101
|
Qin L, Kamash P, Yang Y, Ding Y, Ren C. A narrative review of potential neural repair poststroke: Decoction of Chinese angelica and peony in regulating microglia polarization through the neurosteroid pathway. Brain Circ 2024; 10:5-10. [PMID: 38655444 PMCID: PMC11034443 DOI: 10.4103/bc.bc_45_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/22/2023] [Accepted: 07/13/2023] [Indexed: 04/26/2024] Open
Abstract
Ischemic stroke is a major global health crisis, characterized by high morbidity and mortality rates. Although there have been significant advancements in treating the acute phase of this condition, there remains a pressing need for effective treatments that can facilitate the recovery of neurological functions. Danggui-Shaoyao-San (DSS), also known as the Decoction of Chinese Angelica and Peony, is a traditional Chinese herbal formula. It has demonstrated promising results in the regulation of microglial polarization and modulation of neurosteroid receptor expression, which may make it a potent strategy for promoting the recovery of neurological functions. Microglia, which plays a crucial role in neuroplasticity and functional reconstruction poststroke, is regulated by neurosteroids. This review posits that DSS could facilitate the recovery of neuronal function poststroke by influencing microglial polarization through the neurosteroid receptor pathway. We will further discuss the potential mechanisms by which DSS could enhance neural function in stroke, including the regulation of microglial activation, neurosteroid regulation, and other potential mechanisms.
Collapse
Affiliation(s)
- Linhui Qin
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Peter Kamash
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yong Yang
- Department of Herbal Formula Science, Chinese Medicine College, Beijing University of Chinese Medicine, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
102
|
Jia J, Xu S, Hu J, Gan Y, Sun M, Xia S, Bao X, Zhang M, Xu Y. Growth arrest specific protein 6 alleviated white matter injury after experimental ischemic stroke. J Cereb Blood Flow Metab 2024; 44:77-93. [PMID: 37794790 PMCID: PMC10905636 DOI: 10.1177/0271678x231205078] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023]
Abstract
Ischemic white matter injury leads to long-term neurological deficits and lacks effective medication. Growth arrest specific protein 6 (Gas6) clears myelin debris, which is hypothesized to promote white matter integrity in experimental stroke models. By the middle cerebral artery occlusion (MCAO) stroke model, we observed that Gas6 reduced infarcted volume and behavior deficits 4 weeks after MCAO. Compared with control mice, Gas6-treatment mice represented higher FA values in the ipsilateral external capsules by MRI DTI scan. The SMI32/MBP ratio of the ipsilateral cortex and striatum was profoundly alleviated by Gas6 administration. Gas6-treatment group manifested thicker myelin sheaths than the control group by electron microscopy. We observed that Gas6 mainly promoted OPC maturation, which was closely related to microglia. Mechanically, Gas6 accelerated microglia-mediated myelin debris clearance and cholesterol transport protein expression (abca1, abcg1, apoc1, apoe) in vivo and in vitro, accordingly less myelin debris and lipid deposited in Gas6 treated stroke mice. HX531 (RXR inhibitor) administration mitigated the functions of Gas6 in speeding up debris clearance and cholesterol transport protein expression. Generally, we concluded that Gas6 cleared myelin debris and promoted cholesterol transportation protein expression through activating RXR, which could be one critical mechanism contributing to white matter repair after stroke.
Collapse
Affiliation(s)
- Junqiu Jia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Siyi Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Zhenjiang, China
| | - Jinglong Hu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yonghui Gan
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Sun
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meijuan Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Zhenjiang, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
- Jiangsu Provincial medical key discipline, Nanjing, China
| |
Collapse
|
103
|
Wang S, Li G, Liang X, Wu Z, Chen C, Zhang F, Niu J, Li X, Yan J, Wang N, Li J, Wang Y. Small Extracellular Vesicles Derived from Altered Peptide Ligand-Loaded Dendritic Cell Act as A Therapeutic Vaccine for Spinal Cord Injury Through Eliciting CD4 + T cell-Mediated Neuroprotective Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304648. [PMID: 38037457 PMCID: PMC10797491 DOI: 10.1002/advs.202304648] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/13/2023] [Indexed: 12/02/2023]
Abstract
The balance among different CD4+ T cell subsets is crucial for repairing the injured spinal cord. Dendritic cell (DC)-derived small extracellular vesicles (DsEVs) effectively activate T-cell immunity. Altered peptide ligands (APLs), derived from myelin basic protein (MBP), have been shown to affect CD4+ T cell subsets and reduce neuroinflammation levels. However, the application of APLs is challenging because of their poor stability and associated side effects. Herein, it is demonstrate that DsEVs can act as carriers for APL MBP87-99 A91 (A91-DsEVs) to induce the activation of 2 helper T (Th2) and regulatory T (Treg) cells for spinal cord injury (SCI) in mice. These stimulated CD4+ T cells can efficiently "home" to the lesion area and establish a beneficial microenvironment through inducing the activation of M2 macrophages/microglia, inhibiting the expression of inflammatory cytokines, and increasing the release of neurotrophic factors. The microenvironment mediated by A91-DsEVs may enhance axon regrowth, protect neurons, and promote remyelination, which may support the recovery of motor function in the SCI model mice. In conclusion, using A91-DsEVs as a therapeutic vaccine may help induce neuroprotective immunity in the treatment of SCI.
Collapse
Affiliation(s)
- Sikai Wang
- Department of Orthopedic SurgerySecond Affiliated Hospital of Harbin Medical UniversityNo. 246 Baojian RoadHarbin150086China
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and RegenerationThe Second Affiliated Hospital of Harbin Medical UniversityNo. 246 Baojian RoadHarbin150086China
| | - Guanglei Li
- Department of Orthopedic SurgerySecond Affiliated Hospital of Harbin Medical UniversityNo. 246 Baojian RoadHarbin150086China
| | - Xiongjie Liang
- Department of Orthopedic SurgerySecond Affiliated Hospital of Harbin Medical UniversityNo. 246 Baojian RoadHarbin150086China
| | - Zexuan Wu
- Department of Orthopedic SurgerySecond Affiliated Hospital of Harbin Medical UniversityNo. 246 Baojian RoadHarbin150086China
| | - Chao Chen
- Faculty of Medicine and DentistryUniversity of AlbertaEdmontonT5C 0T2Canada
| | - Fawang Zhang
- Department of Orthopedic SurgerySecond Affiliated Hospital of Harbin Medical UniversityNo. 246 Baojian RoadHarbin150086China
| | - Jiawen Niu
- Department of Orthopedic SurgerySecond Affiliated Hospital of Harbin Medical UniversityNo. 246 Baojian RoadHarbin150086China
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and RegenerationThe Second Affiliated Hospital of Harbin Medical UniversityNo. 246 Baojian RoadHarbin150086China
| | - Xuefeng Li
- Department of Orthopedic SurgerySecond Affiliated Hospital of Harbin Medical UniversityNo. 246 Baojian RoadHarbin150086China
| | - Jinglong Yan
- Department of Orthopedic SurgerySecond Affiliated Hospital of Harbin Medical UniversityNo. 246 Baojian RoadHarbin150086China
| | - Nanxiang Wang
- Department of Orthopedic SurgerySecond Affiliated Hospital of Harbin Medical UniversityNo. 246 Baojian RoadHarbin150086China
| | - Jing Li
- Department of Pathology and Electron MicroscopyFaculty of Basic Medical ScienceHarbin Medical UniversityNo. 157 Baojian RoadHarbin150086China
| | - Yufu Wang
- Department of Orthopedic SurgerySecond Affiliated Hospital of Harbin Medical UniversityNo. 246 Baojian RoadHarbin150086China
| |
Collapse
|
104
|
Church KA, Cardona AE, Hopp SC. Roles in Innate Immunity. ADVANCES IN NEUROBIOLOGY 2024; 37:263-286. [PMID: 39207697 DOI: 10.1007/978-3-031-55529-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia are best known as the resident phagocytes of the central nervous system (CNS). As a resident brain immune cell population, microglia play key roles during the initiation, propagation, and resolution of inflammation. The discovery of resident adaptive immune cells in the CNS has unveiled a relationship between microglia and adaptive immune cells for CNS immune-surveillance during health and disease. The interaction of microglia with elements of the peripheral immune system and other CNS resident cells mediates a fine balance between neuroprotection and tissue damage. In this chapter, we highlight the innate immune properties of microglia, with a focus on how pattern recognition receptors, inflammatory signaling cascades, phagocytosis, and the interaction between microglia and adaptive immune cells regulate events that initiate an inflammatory or neuroprotective response within the CNS that modulates immune-mediated disease exacerbation or resolution.
Collapse
Affiliation(s)
- Kaira A Church
- Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Astrid E Cardona
- Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Department of Pharmacology, Biggs Institute for Alzheimer's and Neurodegenerative Disease, The University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
105
|
Bormann D, Knoflach M, Poreba E, Riedl CJ, Testa G, Orset C, Levilly A, Cottereau A, Jauk P, Hametner S, Golabi B, Copic D, Klas K, Direder M, Kühtreiber H, Salek M, zur Nedden S, Baier-Bitterlich G, Kiechl S, Haider C, Endmayr V, Höftberger R, Ankersmit HJ, Mildner M. Single nucleus RNA sequencing reveals glial cell type-specific responses to ischemic stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.26.573302. [PMID: 38234821 PMCID: PMC10793395 DOI: 10.1101/2023.12.26.573302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Reactive neuroglia critically shape the braińs response to ischemic stroke. However, their phenotypic heterogeneity impedes a holistic understanding of the cellular composition and microenvironment of the early ischemic lesion. Here we generated a single cell resolution transcriptomics dataset of the injured brain during the acute recovery from permanent middle cerebral artery occlusion. This approach unveiled infarction and subtype specific molecular signatures in oligodendrocyte lineage cells and astrocytes, which ranged among the most transcriptionally perturbed cell types in our dataset. Specifically, we characterized and compared infarction restricted proliferating oligodendrocyte precursor cells (OPCs), mature oligodendrocytes and heterogeneous reactive astrocyte populations. Our analyses unveiled unexpected commonalities in the transcriptional response of oligodendrocyte lineage cells and astrocytes to ischemic injury. Moreover, OPCs and reactive astrocytes were involved in a shared immuno-glial cross talk with stroke specific myeloid cells. In situ, osteopontin positive myeloid cells accumulated in close proximity to proliferating OPCs and reactive astrocytes, which expressed the osteopontin receptor CD44, within the perilesional zone specifically. In vitro, osteopontin increased the migratory capacity of OPCs. Collectively, our study highlights molecular cross talk events which might govern the cellular composition and microenvironment of infarcted brain tissue in the early stages of recovery.
Collapse
Affiliation(s)
- Daniel Bormann
- Applied Immunology Laboratory, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Michael Knoflach
- Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
- VASCage, Research Centre on Vascular Ageing and Stroke, 6020 Innsbruck, Austria
| | - Emilia Poreba
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Christian J. Riedl
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Giulia Testa
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Cyrille Orset
- Normandie University, UNICAEN, ESR3P, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
| | - Anthony Levilly
- Normandie University, UNICAEN, ESR3P, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
| | - Andreá Cottereau
- Normandie University, UNICAEN, ESR3P, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
| | - Philipp Jauk
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Bahar Golabi
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Dragan Copic
- Applied Immunology Laboratory, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Katharina Klas
- Applied Immunology Laboratory, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Martin Direder
- Applied Immunology Laboratory, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Hannes Kühtreiber
- Applied Immunology Laboratory, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Melanie Salek
- Applied Immunology Laboratory, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Stephanie zur Nedden
- Institute of Neurobiochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Gabriele Baier-Bitterlich
- Institute of Neurobiochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Stefan Kiechl
- Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
- VASCage, Research Centre on Vascular Ageing and Stroke, 6020 Innsbruck, Austria
| | - Carmen Haider
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Verena Endmayr
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Hendrik J. Ankersmit
- Applied Immunology Laboratory, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
106
|
Zhou B. Bioinformatics analysis identifies potential m6A hub genes in the pathogenesis of intracerebral hemorrhage. J Neuroimmunol 2023; 385:578224. [PMID: 37907028 DOI: 10.1016/j.jneuroim.2023.578224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/14/2023] [Accepted: 10/24/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a type of stroke associated with a high rate of disability and mortality. The role of N6-methyladenosine (m6A) in ICH remains unclear. METHODS Screening of m6A DEGs by differentially expressed genes (DEGs) analysis. m6A hub genes in ICH were identified by protein-protein interaction (PPI) networks. Pearson correlation tests were used to explore the relationship between m6A hub genes and DNA methylation. m6A hub genes were examined by ROC curves for their ability to predict ICH occurrence. Immune cell infiltration and m6A hub gene correlation in ICH were analysed using the CIBERSORT algorithm. Construction of ceRNA networks and enrichment analysis by GO/KEGG. RESULTS A total of 12 m6A regulatory enzymes were differentially expressed after ICH. the PPI network screened three m6A hub genes, including YTHDF2, FTO and HNRNPA2B1. A high expression of YTHDF2 was associated with DNA hypomethylation after ICH and could better predict the development of ICH. yTHDF2 was associated with high infiltration of M1 macrophages after ICH. A ceRNA network was constructed based on the m6A central gene with target genes enriched in transcriptional regulation and the LKB1 signalling pathway. CONCLUSION M6A modifications are involved in the progression of ICH. YTHDF2, an m6A key gene, may regulate ICH progression by promoting infiltration of M1 macrophages or through the ceRNA network.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Neurosurgery, the First People's Hospital of Jiashan County, Jiaxing City, Zhejiang Province, PR China.
| |
Collapse
|
107
|
Lu J, Li H, Zhang G, Yang F, Zhang X, Ping A, Xu Z, Gu Y, Wang R, Ying D, Liu J, Zhang J, Shi L. Age-Related Alterations in Peripheral Immune Landscape with Magnified Impact on Post-Stroke Brain. RESEARCH (WASHINGTON, D.C.) 2023; 6:0287. [PMID: 38090608 PMCID: PMC10712880 DOI: 10.34133/research.0287] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/19/2023] [Indexed: 07/31/2024]
Abstract
Immunosenescence refers to the multifaceted and profound alterations in the immune system brought about by aging, exerting complex influences on the pathophysiological processes of diseases that manifest upon it. Using a combination of single-cell RNA sequencing, cytometry by time of flight, and various immunological assays, we investigated the characteristics of immunosenescence in the peripheral blood of aged mice and its impact on the cerebral immune environment after ischemic stroke. Our results revealed some features of immunosenescence. We observed an increase in neutrophil counts, concurrent with accelerated neutrophil aging, characterized by altered expression of aging-associated markers like CD62L and consequential changes in neutrophil-mediated immune functions. Monocytes/macrophages in aged mice exhibited enhanced antigen-presentation capabilities. T cell profiles shifted from naive to effector or memory states, with a specific rise in T helper 1 cells and T helper 17 cells subpopulations and increased regulatory T cell activation in CD4 T cells. Furthermore, regulatory CD8 T cells marked by Klra decreased with aging, while a subpopulation of exhausted-like CD8 T cells expanded, retaining potent immunostimulatory and proinflammatory functions. Critically, these inherent disparities not only persisted but were further amplified within the ischemic hemispheres following stroke. In summary, our comprehensive insights into the key attributes of peripheral immunosenescence provide a vital theoretical foundation for understanding not only ischemic strokes but also other age-associated diseases.
Collapse
Affiliation(s)
- Jianan Lu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Huaming Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Guoqiang Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Fan Yang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Xiaotao Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - An Ping
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Zhouhan Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Yichen Gu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Rui Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Dan Ying
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Jianjian Liu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
- Brain Research Institute,
Zhejiang University, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Brain Science,
Zhejiang University, Hangzhou, Zhejiang, China
| | - Ligen Shi
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
108
|
Xu C, Zhang Q, Zhang Y, Chen H, Tang T, Wang J, Xia S, Chen G, Zhang J. Lateralized response of skull bone marrow via osteopontin signaling in mice after ischemia reperfusion. J Neuroinflammation 2023; 20:294. [PMID: 38071333 PMCID: PMC10710724 DOI: 10.1186/s12974-023-02980-x] [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: 10/22/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Skull bone marrow is thought to be an immune tissue closely associated with the central nervous system (CNS). Recent studies have focused on the role of skull bone marrow in central nervous system disorders. In this study, we performed single-cell RNA sequencing on ipsilateral and contralateral skull bone marrow cells after experimental stroke and then performed flow cytometry and analysis of cytokine expression. Skull marrow showed lateralization in response to stroke. Lateralization is demonstrated primarily by the proliferation and differentiation of myeloid and lymphoid lineage cells in the skull bone marrow adjacent to the ischemic region, with an increased proportion of neutrophils compared to monocytes. Analysis of chemokines in the skull revealed marked differences in chemotactic signals between the ipsilateral and contralateral skull, whereas sympathetic signals innervating the skull did not affect cranial bone marrow lateralization. Osteopontin (OPN) is involved in region-specific activation of the skull marrow that promotes inflammation in the meninges, and inhibition of OPN expression improves neurological function.
Collapse
Affiliation(s)
- Chaoran Xu
- Department of Neurosurgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Qia Zhang
- Department of Neurosurgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yi Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huaijun Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tianchi Tang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Junjie Wang
- Department of Neurosurgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Siqi Xia
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Gao Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Department of Neurosurgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, China.
- Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
109
|
Lin T, Jiang D, Chen W, Lin JS, Zhang X, Chen C, Hsu C, Lai L, Chen P, Yang K, Sansing LH, Chang C. Trained immunity induced by high-salt diet impedes stroke recovery. EMBO Rep 2023; 24:e57164. [PMID: 37965920 PMCID: PMC10702837 DOI: 10.15252/embr.202357164] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 10/22/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023] Open
Abstract
A high-salt diet (HSD) elicits sustained sterile inflammation and worsens tissue injury. However, how this occurs after stroke, a leading cause of morbidity and mortality, remains unknown. Here, we report that HSD impairs long-term brain recovery after intracerebral hemorrhage, a severe form of stroke, despite salt withdrawal prior to the injury. Mechanistically, HSD induces innate immune priming and training in hematopoietic stem and progenitor cells (HSPCs) by downregulation of NR4a family and mitochondrial oxidative phosphorylation. This training compromises alternative activation of monocyte-derived macrophages (MDMs) without altering the initial inflammatory responses of the stroke brain. Healthy mice transplanted with bone marrow from HSD-fed mice retain signatures of reduced MDM reparative functions, further confirming a persistent form of innate immune memory that originates in the bone marrow. Loss of NR4a1 in macrophages recapitulates HSD-induced negative impacts on stroke outcomes while gain of NR4a1 enables stroke recovery in HSD animals. Together, we provide the first evidence that links HSD-induced innate immune memory to the acquisition of persistent dysregulated inflammatory responses and unveils NR4a1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Tze‐Yen Lin
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Danye Jiang
- Department of NeurologyMcGovern Medical School at the University of Texas Health Science Center in HoustonHoustonTXUSA
| | - Wan‐Ru Chen
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
- School of MedicineNational Taiwan University College of MedicineTaipeiTaiwan
| | - Jhih Syuan Lin
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Xin‐Yu Zhang
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Chih‐Hung Chen
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Chia‐Lang Hsu
- Department of Medical ResearchNational Taiwan University HospitalTaipeiTaiwan
| | - Liang‐Chuan Lai
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Ping‐Hung Chen
- Department and Graduate Institute of Biochemistry and Molecular BiologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Kai‐Chien Yang
- Department and Graduate Institute of PharmacologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Lauren H Sansing
- Department of NeurologyYale University School of MedicineNew HavenCTUSA
| | - Che‐Feng Chang
- Department and Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwan
| |
Collapse
|
110
|
Hu S, Yang B, Shu S, He X, Sang H, Fan X, Zhang H. Targeting Pericytes for Functional Recovery in Ischemic Stroke. Neuromolecular Med 2023; 25:457-470. [PMID: 37166748 DOI: 10.1007/s12017-023-08748-z] [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: 02/24/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023]
Abstract
Pericytes surrounding endothelial cells in the capillaries are emerging as an attractive cell resource, which can show a large variety of functions in ischemic stroke, including preservation of the blood-brain barrier, regulation of immune function, and support for cerebral vasculature. These functions have been fully elucidated in previous studies. However, in recent years, increasing evidence has shown that pericytes play an important role in neurological recovery after ischemic stroke due to their regenerative function which can be summarized in two aspects according to current discoveries, one is that pericytes are thought to be multipotential themselves, and the other is that pericytes can promote the differentiation of oligodendrocyte progenitor cells (OPCs). Considering the neuroprotective treatment for stroke has not been much progressed in recent years, new therapies targeting pericytes may be a future direction. Here, we will review the beneficial effects of pericytes in ischemic stroke from two directions: the barrier and vascular functions and the regenerative functions of pericytes.
Collapse
Affiliation(s)
- Shuqi Hu
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Bingjie Yang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Song Shu
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Xudong He
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Hongfei Sang
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Xuemei Fan
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Hao Zhang
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
111
|
Hui W, Huang W, Zheng Z, Li Y, Li P, Yang H. Ginkgo biloba extract promotes Treg differentiation to ameliorate ischemic stroke via inhibition of HIF-1α/HK2 pathway. Phytother Res 2023; 37:5821-5836. [PMID: 37655539 DOI: 10.1002/ptr.7988] [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: 02/24/2023] [Revised: 07/19/2023] [Accepted: 08/09/2023] [Indexed: 09/02/2023]
Abstract
The ischemic brain can dialogue with peripheral tissues through the immune system. Ginkgo biloba extract (EGb) was used to regulate various neurological disorders; however, the impact of EGb on ischemic stroke is still unclear. Here, we aimed to investigate whether immunomodulation has participated in the beneficial effects of EGb on ischemia/reperfusion (I/R) brain injury. Mice were orally administered with EGb once daily for 7 days before the induction of I/R. Neurobehavioral scores, infarct volume, and brain inflammation were determined. The proportion of CD4+ T cells was detected by flow cytometry. EGb significantly lowered neurobehavioral scores, infarct volume, and the level of inflammatory cytokines in I/R mice. Interestingly, EGb altered the proportion of CD4+ T cells, particularly increasing the proportion of Treg cells. Depletion of Treg cells weakened the neuroprotective effects of EGb on ischemic stroke; furthermore, EGb decreased the expression of HIF-1α and HK2 and promoted the differentiation of Treg cells in vitro. EGb suppressed the HIF-1α/HK2 signaling pathway to promote the differentiation of Treg cells and ameliorate ischemic stroke in mice. The expansion effect of EGb on Treg cells could be exploited as part of future stroke therapy.
Collapse
Affiliation(s)
- Wenyu Hui
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wei Huang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zuguo Zheng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yi Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hua Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
112
|
Liu Y, Dong J, Zhang Z, Liu Y, Wang Y. Regulatory T cells: A suppressor arm in post-stroke immune homeostasis. Neurobiol Dis 2023; 189:106350. [PMID: 37952680 DOI: 10.1016/j.nbd.2023.106350] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/09/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023] Open
Abstract
The activation of the immune system and the onset of pro- and anti-inflammatory responses play crucial roles in the pathophysiological processes of ischaemic stroke (IS). CD4+ regulatory T (Treg) cells is the main immunosuppressive cell population that is studied in the context of peripheral tolerance, autoimmunity, and the development of chronic inflammatory diseases. In recent years, more studies have focused on immune modulation after IS, and Treg cells have been demonstrated to be essential in the remission of inflammation, nerve regeneration, and behavioural recovery. However, the exact effects of Treg cells in the context of IS remain controversial, with some studies suggesting a negative correlation with stroke outcomes. In this review, we aim to provide a comprehensive overview of the current understanding of Treg cell involvement in post-stroke homeostasis. We summarized the literature focusing on the temporal changes in Treg cell populations after IS, the mechanisms of Treg cell-mediated immunomodulation in the brain, and the potential of Treg cell-based therapies for treatment. The purposes of the current article are to address the importance of Treg cells and inspire more studies to help physicians, as well as scientists, understand the whole map of immune responses during IS.
Collapse
Affiliation(s)
- Yiqi Liu
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jing Dong
- Department of Medical Engineering, Tsinghua University Yuquan Hospital, Beijing 100049, China
| | - Ziqing Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yunpeng Liu
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yang Wang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
113
|
Quan H, Zhang R. Microglia dynamic response and phenotype heterogeneity in neural regeneration following hypoxic-ischemic brain injury. Front Immunol 2023; 14:1320271. [PMID: 38094292 PMCID: PMC10716326 DOI: 10.3389/fimmu.2023.1320271] [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: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Hypoxic-ischemic brain injury poses a significant threat to the neural niche within the central nervous system. In response to this pathological process, microglia, as innate immune cells in the central nervous system, undergo rapid morphological, molecular and functional changes. Here, we comprehensively review these dynamic changes in microglial response to hypoxic-ischemic brain injury under pathological conditions, including stroke, chronic intermittent hypoxia and neonatal hypoxic-ischemic brain injury. We focus on the regulation of signaling pathways under hypoxic-ischemic brain injury and further describe the process of microenvironment remodeling and neural tissue regeneration mediated by microglia after hypoxic-ischemic injury.
Collapse
Affiliation(s)
- Hongxin Quan
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Runrui Zhang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| |
Collapse
|
114
|
Ricci A, Liesz A. A tale of two cells: Regulatory T cell-microglia cross-talk in the ischemic brain. Sci Transl Med 2023; 15:eadj0052. [PMID: 37939163 DOI: 10.1126/scitranslmed.adj0052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Regulatory T cells exert a beneficial immunomodulatory effect on poststroke neuroinflammation that is amplified by microglial cells.
Collapse
Affiliation(s)
- Alessio Ricci
- Institute for Stroke and Dementia Research, University Medical Center Munich, Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Medical Center Munich, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
115
|
Wang R, Li H, Ling C, Zhang X, Lu J, Luan W, Zhang J, Shi L. A novel phenotype of B cells associated with enhanced phagocytic capability and chemotactic function after ischemic stroke. Neural Regen Res 2023; 18:2413-2423. [PMID: 37282471 DOI: 10.4103/1673-5374.371365] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
Accumulating evidence has demonstrated the involvement of B cells in neuroinflammation and neuroregeneration. However, the role of B cells in ischemic stroke remains unclear. In this study, we identified a novel phenotype of macrophage-like B cells in brain-infiltrating immune cells expressing a high level of CD45. Macrophage-like B cells characterized by co-expression of B-cell and macrophage markers, showed stronger phagocytic and chemotactic functions compared with other B cells and showed upregulated expression of phagocytosis-related genes. Gene Ontology analysis found that the expression of genes associated with phagocytosis, including phagosome- and lysosome-related genes, was upregulated in macrophage-like B cells. The phagocytic activity of macrophage-like B cells was verified by immunostaining and three-dimensional reconstruction, in which TREM2-labeled macrophage-like B cells enwrapped and internalized myelin debris after cerebral ischemia. Cell-cell interaction analysis revealed that macrophage-like B cells released multiple chemokines to recruit peripheral immune cells mainly via CCL pathways. Single-cell RNA sequencing showed that the transdifferentiation to macrophage-like B cells may be induced by specific upregulation of the transcription factor CEBP family to the myeloid lineage and/or by downregulation of the transcription factor Pax5 to the lymphoid lineage. Furthermore, this distinct B cell phenotype was detected in brain tissues from mice or patients with traumatic brain injury, Alzheimer's disease, and glioblastoma. Overall, these results provide a new perspective on the phagocytic capability and chemotactic function of B cells in the ischemic brain. These cells may serve as an immunotherapeutic target for regulating the immune response of ischemic stroke.
Collapse
Affiliation(s)
- Rui Wang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Huaming Li
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Chenhan Ling
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Xiaotao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jianan Lu
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Weimin Luan
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Brain Research Institute, Zhejiang University; Stroke Research Center for Diagnostic and Therapeutic Technologies of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Ligen Shi
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, Zhejiang Province, China
| |
Collapse
|
116
|
Zhou M, Su P, Liang J, Xiong T. Research progress on the roles of neurovascular unit in stroke-induced immunosuppression. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:662-672. [PMID: 37899404 PMCID: PMC10630064 DOI: 10.3724/zdxbyxb-2023-0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023]
Abstract
A complex pathophysiological mechanism is involved in brain injury following cerebral infarction. The neurovascular unit (NVU) is a complex multi-cellular structure consisting of neurons, endothelial cells, pericyte, astrocyte, microglia and extracellular matrix, etc. The dyshomeostasis of NVU directly participates in the regulation of inflammatory immune process. The components of NVU promote inflammatory overreaction and synergize with the overactivation of autonomic nervous system to initiate stroke-induced immunodepression (SIID). SIID can alleviate the damage caused by inflammation, however, it also makes stroke patients more susceptible to infection, leading to systemic damage. This article reviews the mechanism of SIID and the roles of NVU in SIID, to provide a perspective for reperfusion, prognosis and immunomodulatory therapy of cerebral infarction.
Collapse
Affiliation(s)
- Mengqin Zhou
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou 225009, Jiangsu Province, China.
| | - Peng Su
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou 225009, Jiangsu Province, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225009, Jiangsu Province, China
| | - Jingyan Liang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou 225009, Jiangsu Province, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225009, Jiangsu Province, China
| | - Tianqing Xiong
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou 225009, Jiangsu Province, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225009, Jiangsu Province, China.
| |
Collapse
|
117
|
Wang Y, Liu W, Geng P, Du W, Guo C, Wang Q, Zheng GQ, Jin X. Role of Crosstalk between Glial Cells and Immune Cells in Blood-Brain Barrier Damage and Protection after Acute Ischemic Stroke. Aging Dis 2023; 15:2507-2525. [PMID: 37962453 PMCID: PMC11567273 DOI: 10.14336/ad.2023.1010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/10/2023] [Indexed: 11/15/2023] Open
Abstract
Blood-brain barrier (BBB) damage is the main pathological basis for acute ischemic stroke (AIS)-induced cerebral vasogenic edema and hemorrhagic transformation (HT). Glial cells, including microglia, astrocytes, and oligodendrocyte precursor cells (OPCs)/oligodendrocytes (OLs) play critical roles in BBB damage and protection. Recent evidence indicates that immune cells also have an important role in BBB damage, vasogenic edema and HT. Therefore, regulating the crosstalk between glial cells and immune cells would hold the promise to alleviate AIS-induced BBB damage. In this review, we first introduce the roles of glia cells, pericytes, and crosstalk between glial cells in the damage and protection of BBB after AIS, emphasizing the polarization, inflammatory response and crosstalk between microglia, astrocytes, and other glia cells. We then describe the role of glial cell-derived exosomes in the damage and protection of BBB after AIS. Next, we specifically discuss the crosstalk between glial cells and immune cells after AIS. Finally, we propose that glial cells could be a potential target for alleviating BBB damage after AIS and we discuss some molecular targets and potential strategies to alleviate BBB damage by regulating glial cells after AIS.
Collapse
Affiliation(s)
- Yihui Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Wencao Liu
- Shanxi Provincial People's Hospital, Taiyuan 030001, China.
| | - Panpan Geng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK.
| | - Qian Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Guo-qing Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
118
|
Gao Q, Li X, Li Y, Long J, Pan M, Wang J, Yang F, Zhang Y. Bibliometric analysis of global research trends on regulatory T cells in neurological diseases. Front Neurol 2023; 14:1284501. [PMID: 37900596 PMCID: PMC10603183 DOI: 10.3389/fneur.2023.1284501] [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: 08/28/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
This bibliometric study aimed to summarize and visualize the current research status, emerging trends, and research hotspots of regulatory T (Treg) cells in neurological diseases. Relevant documents were retrieved from the Web of Science Core Collection. Tableau Public, VOSviewer, and CiteSpace software were used to perform bibliometric analysis and network visualization. A total of 2,739 documents were included, and research on Treg cells in neurological diseases is still in a prolific period. The documents included in the research were sourced from 85 countries/regions, with the majority of them originating from the United States, and 2,811 organizations, with a significant proportion of them coming from Harvard Medical School. Howard E Gendelman was the most prolific author in this research area. Considering the number of documents and citations, impact factors, and JCR partitions, Frontiers in Immunology was the most popular journal in this research area. Keywords "multiple sclerosis," "inflammation," "regulatory T cells," "neuroinflammation," "autoimmunity," "cytokines," and "immunomodulation" were identified as high-frequency keywords. Additionally, "gut microbiota" has recently emerged as a new topic of interest. The study of Treg cells in neurological diseases continues to be a hot topic. Immunomodulation, gut microbiota, and cytokines represent the current research hotspots and frontiers in this field. Treg cell-based immunomodulatory approaches have shown immense potential in the treatment of neurological diseases. Modifying gut microbiota or regulating cytokines to boost the numbers and functions of Treg cells represents a promising therapeutic strategy for neurological diseases.
Collapse
Affiliation(s)
- Qian Gao
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xinmin Li
- School of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yan Li
- School of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Junzi Long
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Mengyang Pan
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jing Wang
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Fangjie Yang
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yasu Zhang
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
119
|
Kong G, Xiong W, Li C, Xiao C, Wang S, Li W, Chen X, Wang J, Chen S, Zhang Y, Gu J, Fan J, Jin Z. Treg cells-derived exosomes promote blood-spinal cord barrier repair and motor function recovery after spinal cord injury by delivering miR-2861. J Nanobiotechnology 2023; 21:364. [PMID: 37794487 PMCID: PMC10552208 DOI: 10.1186/s12951-023-02089-6] [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: 05/22/2023] [Accepted: 08/29/2023] [Indexed: 10/06/2023] Open
Abstract
The blood-spinal cord barrier (BSCB) is a physical barrier between the blood and the spinal cord parenchyma. Current evidence suggests that the disruption of BSCB integrity after spinal cord injury can lead to secondary injuries such as spinal cord edema and excessive inflammatory response. Regulatory T (Treg) cells are effective anti-inflammatory cells that can inhibit neuroinflammation after spinal cord injury, and their infiltration after spinal cord injury exhibits the same temporal and spatial characteristics as the automatic repair of BSCB. However, few studies have assessed the relationship between Treg cells and spinal cord injury, emphasizing BSCB integrity. This study explored whether Treg affects the recovery of BSCB after SCI and the underlying mechanism. We confirmed that spinal cord angiogenesis and Treg cell infiltration occurred simultaneously after SCI. Furthermore, we observed significant effects on BSCB repair and motor function in mice by Treg cell knockout and overexpression. Subsequently, we demonstrated the presence and function of exosomes in vitro. In addition, we found that Treg cell-derived exosomes encapsulated miR-2861, and miR-2861 regulated the expression of vascular tight junction (TJs) proteins. The luciferase reporter assay confirmed the negative regulation of IRAK1 by miR-2861, and a series of rescue experiments validated the biological function of IRAKI in regulating BSCB. In summary, we demonstrated that Treg cell-derived exosomes could package and deliver miR-2861 and regulate the expression of IRAK1 to affect BSCB integrity and motor function after SCI in mice, which provides novel insights for functional repair and limiting inflammation after SCI.
Collapse
Affiliation(s)
- Guang Kong
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wu Xiong
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cong Li
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenyu Xiao
- Nanjing Medical University, Nanjing, Jiangsu, China
- Department of human anatomy, School of Basic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Siming Wang
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenbo Li
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiangjun Chen
- Nanjing Medical University, Nanjing, Jiangsu, China
- Department of human anatomy, School of Basic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Juan Wang
- Nanjing Medical University, Nanjing, Jiangsu, China
- Department of human anatomy, School of Basic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sheng Chen
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yongjie Zhang
- Nanjing Medical University, Nanjing, Jiangsu, China.
- Department of human anatomy, School of Basic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jun Gu
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China.
| | - Jin Fan
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Zhengshuai Jin
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China.
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
120
|
Zhu H, Guan A, Liu J, Peng L, Zhang Z, Wang S. Noteworthy perspectives on microglia in neuropsychiatric disorders. J Neuroinflammation 2023; 20:223. [PMID: 37794488 PMCID: PMC10548593 DOI: 10.1186/s12974-023-02901-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023] Open
Abstract
Microglia are so versatile that they not only provide immune surveillance for central nervous system, but participate in neural circuitry development, brain blood vessels formation, blood-brain barrier architecture, and intriguingly, the regulation of emotions and behaviors. Microglia have a profound impact on neuronal survival, brain wiring and synaptic plasticity. As professional phagocytic cells in the brain, they remove dead cell debris and neurotoxic agents via an elaborate mechanism. The functional profile of microglia varies considerately depending on age, gender, disease context and other internal or external environmental factors. Numerous studies have demonstrated a pivotal involvement of microglia in neuropsychiatric disorders, including negative affection, social deficit, compulsive behavior, fear memory, pain and other symptoms associated with major depression disorder, anxiety disorder, autism spectrum disorder and schizophrenia. In this review, we summarized the latest discoveries regarding microglial ontogeny, cell subtypes or state spectrum, biological functions and mechanistic underpinnings of emotional and behavioral disorders. Furthermore, we highlight the potential of microglia-targeted therapies of neuropsychiatric disorders, and propose outstanding questions to be addressed in future research of human microglia.
Collapse
Affiliation(s)
- Hongrui Zhu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Ao Guan
- School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Jiayuan Liu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Li Peng
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Zhi Zhang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Sheng Wang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
121
|
Lv M, Zhang Z, Cui Y. Unconventional T cells in brain homeostasis, injury and neurodegeneration. Front Immunol 2023; 14:1273459. [PMID: 37854609 PMCID: PMC10579804 DOI: 10.3389/fimmu.2023.1273459] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023] Open
Abstract
The interaction between peripheral immune cells and the brain is an important component of the neuroimmune axis. Unconventional T cells, which include natural killer T (NKT) cells, mucosal-associated invariant T (MAIT) cells, γδ T cells, and other poorly defined subsets, are a special group of T lymphocytes that recognize a wide range of nonpolymorphic ligands and are the connection between adaptive and innate immunity. Recently, an increasing number of complex functions of these unconventional T cells in brain homeostasis and various brain disorders have been revealed. In this review, we describe the classification and effector function of unconventional T cells, review the evidence for the involvement of unconventional T cells in the regulation of brain homeostasis, summarize the roles and mechanisms of unconventional T cells in the regulation of brain injury and neurodegeneration, and discuss immunotherapeutic potential as well as future research goals. Insight of these processes can shed light on the regulation of T cell immunity on brain homeostasis and diseases and provide new clues for therapeutic approaches targeting brain injury and neurodegeneration.
Collapse
Affiliation(s)
- Mengfei Lv
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zhaolong Zhang
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yu Cui
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
122
|
Hajjar S, Zhou X. pH sensing at the intersection of tissue homeostasis and inflammation. Trends Immunol 2023; 44:807-825. [PMID: 37714775 PMCID: PMC10543622 DOI: 10.1016/j.it.2023.08.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/13/2023] [Accepted: 08/13/2023] [Indexed: 09/17/2023]
Abstract
pH is tightly maintained at cellular, tissue, and systemic levels, and altered pH - particularly in the acidic range - is associated with infection, injury, solid tumors, and physiological and pathological inflammation. However, how pH is sensed and regulated and how it influences immune responses remain poorly understood at the tissue level. Applying conceptual frameworks of homeostatic and inflammatory circuitries, we categorize cellular and tissue components engaged in pH regulation, drawing parallels from established cases in physiology. By expressing various intracellular (pHi) and extracellular pH (pHe)-sensing receptors, the immune system may integrate information on tissue and cellular states into the regulation of homeostatic and inflammatory programs. We introduce the novel concept of resistance and adaptation responses to rationalize pH-dependent immunomodulation intertwined with homeostatic equilibrium and inflammatory control. We discuss emerging challenges and opportunities in understanding the immunological roles of pH sensing, which might reveal new strategies to combat inflammation and restore tissue homeostasis.
Collapse
Affiliation(s)
- Stephanie Hajjar
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, 300 Longwood Ave, Boston, MA 02115, USA
| | - Xu Zhou
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, 300 Longwood Ave, Boston, MA 02115, USA.
| |
Collapse
|
123
|
Li H, Ghorbani S, Ling CC, Yong VW, Xue M. The extracellular matrix as modifier of neuroinflammation and recovery in ischemic stroke and intracerebral hemorrhage. Neurobiol Dis 2023; 186:106282. [PMID: 37683956 DOI: 10.1016/j.nbd.2023.106282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Stroke is the second leading cause of death worldwide and has two major subtypes: ischemic stroke and hemorrhagic stroke. Neuroinflammation is a pathological hallmark of ischemic stroke and intracerebral hemorrhage (ICH), contributing to the extent of brain injury but also in its repair. Neuroinflammation is intricately linked to the extracellular matrix (ECM), which is profoundly altered after brain injury and in aging. In the early stages after ischemic stroke and ICH, immune cells are involved in the deposition and remodeling of the ECM thereby affecting processes such as blood-brain barrier and cellular integrity. ECM components regulate leukocyte infiltration into the central nervous system, activate a variety of immune cells, and induce the elevation of matrix metalloproteinases (MMPs) after stroke. In turn, excessive MMPs may degrade ECM into components that are pro-inflammatory and injurious. Conversely, in the later stages after stroke, several ECM molecules may contribute to tissue recovery. For example, thrombospondin-1 and biglycan may promote activity of regulatory T cells, inhibit the synthesis of proinflammatory cytokines, and aid regenerative processes. We highlight these roles of the ECM in ischemic stroke and ICH and discuss their potential cellular and molecular mechanisms. Finally, we discuss therapeutics that could be considered to normalize the ECM in stroke. Our goal is to spur research on the ECM in order to improve the prognosis of ischemic stroke and ICH.
Collapse
Affiliation(s)
- Hongmin Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China; Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Samira Ghorbani
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Chang-Chun Ling
- Department of Chemistry, University of Calgary, Alberta, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada.
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
124
|
Zheng P, Xiu Y, Chen Z, Yuan M, Li Y, Wang N, Zhang B, Zhao X, Li M, Liu Q, Shi FD, Jin WN. Group 2 innate lymphoid cells resolve neuroinflammation following cerebral ischaemia. Stroke Vasc Neurol 2023; 8:424-434. [PMID: 37072337 PMCID: PMC10647866 DOI: 10.1136/svn-2022-001919] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 03/02/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Acute brain ischaemia elicits pronounced inflammation, which aggravates neural injury. However, the mechanisms governing the resolution of acute neuroinflammation remain poorly understood. In contrast to regulatory T and B cells, group 2 innate lymphoid cells (ILC2s) are immunoregulatory cells that can be swiftly mobilised without antigen presentation; whether and how these ILC2s participate in central nervous system inflammation following brain ischaemia is still unknown. METHODS Leveraging brain tissues from patients who had an ischaemic stroke and a mouse model of focal ischaemia, we characterised the presence and cytokine release of brain-infiltrating ILC2s. The impact of ILC2s on neural injury was evaluated through antibody depletion and ILC2 adoptive transfer experiments. Using Rag2-/-γc-/- mice receiving passive transfer of IL-4-/- ILC2s, we further assessed the contribution of interleukin (IL)-4, produced by ILC2s, in ischaemic brain injury. RESULTS We demonstrate that ILC2s accumulate in the areas surrounding the infarct in brain tissues of patients with cerebral ischaemia, as well as in mice subjected to focal cerebral ischaemia. Oligodendrocytes were a major source of IL-33, which contributed to ILC2s mobilisation. Adoptive transfer and expansion of ILC2s reduced brain infarction. Importantly, brain-infiltrating ILC2s reduced the magnitude of stroke injury severity through the production of IL-4. CONCLUSIONS Our findings revealed that brain ischaemia mobilises ILC2s to curb neuroinflammation and brain injury, expanding the current understanding of inflammatory networks following stroke.
Collapse
Affiliation(s)
- Pei Zheng
- Center for Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuwhen Xiu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhili Chen
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Meng Yuan
- Center for Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yan Li
- Center for Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ningning Wang
- Center for Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bohao Zhang
- Department of Neurology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xin Zhao
- Department of Neurology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Minshu Li
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Liu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Fu-Dong Shi
- Center for Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei-Na Jin
- Center for Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
125
|
Fan PL, Wang SS, Chu SF, Chen NH. Time-dependent dual effect of microglia in ischemic stroke. Neurochem Int 2023; 169:105584. [PMID: 37454817 DOI: 10.1016/j.neuint.2023.105584] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Stroke, the third leading cause of death and disability worldwide, is classified into ischemic or hemorrhagic, in which approximately 85% of strokes are ischemic. Ischemic stroke occurs as a result of arterial occlusion due to embolus or thrombus, with ischemia in the perfusion territory supplied by the occluded artery. The traditional concept that ischemic stroke is solely a vascular occlusion disorder has been expanded to include the dynamic interaction between microglia, astrocytes, neurons, vascular cells, and matrix components forming the "neurovascular unit." Acute ischemic stroke triggers a wide spectrum of neurovascular disturbances, glial activation, and secondary neuroinflammation that promotes further injury, ultimately resulting in neuronal death. Microglia, as the resident macrophages in the central nervous system, is one of the first responders to ischemic injury and plays a significant role in post-ischemic neuroinflammation. In this review, we reviewed the mechanisms of microglia in multiple stages of post-ischemic neuroinflammation development, including acute, sub-acute and chronic phases of stroke. A comprehensive understanding of the dynamic variation and the time-dependent role of microglia in post-stroke neuroinflammation could aid in the search for more effective therapeutics and diagnostic strategies for ischemic stroke.
Collapse
Affiliation(s)
- Ping-Long Fan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Sha-Sha Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Nai-Hong Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
126
|
Liu S, Zhang Z, He Y, Kong L, Jin Q, Qi X, Qi D, Gao Y. Inhibiting leukocyte-endothelial cell interactions by Chinese medicine Tongxinluo capsule alleviates no-reflow after arterial recanalization in ischemic stroke. CNS Neurosci Ther 2023; 29:3014-3030. [PMID: 37122157 PMCID: PMC10493667 DOI: 10.1111/cns.14242] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/12/2023] [Accepted: 04/16/2023] [Indexed: 05/02/2023] Open
Abstract
AIMS Despite successful vascular recanalization in stroke, one-fourth of patients have an unfavorable outcome due to no-reflow. The pathogenesis of no-reflow is fully unclear, and therapeutic strategies are lacking. Upon traditional Chinese medicine, Tongxinluo capsule (TXL) is a potential therapeutic agent for no-reflow. Thus, this study is aimed to investigate the pathogenesis of no-reflow in stroke, and whether TXL could alleviate no-reflow as well as its potential mechanisms of action. METHODS Mice were orally administered with TXL (3.0 g/kg/d) after transient middle cerebral artery occlusion. We examined the following parameters: neurological function, no-reflow, leukocyte-endothelial cell interactions, HE staining, leukocyte subtypes, adhesion molecules, and chemokines. RESULTS Our results showed stroke caused neurological deficits, neuron death, and no-reflow. Adherent and aggregated leukocytes obstructed microvessels as well as leukocyte infiltration in ischemic brain. Leukocyte subtypes changed after stroke mainly including neutrophils, lymphocytes, regulatory T cells, suppressor T cells, helper T type 1 (Th1) cells, Th2 cells, B cells, macrophages, natural killer cells, and dendritic cells. Stroke resulted in upregulated expression of adhesion molecules (P-selectin, E-selectin, and ICAM-1) and chemokines (CC-chemokine ligand (CCL)-2, CCL-3, CCL-4, CCL-5, and chemokine C-X-C ligand 1 (CXCL-1)). Notably, TXL improved neurological deficits, protected neurons, alleviated no-reflow and leukocyte-endothelial cell interactions, regulated multiple leukocyte subtypes, and inhibited the expression of various inflammatory mediators. CONCLUSION Leukocyte-endothelial cell interactions mediated by multiple inflammatory factors are an important cause of no-reflow in stroke. Accordingly, TXL could alleviate no-reflow via suppressing the interactions through modulating various leukocyte subtypes and inhibiting the expression of multiple inflammatory mediators.
Collapse
Affiliation(s)
- Shen Liu
- Department of Neurology of TCM, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
- Department of Rehabilitation MedicineThe First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan HospitalJinanChina
| | - Zhaoxu Zhang
- Department of NeurologyPeking University People's HospitalBeijingChina
| | - Yannan He
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
| | - Lingbo Kong
- Department of Neurology of TCM, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
| | - Qiushuo Jin
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
| | - Xiangjia Qi
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan HospitalJinanChina
| | - Dahe Qi
- Department of Neurology of TCM, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
| | - Ying Gao
- Department of Neurology of TCM, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
- Institute for Brain Disorders, Beijing University of Chinese MedicineBeijingChina
| |
Collapse
|
127
|
Touil H, Li R, Zuroff L, Moore CS, Healy L, Cignarella F, Piccio L, Ludwin S, Prat A, Gommerman J, Bennett FC, Jacobs D, Benjamins JA, Lisak RP, Antel JP, Bar-Or A. Cross-talk between B cells, microglia and macrophages, and implications to central nervous system compartmentalized inflammation and progressive multiple sclerosis. EBioMedicine 2023; 96:104789. [PMID: 37703640 PMCID: PMC10505984 DOI: 10.1016/j.ebiom.2023.104789] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/07/2023] [Accepted: 08/22/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND B cells can be enriched within meningeal immune-cell aggregates of multiple sclerosis (MS) patients, adjacent to subpial cortical demyelinating lesions now recognized as important contributors to progressive disease. This subpial demyelination is notable for a 'surface-in' gradient of neuronal loss and microglial activation, potentially reflecting the effects of soluble factors secreted into the CSF. We previously demonstrated that MS B-cell secreted products are toxic to oligodendrocytes and neurons. The potential for B-cell-myeloid cell interactions to propagate progressive MS is of considerable interest. METHODS Secreted products of MS-implicated pro-inflammatory effector B cells or IL-10-expressing B cells with regulatory potential were applied to human brain-derived microglia or monocyte-derived macrophages, with subsequent assessment of myeloid phenotype and function through measurement of their expression of pro-inflammatory, anti-inflammatory and homeostatic/quiescent molecules, and phagocytosis (using flow cytometry, ELISA and fluorescently-labeled myelin). Effects of secreted products of differentially activated microglia on B-cell survival and activation were further studied. FINDINGS Secreted products of MS-implicated pro-inflammatory B cells (but not IL-10 expressing B cells) substantially induce pro-inflammatory cytokine (IL-12, IL-6, TNFα) expression by both human microglia and macrophage (in a GM-CSF dependent manner), while down-regulating their expression of IL-10 and of quiescence-associated molecules, and suppressing their myelin phagocytosis. In contrast, secreted products of IL-10 expressing B cells upregulate both human microglia and macrophage expression of quiescence-associated molecules and enhance their myelin phagocytosis. Secreted factors from pro-inflammatory microglia enhance B-cell activation. INTERPRETATION Potential cross-talk between disease-relevant human B-cell subsets and both resident CNS microglia and infiltrating macrophages may propagate CNS-compartmentalized inflammation and injury associated with MS disease progression. These interaction represents an attractive therapeutic target for agents such as Bruton's tyrosine kinase inhibitors (BTKi) that modulate responses of both B cells and myeloid cells. FUNDING Stated in Acknowledgments section of manuscript.
Collapse
Affiliation(s)
- Hanane Touil
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rui Li
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leah Zuroff
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Craig S Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Luke Healy
- Neuroimmunology Unit, Montréal Neurological Institute, McGill University, Canada
| | - Francesca Cignarella
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, USA
| | - Laura Piccio
- Charles Perkins Centre and School of Medical Sciences, The University of Sydney, Camperdown, NSW, Australia
| | - Samuel Ludwin
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Alexandre Prat
- Université de Montréal Centre de Recherche du CHUM (CRCHUM) and Department of Neuroscience, Université de Montréal, 900 Saint Denis Street, Montréal, QC, H2X 0A9, Canada
| | - Jennifer Gommerman
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Frederick C Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dina Jacobs
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joyce A Benjamins
- Departments of Neurology and Biochemistry, Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robert P Lisak
- Departments of Neurology and Biochemistry, Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jack P Antel
- Neuroimmunology Unit, Montréal Neurological Institute, McGill University, Canada
| | - Amit Bar-Or
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
128
|
Guo G, Fan L, Yan Y, Xu Y, Deng Z, Tian M, Geng Y, Xia Z, Xu Y. Shared metabolic shifts in endothelial cells in stroke and Alzheimer's disease revealed by integrated analysis. Sci Data 2023; 10:666. [PMID: 37775708 PMCID: PMC10542331 DOI: 10.1038/s41597-023-02512-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 08/30/2023] [Indexed: 10/01/2023] Open
Abstract
Since metabolic dysregulation is a hallmark of both stroke and Alzheimer's disease (AD), mining shared metabolic patterns in these diseases will help to identify their possible pathogenic mechanisms and potential intervention targets. However, a systematic integration analysis of the metabolic networks of the these diseases is still lacking. In this study, we integrated single-cell RNA sequencing datasets of ischemic stroke (IS), hemorrhagic stroke (HS) and AD models to construct metabolic flux profiles at the single-cell level. We discovered that the three disorders cause shared metabolic shifts in endothelial cells. These altered metabolic modules were mainly enriched in the transporter-related pathways and were predicted to potentially lead to a decrease in metabolites such as pyruvate and fumarate. We further found that Lef1, Elk3 and Fosl1 may be upstream transcriptional regulators causing metabolic shifts and may be possible targets for interventions that halt the course of neurodegeneration.
Collapse
Affiliation(s)
- Guangyu Guo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liyuan Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Yingxue Yan
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Yunhao Xu
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Zhifen Deng
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Miaomiao Tian
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaoqi Geng
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zongping Xia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, Zhengzhou, China.
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, Zhengzhou, China.
| |
Collapse
|
129
|
Liu ZZ, Huang Y, Hong CG, Wang X, Duan R, Liu JY, He JL, Duan D, Xie H, Lu M. Autologous olfactory mucosa mesenchymal stem cells treatment improves the neural network in chronic refractory epilepsy. Stem Cell Res Ther 2023; 14:237. [PMID: 37674249 PMCID: PMC10483711 DOI: 10.1186/s13287-023-03458-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/18/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND AND AIMS Refractory epilepsy is also known as drug-resistant epilepsy with limited clinical treatment. Benefitting from its safety and easy availability, olfactory mucosa mesenchymal stem cells (OM-MSCs) are considered a preferable MSC source for clinical application. This study aims to investigate whether OM-MSCs are a promising alternative source for treating refractory epilepsy clinically and uncover the mechanism by OM-MSCs administration on an epileptic mouse model. METHODS OM-MSCs were isolated from turbinal and characterized by flow cytometry. Autologous human OM-MSCs treatment on a patient was carried out using intrathecal administration. Epileptic mouse model was established by 1 mg/kg scopolamine and 300 mg/kg pilocarpine treatment (intraperitoneal). Stereotaxic microinjection was employed to deliver the mouse OM-MSCs. Mouse electroencephalograph recording was used to investigate the seizures. Brain structure was evaluated by magnetic resonance imaging (MRI). Immunohistochemical and immunofluorescent staining of GFAP, IBA1, MAP2, TUBB3, OLIG2, CD4, CD25, and FOXP3 was carried out to investigate the neural cells and Treg cells. QRT-PCR and ELISA were performed to determine the cytokines (Il1b, Il6, Tnf, Il10) on mRNA and protein level. Y-maze, the object location test, and novel object recognition test were performed to measure the cognitive function. Footprint test, rotarod test, balance beam test, and grip strength test were conducted to evaluate the locomotive function. Von Frey testing was carried out to assess the mechanical allodynia. RESULTS Many beneficial effects of the OM-MSC treatment on disease status, including seizure type, frequency, severity, duration, and cognitive function, and no apparent adverse effects were observed at the 8-year follow-up case. Brain MRI indicated that autologous OM-MSC treatment alleviated brain atrophy in epilepsy patients. A study in an epileptic mouse model revealed that OM-MSC treatment recruited Treg cells to the brain, inhibited inflammation, rebuilt the neural network, and improved the cognitive, locomotive, and perceptive functions of epileptic mice. CONCLUSIONS Autologous OM-MSC treatment is efficacious for improving chronic refractory epilepsy, suggesting a future therapeutic candidate for epilepsy. TRIAL REGISTRATION The study was registered with Chinese Clinical Trial Registry (ChiCTR2200055357).
Collapse
Affiliation(s)
- Zheng-Zhao Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Organ Injury, Aging and Regenerative Medicine, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Bone Joint Degeneration and Injury, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yan Huang
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, Hunan, China
- Hunan Provincial Key Laboratory of Neurorestoration, Hunan Normal University, Changsha, 410081, Hunan, China
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, 410219, Hunan, China
- First Clinical Department of Changsha Medical University, Changsha, 410081, Hunan, China
| | - Chun-Gu Hong
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xin Wang
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ran Duan
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jian-Yang Liu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jia-Lin He
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Da Duan
- Department of Neurosurgery, the 921st Hospital of PLA (Second Affiliated Hospital of Hunan Normal University), Changsha, 410081, Hunan, China
| | - Hui Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Organ Injury, Aging and Regenerative Medicine, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Bone Joint Degeneration and Injury, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Ming Lu
- Department of Neurosurgery, the 921st Hospital of PLA (Second Affiliated Hospital of Hunan Normal University), Changsha, 410081, Hunan, China.
- Hunan Provincial Key Laboratory of Neurorestoration, Hunan Normal University, Changsha, 410081, Hunan, China.
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, 410219, Hunan, China.
| |
Collapse
|
130
|
Gao X, Tang Y, Kong L, Fan Y, Wang C, Wang R. Treg cell: Critical role of regulatory T-cells in depression. Pharmacol Res 2023; 195:106893. [PMID: 37611836 DOI: 10.1016/j.phrs.2023.106893] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/28/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023]
Abstract
Depression is a highly prevalent disorder of the central nervous system. The neuropsychiatric symptoms of clinical depression are persistent and include fatigue, anorexia, weight loss, altered sleep patterns, hyperalgesia, melancholia, anxiety, and impaired social behaviours. Mounting evidences suggest that neuroinflammation triggers dysregulated cellular immunity and increases susceptibility to psychiatric diseases. Neuroimmune responses have transformed the clinical approach to depression because of their roles in its pathophysiology and their therapeutic potential. In particular, activated regulatory T (Treg) cells play an increasingly evident role in the inflammatory immune response. In this review, we summarized the available data and discussed in depth the fundamental roles of Tregs in the pathogenesis of depression, as well as the clinical therapeutic potential of Tregs. We aimed to provide recent information regarding the potential of Tregs as immune-modulating biologics for the treatment and prevention of long-term neuropsychiatric symptoms of depression.
Collapse
Affiliation(s)
- Xiao Gao
- Department of Geriatrics, Qingdao Mental Health Center, 26600 Qingdao, Shandong Province, China
| | - Yuru Tang
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, 26600 Qingdao, Shandong Province, China
| | - Lingli Kong
- Department of Geriatrics, Qingdao Mental Health Center, 26600 Qingdao, Shandong Province, China
| | - Yong Fan
- Department of Geriatrics, Qingdao Mental Health Center, 26600 Qingdao, Shandong Province, China
| | - Chunxia Wang
- Department of Geriatrics, Qingdao Mental Health Center, 26600 Qingdao, Shandong Province, China.
| | - Rui Wang
- Department of Pain Management, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), 26600 Qingdao, Shandong Province, China.
| |
Collapse
|
131
|
He N, Mao XJ, Ding YM, Zuo T, Chen YY, Wang LL. New insights into the biological roles of immune cells in neural stem cells in post-traumatic injury of the central nervous system. Neural Regen Res 2023; 18:1908-1916. [PMID: 36926707 PMCID: PMC10233778 DOI: 10.4103/1673-5374.367836] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/21/2022] [Accepted: 12/22/2022] [Indexed: 01/22/2023] Open
Abstract
Traumatic injuries in the central nervous system, such as traumatic brain injury and spinal cord injury, are associated with tissue inflammation and the infiltration of immune cells, which simultaneously affect the self-renewal and differentiation of neural stem cells. However, the tissue repair process instigated by endogenous neural stem cells is incapable of restoring central nervous system injuries without external intervention. Recently, resident/peripheral immune cells have been demonstrated to exert significant effects on neural stem cells. Thus, the restoration of traumatic injuries in the central nervous system by the immune intervention in neural stem cells represents a potential therapeutic method. In this review, we discuss the roles and possible mechanisms of immune cells on the self-renewal and differentiation of neural stem cells along with the prognosis of central nervous system injuries based on immune intervention. Finally, we discuss remaining research challenges that need to be considered in the future. Further elucidation of these challenges will facilitate the successful application of neural stem cells in central nervous system injuries.
Collapse
Affiliation(s)
- Ning He
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Xing-Jia Mao
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yue-Min Ding
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang Province, China
| | - Tong Zuo
- University of Chicago College, University of Chicago, Chicago, IL, USA
| | - Ying-Ying Chen
- Department of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Lin-Lin Wang
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
132
|
von Baumgarten L, Stauss HJ, Lünemann JD. Synthetic Cell-Based Immunotherapies for Neurologic Diseases. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200139. [PMID: 37385738 PMCID: PMC10474853 DOI: 10.1212/nxi.0000000000200139] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/11/2023] [Indexed: 07/01/2023]
Abstract
The therapeutic success and widespread approval of genetically engineered T cells for a variety of hematologic malignancies spurred the development of synthetic cell-based immunotherapies for CNS lymphoma, primary brain tumors, and a growing spectrum of nononcologic disease conditions of the nervous system. Chimeric antigen receptor effector T cells bear the potential to deplete target cells with higher efficacy, better tissue penetration, and greater depth than antibody-based cell depletion therapies. In multiple sclerosis and other autoimmune disorders, engineered T-cell therapies are being designed and currently tested in clinical trials for their safety and efficacy to eliminate pathogenic B-lineage cells. Chimeric autoantibody receptor T cells expressing a disease-relevant autoantigen as cell surface domains are designed to selectively deplete autoreactive B cells. Alternative to cell depletion, synthetic antigen-specific regulatory T cells can be engineered to locally restrain inflammation, support immune tolerance, or efficiently deliver neuroprotective factors in brain diseases in which current therapeutic options are very limited. In this article, we illustrate prospects and bottlenecks for the clinical development and implementation of engineered cellular immunotherapies in neurologic diseases.
Collapse
Affiliation(s)
- Louisa von Baumgarten
- From the Department of Neurosurgery (L.v.B.), University Hospital, Ludwig-Maximilians-Universität Munich, Germany; Division of Infection & Immunity (H.J.S.), UCL Institute of Immunity & Transplantation, London, UK; and Department of Neurology with Institute of Translational Neurology (J.D.L.), University Hospital Münster, Germany
| | - Hans J Stauss
- From the Department of Neurosurgery (L.v.B.), University Hospital, Ludwig-Maximilians-Universität Munich, Germany; Division of Infection & Immunity (H.J.S.), UCL Institute of Immunity & Transplantation, London, UK; and Department of Neurology with Institute of Translational Neurology (J.D.L.), University Hospital Münster, Germany
| | - Jan D Lünemann
- From the Department of Neurosurgery (L.v.B.), University Hospital, Ludwig-Maximilians-Universität Munich, Germany; Division of Infection & Immunity (H.J.S.), UCL Institute of Immunity & Transplantation, London, UK; and Department of Neurology with Institute of Translational Neurology (J.D.L.), University Hospital Münster, Germany.
| |
Collapse
|
133
|
Li TT, Zhao DM, Wei YT, Li JB, Li XF, Wan Q, Zhang X, Liu XN, Yang WC, Li WZ. Effect and Mechanism of Sodium Butyrate on Neuronal Recovery and Prognosis in Diabetic Stroke. J Neuroimmune Pharmacol 2023; 18:366-382. [PMID: 37318680 DOI: 10.1007/s11481-023-10071-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 05/18/2023] [Indexed: 06/16/2023]
Abstract
Ischemic stroke is a cerebrovascular lesion caused by local ischemia and hypoxia. Diabetes mellitus (DM) is a chronic inflammatory disease that disturbs immune homeostasis and predisposes patients to ischemic stroke. The mechanism by which DM exacerbates stroke remains unclear, although it may involve disturbances in immune homeostasis. Regulatory T cells (Tregs) play a regulatory role in many diseases, but the mechanism of Tregs in diabetes complicated by stroke remains unclear. Sodium butyrate is a short-chain fatty acid that increases Treg levels. This study examined the role of sodium butyrate in the prognosis of neurological function in diabetic stroke and the mechanism by which Tregs are amplified in the bilateral cerebral hemispheres. We evaluated the brain infarct volume, observed 48-h neuronal injury and 28-day behavioral changes, and calculated the 28-day survival rate in mice. We also measured Treg levels in peripheral blood and brain tissue, recorded changes in the blood‒brain barrier and water channel proteins and neurotrophic changes in mice, measured cytokine levels and peripheral B-cell distribution in bilateral hemispheres and peripheral blood, and examined the polarization of microglia and the distribution of peripheral T-cell subpopulations in bilateral hemispheres. Diabetes significantly exacerbated the poor prognosis and neurological deficits in mice with stroke, and sodium butyrate significantly improved infarct volume, prognosis, and neurological function and showed different mechanisms in brain tissue and peripheral blood. The potential regulatory mechanism in brain tissue involved modulating Tregs/TGF-β/microglia to suppress neuroinflammation, while that in peripheral blood involved improving the systemic inflammatory response through Tregs/TGF-β/T cells.
Collapse
Affiliation(s)
- Ting-Ting Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Deng-Ming Zhao
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Yu-Ting Wei
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Jing-Bo Li
- The Heilongjiang Key Laboratory of Anesthesia and Intensive Care Research, Harbin Medical University, 150081, Heilongjiang Province, Harbin, China
| | - Xue-Fei Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Qiang Wan
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, 650000, Yunnan Province, Kunming, China
| | - Xin Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Xiang-Nan Liu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Wan-Chao Yang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Wen-Zhi Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China.
- The Heilongjiang Key Laboratory of Anesthesia and Intensive Care Research, Harbin Medical University, 150081, Heilongjiang Province, Harbin, China.
| |
Collapse
|
134
|
Zhou J, Yang F, Li H, Xu P, Wang Z, Shao F, Shao A, Zhang J. Regulatory T Cells Secrete IL10 to Suppress Neuroinflammation in Early Stage after Subarachnoid Hemorrhage. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1317. [PMID: 37512128 PMCID: PMC10383056 DOI: 10.3390/medicina59071317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/09/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023]
Abstract
Objective: Accumulating evidence supports neuroprotective effects of regulatory T cells (Tregs) in response to brain injury. However, the precise mechanisms underlying the beneficial effects of Tregs on suppressing neuroinflammation after subarachnoid hemorrhage (SAH) remain unclear. Methods: We performed flow cytometry to detect the infiltration of Tregs into the brain at different time points after SAH. Behavioral tests, including Adhesive and Rotarod, were performed to assess neurological deficits in mice after SAH. Bulk RNA sequencing was used to investigate the transcriptomic change of Tregs infiltrating into the brain after SAH. qPCR was performed to verify the variation of inflammatory cytokines expression in the brain after Tregs exogenous infusion. FoxP3-DTR mice and Il10 gene KO mice were used to explore the mechanism of Tregs inhibiting neuron apoptosis after infiltrating the brain following SAH onset. Results: Peripheral Tregs infiltrated into the brain one day after SAH and gradually accumulated in the hemorrhagic hemisphere. An exogenous infusion of Tregs significantly improved the neurological function of mice after SAH, while poor recovery of neurological function was observed in Tregs depletion mice. Transcriptome sequencing data suggested that the immunosuppressive function of brain-infiltrated Tregs was significantly upregulated. qPCR showed that the expression of pro-inflammatory cytokines decreased in the brain of SAH mice after exogenous Tregs infusion. Bioinformatic analysis revealed that IL-10 and other cytokines secreted by brain-infiltrated Tregs were upregulated after SAH. Moreover, exogenous infusion of Il10 gene KO Tregs did not totally improve neurological function in SAH mice. Conclusions: Tregs infiltrated into the brain in the early stage after SAH and exerted neuroprotective effect by secreting IL-10 to suppress neuroinflammation and reduce neuron apoptosis.
Collapse
Affiliation(s)
- Jingyi Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310006, China
| | - Fan Yang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310006, China
| | - Huaming Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310006, China
| | - Penglei Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310006, China
| | - Zefeng Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310006, China
| | - Fangjie Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310006, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310006, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310006, China
- Brain Research Institute, Zhejiang University, Hangzhou 310058, China
- Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
135
|
Tang Q. Regulatory T cells aid stem-cell therapy for Parkinson's disease. Nature 2023:10.1038/d41586-023-02177-5. [PMID: 37438628 DOI: 10.1038/d41586-023-02177-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
|
136
|
Hu M, Li T, Ma X, Liu S, Li C, Huang Z, Lin Y, Wu R, Wang S, Lu D, Lu T, Men X, Shen S, Huang H, Liu Y, Song K, Jian B, Jiang Y, Qiu W, Liu Q, Lu Z, Cai W. Macrophage lineage cells-derived migrasomes activate complement-dependent blood-brain barrier damage in cerebral amyloid angiopathy mouse model. Nat Commun 2023; 14:3945. [PMID: 37402721 DOI: 10.1038/s41467-023-39693-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 06/16/2023] [Indexed: 07/06/2023] Open
Abstract
Accumulation of amyloid beta protein (Aβ) in brain vessels damages blood brain barrier (BBB) integrity in cerebral amyloid angiopathy (CAA). Macrophage lineage cells scavenge Aβ and produce disease-modifying mediators. Herein, we report that Aβ40-induced macrophage-derived migrasomes are sticky to blood vessels in skin biopsy samples from CAA patients and brain tissue from CAA mouse models (Tg-SwDI/B and 5xFAD mice). We show that CD5L is packed in migrasomes and docked to blood vessels, and that enrichment of CD5L impairs the resistance to complement activation. Increased migrasome-producing capacity of macrophages and membrane attack complex (MAC) in blood are associated with disease severity in both patients and Tg-SwDI/B mice. Of note, complement inhibitory treatment protects against migrasomes-mediated blood-brain barrier injury in Tg-SwDI/B mice. We thus propose that macrophage-derived migrasomes and the consequent complement activation are potential biomarkers and therapeutic targets in CAA.
Collapse
Affiliation(s)
- Mengyan Hu
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, 510630, China
- Center of Clinical Immunology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Tiemei Li
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xiaomeng Ma
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Sanxin Liu
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Chunyi Li
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Zhenchao Huang
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yinyao Lin
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Ruizhen Wu
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Shisi Wang
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Danli Lu
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Tingting Lu
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xuejiao Men
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Shishi Shen
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Huipeng Huang
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yuxin Liu
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Kangyu Song
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Banghao Jian
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yuxuan Jiang
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Wei Qiu
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Quentin Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.
| | - Zhengqi Lu
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Wei Cai
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, 510630, China.
- Center of Clinical Immunology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
137
|
Nie L, Yao D, Chen S, Wang J, Pan C, Wu D, Liu N, Tang Z. Directional induction of neural stem cells, a new therapy for neurodegenerative diseases and ischemic stroke. Cell Death Discov 2023; 9:215. [PMID: 37393356 DOI: 10.1038/s41420-023-01532-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023] Open
Abstract
Due to the limited capacity of the adult mammalian brain to self-repair and regenerate, neurological diseases, especially neurodegenerative disorders and stroke, characterized by irreversible cellular damage are often considered as refractory diseases. Neural stem cells (NSCs) play a unique role in the treatment of neurological diseases for their abilities to self-renew and form different neural lineage cells, such as neurons and glial cells. With the increasing understanding of neurodevelopment and advances in stem cell technology, NSCs can be obtained from different sources and directed to differentiate into a specific neural lineage cell phenotype purposefully, making it possible to replace specific cells lost in some neurological diseases, which provides new approaches to treat neurodegenerative diseases as well as stroke. In this review, we outline the advances in generating several neuronal lineage subtypes from different sources of NSCs. We further summarize the therapeutic effects and possible therapeutic mechanisms of these fated specific NSCs in neurological disease models, with special emphasis on Parkinson's disease and ischemic stroke. Finally, from the perspective of clinical translation, we compare the strengths and weaknesses of different sources of NSCs and different methods of directed differentiation, and propose future research directions for directed differentiation of NSCs in regenerative medicine.
Collapse
Affiliation(s)
- Luwei Nie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dabao Yao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Shiling Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jingyi Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chao Pan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dongcheng Wu
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, 430030, China
- Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, 430030, China
| | - Na Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
138
|
He H, Chen S, Fan Z, Dong Y, Wang Y, Li S, Sun X, Song Y, Yang J, Cao Q, Jiang J, Wang X, Wen W, Wang H. Multi-dimensional single-cell characterization revealed suppressive immune microenvironment in AFP-positive hepatocellular carcinoma. Cell Discov 2023; 9:60. [PMID: 37336873 PMCID: PMC10279759 DOI: 10.1038/s41421-023-00563-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/10/2023] [Indexed: 06/21/2023] Open
Abstract
Alpha-fetoprotein (AFP)-secreting hepatocellular carcinoma (HCC), which accounts for ~75% of HCCs, is more aggressive with a worse prognosis than those without AFP production. The mechanism through which the interaction between tumors and the microenvironment leads to distinct phenotypes is not yet clear. Therefore, our study aims to identify the characteristic features and potential treatment targets of AFP-negative HCC (ANHC) and AFP-positive HCC (APHC). We utilized single-cell RNA sequencing to analyze 6 ANHC, 6 APHC, and 4 adjacent normal tissues. Integrated multi-omics analysis together with survival analysis were also performed. Further validation was conducted via cytometry time-of-flight on 30 HCCs and multiplex immunohistochemistry on additional 59 HCCs. Our data showed that the genes related to antigen processing and interferon-γ response were abundant in tumor cells of APHC. Meanwhile, APHC was associated with multifaceted immune distortion, including exhaustion of diverse T cell subpopulations, and the accumulation of tumor-associated macrophages (TAMs). Notably, TAM-SPP1+ was highly enriched in APHC, as was its receptor CD44 on T cells and tumor cells. Targeting the Spp1-Cd44 axis restored T cell function in vitro and significantly reduced tumor burden when treated with either anti-Spp1 or anti-Cd44 antibody alone or in combination with anti-Pd-1 antibody in the mouse model. Furthermore, elevated IL6 and TGF-β1 signaling contributed to the enrichment of TAM-SPP1+ in APHC. In conclusion, this study uncovered a highly suppressive microenvironment in APHC and highlighted the role of TAM-SPP1+ in regulating the immune microenvironment, thereby revealing the SPP1-CD44 axis as a promising target for achieving a more favorable immune response in APHC treatment.
Collapse
Affiliation(s)
- Huisi He
- Third Affiliated Hospital of Naval Medical University, National Center for Liver Cancer, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Shuzhen Chen
- Third Affiliated Hospital of Naval Medical University, National Center for Liver Cancer, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Zhecai Fan
- Third Affiliated Hospital of Naval Medical University, National Center for Liver Cancer, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yaping Dong
- Third Affiliated Hospital of Naval Medical University, National Center for Liver Cancer, Shanghai, China
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Wang
- Department of Laboratory Diagnosis, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Shiyao Li
- Third Affiliated Hospital of Naval Medical University, National Center for Liver Cancer, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xiaojuan Sun
- Department of Laboratory Diagnosis, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yuting Song
- Third Affiliated Hospital of Naval Medical University, National Center for Liver Cancer, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jinxian Yang
- Third Affiliated Hospital of Naval Medical University, National Center for Liver Cancer, Shanghai, China
- Department of Laboratory Diagnosis, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Qiqi Cao
- Third Affiliated Hospital of Naval Medical University, National Center for Liver Cancer, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jie Jiang
- Third Affiliated Hospital of Naval Medical University, National Center for Liver Cancer, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xianming Wang
- Third Affiliated Hospital of Naval Medical University, National Center for Liver Cancer, Shanghai, China
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen Wen
- Third Affiliated Hospital of Naval Medical University, National Center for Liver Cancer, Shanghai, China.
- Department of Laboratory Diagnosis, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China.
| | - Hongyang Wang
- Third Affiliated Hospital of Naval Medical University, National Center for Liver Cancer, Shanghai, China.
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China.
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
139
|
Szepanowski RD, Haupeltshofer S, Vonhof SE, Frank B, Kleinschnitz C, Casas AI. Thromboinflammatory challenges in stroke pathophysiology. Semin Immunopathol 2023:10.1007/s00281-023-00994-4. [PMID: 37273022 DOI: 10.1007/s00281-023-00994-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/27/2023] [Indexed: 06/06/2023]
Abstract
Despite years of encouraging translational research, ischemic stroke still remains as one of the highest unmet medical needs nowadays, causing a tremendous burden to health care systems worldwide. Following an ischemic insult, a complex signaling pathway emerges leading to highly interconnected thrombotic as well as neuroinflammatory signatures, the so-called thromboinflammatory cascade. Here, we thoroughly review the cell-specific and time-dependent role of different immune cell types, i.e., neutrophils, macrophages, T and B cells, as key thromboinflammatory mediators modulating the neuroinflammatory response upon stroke. Similarly, the relevance of platelets and their tight crosstalk with a variety of immune cells highlights the relevance of this cell-cell interaction during microvascular dysfunction, neovascularization, and cellular adhesion. Ultimately, we provide an up-to-date overview of therapeutic approaches mechanistically targeting thromboinflammation currently under clinical translation, especially focusing on phase I to III clinical trials.
Collapse
Affiliation(s)
- R D Szepanowski
- Department of Neurology, University Hospital Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen, Germany
| | - S Haupeltshofer
- Department of Neurology, University Hospital Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen, Germany
| | - S E Vonhof
- Department of Neurology, University Hospital Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen, Germany
| | - B Frank
- Department of Neurology, University Hospital Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen, Germany
| | - C Kleinschnitz
- Department of Neurology, University Hospital Essen, Essen, Germany.
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen, Germany.
| | - A I Casas
- Department of Neurology, University Hospital Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen, Germany
- Department of Pharmacology and Personalised Medicine, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
140
|
Shan Y, Wang L, Sun J, Chang S, Di W, Lv H. Exercise preconditioning attenuates cerebral ischemia-induced neuronal apoptosis, Th17/Treg imbalance, and inflammation in rats by inhibiting the JAK2/STAT3 pathway. Brain Behav 2023; 13:e3030. [PMID: 37143406 PMCID: PMC10275560 DOI: 10.1002/brb3.3030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Exercise preconditioning (EP) is essential for preventing ischemic stroke. Recent studies have shown that EP exerts neuroprotective effects in the cerebral ischemia-reperfusion injury model. Nonetheless, there have been few reports on the relationship between EP and the Th17/Treg balance. Moreover, it is unclear whether the JAK2/STAT3 pathway is responsible for the neuroprotective effect of EP. Therefore, we aimed to explore the impact of EP, other than the anti-inflammatory and antiapoptotic functions, on the Th17/Treg balance via the JAK2/STAT3 pathway in a middle cerebral artery occlusion (MCAO)-induced model. RESULTS Fifty rats were randomly allocated into five groups, including the sham group (n = 10), EP+sham group (n = 10), MCAO group (n = 10), EP+MCAO group (n = 10), and EP+MCAO+JAK2/STAT3 pathway agonist (coumermycin A1, CA1) group (n = 10). The results indicated that EP alleviated neurological deficits, reduced infarct volume, and ameliorated neuronal apoptosis induced by MCAO. Additionally, the MCAO-induced Th17/Treg imbalance could be rectified by EP. The decreased levels of IL-10 and Foxp3 and increased IL-17 and RORα in the MCAO group were reversed by EP treatment. Regarding inflammation, EP reduced the concentrations of IL-6 and IL-17 and elevated those of IL-10 and TGF-β. The neuroprotective effects of EP were accompanied by decreased phosphorylation of JAK2 and STAT3. Furthermore, CA1 pretreatment diminished all the beneficial effects of EP partially. CONCLUSION Our findings suggest that EP contributes to attenuating neuronal apoptosis, Th17/Treg imbalance, and inflammation induced by MCAO via inhibiting the JAK2/STAT3 pathway, indicating its therapeutic potential in ischemic stroke.
Collapse
Affiliation(s)
- Yuan Shan
- Department of NeurologyShaanxi Provincial People's HospitalXi'anChina
| | - Le Wang
- Department of NeurologyShaanxi Provincial People's HospitalXi'anChina
| | - Jingying Sun
- Central Research LaboratoryShaanxi Provincial People's HospitalXi'anChina
| | - Sha Chang
- Department of NeurologyShaanxi Provincial People's HospitalXi'anChina
| | - Wei Di
- Department of NeurologyShaanxi Provincial People's HospitalXi'anChina
| | - Hua Lv
- Department of NeurologyShaanxi Provincial People's HospitalXi'anChina
| |
Collapse
|
141
|
Wang W, Ye CH, Deng ZF, Wang JL, Zhang L, Bao L, Xu BH, Zhu H, Guo Y, Wen Z. CD4 +CD25 + regulatory T cells decreased future liver remnant after associating liver partition and portal vein ligation for staged hepatectomy. World J Gastrointest Surg 2023; 15:917-930. [PMID: 37342857 PMCID: PMC10277939 DOI: 10.4240/wjgs.v15.i5.917] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/22/2022] [Accepted: 04/04/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Associating liver partition and portal vein ligation for staged hepatectomy (ALPPS) is an innovative surgical approach for the treatment of massive hepatocellular carcinoma (HCC), the key to successful planned stage 2 ALPPS is future liver remnant (FLR) volume growth, but the exact mechanism has not been elucidated. The correlation between regulatory T cells (Tregs) and postoperative FLR regeneration has not been reported. AIM To investigate the effect of CD4+CD25+ Tregs on FLR regeneration after ALPPS. METHODS Clinical data and specimens were collected from 37 patients who developed massive HCC treated with ALPPS. Flow cytometry was performed to detect changes in the proportion of CD4+CD25+ Tregs to CD4+ T cells in peripheral blood before and after ALPPS. To analyze the relationship between peripheral blood CD4+CD25+ Treg proportion and clinicopathological information and liver volume. RESULTS The postoperative CD4+CD25+ Treg proportion in stage 1 ALPPS was negatively correlated with the amount of proliferation volume, proliferation rate, and kinetic growth rate (KGR) of the FLR after stage 1 ALPPS. Patients with low Treg proportion had significantly higher KGR than those with high Treg proportion (P = 0.006); patients with high Treg proportion had more severe postoperative pathological liver fibrosis than those with low Treg proportion (P = 0.043). The area under the receiver operating characteristic curve between the percentage of Tregs and proliferation volume, proliferation rate, and KGR were all greater than 0.70. CONCLUSION CD4+CD25+ Tregs in the peripheral blood of patients with massive HCC at stage 1 ALPPS were negatively correlated with indicators of FLR regeneration after stage 1 ALPPS and may influence the degree of fibrosis in patients' livers. Treg percentage was highly accurate in predicting the FLR regeneration after stage 1 ALPPS.
Collapse
Affiliation(s)
- Wei Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Chun-Hui Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhen-Feng Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Ji-Long Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Ling Zhang
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Li Bao
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300000, China
| | - Bang-Hao Xu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Hai Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Ya Guo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhang Wen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
142
|
Qin C, Yang S, Chen M, Dong MH, Zhou LQ, Chu YH, Shen ZX, Bosco DB, Wu LJ, Tian DS, Wang W. Modulation of microglial metabolism facilitates regeneration in demyelination. iScience 2023; 26:106588. [PMID: 37138776 PMCID: PMC10149336 DOI: 10.1016/j.isci.2023.106588] [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: 09/09/2022] [Revised: 01/28/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Microglia exhibit diverse phenotypes in various central nervous system disorders and metabolic pathways exert crucial effects on microglial activation and effector functions. Here, we discovered two novel distinct microglial clusters, functionally associated with enhanced phagocytosis (PEMs) and myelination (MAMs) respectively, in human patients with multiple sclerosis by integrating public snRNA-seq data. Microglia adopt a PEMs phenotype during the early phase of demyelinated lesions, predominated in pro-inflammatory responses and aggravated glycolysis, while MAMs mainly emerged during the later phase, with regenerative signatures and enhanced oxidative phosphorylation. In addition, microglial triggering receptor expressed on myeloid cells 2 (Trem2) was greatly involved in the phenotype transition in demyelination, but not indispensable for microglia transition toward PEMs. Rosiglitazone could promote microglial phenotype conversion from PEMs to MAMs, thus favoring myelin repair. Taken together, these findings provide insights into therapeutic interventions targeting immunometabolism to switch microglial phenotypes and facilitate regenerative capacity in demyelination.
Collapse
Affiliation(s)
- Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ming-Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhu-Xia Shen
- Department of Cardiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai 200040, China
| | - Dale B. Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Corresponding author
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Corresponding author
| |
Collapse
|
143
|
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
|
144
|
Lemaitre P, Tareen SHK, Pasciuto E, Mascali L, Martirosyan A, Callaerts‐Vegh Z, Poovathingal S, Dooley J, Holt MG, Yshii L, Liston A. Molecular and cognitive signatures of ageing partially restored through synthetic delivery of IL2 to the brain. EMBO Mol Med 2023; 15:e16805. [PMID: 36975362 PMCID: PMC10165365 DOI: 10.15252/emmm.202216805] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
Cognitive decline is a common pathological outcome during aging, with an ill-defined molecular and cellular basis. In recent years, the concept of inflammaging, defined as a low-grade inflammation increasing with age, has emerged. Infiltrating T cells accumulate in the brain with age and may contribute to the amplification of inflammatory cascades and disruptions to the neurogenic niche observed with age. Recently, a small resident population of regulatory T cells has been identified in the brain, and the capacity of IL2-mediated expansion of this population to counter neuroinflammatory disease has been demonstrated. Here, we test a brain-specific IL2 delivery system for the prevention of neurological decline in aging mice. We identify the molecular hallmarks of aging in the brain glial compartments and identify partial restoration of this signature through IL2 treatment. At a behavioral level, brain IL2 delivery prevented the age-induced defect in spatial learning, without improving the general decline in motor skill or arousal. These results identify immune modulation as a potential path to preserving cognitive function for healthy aging.
Collapse
Affiliation(s)
- Pierre Lemaitre
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of Microbiology, Immunology and TransplantationKU LeuvenLeuvenBelgium
| | | | - Emanuela Pasciuto
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of Microbiology, Immunology and TransplantationKU LeuvenLeuvenBelgium
| | - Loriana Mascali
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of Microbiology, Immunology and TransplantationKU LeuvenLeuvenBelgium
| | - Araks Martirosyan
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of NeurosciencesKU LeuvenLeuvenBelgium
| | | | | | - James Dooley
- Immunology ProgrammeThe Babraham InstituteBabrahamUK
- Department of PathologyThe University of CambridgeCambridgeUK
| | - Matthew G Holt
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of NeurosciencesKU LeuvenLeuvenBelgium
- Instituto de Investigaçāo e Inovaçāo em Saúde (i3S)University of PortoPortoPortugal
| | - Lidia Yshii
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of Microbiology, Immunology and TransplantationKU LeuvenLeuvenBelgium
- Department of NeurosciencesKU LeuvenLeuvenBelgium
| | - Adrian Liston
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of Microbiology, Immunology and TransplantationKU LeuvenLeuvenBelgium
- Immunology ProgrammeThe Babraham InstituteBabrahamUK
- Department of PathologyThe University of CambridgeCambridgeUK
| |
Collapse
|
145
|
Gao J, Liu J, Li Y, Liu J, Wang H, Chai M, Dong Y, Zhang Z, Su G, Wang M. Targeting p53 for neuroinflammation: New therapeutic strategies in ischemic stroke. J Neurosci Res 2023. [PMID: 37156641 DOI: 10.1002/jnr.25200] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/10/2023] [Accepted: 04/21/2023] [Indexed: 05/10/2023]
Abstract
Ischemic stroke (IS) is characterized by high incidence, high recurrence, and high mortality and places a heavy burden on society and families. The pathological mechanisms of IS are complex, among which secondary neurological impairment mediated by neuroinflammation is considered to be the main factor in cerebral ischemic injury. At present, there is still a lack of specific therapies to treat neuroinflammation. The tumor suppressor protein p53 has long been regarded as a key substance in the regulation of the cell cycle and apoptosis in the past. Recently, studies have found that p53 also plays an important role in neuroinflammatory diseases, such as IS. Therefore, p53 may be a crucial target for the regulation of the neuroinflammatory response. Here, we provide a comprehensive review of the potential of targeting p53 in the treatment of neuroinflammation after IS. We describe the function of p53, the major immune cells involved in neuroinflammation, and the role of p53 in inflammatory responses mediated by these cells. Finally, we summarize the therapeutic strategies of targeting p53 in regulating the neuroinflammatory response after IS to provide new directions and ideas for the treatment of ischemic brain injury.
Collapse
Affiliation(s)
- Juan Gao
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jifei Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yonghong Li
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China
| | - Junxi Liu
- Chinese Academy of Sciences Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, China
| | - He Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Miao Chai
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Ying Dong
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhenchang Zhang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Gang Su
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
146
|
Lu W, Ji H, Wu D. SIRT2 plays complex roles in neuroinflammation neuroimmunology-associated disorders. Front Immunol 2023; 14:1174180. [PMID: 37215138 PMCID: PMC10196137 DOI: 10.3389/fimmu.2023.1174180] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Neuroinflammation and neuroimmunology-associated disorders, including ischemic stroke and neurodegenerative disease, commonly cause severe neurologic function deficits, including bradypragia, hemiplegia, aphasia, and cognitive impairment, and the pathological mechanism is not completely clear. SIRT2, an NAD+-dependent deacetylase predominantly localized in the cytoplasm, was proven to play an important and paradoxical role in regulating ischemic stroke and neurodegenerative disease. This review summarizes the comprehensive mechanism of the crucial pathological functions of SIRT2 in apoptosis, necroptosis, autophagy, neuroinflammation, and immune response. Elaborating on the mechanism by which SIRT2 participates in neuroinflammation and neuroimmunology-associated disorders is beneficial to discover novel effective drugs for diseases, varying from vascular disorders to neurodegenerative diseases.
Collapse
|
147
|
Cheru N, Hafler DA, Sumida TS. Regulatory T cells in peripheral tissue tolerance and diseases. Front Immunol 2023; 14:1154575. [PMID: 37197653 PMCID: PMC10183596 DOI: 10.3389/fimmu.2023.1154575] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/13/2023] [Indexed: 05/19/2023] Open
Abstract
Maintenance of peripheral tolerance by CD4+Foxp3+ regulatory T cells (Tregs) is essential for regulating autoreactive T cells. The loss of function of Foxp3 leads to autoimmune disease in both animals and humans. An example is the rare, X-linked recessive disorder known as IPEX (Immune Dysregulation, Polyendocrinopathy, Enteropathy X-linked) syndrome. In more common human autoimmune diseases, defects in Treg function are accompanied with aberrant effector cytokines such as IFNγ. It has recently become appreciated that Tregs plays an important role in not only maintaining immune homeostasis but also in establishing the tissue microenvironment and homeostasis of non-lymphoid tissues. Tissue resident Tregs show profiles that are unique to their local environments which are composed of both immune and non-immune cells. Core tissue-residence gene signatures are shared across different tissue Tregs and are crucial to homeostatic regulation and maintaining the tissue Treg pool in a steady state. Through interaction with immunocytes and non-immunocytes, tissue Tregs exert a suppressive function via conventional ways involving contact dependent and independent processes. In addition, tissue resident Tregs communicate with other tissue resident cells which allows Tregs to adopt to their local microenvironment. These bidirectional interactions are dependent on the specific tissue environment. Here, we summarize the recent advancements of tissue Treg studies in both human and mice, and discuss the molecular mechanisms that maintain tissue homeostasis and prevent pathogenesis.
Collapse
Affiliation(s)
- Nardos Cheru
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - David A. Hafler
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Tomokazu S. Sumida
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
148
|
Jian L, Hu Y, Gao M, Shu L. Promoted Generation of T Helper 1-Like Regulatory T Cells After Transient Middle Cerebral Artery Occlusion in Type-2 Diabetic Mice. Immunol Invest 2023:1-17. [PMID: 37076315 DOI: 10.1080/08820139.2023.2197009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
BACKGROUND Regulatory T cells (Tregs) play a remarkable role in modulating post-ischemic neuroinflammation. However, the characteristics of Tregs in diabetic ischemic stroke remain unknown. METHODS Transient middle cerebral artery occlusion (MCAO) was conducted on leptin receptor-mutated db/db mice and db/+ mice. The number, cytokine production, and signaling features of Tregs in peripheral blood and ipsilateral hemispheres were evaluated by flow cytometry. Treg plasticity was assessed by the adoptive transfer of splenic Tregs into mice. The effect of ipsilateral macrophages/microglia on Treg plasticity was determined by in vitro co-culture analysis. RESULTS db/db mice had more infiltrating Tregs in their ipsilateral hemispheres than db/+ mice. Infiltrating Tregs in db/db mice expressed higher transforming growth factor-β (TGF-β), interleukin-10 (IL-10), forkhead box P3 (Foxp3), interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and T-box expressed in T cells (T-bet) in comparison to infiltrating Tregs in db/+ mice, suggesting promoted generation of T helper 1 (Th1)-like Tregs in the brains of db/db mice after stroke. The post-ischemic brain microenvironment of db/db mice significantly up-regulated IFN-γ, TNF-α, T-bet, IL-10, and TGF-β in infiltrating Tregs. Moreover, ipsilateral macrophages/microglia remarkably enhanced the expression of IFN-γ, TNF-α, and T-bet but not IL-10 and TGF-β in Tregs. db/db macrophages/microglia were more potent in up-regulating IFN-γ, TNF-α, and T-bet than db/+ macrophages/microglia. Interleukin-12 (IL-12) blockage partially abolished the modulatory effect of macrophages/microglia on Tregs. CONCLUSION The generation of Th1-like Tregs was promoted in the brains of type 2 diabetic mice after stroke. Our study reveals significant Treg plasticity in diabetic stroke.Abbreviations: Foxp3: forkhead box P3; IFN-γ: interferon-γ; IL-10: interleukin-10; IL-12: interleukin-12; MCAO: middle cerebral artery occlusion; PBS: phosphate-buffered saline; STAT1: Signal transducer and activator of transcription 1; STAT5: Signal transducer and activator of transcription 1; T-bet: T-box expressed in T cells; TGF-β: transforming growth factor-β; Th1: T helper 1; TNF-α: tumor necrosis factor-α; Tregs: regulatory T cells. Foxp3: forkhead box P3; IFN-γ: interferon-γ; IL-10: interleukin-10; IL-12: interleukin-12; MCAO: middle cerebral artery occlusion; PBS: phosphate-buffered saline; STAT1: Signal transducer and activator of transcription 1; STAT5: Signal transducer and activator of transcription 1; T-bet: T-box expressed in T cells; TGF-β: transforming growth factor-β; Th1: T helper 1; TNF-α: tumor necrosis factor-α; Tregs: regulatory T cells.
Collapse
Affiliation(s)
- Lei Jian
- The department of Endocrinology, Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province, China
| | - Yanqi Hu
- The department of Endocrinology, Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province, China
| | - Mingjie Gao
- The Department of Neurology, Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province, China
| | - Long Shu
- The Department of Neurology, Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province, China
| |
Collapse
|
149
|
Shen X, Li M, Shao K, Li Y, Ge Z. Post-ischemic inflammatory response in the brain: Targeting immune cell in ischemic stroke therapy. Front Mol Neurosci 2023; 16:1076016. [PMID: 37078089 PMCID: PMC10106693 DOI: 10.3389/fnmol.2023.1076016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
An ischemic stroke occurs when the blood supply is obstructed to the vascular basin, causing the death of nerve cells and forming the ischemic core. Subsequently, the brain enters the stage of reconstruction and repair. The whole process includes cellular brain damage, inflammatory reaction, blood–brain barrier destruction, and nerve repair. During this process, the proportion and function of neurons, immune cells, glial cells, endothelial cells, and other cells change. Identifying potential differences in gene expression between cell types or heterogeneity between cells of the same type helps to understand the cellular changes that occur in the brain and the context of disease. The recent emergence of single-cell sequencing technology has promoted the exploration of single-cell diversity and the elucidation of the molecular mechanism of ischemic stroke, thus providing new ideas and directions for the diagnosis and clinical treatment of ischemic stroke.
Collapse
Affiliation(s)
- Xueyang Shen
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Mingming Li
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Gansu Provincial Neurology Clinical Medical Research Center, The Second Hospital of Lanzhou University, Lanzhou, China
- Expert Workstation of Academician Wang Longde, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Kangmei Shao
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Yongnan Li,
| | - Zhaoming Ge
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Gansu Provincial Neurology Clinical Medical Research Center, The Second Hospital of Lanzhou University, Lanzhou, China
- Expert Workstation of Academician Wang Longde, The Second Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Zhaoming Ge,
| |
Collapse
|
150
|
Garcia-Bonilla L, Shahanoor Z, Sciortino R, Nazarzoda O, Racchumi G, Iadecola C, Anrather J. Brain and blood single-cell transcriptomics in acute and subacute phases after experimental stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.31.535150. [PMID: 37066298 PMCID: PMC10103945 DOI: 10.1101/2023.03.31.535150] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Cerebral ischemia triggers a powerful inflammatory reaction involving both peripheral leukocytes and brain resident cells. Recent evidence indicates that their differentiation into a variety of functional phenotypes contributes to both tissue injury and repair. However, the temporal dynamics and diversity of post-stroke immune cell subsets remain poorly understood. To address these limitations, we performed a longitudinal single-cell transcriptomic study of both brain and mouse blood to obtain a composite picture of brain-infiltrating leukocytes, circulating leukocytes, microglia and endothelium diversity over the ischemic/reperfusion time. Brain cells and blood leukocytes isolated from mice 2 or 14 days after transient middle cerebral artery occlusion or sham surgery were purified by FACS sorting and processed for droplet-based single-cell transcriptomics. The analysis revealed a strong divergence of post-ischemic microglia, macrophages, and neutrophils over time, while such diversity was less evident in dendritic cells, B, T and NK cells. Conversely, brain endothelial cells and brain associated-macrophages showed altered transcriptomic signatures at 2 days post-stroke, but low divergence from sham at day 14. Pseudotime trajectory inference predicted the in-situ longitudinal progression of monocyte-derived macrophages from their blood precursors into day 2 and day 14 phenotypes, while microglia phenotypes at these two time points were not connected. In contrast to monocyte-derived macrophages, neutrophils were predicted to be continuously de-novo recruited from the blood. Brain single-cell transcriptomics from both female and male aged mice did not show major changes in respect to young mice, but aged and young brains differed in their immune cell composition. Furthermore, blood leukocyte analysis also revealed altered transcriptomes after stroke. However, brain-infiltrating leukocytes displayed higher transcriptomic divergence than their circulating counterparts, indicating that phenotypic diversification into cellular subsets occurs within the brain in the early and the recovery phase of ischemic stroke. In addition, this resource report contains a searchable database https://anratherlab.shinyapps.io/strokevis/ to allow user-friendly access to our data. The StrokeVis tool constitutes a comprehensive gene expression atlas that can be interrogated at the gene and cell type level to explore the transcriptional changes of endothelial and immune cell subsets from mouse brain and blood after stroke.
Collapse
Affiliation(s)
- Lidia Garcia-Bonilla
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| | - Ziasmin Shahanoor
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| | - Rose Sciortino
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| | - Omina Nazarzoda
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| | - Gianfranco Racchumi
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| | - Costantino Iadecola
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| | - Josef Anrather
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021
| |
Collapse
|