < 0.05), TNF-α (P < 0.01), IFN-γ (P < 0.01), COX-2 (P < 0.01) and MIP-2 (P < 0.01) in peripheral blood of VD rats. CONCLUSION This study indicated that Huangdisan grain could decrease the activation of microglia/macrophages, regulate the proportions of lymphocyte subsets and the level of cytokines, which could adjust the immunologic abnormalities of VD rats, and ultimately improve cognitive function.
Collapse
Affiliation(s)
- Yao Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300380, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300380, China
| | - Hui Zhi
- The Ninth People's Hospital of Chongqing, Chongqing 400799, China
| | - Xuezhu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300380, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300380, China.
| |
Collapse
|
23
|
Pinosanu LR, Capitanescu B, Glavan D, Godeanu S, Cadenas IF, Doeppner TR, Hermann DM, Balseanu AT, Bogdan C, Popa-Wagner A. Neuroglia Cells Transcriptomic in Brain Development, Aging and Neurodegenerative Diseases. Aging Dis 2023; 14:63-83. [PMID: 36818562 PMCID: PMC9937697 DOI: 10.14336/ad.2022.0621] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
Glia cells are essential for brain functioning during development, aging and disease. However, the role of astroglia plays during brain development is quite different from the role played in the adult lesioned brain. Therefore, a deeper understanding of pathomechanisms underlying astroglia activity in the aging brain and cerebrovascular diseases is essential to guide the development of new therapeutic strategies. To this end, this review provides a comparison between the transcriptomic activity of astroglia cells during development, aging and neurodegenerative diseases, including cerebral ischemia. During fetal brain development, astrocytes and microglia often affect the same developmental processes such as neuro-/gliogenesis, angiogenesis, axonal outgrowth, synaptogenesis, and synaptic pruning. In the adult brain astrocytes are a critical player in the synapse remodeling by mediating synapse elimination while microglia activity has been associated with changes in synaptic plasticity and remove cell debris by constantly sensing the environment. However, in the lesioned brain astrocytes proliferate and play essential functions with regard to energy supply to the neurons, neurotransmission and buildup of a protective scar isolating the lesion site from the surroundings. Inflammation, neurodegeneration, or loss of brain homeostasis induce changes in microglia gene expression, morphology, and function, generally referred to as "primed" microglia. These changes in gene expression are characterized by an enrichment of phagosome, lysosome, and antigen presentation signaling pathways and is associated with an up-regulation of genes encoding cell surface receptors. In addition, primed microglia are characterized by upregulation of a network of genes in response to interferon gamma. Conclusion. A comparison of astroglia cells transcriptomic activity during brain development, aging and neurodegenerative disorders might provide us with new therapeutic strategies with which to protect the aging brain and improve clinical outcome.
Collapse
Affiliation(s)
- Leonard Radu Pinosanu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.
| | - Bogdan Capitanescu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.
| | - Daniela Glavan
- Psychiatric clinic, University of Medicine and Pharmacy Craiova, Craiova, Romania.
| | - Sanziana Godeanu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.
| | - Israel Ferna´ndez Cadenas
- Stroke Pharmacogenomics and Genetics group, Sant Pau Hospital Institute of Research, Barcelona, Spain.
| | - Thorsten R. Doeppner
- Department of Neurology, University Hospital Giessen, Giessen, Germany.,University of Göttingen Medical School, Department of Neurology, Göttingen, Germany.
| | - Dirk M. Hermann
- Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, Germany.
| | - Adrian-Tudor Balseanu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.
| | - Catalin Bogdan
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.,Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, Germany.,Correspondence should be addressed to: Dr. Aurel Popa-Wagner () and Dr. Catalin Bogdan (), University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Aurel Popa-Wagner
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.,Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, Germany.,Correspondence should be addressed to: Dr. Aurel Popa-Wagner () and Dr. Catalin Bogdan (), University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany
| |
Collapse
|
24
|
Zhou SY, Guo ZN, Yang Y, Qu Y, Jin H. Gut-brain axis: Mechanisms and potential therapeutic strategies for ischemic stroke through immune functions. Front Neurosci 2023; 17:1081347. [PMID: 36777635 PMCID: PMC9911679 DOI: 10.3389/fnins.2023.1081347] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
After an ischemic stroke (IS) occurs, immune cells begin traveling to the brain and immune system from the gut and gastrointestinal tract, where most of them typically reside. Because the majority of the body's macrophages and more than 70% of the total immune cell pool are typically found within the gut and gastrointestinal tract, inflammation and immune responses in the brain and immune organs require the mobilization of a large number of immune cells. The bidirectional communication pathway between the brain and gut is often referred to as the gut-brain axis. IS usually leads to intestinal motility disorders, dysbiosis of intestinal microbiota, and a leaky gut, which are often associated with poor prognosis in patients with IS. In recent years, several studies have suggested that intestinal inflammation and immune responses play key roles in the development of IS, and thus may become potential therapeutic targets that can drive new therapeutic strategies. However, research on gut inflammation and immune responses after stroke remains in its infancy. A better understanding of gut inflammation and immune responses after stroke may be important for developing effective therapies. This review discusses the immune-related mechanisms of the gut-brain axis after IS and compiles potential therapeutic targets to provide new ideas and strategies for the future effective treatment of IS.
Collapse
Affiliation(s)
- Sheng-Yu Zhou
- Department of Neurology, Stroke Center, The First Hospital of Jilin University, Changchun, China
| | - Zhen-Ni Guo
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
| | - Yi Yang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
| | - Yang Qu
- Department of Neurology, Stroke Center, The First Hospital of Jilin University, Changchun, China
| | - Hang Jin
- Department of Neurology, Stroke Center, The First Hospital of Jilin University, Changchun, China,*Correspondence: Hang Jin,
| |
Collapse
|
25
|
Xu B, Shimauchi-Ohtaki H, Yoshimoto Y, Sadakata T, Ishizaki Y. Transplanted human iPSC-derived vascular endothelial cells promote functional recovery by recruitment of regulatory T cells to ischemic white matter in the brain. J Neuroinflammation 2023; 20:11. [PMID: 36650518 PMCID: PMC9847196 DOI: 10.1186/s12974-023-02694-0] [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: 10/26/2022] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Ischemic stroke in white matter of the brain induces not only demyelination, but also neuroinflammation. Peripheral T lymphocytes, especially regulatory T cells (Tregs), are known to infiltrate into ischemic brain and play a crucial role in modulation of inflammatory response there. We previously reported that transplantation of vascular endothelial cells generated from human induced pluripotent stem cells (iVECs) ameliorated white matter infarct. The aim of this study is to investigate contribution of the immune system, especially Tregs, to the mechanism whereby iVEC transplantation ameliorates white matter infarct. METHODS iVECs and human Tregs were transplanted into the site of white matter lesion seven days after induction of ischemia. The egress of T lymphocytes from lymph nodes was sequestered by treating the animals with fingolimod (FTY720). The infarct size was evaluated by magnetic resonance imaging. Immunohistochemistry was performed to detect the activated microglia and macrophages, T cells, Tregs, and oligodendrocyte lineage cells. Remyelination was examined by Luxol fast blue staining. RESULTS iVEC transplantation reduced ED-1+ inflammatory cells and CD4+ T cells, while increased Tregs in the white matter infarct. Treatment of the animals with FTY720 suppressed neuroinflammation and reduced the number of both CD4+ T cells and Tregs in the lesion, suggesting the importance of infiltration of these peripheral immune cells into the lesion in aggravation of neuroinflammation. Suppression of neuroinflammation by FTY720 per se, however, did not promote remyelination in the infarct. FTY720 treatment negated the increase in the number of Tregs by iVEC transplantation in the infarct, and attenuated remyelination promoted by transplanted iVECs, while it did not affect the number of oligodendrocyte lineage cells increased by iVEC transplantation. Transplantation of Tregs together with iVECs into FTY720-treated ischemic white matter did not affect the number of oligodendrocyte lineage cells, while it remarkably promoted myelin regeneration. CONCLUSIONS iVEC transplantation suppresses neuroinflammation, but suppression of neuroinflammation per se does not promote remyelination. Recruitment of Tregs by transplanted iVECs contributes significantly to promotion of remyelination in the injured white matter.
Collapse
Affiliation(s)
- Bin Xu
- grid.256642.10000 0000 9269 4097Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, 3-39-22 Showa-Machi, Maebashi, Gunma 371-8511 Japan ,grid.452661.20000 0004 1803 6319Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Hiroya Shimauchi-Ohtaki
- grid.256642.10000 0000 9269 4097Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Gunma Japan
| | - Yuhei Yoshimoto
- grid.256642.10000 0000 9269 4097Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Gunma Japan
| | - Tetsushi Sadakata
- grid.256642.10000 0000 9269 4097Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi, Gunma Japan
| | - Yasuki Ishizaki
- grid.256642.10000 0000 9269 4097Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, 3-39-22 Showa-Machi, Maebashi, Gunma 371-8511 Japan
| |
Collapse
|
26
|
Qin X, Yi S, Rong J, Lu H, Ji B, Zhang W, Ding R, Wu L, Chen Z. Identification of anoikis-related genes classification patterns and immune infiltration characterization in ischemic stroke based on machine learning. Front Aging Neurosci 2023; 15:1142163. [PMID: 37032832 PMCID: PMC10076550 DOI: 10.3389/fnagi.2023.1142163] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Ischemic stroke (IS) is a type of stroke that leads to high mortality and disability. Anoikis is a form of programmed cell death. When cells detach from the correct extracellular matrix, anoikis disrupts integrin junctions, thus preventing abnormal proliferating cells from growing or attaching to an inappropriate matrix. Although there is growing evidence that anoikis regulates the immune response, which makes a great contribution to the development of IS, the role of anoikis in the pathogenesis of IS is rarely explored. Methods First, we downloaded GSE58294 set and GSE16561 set from the NCBI GEO database. And 35 anoikis-related genes (ARGs) were obtained from GSEA website. The CIBERSORT algorithm was used to estimate the relative proportions of 22 infiltrating immune cell types. Next, consensus clustering method was used to classify ischemic stroke samples. In addition, we used least absolute shrinkage and selection operator (LASSO), support vector machine-recursive feature elimination (SVM-RFE) and random forest (RF) algorithms to screen the key ARGs in ischemic stroke. Next, we performed receiver operating characteristics (ROC) analysis to assess the accuracy of each diagnostic gene. At the same time, the nomogram was constructed to diagnose IS by integrating trait genes. Then, we analyzed the correlation between gene expression and immune cell infiltration of the diagnostic genes in the combined database. And gene ontology (GO) and kyoto encyclopedia of genes and genomes (KEGG) analysis were performed on these genes to explore differential signaling pathways and potential functions, as well as the construction and visualization of regulatory networks using NetworkAnalyst and Cytoscape. Finally, we investigated the expression pattern of ARGs in IS patients across age or gender. Results Our study comprehensively analyzed the role of ARGs in IS for the first time. We revealed the expression profile of ARGs in IS and the correlation with infiltrating immune cells. And The results of consensus clustering analysis suggested that we can classify IS patients into two clusters. The machine learning analysis screened five signature genes, including AKT1, BRMS1, PTRH2, TFDP1 and TLE1. We also constructed nomogram models based on the five risk genes and evaluated the immune infiltration correlation, gene-miRNA, gene-TF and drug-gene interaction regulatory networks of these signature genes. The expression of ARGs did not differ by sex or age. Discussion This study may provide a beneficial reference for further elucidating the pathogenesis of IS, and render new ideas for drug screening, individualized therapy and immunotherapy of IS.
Collapse
Affiliation(s)
- Xiaohong Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shangfeng Yi
- Department of Neurosurgery, Enshi Center Hospital, Enshi, Hubei, China
| | - Jingtong Rong
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Haoran Lu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Baowei Ji
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wenfei Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Rui Ding
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Liquan Wu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- *Correspondence: Liquan Wu,
| | - Zhibiao Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Zhibiao Chen,
| |
Collapse
|
27
|
Weber RZ, Mulders G, Perron P, Tackenberg C, Rust R. Molecular and anatomical roadmap of stroke pathology in immunodeficient mice. Front Immunol 2022; 13:1080482. [PMID: 36569903 PMCID: PMC9785704 DOI: 10.3389/fimmu.2022.1080482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Background Stroke remains a leading cause of disability and death worldwide. It has become apparent that inflammation and immune mediators have a pre-dominant role in initial tissue damage and long-term recovery. Still, different immunosuppressed mouse models are necessary in stroke research e.g., to evaluate therapies using human cell grafts. Despite mounting evidence delineating the importance of inflammation in the stroke pathology, it is poorly described to what extent immune deficiency influences overall stroke outcome. Methods Here, we assessed the stroke pathology of popular genetic immunodeficient mouse models, i.e., NOD scid gamma (NSG) and recombination activating gene 2 (Rag2-/-) mice as well as pharmacologically immunosuppressed mice and compared them to immune competent, wildtype (WT) C57BL/6J mice three weeks after injury. We performed histology, gene expression, blood serum and behavioural analysis to identify the impact of immunosuppression on stroke progression. Results We detected changes in microglia activation/macrophage infiltration, scar-forming and vascular repair in immune-suppressed mice three weeks after injury. Transcriptomic analysis of stroked tissue revealed the strongest deviation from WT was observed in NSG mice affecting immunological and angiogenic pathways. Pharmacological immunosuppression resulted in the least variation in gene expression compared with the WT. These anatomical and genetic changes did not affect functional recovery in a time course of three weeks. To determine whether timing of immunosuppression is critical, we compared mice with acute and delayed pharmacological immunosuppression after stroke. Mice with delayed immunosuppression (7d) showed increased inflammatory and scarring responses compared to animals acutely treated with tacrolimus, thus more closely resembling WT pathology. Transplantation of human cells in the brains of immunosuppressed mice led to prolonged cell survival in all immunosuppressed mouse models, which was most consistent in NSG and Rag2-/- mice. Conclusions We detected distinct anatomical and molecular changes in the stroke pathology between individual immunosuppressed mouse models that should be considered when selecting an appropriate mouse model for stroke research.
Collapse
Affiliation(s)
- Rebecca Z. Weber
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland,Neuroscience Center Zurich, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Geertje Mulders
- Department of Health Sciences and Technology, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Patrick Perron
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
| | - Christian Tackenberg
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland,Neuroscience Center Zurich, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Ruslan Rust
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland,Neuroscience Center Zurich, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland,*Correspondence: Ruslan Rust,
| |
Collapse
|
28
|
The immunopathology of B lymphocytes during stroke-induced injury and repair. Semin Immunopathol 2022:10.1007/s00281-022-00971-3. [PMID: 36446955 PMCID: PMC9708141 DOI: 10.1007/s00281-022-00971-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/28/2022] [Indexed: 11/30/2022]
Abstract
B cells, also known as B lymphocytes or lymphoid lineage cells, are a historically understudied cell population with regard to brain-related injuries and diseases. However, an increasing number of publications have begun to elucidate the different phenotypes and roles B cells can undertake during central nervous system (CNS) pathology, including following ischemic and hemorrhagic stroke. B cell phenotype is intrinsically linked to function following stroke, as they may be beneficial or detrimental depending on the subset, timing, and microenvironment. Factors such as age, sex, and presence of co-morbidity also influence the behavior of post-stroke B cells. The following review will briefly describe B cells from origination to senescence, explore B cell function by integrating decades of stroke research, differentiate between the known B cell subtypes and their respective activity, discuss some of the physiological influences on B cells as well as the influence of B cells on certain physiological functions, and highlight the differences between B cells in healthy and disease states with particular emphasis in the context of ischemic stroke.
Collapse
|
29
|
Shi S, Zhang Q, Qu C, Tang Y, Qu Y, Wen S, Sun R, Pan Y. Identification of pyroptosis-related immune signature and drugs for ischemic stroke. Front Genet 2022; 13:909482. [PMID: 36238162 PMCID: PMC9552296 DOI: 10.3389/fgene.2022.909482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/23/2022] [Indexed: 11/19/2022] Open
Abstract
Background: Ischemic stroke (IS) is a common and serious neurological disease, and multiple pathways of cell apoptosis are implicated in its pathogenesis. Recently, extensive studies have indicated that pyroptosis is involved in various diseases, especially cerebrovascular diseases. However, the exact mechanism of interaction between pyroptosis and IS is scarcely understood. Thus, we aimed to investigate the impact of pyroptosis on IS-mediated systemic inflammation. Methods: First, the RNA regulation patterns mediated by 33 pyroptosis-related genes identified in 20 IS samples and 20 matched-control samples were systematically evaluated. Second, a series of bioinformatics algorithms were used to investigate the contribution of PRGs to IS pathogenesis. We determined three composition classifiers of PRGs which potentially distinguished healthy samples from IS samples according to the risk score using single-variable logistic regression, LASSO-Cox regression, and multivariable logistic regression analyses. Third, 20 IS patients were classified by unsupervised consistent cluster analysis in relation to pyroptosis. The association between pyroptosis and systemic inflammation characteristics was explored, which was inclusive of immune reaction gene sets, infiltrating immunocytes and human leukocyte antigen genes. Results: We identified that AIM2, SCAF11, and TNF can regulate immuno-inflammatory responses after strokes via the production of inflammatory factors and activation of the immune cells. Meanwhile, we identified distinct expression patterns mediated by pyroptosis and revealed their immune characteristics, differentially expressed genes, signaling pathways, and target drugs. Conclusion: Our findings lay a foundation for further research on pyroptosis and IS systemic inflammation, to improve IS prognosis and its responses to immunotherapy.
Collapse
Affiliation(s)
- Shanshan Shi
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgey Ministry of Education, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Qi Zhang
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Changda Qu
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yushi Tang
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yewei Qu
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Shirong Wen
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
- *Correspondence: Shirong Wen, ; Ruohan Sun, ; Yujun Pan,
| | - Ruohan Sun
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
- *Correspondence: Shirong Wen, ; Ruohan Sun, ; Yujun Pan,
| | - Yujun Pan
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
- *Correspondence: Shirong Wen, ; Ruohan Sun, ; Yujun Pan,
| |
Collapse
|
30
|
Korf JM, Honarpisheh P, Mohan EC, Banerjee A, Blasco-Conesa MP, Honarpisheh P, Guzman GU, Khan R, Ganesh BP, Hazen AL, Lee J, Kumar A, McCullough LD, Chauhan A. CD11b high B Cells Increase after Stroke and Regulate Microglia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:288-300. [PMID: 35732342 PMCID: PMC9446461 DOI: 10.4049/jimmunol.2100884] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 04/22/2022] [Indexed: 06/02/2023]
Abstract
Recent studies have highlighted the deleterious contributions of B cells to post-stroke recovery and cognitive decline. Different B cell subsets have been proposed on the basis of expression levels of transcription factors (e.g., T-bet) as well as specific surface proteins. CD11b (α-chain of integrin) is expressed by several immune cell types and is involved in regulation of cell motility, phagocytosis, and other essential functions of host immunity. Although B cells express CD11b, the CD11bhigh subset of B cells has not been well characterized, especially in immune dysregulation seen with aging and after stroke. Here, we investigate the role of CD11bhigh B cells in immune responses after stroke in young and aged mice. We evaluated the ability of CD11bhigh B cells to influence pro- and anti-inflammatory phenotypes of young and aged microglia (MG). We hypothesized that CD11bhigh B cells accumulate in the brain and contribute to neuroinflammation in aging and after stroke. We found that CD11bhigh B cells are a heterogeneous subpopulation of B cells predominantly present in naive aged mice. Their frequency increases in the brain after stroke in young and aged mice. Importantly, CD11bhigh B cells regulate MG phenotype and increase MG phagocytosis in both ex vivo and in vivo settings, likely by production of regulatory cytokines (e.g., TNF-α). As both APCs and adaptive immune cells with long-term memory function, B cells are uniquely positioned to regulate acute and chronic phases of the post-stroke immune response, and their influence is subset specific.
Collapse
Affiliation(s)
- Janelle M Korf
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX
- University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX
| | - Pedram Honarpisheh
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX
- University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX
| | - Eric C Mohan
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX
| | - Anik Banerjee
- University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX
| | | | - Parisa Honarpisheh
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX
| | - Gary U Guzman
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX
| | - Romeesa Khan
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX
| | - Bhanu P Ganesh
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX
| | - Amy L Hazen
- University of Texas McGovern Medical School, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, Houston, TX
| | - Juneyoung Lee
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX
| | - Aditya Kumar
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX
| | - Louise D McCullough
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX
| | - Anjali Chauhan
- Department of Neurology, University of Texas McGovern Medical School, Houston, TX;
| |
Collapse
|
31
|
Mao R, Zong N, Hu Y, Chen Y, Xu Y. Neuronal Death Mechanisms and Therapeutic Strategy in Ischemic Stroke. Neurosci Bull 2022; 38:1229-1247. [PMID: 35513682 PMCID: PMC9554175 DOI: 10.1007/s12264-022-00859-0] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/18/2022] [Indexed: 12/17/2022] Open
Abstract
Ischemic stroke caused by intracranial vascular occlusion has become increasingly prevalent with considerable mortality and disability, which gravely burdens the global economy. Current relatively effective clinical treatments are limited to intravenous alteplase and thrombectomy. Even so, patients still benefit little due to the short therapeutic window and the risk of ischemia/reperfusion injury. It is therefore urgent to figure out the neuronal death mechanisms following ischemic stroke in order to develop new neuroprotective strategies. Regarding the pathogenesis, multiple pathological events trigger the activation of cell death pathways. Particular attention should be devoted to excitotoxicity, oxidative stress, and inflammatory responses. Thus, in this article, we first review the principal mechanisms underlying neuronal death mediated by these significant events, such as intrinsic and extrinsic apoptosis, ferroptosis, parthanatos, pyroptosis, necroptosis, and autophagic cell death. Then, we further discuss the possibility of interventions targeting these pathological events and summarize the present pharmacological achievements.
Collapse
Affiliation(s)
- Rui Mao
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Ningning Zong
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Yujie Hu
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Ying Chen
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Yun Xu
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China.
- The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, 210008, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China.
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, China.
- Nanjing Neurology Clinic Medical Center, Nanjing, 210008, China.
| |
Collapse
|
32
|
Endres M, Moro MA, Nolte CH, Dames C, Buckwalter MS, Meisel A. Immune Pathways in Etiology, Acute Phase, and Chronic Sequelae of Ischemic Stroke. Circ Res 2022; 130:1167-1186. [PMID: 35420915 DOI: 10.1161/circresaha.121.319994] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inflammation and immune mechanisms are crucially involved in the pathophysiology of the development, acute damage cascades, and chronic course after ischemic stroke. Atherosclerosis is an inflammatory disease, and, in addition to classical risk factors, maladaptive immune mechanisms lead to an increased risk of stroke. Accordingly, individuals with signs of inflammation or corresponding biomarkers have an increased risk of stroke. Anti-inflammatory drugs, such as IL (interleukin)-1β blockers, methotrexate, or colchicine, represent attractive treatment strategies to prevent vascular events and stroke. Lately, the COVID-19 pandemic shows a clear association between SARS-CoV2 infections and increased risk of cerebrovascular events. Furthermore, mechanisms of both innate and adaptive immune systems influence cerebral damage cascades after ischemic stroke. Neutrophils, monocytes, and microglia, as well as T and B lymphocytes each play complex interdependent roles that synergize to remove dead tissue but also can cause bystander injury to intact brain cells and generate maladaptive chronic inflammation. Chronic systemic inflammation and comorbid infections may unfavorably influence both outcome after stroke and recurrence risk for further stroke. In addition, stroke triggers specific immune depression, which in turn can promote infections. Recent research is now increasingly addressing the question of the extent to which immune mechanisms may influence long-term outcome after stroke and, in particular, cause specific complications such as poststroke dementia or even poststroke depression.
Collapse
Affiliation(s)
- Matthias Endres
- Klinik für Neurologie mit Experimenteller Neurologie (M.E., C.H.N., A.M.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany.,Center for Stroke Research Berlin (M.E., C.H.N., C.D., A.M.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany.,Excellence Cluster NeuroCure (M.E.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany.,German Center for Neurodegenerative Diseases, Partner Site Berlin, Germany (M.E.).,German Centre for Cardiovascular Research, Partner Site Berlin, Germany (M.E., C.H.N.)
| | - Maria A Moro
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (M.A.M.).,Departamento de Farmacología yToxicología, Unidad de Investigación Neurovascular, Universidad Complutense de Madrid, Madrid, Spain (M.A.M.).,Instituto Universitario de Investigación en Neuroquímica, UCM, Madrid, Spain (M.A.M.).,Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain (M.A.M.)
| | - Christian H Nolte
- Klinik für Neurologie mit Experimenteller Neurologie (M.E., C.H.N., A.M.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany.,Center for Stroke Research Berlin (M.E., C.H.N., C.D., A.M.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Germany (M.E., C.H.N.)
| | - Claudia Dames
- Center for Stroke Research Berlin (M.E., C.H.N., C.D., A.M.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany.,Institute for Medical Immunology (C.D.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, CA (M.S.B.).,Wu Tsai Neurosciences Institute, Stanford University, CA (M.S.B.)
| | - Andreas Meisel
- Klinik für Neurologie mit Experimenteller Neurologie (M.E., C.H.N., A.M.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany.,Center for Stroke Research Berlin (M.E., C.H.N., C.D., A.M.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany.,NeuroCure Clinical Research Center (A.M.), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| |
Collapse
|
33
|
Becktel DA, Zbesko JC, Frye JB, Chung AG, Hayes M, Calderon K, Grover JW, Li A, Garcia FG, Tavera-Garcia MA, Schnellmann RG, Wu HJJ, Nguyen TVV, Doyle KP. Repeated Administration of 2-Hydroxypropyl-β-Cyclodextrin (HPβCD) Attenuates the Chronic Inflammatory Response to Experimental Stroke. J Neurosci 2022; 42:325-348. [PMID: 34819339 PMCID: PMC8802936 DOI: 10.1523/jneurosci.0933-21.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/24/2021] [Accepted: 11/16/2021] [Indexed: 11/30/2022] Open
Abstract
Globally, more than 67 million people are living with the effects of ischemic stroke. Importantly, many stroke survivors develop a chronic inflammatory response that may contribute to cognitive impairment, a common and debilitating sequela of stroke that is insufficiently studied and currently untreatable. 2-Hydroxypropyl-β-cyclodextrin (HPβCD) is an FDA-approved cyclic oligosaccharide that can solubilize and entrap lipophilic substances. The goal of the present study was to determine whether the repeated administration of HPβCD curtails the chronic inflammatory response to stroke by reducing lipid accumulation within stroke infarcts in a distal middle cerebral artery occlusion mouse model of stroke. To achieve this goal, we subcutaneously injected young adult and aged male mice with vehicle or HPβCD 3 times per week, with treatment beginning 1 week after stroke. We evaluated mice at 7 weeks following stroke using immunostaining, RNA sequencing, lipidomic, and behavioral analyses. Chronic stroke infarct and peri-infarct regions of HPβCD-treated mice were characterized by an upregulation of genes involved in lipid metabolism and a downregulation of genes involved in innate and adaptive immunity, reactive astrogliosis, and chemotaxis. Correspondingly, HPβCD reduced the accumulation of lipid droplets, T lymphocytes, B lymphocytes, and plasma cells in stroke infarcts. Repeated administration of HPβCD also preserved NeuN immunoreactivity in the striatum and thalamus and c-Fos immunoreactivity in hippocampal regions. Additionally, HPβCD improved recovery through the protection of hippocampal-dependent spatial working memory and reduction of impulsivity. These results indicate that systemic HPβCD treatment following stroke attenuates chronic inflammation and secondary neurodegeneration and prevents poststroke cognitive decline.SIGNIFICANCE STATEMENT Dementia is a common and debilitating sequela of stroke. Currently, there are no available treatments for poststroke dementia. Our study shows that lipid metabolism is disrupted in chronic stroke infarcts, which causes an accumulation of uncleared lipid debris and correlates with a chronic inflammatory response. To our knowledge, these substantial changes in lipid homeostasis have not been previously recognized or investigated in the context of ischemic stroke. We also provide a proof of principle that solubilizing and entrapping lipophilic substances using HPβCD could be an effective strategy for treating chronic inflammation after stroke and other CNS injuries. We propose that using HPβCD for the prevention of poststroke dementia could improve recovery and increase long-term quality of life in stroke sufferers.
Collapse
Affiliation(s)
- Danielle A Becktel
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Jacob C Zbesko
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Jennifer B Frye
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Amanda G Chung
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Megan Hayes
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Kylie Calderon
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Jeffrey W Grover
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona 85719
| | - Anna Li
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
- Arizona Arthritis Center, University of Arizona, Tucson, Arizona 85719
| | - Frankie G Garcia
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | | | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85719
| | - Hsin-Jung Joyce Wu
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
- Arizona Arthritis Center, University of Arizona, Tucson, Arizona 85719
| | - Thuy-Vi V Nguyen
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
- Department of Neurology, University of Arizona, Tucson, Arizona 85719
| | - Kristian P Doyle
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
- Department of Neurology, University of Arizona, Tucson, Arizona 85719
- BIO5 Institute, University of Arizona, Tucson, Arizona 85719
- Arizona Center on Aging, University of Arizona, Tucson, Arizona 85719
- Department of Psychology, University of Arizona, Tucson, Arizona 85719
- Department of Neurosurgery, University of Arizona, Tucson, Arizona 85719
| |
Collapse
|
34
|
Hermann DM, Liesz A, Dzyubenko E. Implications of immune responses for ischemic brain injury and stroke recovery. Brain Behav Immun 2021; 96:292-294. [PMID: 34023355 DOI: 10.1016/j.bbi.2021.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Dirk M Hermann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, Ludwig Maximilians University, Munich and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Egor Dzyubenko
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
35
|
Selvaraj UM, Ujas TA, Kong X, Kumar A, Plautz EJ, Zhang S, Xing C, Sudduth TL, Wilcock DM, Turchan-Cholewo J, Goldberg MP, Stowe AM. Delayed diapedesis of CD8 T cells contributes to long-term pathology after ischemic stroke in male mice. Brain Behav Immun 2021; 95:502-513. [PMID: 33964435 PMCID: PMC8221572 DOI: 10.1016/j.bbi.2021.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 04/14/2021] [Accepted: 05/03/2021] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Stroke is a debilitating disorder with significant annual mortality and morbidity rates worldwide. Immune cells are recruited to the injured brain within hours after stroke onset and can exhibit either protective or detrimental effects on recovery. However, immune cells, including CD8 T cells, persist in the injured brain for weeks, suggesting a longer-term role for the adaptive immune system during functional recovery. The aim of this study was to determine if the delayed secondary diapedesis of CD8 T cells into the ischemic brain negatively impacts functional recovery after transient ischemic stroke in male mice. RESULTS Mice exhibited an increased number of leukocytes in the ipsilesional hemispheres at 14 days (3-fold; p < 0.001) and 30 days (2.2-fold; p = 0.02) after transient middle cerebral artery occlusion (tMCAo) compared to 8 days post-tMCAo, at which time acute neuroinflammation predominantly resolves. Moreover, mice with higher ipsilesional CD8 T cells at 30 days (R2 = 0.52, p < 0.01) exhibited worse functional recovery. To confirm a detrimental role of chronic CD8 T cell diapedesis on recovery, peripheral CD8 T cells were depleted beginning 10 days post-tMCAo. Delayed CD8 T cell depletion improved motor recovery on the rotarod (F(1,28) = 4.264; p = 0.048) compared to isotype control-treated mice. CD8 T cell-depleted mice also exhibited 2-fold (p < 0.001) reduced leukocyte infiltration at 30 days post-tMCAo. Specifically, macrophage, neutrophil, and CD4 T cell numbers were reduced in the ipsilesional hemisphere of the CD8 T cell-depleted mice independent of inflammatory status of the post-stroke CNS (e.g. microglial phenotype and cytokine production). RNAseq identified a unique profile for brain infiltrating CD8 T cells at 30 days post-tMCAo, with 46 genes differentially expressed relative to CD8 T cells at 3 days post-tMCAo. CONCLUSION Our data reveal a role for CD8 T cells in the chronic phase post-stroke that can be therapeutically targeted. We demonstrate long-term CD8 T cell recruitment into the ipsilesional hemisphere that affects both immune cell numbers present in the injured brain and functional recovery through one month after stroke onset.
Collapse
Affiliation(s)
- Uma Maheswari Selvaraj
- Department of Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Thomas A Ujas
- Department of Neurology, Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, United States
| | - Xiangmei Kong
- Department of Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ashwani Kumar
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Erik J Plautz
- Department of Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Shanrong Zhang
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, United States; Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, United States; Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Tiffany L Sudduth
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Donna M Wilcock
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Jadwiga Turchan-Cholewo
- Department of Neurology, Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, United States
| | - Mark P Goldberg
- Department of Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ann M Stowe
- Department of Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States; Department of Neurology, Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
36
|
Domblides C, Rochefort J, Riffard C, Panouillot M, Lescaille G, Teillaud JL, Mateo V, Dieu-Nosjean MC. Tumor-Associated Tertiary Lymphoid Structures: From Basic and Clinical Knowledge to Therapeutic Manipulation. Front Immunol 2021; 12:698604. [PMID: 34276690 PMCID: PMC8279885 DOI: 10.3389/fimmu.2021.698604] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022] Open
Abstract
The tumor microenvironment is a complex ecosystem almost unique to each patient. Most of available therapies target tumor cells according to their molecular characteristics, angiogenesis or immune cells involved in tumor immune-surveillance. Unfortunately, only a limited number of patients benefit in the long-term of these treatments that are often associated with relapses, in spite of the remarkable progress obtained with the advent of immune checkpoint inhibitors (ICP). The presence of “hot” tumors is a determining parameter for selecting therapies targeting the patient immunity, even though some of them still do not respond to treatment. In human studies, an in-depth analysis of the organization and interactions of tumor-infiltrating immune cells has revealed the presence of an ectopic lymphoid organization termed tertiary lymphoid structures (TLS) in a large number of tumors. Their marked similarity to secondary lymphoid organs has suggested that TLS are an “anti-tumor school” and an “antibody factory” to fight malignant cells. They are effectively associated with long-term survival in most solid tumors, and their presence has been recently shown to predict response to ICP inhibitors. This review discusses the relationship between TLS and the molecular characteristics of tumors and the presence of oncogenic viruses, as well as their role when targeted therapies are used. Also, we present some aspects of TLS biology in non-tumor inflammatory diseases and discuss the putative common characteristics that they share with tumor-associated TLS. A detailed overview of the different pre-clinical models available to investigate TLS function and neogenesis is also presented. Finally, new approaches aimed at a better understanding of the role and function of TLS such as the use of spheroids and organoids and of artificial intelligence algorithms, are also discussed. In conclusion, increasing our knowledge on TLS will undoubtedly improve prognostic prediction and treatment selection in cancer patients with key consequences for the next generation immunotherapy.
Collapse
Affiliation(s)
- Charlotte Domblides
- Faculté de Médecine Sorbonne Université, Sorbonne Université, UMRS 1135, Paris, France.,Faculté de Médecine Sorbonne Université, INSERM U1135, Paris, France.,Laboratory "Immune microenvironment and immunotherapy", Centre d'Immunologie et des Maladies Infectieuses Paris (CIMI-Paris), Paris, France
| | - Juliette Rochefort
- Faculté de Médecine Sorbonne Université, Sorbonne Université, UMRS 1135, Paris, France.,Faculté de Médecine Sorbonne Université, INSERM U1135, Paris, France.,Laboratory "Immune microenvironment and immunotherapy", Centre d'Immunologie et des Maladies Infectieuses Paris (CIMI-Paris), Paris, France.,Université de Paris, Faculté de Santé, UFR Odontologie, Paris, France.,Service Odontologie, Assistance Publique Hôpitaux de Paris (AP-HP), La Pitié-Salpêtrière, Paris, France
| | - Clémence Riffard
- Faculté de Médecine Sorbonne Université, Sorbonne Université, UMRS 1135, Paris, France.,Faculté de Médecine Sorbonne Université, INSERM U1135, Paris, France.,Laboratory "Immune microenvironment and immunotherapy", Centre d'Immunologie et des Maladies Infectieuses Paris (CIMI-Paris), Paris, France
| | - Marylou Panouillot
- Faculté de Médecine Sorbonne Université, Sorbonne Université, UMRS 1135, Paris, France.,Faculté de Médecine Sorbonne Université, INSERM U1135, Paris, France.,Laboratory "Immune microenvironment and immunotherapy", Centre d'Immunologie et des Maladies Infectieuses Paris (CIMI-Paris), Paris, France
| | - Géraldine Lescaille
- Faculté de Médecine Sorbonne Université, Sorbonne Université, UMRS 1135, Paris, France.,Faculté de Médecine Sorbonne Université, INSERM U1135, Paris, France.,Laboratory "Immune microenvironment and immunotherapy", Centre d'Immunologie et des Maladies Infectieuses Paris (CIMI-Paris), Paris, France.,Université de Paris, Faculté de Santé, UFR Odontologie, Paris, France.,Service Odontologie, Assistance Publique Hôpitaux de Paris (AP-HP), La Pitié-Salpêtrière, Paris, France
| | - Jean-Luc Teillaud
- Faculté de Médecine Sorbonne Université, Sorbonne Université, UMRS 1135, Paris, France.,Faculté de Médecine Sorbonne Université, INSERM U1135, Paris, France.,Laboratory "Immune microenvironment and immunotherapy", Centre d'Immunologie et des Maladies Infectieuses Paris (CIMI-Paris), Paris, France
| | - Véronique Mateo
- Faculté de Médecine Sorbonne Université, Sorbonne Université, UMRS 1135, Paris, France.,Faculté de Médecine Sorbonne Université, INSERM U1135, Paris, France.,Laboratory "Immune microenvironment and immunotherapy", Centre d'Immunologie et des Maladies Infectieuses Paris (CIMI-Paris), Paris, France
| | - Marie-Caroline Dieu-Nosjean
- Faculté de Médecine Sorbonne Université, Sorbonne Université, UMRS 1135, Paris, France.,Faculté de Médecine Sorbonne Université, INSERM U1135, Paris, France.,Laboratory "Immune microenvironment and immunotherapy", Centre d'Immunologie et des Maladies Infectieuses Paris (CIMI-Paris), Paris, France
| |
Collapse
|
37
|
Zbesko JC, Frye JB, Becktel DA, Gerardo DK, Stokes J, Calderon K, Nguyen TVV, Bhattacharya D, Doyle KP. IgA natural antibodies are produced following T-cell independent B-cell activation following stroke. Brain Behav Immun 2021; 91:578-586. [PMID: 32956832 PMCID: PMC8279117 DOI: 10.1016/j.bbi.2020.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022] Open
Abstract
Up to 30% of stroke patients experience cognitive decline within one year of their stroke. There are currently no FDA-approved drugs that can prevent post-stroke cognitive decline, in part due to a poor understanding of the mechanisms involved. We have previously demonstrated that a B-lymphocyte response to stroke, marked by IgA + cells, can cause delayed cognitive dysfunction in mice and that a similar adaptive immune response occurs in the brains of some human stroke patients that suffer from vascular dementia. The stimuli which trigger B-lymphocyte activation following stroke, and their target antigens, are still unknown. Therefore, to learn more about the mechanisms by which B-lymphocytes become activated following stroke we first characterized the temporal kinetics of the B-lymphocyte, T-lymphocyte, and plasma cell (PC) response to stroke in the brain by immunohistochemistry (IHC). We discovered that B-lymphocyte, T-lymphocyte, and plasma cell infiltration within the infarct progressively increases between 2 and 7 weeks after stroke. We then compared the B-lymphocyte response to stroke in WT, MHCII-/-, CD4-/-, and MyD88-/- mice to determine if B-lymphocytes mature into IgA + PCs through a T-lymphocyte and MyD88 dependent mechanism. Our data from a combination of IHC and flow cytometry indicate that following stroke, a population of IgA + PCs develops independently of CD4 + helper T-lymphocytes and MyD88 signaling. Subsequent sequencing of immunoglobulin genes of individual IgA + PCs present within the infarct identified a novel population of natural antibodies with few somatic mutations in complementarity-determining regions. These findings indicate that a population of IgA + PCs develops in the infarct following stroke by B-lymphocytes interacting with one or more thymus independent type 2 (TI-2) antigens, and that they produce IgA natural antibodies.
Collapse
Affiliation(s)
- Jacob C. Zbesko
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | | | | | - Diana K. Gerardo
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | - Jessica Stokes
- Department of Pediatrics, University of Arizona, Tucson, AZ 85719, USA
| | - Kylie Calderon
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA
| | - Thuy-Vi V. Nguyen
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA,Department of Neurology, University of Arizona, Tucson, AZ 85719, USA
| | | | - Kristian P. Doyle
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719, USA,Department of Neurology, University of Arizona, Tucson, AZ 85719, USA,Arizona Center on Aging, University of Arizona, Tucson, AZ 85719, USA,Corresponding author at: Department of Immunobiology, University of Arizona, Tucson, 1656 E. Mabel Street, Tucson, Arizona 85719, USA. (K.P. Doyle)
| |
Collapse
|
38
|
Honarpisheh P, Lee J, Banerjee A, Blasco-Conesa MP, Honarpisheh P, d'Aigle J, Mamun AA, Ritzel RM, Chauhan A, Ganesh BP, McCullough LD. Potential caveats of putative microglia-specific markers for assessment of age-related cerebrovascular neuroinflammation. J Neuroinflammation 2020; 17:366. [PMID: 33261619 PMCID: PMC7709276 DOI: 10.1186/s12974-020-02019-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/29/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The ability to distinguish resident microglia from infiltrating myeloid cells by flow cytometry-based surface phenotyping is an important technique for examining age-related neuroinflammation. The most commonly used surface markers for the identification of microglia include CD45 (low-intermediate expression), CD11b, Tmem119, and P2RY12. METHODS In this study, we examined changes in expression levels of these putative microglia markers in in vivo animal models of stroke, cerebral amyloid angiopathy (CAA), and aging as well as in an ex vivo LPS-induced inflammation model. RESULTS We demonstrate that Tmem119 and P2RY12 expression is evident within both CD45int and CD45high myeloid populations in models of stroke, CAA, and aging. Interestingly, LPS stimulation of FACS-sorted adult microglia suggested that these brain-resident myeloid cells can upregulate CD45 and downregulate Tmem119 and P2RY12, making them indistinguishable from peripherally derived myeloid populations. Importantly, our findings show that these changes in the molecular signatures of microglia can occur without a contribution from the other brain-resident or peripherally sourced immune cells. CONCLUSION We recommend future studies approach microglia identification by flow cytometry with caution, particularly in the absence of the use of a combination of markers validated for the specific neuroinflammation model of interest. The subpopulation of resident microglia residing within the "infiltrating myeloid" population, albeit small, may be functionally important in maintaining immune vigilance in the brain thus should not be overlooked in neuroimmunological studies.
Collapse
Affiliation(s)
- Pedram Honarpisheh
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA.,UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Juneyoung Lee
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - Anik Banerjee
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA.,UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Maria P Blasco-Conesa
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - Parisa Honarpisheh
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - John d'Aigle
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - Abdullah A Mamun
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - Rodney M Ritzel
- Department of Anesthesiology, Center for Shock, Trauma, and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Anjali Chauhan
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - Bhanu P Ganesh
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA
| | - Louise D McCullough
- Department of Neurology, University of Texas John P. and Kathrine G. McGovern Medical School, Houston, TX, USA.
| |
Collapse
|