1
|
Wu Q, Xie Z, Cao X, Hu D, Sheng L, Guo X, Yan D, Ding C, Li C, Xiao J, Liu C, Wu K, Gong Y, Fan Q, Wang Q, Liu J, Liu Y. Chaihu-Shugan-San alleviates post-stroke depression in mice: Mechanistic insights into exosome-mediated neuroprotection. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119700. [PMID: 40154896 DOI: 10.1016/j.jep.2025.119700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Post-stroke depression (PSD) is common among stroke survivors and negatively impacts recovery. Chaihu-Shugan-San (CSS), a traditional Chinese medicine, has shown therapeutic potential for mood disorders, particularly PSD. Recent studies suggest that CSS's effects may be mediated by exosomes, but the mechanisms remain unclear. AIM OF STUDY This study aimed to evaluate the therapeutic effects of CSS on PSD in mice and investigate the underlying mechanisms, particularly the role of exosomes. MATERIALS AND METHODS Active compounds in CSS were identified from rat serum using liquid chromatography-mass spectrometry (LC-MS) and analyzed through network pharmacology. In vitro, an oxygen-glucose deprivation/reperfusion (OGD/R) BV2 microglia model was used to assess the effects of CSS-containing serum (CSS-S). Exosomes from OGD/R-treated BV2 microglia were isolated, labeled with PKH26, and analyzed using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). In vivo, a photothrombotic stroke (PT) model combined with chronic unpredictable mild stress (CUMS) was used to induce PSD in mice. Behavioral assessments and histological analysis were performed, along with immunofluorescence (IF), ELISA and q-PCR to measure key protein and miR-146 expression in the hippocampus. RESULTS CSS treatment significantly alleviated depressive-like behaviors in the PSD mouse model. Mice treated with high-dose CSS (4.2 g/kg) exhibited increased sucrose preference, reduced immobility in the tail suspension test (TST) and forced swimming test (FST), and enhanced exploratory activity in the open field test (OFT). Histological analysis demonstrated that CSS treatment improved brain tissue integrity, alleviating neuronal damage and reducing neuroinflammation. Exosome analysis revealed that CSS increased the expression of microglia-derived exosomes in the hippocampus, which were shown to carry miR-146. Further examination of miR-146 isoforms in the hippocampal tissue revealed significant changes: miR-146b-3p and miR-146a-5p were upregulated, while miR-146a-3p and miR-146b-5p were downregulated in PSD mice. Treatment with CSS reversed the altered miRNA expression, indicating a potential mechanism for its neuroprotective effects. Additionally, CSS treatment reduced the expression of inflammatory cytokines such as S100A8, IL1β, IL6, and TNF-α, while restoring the levels of angiogenic factors VEGFC and VEGFR3. ELISA measurements showed significant decreases in cyclic AMP response element-binding protein (CREB), brain-derived neurotrophic factor (BDNF), 5-hydroxytryptamine (5-HT), dopamine (DA), and noradrenaline (NE) in PSD mice; high-dose CSS notably elevated CREB and BDNF levels and showed comparable effects to fluoxetine in restoring 5-HT and DA levels. Additionally, the calcium signaling pathway was implicated, with altered mRNA expressions of CaMKIIα, CREB, phosphorylated CREB (p-CREB), PDE4D, and BDNF, although fluoxetine demonstrated stronger modulatory effects than CSS. CONCLUSIONS CSS alleviates PSD in mice by modulating exosome-mediated signaling, particularly through the regulation of miR-146. The treatment reversed abnormal miRNA expression, reduced neuroinflammation, and improved synaptic function. These findings highlight CSS's potential as an effective therapeutic strategy for PSD by targeting exosome-mediated neuroprotection and miR-146 regulation.
Collapse
Affiliation(s)
- Qiqing Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Zhouyuan Xie
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Xinyue Cao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Dan Hu
- Department of Neurology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, 210017, China.
| | - Lei Sheng
- Department of Neurology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, 210017, China.
| | - Xueyan Guo
- Department of Neurology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, 210017, China.
| | - Dong Yan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Caixia Ding
- Department of Neurology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, 210017, China.
| | - Chuanyou Li
- Department of Neurology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, 210017, China.
| | - Jing Xiao
- Department of Neurology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, 210017, China.
| | - Chunyu Liu
- Department of Neurology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, 210017, China.
| | - Ke Wu
- Department of Neurology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, 210017, China.
| | - Yue Gong
- Department of Neurology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, 210017, China.
| | - Qiqi Fan
- Central Laboratory, Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen, 529099, China.
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Jinman Liu
- Central Laboratory, Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen, 529099, China.
| | - Yuanyue Liu
- Department of Neurology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, 210017, China.
| |
Collapse
|
2
|
Chen S, Han C, Shi Z, Guan X, Cheng L, Wang L, Zou W, Liu J. Umbilical mesenchymal stem cells mitigate T-cell compartments shift and Th17/Treg imbalance in acute ischemic stroke via mitochondrial transfer. Stem Cell Res Ther 2025; 16:134. [PMID: 40075467 PMCID: PMC11905603 DOI: 10.1186/s13287-025-04224-6] [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: 09/24/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Acute ischemic stroke (AIS) initiates secondary injuries that worsen neurological damage and hinder recovery. While peripheral immune responses play a key role in stroke outcomes, clinical results from immunotherapy have been suboptimal, with limited focus on T-cell dynamics. Umbilical mesenchymal stem cells (UMSCs) offer therapeutic potential due to their immunomodulatory properties. They can regulate immune responses and reduce neuroinflammation, potentially enhancing recovery by fostering a pro-regenerative peripheral immune environment. However, the effect of UMSCs on T-cell dynamics in AIS remains underexplored. This study investigates T-cell dynamics following AIS and examines how UMSCs may mitigate immune dysregulation to develop better treatment strategies. METHODS AIS patients (NIHSS scores 0-15) were recruited within 72 h of stroke onset, with peripheral blood samples collected on Day 0 (enrollment) and Day 7. T-cell compartments were identified by flow cytometry, and plasma cytokine levels were quantified using a cytometric bead array (CBA). Mitochondria in UMSCs were labeled with MitoTracker. Peripheral blood mononuclear cells from patients were isolated, treated with lipopolysaccharide (LPS), and cocultured with UMSCs in both direct contact and Transwell systems. Flow cytometry, CBA, RT-qPCR, and immunofluorescence assays were used to detect T-cell compartments, gene expression markers for helper T (Th) cell differentiation, cytokine profiles, mitochondrial transfer, reactive oxygen species (ROS) production, and mitochondrial membrane potential. Additionally, mitochondrial DNA in UMSCs was depleted. The effects of UMSCs and mitochondria-depleted UMSCs on ischemic stroke mice were compared through behavioral assessments and analysis of the peripheral immune microenvironment. RESULTS In AIS, T-cell compartments underwent a phenotypic shift from naïve to effector or memory states, with a specific increase in Th17 cells and a decrease in regulatory T cells, leading to alterations in T-cell-mediated immune functions. In an ex vivo co-culture system, LPS stimulation further amplified these disparities, inducing mitochondrial dysfunction and oxidative stress in T cells. Notably, UMSCs restored mitochondrial function and reversed the shift in T-cell compartments through mitochondrial transfer. Critically, UMSC treatment significantly improved both neurological deficits and peripheral immune disorders in ischemic stroke mice, whereas mitochondria-depleted UMSCs failed to produce this effect. CONCLUSIONS Our comprehensive insights into the key attributes of T-cell compartments in acute ischemic stroke and the immune regulatory mechanisms of UMSCs provide a crucial theoretical foundation for understanding peripheral immune disorders in ischemic stroke and the therapeutic potential of UMSC treatment.
Collapse
Affiliation(s)
- Shuna Chen
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Chao Han
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China
- College of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, People's Republic of China
| | - Zihan Shi
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China
- College of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, People's Republic of China
| | - Xin Guan
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China
| | - Liyuan Cheng
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Liang Wang
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China
- College of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, People's Republic of China
| | - Wei Zou
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China
| | - Jing Liu
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China.
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China.
| |
Collapse
|
3
|
Zeng X, Ma C, Fu W, Xu Y, Wang R, Liu D, Zhang L, Hu N, Li D, Li W. Changes in Type 1 Diabetes-Associated Gut Microbiota Aggravate Brain Ischemia Injury by Affecting Microglial Polarization Via the Butyrate-MyD88 Pathway in Mice. Mol Neurobiol 2025; 62:3764-3780. [PMID: 39322832 DOI: 10.1007/s12035-024-04514-9] [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: 02/25/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
People with type 1 diabetes (T1D) have a significantly elevated risk of stroke, but the mechanism through which T1D worsens ischemic stroke remains unclear. This study was aimed at investigating the roles of T1D-associated changes in the gut microbiota in aggravating ischemic stroke and the underlying mechanism. Fecal 16SrRNA sequencing indicated that T1D mice and mice with transplantation of T1D mouse gut microbiota had lower relative abundance of butyric acid producers, f_Erysipelotrichaceae and g_Allobaculum, and lower content of butyric acid in feces. After middle cerebral artery occlusion (MCAO), these mice had poorer neurological outcomes and more severe inflammation, but higher expression of myeloid differentiation factor 88 (MyD88) in the ischemic penumbra; moreover, the microglia were inclined to polarize toward the pro-inflammatory type. Administration of butyrate to T1D mice in the drinking water alleviated the neurological damage after MCAO. Butyrate influenced the response and polarization of BV2 and decreased the production of inflammatory cytokines via MyD88 after oxygen-glucose deprivation/reoxygenation. Knocking down MyD88 in the brain alleviated neurological outcomes and decreased the concentrations of inflammatory cytokines in the brain after stroke in mice with transplantation of T1D mouse gut microbiota. Poor neurological outcomes and aggravated inflammatory responses of T1D mice after ischemic stroke may be partly due to differences in microglial polarization mediated by the gut microbiota-butyrate-MyD88 pathway. These findings provide new ideas and potential intervention targets for alleviating neurological damage after ischemic stroke in T1D.
Collapse
Affiliation(s)
- Xianzhang Zeng
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Can Ma
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Wenchao Fu
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Yongmei Xu
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Rui Wang
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Dan Liu
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Lijuan Zhang
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Narisu Hu
- Oral Implant Center, Second Affiliated Hospital, Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Dongmei Li
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Wenzhi Li
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, 246 Xuefu Road, Harbin, 150001, Heilongjiang, People's Republic of China.
| |
Collapse
|
4
|
Li Y, Guan X, Lan T, Zhang ZR, Zhang Y, Jiang S, Li M, Shi FD, Jin WN. The miR-451a facilitates natural killer cell-associated immune deficiency after ischemic stroke. J Cereb Blood Flow Metab 2025:271678X251321641. [PMID: 39985210 PMCID: PMC11846095 DOI: 10.1177/0271678x251321641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/16/2024] [Accepted: 01/31/2025] [Indexed: 02/24/2025]
Abstract
Ischemic stroke is a devastating neurological disease. Brain ischemia impairs systemic immune responses and heightens susceptibility to infections, though the underlying mechanisms remain incompletely understood. Natural killer (NK) cells exhibited decreased frequency and compromised function after acute stage of stroke, resulting in NK cell-associated immune deficiency and increased risk of infection. MicroRNAs (miRNAs) are post-transcriptional molecular modulators. Our previous study revealed a significant upregulation of miR-451a in circulating NK cells from patients with ischemic stroke, but its effects and precise mechanism on immune defense remain elusive. In this study, we observed a substantial elevation of miR-451a level in brain and splenic NK cells in murine model of ischemic stroke miR-451a mimics suppressed NK cell activation and cytotoxicity within the ischemic brain and periphery, including a downregulation of activation marker CD69, and reduced production of effector molecules IFN-γ and perforin. Conversely, miR-451a inhibitor preserved NK cell activation and cytotoxicity, rescuing local inflammation, and reducing bacterial burden in the lung. Pharmacological inhibition of Akt-mTOR pathway with AZD8055 effectively blocked the impacts of miR-451a on NK cell functions. Collectively, these findings suggest miR-451a negatively regulates NK cell cytotoxicity in both the brain and periphery, which could be re-addressed by modulating the Akt-mTOR signaling pathway.
Collapse
Affiliation(s)
- Yan Li
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiuchen Guan
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Tian Lan
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhuo-ran Zhang
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Ying Zhang
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shihe Jiang
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Minshu Li
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Fu-Dong Shi
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei-Na Jin
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
He JQ, Yuan RL, Jiang YT, Peng Y, Ye JR, Wang SS, Li LQ, Ruan Y, Li PY, Yan X, He WB, Li G, Chu SF, Zhang Z, Chen NH. Esculetin facilitates post-stroke rehabilitation by inhibiting CKLF1-mediated neutrophil infiltration. Acta Pharmacol Sin 2025; 46:52-65. [PMID: 39112771 PMCID: PMC11697434 DOI: 10.1038/s41401-024-01352-2] [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: 01/11/2024] [Accepted: 07/02/2024] [Indexed: 01/04/2025]
Abstract
Esculetin (ESC) is a coumarin-derived phytochemical prevalent in traditional Chinese medicine that exhibits anti-acute ischemic stroke activities. Our previous studies demonstrate that CKLF1 is a potential anti-stroke target for coumarin-derived compound. In this study we investigated whether CKLF1 was involved in the neuroprotective effects of ESC against photothrombotic stroke in mice. The mice were treated with ESC (20, 40 or 80 mg·kg-1·d-1, i.g.) for two weeks. The therapeutic effect of ESC was assessed using MRI, neurological function evaluation, and a range of behavioral tests on D1, 3, 7 and 14 of ESC administration. We showed that oral administration of ESC dose-dependently reduced the cerebral infarction volume within one week after stroke, improved behavioral performance, and alleviated neuropathological damage within two weeks. Functional MRI revealed that ESC significantly enhanced the abnormal low-frequency fluctuation (ALFF) value of the motor cortex and promoted functional connectivity between the supplementary motor area (SMA) and multiple brain regions. We demonstrated that ESC significantly reduced the protein levels of CKLF1 and CCR5, as well as the CKLF1/CCR5 protein complex in the peri-infarcted area. We showed that ESC (0.1-10 μM) dose-dependently blocked CKLF1-induced chemotactic movement of neutrophils in the Transwell assay, reducing the interaction of CKLF1/CCR5 on the surface of neutrophils, thereby reducing neutrophil infiltration, and decreasing the expression of ICAM-1, VCAM-1 and MMP-9 in the peri-infarct tissue. Knockout of CKLF1 reduced brain infarction volume and motor dysfunction after stroke but also negated the anti-stroke efficacy and neutrophil infiltration of ESC. These results suggest that the efficacy of ESC in promoting post-stroke neural repair depends on its inhibition on CKLF1-mediated neutrophil infiltration, which offering novel perspectives for elucidating the therapeutic properties of coumarins.
Collapse
Affiliation(s)
- Jia-Qi He
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ruo-Lan Yuan
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yu-Tong Jiang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ye Peng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jun-Rui Ye
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Sha-Sha Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Li-Qing Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yuan Ruan
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Pei-Yi Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xu Yan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wen-Bin He
- National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China
| | - Gang Li
- Graduate school, Inner Mongolian Medical University, Hohhot, 010110, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Nai-Hong Chen
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
6
|
Rust R, Nih LR, Liberale L, Yin H, El Amki M, Ong LK, Zlokovic BV. Brain repair mechanisms after cell therapy for stroke. Brain 2024; 147:3286-3305. [PMID: 38916992 PMCID: PMC11449145 DOI: 10.1093/brain/awae204] [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: 03/11/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Cell-based therapies hold great promise for brain repair after stroke. While accumulating evidence confirms the preclinical and clinical benefits of cell therapies, the underlying mechanisms by which they promote brain repair remain unclear. Here, we briefly review endogenous mechanisms of brain repair after ischaemic stroke and then focus on how different stem and progenitor cell sources can promote brain repair. Specifically, we examine how transplanted cell grafts contribute to improved functional recovery either through direct cell replacement or by stimulating endogenous repair pathways. Additionally, we discuss recently implemented preclinical refinement methods, such as preconditioning, microcarriers, genetic safety switches and universal (immune evasive) cell transplants, as well as the therapeutic potential of these pharmacologic and genetic manipulations to further enhance the efficacy and safety of cell therapies. By gaining a deeper understanding of post-ischaemic repair mechanisms, prospective clinical trials may be further refined to advance post-stroke cell therapy to the clinic.
Collapse
Affiliation(s)
- Ruslan Rust
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
| | - Lina R Nih
- Department of Brain Health, University of Nevada, Las Vegas, NV 89154, USA
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Lin Kooi Ong
- School of Health and Medical Sciences & Centre for Health Research, University of Southern Queensland, Toowoomba, QLD 4350, Australia
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
7
|
Dinakis E, O'Donnell JA, Marques FZ. The gut-immune axis during hypertension and cardiovascular diseases. Acta Physiol (Oxf) 2024; 240:e14193. [PMID: 38899764 DOI: 10.1111/apha.14193] [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: 02/02/2024] [Revised: 05/04/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
The gut-immune axis is a relatively novel phenomenon that provides mechanistic links between the gut microbiome and the immune system. A growing body of evidence supports it is key in how the gut microbiome contributes to several diseases, including hypertension and cardiovascular diseases (CVDs). Evidence over the past decade supports a causal link of the gut microbiome in hypertension and its complications, including myocardial infarction, atherosclerosis, heart failure, and stroke. Perturbations in gut homeostasis such as dysbiosis (i.e., alterations in gut microbial composition) may trigger immune responses that lead to chronic low-grade inflammation and, ultimately, the development and progression of these conditions. This is unsurprising, as the gut harbors one of the largest numbers of immune cells in the body, yet is a phenomenon not entirely understood in the context of cardiometabolic disorders. In this review, we discuss the role of the gut microbiome, the immune system, and inflammation in the context of hypertension and CVD, and consolidate current evidence of this complex interplay, whilst highlighting gaps in the literature. We focus on diet as one of the major modulators of the gut microbiota, and explain key microbial-derived metabolites (e.g., short-chain fatty acids, trimethylamine N-oxide) as potential mediators of the communication between the gut and peripheral organs such as the heart, arteries, kidneys, and the brain via the immune system. Finally, we explore the dual role of both the gut microbiome and the immune system, and how they work together to not only contribute, but also mitigate hypertension and CVD.
Collapse
Affiliation(s)
- Evany Dinakis
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Maida CD, Norrito RL, Rizzica S, Mazzola M, Scarantino ER, Tuttolomondo A. Molecular Pathogenesis of Ischemic and Hemorrhagic Strokes: Background and Therapeutic Approaches. Int J Mol Sci 2024; 25:6297. [PMID: 38928006 PMCID: PMC11203482 DOI: 10.3390/ijms25126297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Stroke represents one of the neurological diseases most responsible for death and permanent disability in the world. Different factors, such as thrombus, emboli and atherosclerosis, take part in the intricate pathophysiology of stroke. Comprehending the molecular processes involved in this mechanism is crucial to developing new, specific and efficient treatments. Some common mechanisms are excitotoxicity and calcium overload, oxidative stress and neuroinflammation. Furthermore, non-coding RNAs (ncRNAs) are critical in pathophysiology and recovery after cerebral ischemia. ncRNAs, particularly microRNAs, and long non-coding RNAs (lncRNAs) are essential for angiogenesis and neuroprotection, and they have been suggested to be therapeutic, diagnostic and prognostic tools in cerebrovascular diseases, including stroke. This review summarizes the intricate molecular mechanisms underlying ischemic and hemorrhagic stroke and delves into the function of miRNAs in the development of brain damage. Furthermore, we will analyze new perspectives on treatment based on molecular mechanisms in addition to traditional stroke therapies.
Collapse
Affiliation(s)
- Carlo Domenico Maida
- Department of Internal Medicine, S. Elia Hospital, 93100 Caltanissetta, Italy;
- Molecular and Clinical Medicine Ph.D. Programme, University of Palermo, 90133 Palermo, Italy
| | - Rosario Luca Norrito
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90133 Palermo, Italy; (R.L.N.); (M.M.); (A.T.)
| | - Salvatore Rizzica
- Department of Internal Medicine, S. Elia Hospital, 93100 Caltanissetta, Italy;
| | - Marco Mazzola
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90133 Palermo, Italy; (R.L.N.); (M.M.); (A.T.)
| | - Elisa Rita Scarantino
- Division of Geriatric and Intensive Care Medicine, Azienda Ospedaliera Universitaria Careggi, University of Florence, 50134 Florence, Italy;
| | - Antonino Tuttolomondo
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90133 Palermo, Italy; (R.L.N.); (M.M.); (A.T.)
| |
Collapse
|
9
|
Prescott K, Cothren TO, Holsten JT, Evonko CJ, Doyle EC, Bullock FE, Marron PT, Staton JG, Hatvany LS, Flack JW, Beuschel SL, MacQueen DA, Peterson TC. Increased sensitivity in detection of deficits following two commonly used animal models of stroke. Behav Brain Res 2024; 467:114991. [PMID: 38614209 DOI: 10.1016/j.bbr.2024.114991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/18/2024] [Accepted: 04/04/2024] [Indexed: 04/15/2024]
Abstract
Stroke is a leading cause of death and disability in the United States. Most strokes are ischemic, resulting in both cognitive and motor impairments. Animal models of ischemic stroke such as the distal middle cerebral artery occlusion (dMCAO) and photothrombotic stroke (PTS) procedures have become invaluable tools, with their own advantages and disadvantages. The dMCAO model is clinically relevant as it occludes the artery most affected in humans, but yields variability in the infarct location as well as the behavioral and cognitive phenotypes disrupted. The PTS model has the advantage of allowing for targeted location of infarct, but is less clinically relevant. The present study evaluates phenotype disruption over time in mice subjected to either dMCAO, PTS, or a sham surgery. Post-surgery, animals were tested over 28 days on standard motor tasks (grid walk, cylinder, tapered beam, and rotating beam), as well as a novel odor-based operant task; the 5:1 Odor Discrimination Task (ODT). Results demonstrate a significantly greater disturbance of motor control with PTS as compared with Sham and dMCAO. Disruption of the PTS group was detected up to 28 days post-stroke on the grid walk, and up to 7 days on the rotating and tapered beam tasks. PTS also led to significant short-term disruption of ODT performance (1-day post-surgery), exclusively in males, which appeared to be driven by motoric disruption of the lick response. Together, this data provides critical insights into the selection and optimization of animal models for ischemic stroke research. Notably, the PTS procedure was best suited for producing disruptions of motor behavior that can be detected with common behavioral assays and are relatively enduring, as is observed in human stroke.
Collapse
Affiliation(s)
- Kimberly Prescott
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| | - Taitum O Cothren
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| | - John T Holsten
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| | - Christopher J Evonko
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| | - Elan C Doyle
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| | - Faith E Bullock
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| | - Paul T Marron
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| | - Julia G Staton
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| | - Laura S Hatvany
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| | - Justin W Flack
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| | - Stacie L Beuschel
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| | - David A MacQueen
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| | - Todd C Peterson
- Department of Psychology, University of North Carolina Wilmington, 601 College Road, Wilmington, NC 28428, United States.
| |
Collapse
|
10
|
Duan M, Xu Y, Li Y, Feng H, Chen Y. Targeting brain-peripheral immune responses for secondary brain injury after ischemic and hemorrhagic stroke. J Neuroinflammation 2024; 21:102. [PMID: 38637850 PMCID: PMC11025216 DOI: 10.1186/s12974-024-03101-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024] Open
Abstract
The notion that the central nervous system is an immunologically immune-exempt organ has changed over the past two decades, with increasing evidence of strong links and interactions between the central nervous system and the peripheral immune system, both in the healthy state and after ischemic and hemorrhagic stroke. Although primary injury after stroke is certainly important, the limited therapeutic efficacy, poor neurological prognosis and high mortality have led researchers to realize that secondary injury and damage may also play important roles in influencing long-term neurological prognosis and mortality and that the neuroinflammatory process in secondary injury is one of the most important influences on disease progression. Here, we summarize the interactions of the central nervous system with the peripheral immune system after ischemic and hemorrhagic stroke, in particular, how the central nervous system activates and recruits peripheral immune components, and we review recent advances in corresponding therapeutic approaches and clinical studies, emphasizing the importance of the role of the peripheral immune system in ischemic and hemorrhagic stroke.
Collapse
Affiliation(s)
- Mingxu Duan
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ya Xu
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yuanshu Li
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yujie Chen
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
11
|
Khassafi N, Azami Tameh A, Mirzaei H, Rafat A, Barati S, Khassafi N, Vahidinia Z. Crosstalk between Nrf2 signaling pathway and inflammation in ischemic stroke: Mechanisms of action and therapeutic implications. Exp Neurol 2024; 373:114655. [PMID: 38110142 DOI: 10.1016/j.expneurol.2023.114655] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/03/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023]
Abstract
One of the major causes of long-term disability and mortality is ischemic stroke that enjoys limited treatment approaches. On the one hand, oxidative stress, induced by excessive generation of reactive oxygen species (ROS), plays a critical role in post-stroke inflammatory response. Increased ROS generation is one of the basic factors in the progression of stroke-induced neuroinflammation. Moreover, intravenous (IV) thrombolysis using recombinant tissue plasminogen activator (rtPA) as the only medication approved for patients with acute ischemic stroke who suffer from some clinical restrictions it could not cover the complicated episodes that happen after stroke. Thus, identifying novel therapeutic targets is crucial for successful preparation of new medicines. Recent evidence indicates that the transcription factor Nuclear factor erythroid 2-related factor 2 (Nrf2) contributes significantly to regulating the antioxidant production in cytosol, which causes antiinflammatory effects on neurons. New findings have shown a relationship between activation of the Nrf2 and glial cells, nuclear factor kappa B (NF-κB) pathway, the nucleotide-binding domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome signaling, and expression of inflammatory markers, suggesting induction of Nrf2 activation can represent a promising therapeutic alternative as the modulators of Nrf2 dependent pathways for targeting inflammatory responses in neural tissue. Hence, this review addresses the relationship of Nrf2 signaling with inflammation and Nrf2 activators' potential as therapeutic agents. This review helps to improve required knowledge for focused therapy and the creation of modern and improved treatment choices for patients with ischemic stroke.
Collapse
Affiliation(s)
- Negar Khassafi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Rafat
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran
| | - Negin Khassafi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zeinab Vahidinia
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
12
|
Gajghate S, Li H, Rom S. GPR55 Inactivation Diminishes Splenic Responses and Improves Neurological Outcomes in the Mouse Ischemia/Reperfusion Stroke Model. Cells 2024; 13:280. [PMID: 38334672 PMCID: PMC10855118 DOI: 10.3390/cells13030280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
Although strokes are frequent and severe, treatment options are scarce. Plasminogen activators, the only FDA-approved agents for clot treatment (tissue plasminogen activators (tPAs)), are used in a limited patient group. Moreover, there are few approaches for handling the brain's inflammatory reactions to a stroke. The orphan G protein-coupled receptor 55 (GPR55)'s connection to inflammatory processes has been recently reported; however, its role in stroke remains to be discovered. Post-stroke neuroinflammation involves the central nervous system (CNS)'s resident microglia activation and the infiltration of leukocytes from circulation into the brain. Additionally, splenic responses have been shown to be detrimental to stroke recovery. While lymphocytes enter the brain in small numbers, they regularly emerge as a very influential leukocyte subset that causes secondary inflammatory cerebral damage. However, an understanding of how this limited lymphocyte presence profoundly impacts stroke outcomes remains largely unclear. In this study, a mouse model for transient middle cerebral artery occlusion (tMCAO) was used to mimic ischemia followed by a reperfusion (IS/R) stroke. GPR55 inactivation, with a potent GPR55-specific antagonist, ML-193, starting 6 h after tMCAO or the absence of the GPR55 in mice (GPR55 knock out (GPR55ko)) resulted in a reduced infarction volume, improved neurological outcomes, and decreased splenic responses. The inhibition of GPR55 with ML-193 diminished CD4+T-cell spleen egress and attenuated CD4+T-cell brain infiltration. Additionally, ML-193 treatment resulted in an augmented number of regulatory T cells (Tregs) in the brain post-tMCAO. Our report offers documentation and the functional evaluation of GPR55 in the brain-spleen axis and lays the foundation for refining therapeutics for patients after ischemic attacks.
Collapse
Affiliation(s)
- Sachin Gajghate
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Hongbo Li
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Slava Rom
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| |
Collapse
|
13
|
Liu F, Cheng X, Zhao C, Zhang X, Liu C, Zhong S, Liu Z, Lin X, Qiu W, Zhang X. Single-Cell Mapping of Brain Myeloid Cell Subsets Reveals Key Transcriptomic Changes Favoring Neuroplasticity after Ischemic Stroke. Neurosci Bull 2024; 40:65-78. [PMID: 37755676 PMCID: PMC10774469 DOI: 10.1007/s12264-023-01109-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/27/2023] [Indexed: 09/28/2023] Open
Abstract
Interactions between brain-resident and peripheral infiltrated immune cells are thought to contribute to neuroplasticity after cerebral ischemia. However, conventional bulk sequencing makes it challenging to depict this complex immune network. Using single-cell RNA sequencing, we mapped compositional and transcriptional features of peri-infarct immune cells. Microglia were the predominant cell type in the peri-infarct region, displaying a more diverse activation pattern than the typical pro- and anti-inflammatory state, with axon tract-associated microglia (ATMs) being associated with neuronal regeneration. Trajectory inference suggested that infiltrated monocyte-derived macrophages (MDMs) exhibited a gradual fate trajectory transition to activated MDMs. Inter-cellular crosstalk between MDMs and microglia orchestrated anti-inflammatory and repair-promoting microglia phenotypes and promoted post-stroke neurogenesis, with SOX2 and related Akt/CREB signaling as the underlying mechanisms. This description of the brain's immune landscape and its relationship with neurogenesis provides new insight into promoting neural repair by regulating neuroinflammatory responses.
Collapse
Affiliation(s)
- Fangxi Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xi Cheng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Chuansheng Zhao
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
- Stroke Center, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaoqian Zhang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Chang Liu
- Stroke Center, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Shanshan Zhong
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Zhouyang Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xinyu Lin
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Xiuchun Zhang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
14
|
Hood RJ, Sanchez-Bezanilla S, Beard DJ, Rust R, Turner RJ, Stuckey SM, Collins-Praino LE, Walker FR, Nilsson M, Ong LK. Leakage beyond the primary lesion: A temporal analysis of cerebrovascular dysregulation at sites of hippocampal secondary neurodegeneration following cortical photothrombotic stroke. J Neurochem 2023; 167:733-752. [PMID: 38010732 DOI: 10.1111/jnc.16008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/29/2023]
Abstract
We have previously demonstrated that a cortical stroke causes persistent impairment of hippocampal-dependent cognitive tasks concomitant with secondary neurodegenerative processes such as amyloid-β accumulation in the hippocampus, a region remote from the primary infarct. Interestingly, there is emerging evidence suggesting that deposition of amyloid-β around cerebral vessels may lead to cerebrovascular structural changes, neurovascular dysfunction, and disruption of blood-brain barrier integrity. However, there is limited knowledge about the temporal changes of hippocampal cerebrovasculature after cortical stroke. In the current study, we aimed to characterise the spatiotemporal cerebrovascular changes after cortical stroke. This was done using the photothrombotic stroke model targeting the motor and somatosensory cortices of mice. Cerebrovascular morphology as well as the co-localisation of amyloid-β with vasculature and blood-brain barrier integrity were assessed in the cortex and hippocampal regions at 7, 28 and 84 days post-stroke. Our findings showed transient cerebrovascular remodelling in the peri-infarct area up to 28 days post-stroke. Importantly, the cerebrovascular changes were extended beyond the peri-infarct region to the ipsilateral hippocampus and were sustained out to 84 days post-stroke. When investigating vessel diameter, we showed a decrease at 84 days in the peri-infarct and CA1 regions that were exacerbated in vessels with amyloid-β deposition. Lastly, we showed sustained vascular leakage in the peri-infarct and ipsilateral hippocampus, indicative of a compromised blood-brain-barrier. Our findings indicate that hippocampal vasculature may represent an important therapeutic target to mitigate the progression of post-stroke cognitive impairment.
Collapse
Affiliation(s)
- Rebecca J Hood
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Sonia Sanchez-Bezanilla
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Daniel J Beard
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Ruslan Rust
- Institute for Regenerative Medicine (IREM), University of Zurich, Schlieren, Switzerland
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Renée J Turner
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Shannon M Stuckey
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Lyndsey E Collins-Praino
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Frederick R Walker
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Centre for Rehab Innovations, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Michael Nilsson
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Centre for Rehab Innovations, The University of Newcastle, Callaghan, New South Wales, Australia
- School of Medicine and Public Health, The University of Newcastle, Callaghan, New South Wales, Australia
- LKC School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Lin Kooi Ong
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
- School of Health and Medical Sciences & Centre for Health Research, University of Southern Queensland, Toowoomba, Queensland, Australia
| |
Collapse
|
15
|
Waseem A, Saudamini, Haque R, Janowski M, Raza SS. Mesenchymal stem cell-derived exosomes: Shaping the next era of stroke treatment. NEUROPROTECTION 2023; 1:99-116. [PMID: 38283953 PMCID: PMC10811806 DOI: 10.1002/nep3.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/05/2023] [Accepted: 11/10/2023] [Indexed: 01/30/2024]
Abstract
Exosome-based treatments are gaining traction as a viable approach to addressing the various issues faced by an ischemic stroke. These extracellular vesicles, mainly produced by Mesenchymal Stem Cells (MSCs), exhibit many properties with substantial therapeutic potential. Exosomes are particularly appealing for stroke therapy because of their low immunogenicity, effective cargo transport, and ability to cross the blood-brain barrier. Their diverse effects include neuroprotection, angiogenesis stimulation, inflammatory response modulation, and cell death pathway attenuation, synergistically promoting neuronal survival, tissue regeneration, and functional recovery. Exosomes also show potential as diagnostic indicators for early stroke identification and customized treatment options. Despite these promising qualities, current exosome-based therapeutics have some limitations. The heterogeneity of exosome release among cell types, difficulty in standardization and isolation techniques, and complications linked to dosage and targeted administration necessitates extensive investigation. It is critical to thoroughly understand exosomal processes and their complicated interactions within the cellular milieu. To improve the practicality and efficacy of exosome-based medicines, research efforts must focus on improving production processes, developing robust evaluation criteria, and developing large-scale isolation techniques. Altogether, exosomes' multifunctional properties offer a new route for transforming stroke treatment and significantly improving patient outcomes.
Collapse
Affiliation(s)
- Arshi Waseem
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College HospitalEra University, SarfarazganjLucknowIndia
| | - Saudamini
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College HospitalEra University, SarfarazganjLucknowIndia
- Department of BiotechnologyCentral University of South BiharGayaIndia
| | - Rizwanul Haque
- Department of BiotechnologyCentral University of South BiharGayaIndia
| | - Miroslaw Janowski
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear MedicineUniversity of MarylandBaltimoreMarylandUSA
| | - Syed S. Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College HospitalEra University, SarfarazganjLucknowIndia
- Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College HospitalEra University, SarfarazganjLucknowIndia
| |
Collapse
|
16
|
Brunelli S, Giannella E, Bizzaglia M, De Angelis D, Sancesario GM. Secondary neurodegeneration following Stroke: what can blood biomarkers tell us? Front Neurol 2023; 14:1198216. [PMID: 37719764 PMCID: PMC10502514 DOI: 10.3389/fneur.2023.1198216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/14/2023] [Indexed: 09/19/2023] Open
Abstract
Stroke is one of the leading causes of death and the primary source of disability in adults, resulting in neuronal necrosis of ischemic areas, and in possible secondary degeneration of regions surrounding or distant to the initial damaged area. Secondary neurodegeneration (SNDG) following stroke has been shown to have different pathogenetic origins including inflammation, neurovascular response and cytotoxicity, but can be associated also to regenerative processes. Aside from focal neuronal loss, ipsilateral and contralateral effects distal to the lesion site, disruptions of global functional connectivity and a transcallosal diaschisis have been reported in the chronic stages after stroke. Furthermore, SNDG can be observed in different areas not directly connected to the primary lesion, such as thalamus, hippocampus, amygdala, substantia nigra, corpus callosum, bilateral inferior fronto-occipital fasciculus and superior longitudinal fasciculus, which can be highlighted by neuroimaging techniques. Although the clinical relevance of SNDG following stroke has not been well understood, the identification of specific biomarkers that reflect the brain response to the damage, is of paramount importance to investigate in vivo the different phases of stroke. Actually, brain-derived markers, particularly neurofilament light chain, tau protein, S100b, in post-stroke patients have yielded promising results. This review focuses on cerebral morphological modifications occurring after a stroke, on associated cellular and molecular changes and on state-of-the-art of biomarkers in acute and chronic phase. Finally, we discuss new perspectives regarding the implementation of blood-based biomarkers in clinical practice to improve the rehabilitation approaches and post stroke recovery.
Collapse
Affiliation(s)
- Stefano Brunelli
- NeuroRehabilitation Unit 4, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Emilia Giannella
- Clinical Neurochemistry Unit and Biobank, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Mirko Bizzaglia
- Radiology and Diagnostic Imaging Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | | | | |
Collapse
|
17
|
Maheshwari S, Dwyer LJ, Sîrbulescu RF. Inflammation and immunomodulation in central nervous system injury - B cells as a novel therapeutic opportunity. Neurobiol Dis 2023; 180:106077. [PMID: 36914074 PMCID: PMC10758988 DOI: 10.1016/j.nbd.2023.106077] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 03/13/2023] Open
Abstract
Acute injury to the central nervous system (CNS) remains a complex and challenging clinical need. CNS injury initiates a dynamic neuroinflammatory response, mediated by both resident and infiltrating immune cells. Following the primary injury, dysregulated inflammatory cascades have been implicated in sustaining a pro-inflammatory microenvironment, driving secondary neurodegeneration and the development of lasting neurological dysfunction. Due to the multifaceted nature of CNS injury, clinically effective therapies for conditions such as traumatic brain injury (TBI), spinal cord injury (SCI), and stroke have proven challenging to develop. No therapeutics that adequately address the chronic inflammatory component of secondary CNS injury are currently available. Recently, B lymphocytes have gained increasing appreciation for their role in maintaining immune homeostasis and regulating inflammatory responses in the context of tissue injury. Here we review the neuroinflammatory response to CNS injury with particular focus on the underexplored role of B cells and summarize recent results on the use of purified B lymphocytes as a novel immunomodulatory therapeutic for tissue injury, particularly in the CNS.
Collapse
Affiliation(s)
- Saumya Maheshwari
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Liam J Dwyer
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruxandra F Sîrbulescu
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Wang YR, Cui WQ, Wu HY, Xu XD, Xu XQ. The role of T cells in acute ischemic stroke. Brain Res Bull 2023; 196:20-33. [PMID: 36906042 DOI: 10.1016/j.brainresbull.2023.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
Acute ischemic stroke (AIS) is associated with high rates of disability and mortality, exerting a substantial impact on overall survival and health-related quality of life. Treatment of AIS remains challenging given that the underlying pathologic mechanisms remain unclear. However, recent research has demonstrated that the immune system plays a key role in the development of AIS. Numerous studies have reported infiltration of T cells into ischemic brain tissue. While some types of T cells can promote the development of inflammatory responses and aggravate ischemic damage in patients with AIS, other T cells appear to exert neuroprotective effects via immunosuppression and other mechanisms. In this review, we discuss the recent findings regarding the infiltration of T cells into ischemic brain tissue, and the mechanisms governing how T cells can facilitate tissue injury or neuroprotection in AIS. Factors influencing the function of T cells, such as intestinal microflora and sex differences, are also discussed. We also explore the recent research on the effect of non-coding RNA on T cells after stroke, as well as the potential for specifically targeting T cells in the treatment of stroke patients.
Collapse
Affiliation(s)
- Yi-Ran Wang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wen-Qiang Cui
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China; First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong-Yun Wu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China; First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiang-Dong Xu
- Experimental Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiang-Qing Xu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China; First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
19
|
Fang Z, Chen M, Qian J, Wang C, Zhang J. The Bridge Between Ischemic Stroke and Gut Microbes: Short-Chain Fatty Acids. Cell Mol Neurobiol 2023; 43:543-559. [PMID: 35347532 PMCID: PMC11415173 DOI: 10.1007/s10571-022-01209-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/17/2022] [Indexed: 11/03/2022]
Abstract
Short-chain fatty acids (SCFAs) are monocarboxylates produced by the gut microbiota (GM) and result from the interaction between diet and GM. An increasing number of studies about the microbiota-gut-brain axis (MGBA) indicated that SCFAs may be a crucial mediator in the MGBA, but their roles have not been fully clarified. In addition, there are few studies directly exploring the role of SCFAs as a potential regulator of microbial targeted interventions in ischemic stroke, especially for clinical studies. This review summarizes the recent studies concerning the relationship between ischemic stroke and GM and outlines the role of SCFAs as a bridge between them. The potential mechanisms by which SCFAs affect ischemic stroke are described. Finally, the beneficial effects of SFCAs-mediated therapeutic measures such as diet, dietary supplements (e.g., probiotics and prebiotics), fecal microbiota transplantation, and drugs on ischemic brain injury are also discussed.
Collapse
Affiliation(s)
- Zongwei Fang
- Department of Pharmacy, Fujian Medical University Union Hospital, #29 Xinquan Road, Fuzhou, China
| | - Mingrong Chen
- Department of Pharmacy, Fujian Medical University Union Hospital, #29 Xinquan Road, Fuzhou, China
| | - Jiafen Qian
- Department of Pharmacy, Fujian Medical University Union Hospital, #29 Xinquan Road, Fuzhou, China
| | - Chunhua Wang
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jinhua Zhang
- Department of Pharmacy, Fujian Medical University Union Hospital, #29 Xinquan Road, Fuzhou, China.
| |
Collapse
|
20
|
Truong SHT, Bonnici B, Rupasinghe S, Kemp-Harper BK, Samuel CS, Broughton BRS. Post-stroke administration of H2 relaxin reduces functional deficits, neuronal apoptosis and immune cell infiltration into the mouse brain. Pharmacol Res 2023; 187:106611. [PMID: 36526079 DOI: 10.1016/j.phrs.2022.106611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/11/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022]
Abstract
Brain inflammation and apoptosis contribute to neuronal damage and loss following ischaemic stroke, leading to cognitive and functional disability. It is well-documented that the human gene-2 (H2)-relaxin hormone exhibits pleiotropic properties via its cognate receptor, Relaxin Family Peptide Receptor 1 (RXFP1), including anti-inflammatory and anti-apoptotic effects, thus making it a potential therapeutic for stroke. Hence, the current study investigated whether post-stroke H2-relaxin administration could improve functional and histological outcomes. 8-12-week-old male C57BL/6 mice were subjected to sham operation or photothrombotic stroke and intravenously-administered with either saline (vehicle) or 0.02, 0.2 or 2 mg/kg doses of recombinant H2-relaxin at 6, 24 and 48 h post-stroke. Motor function was assessed using the hanging wire and cylinder test pre-surgery, and at 24 and 72 h post-stroke. Brains were removed after 72 h and infarct volume was assessed via thionin staining, and RXFP1 expression, leukocyte infiltration and apoptosis were determined by immunofluorescence. RXFP1 was identified on neurons, astrocytes and macrophages, and increased post-stroke. Whilst H2-relaxin did not alter infarct volume, it did cause a dose-dependent improvement in motor function at 24 and 72 h post-stroke. Moreover, 2 mg/kg H2-relaxin significantly decreased the number of apoptotic cells as well as macrophages and neutrophils within the ischaemic hemisphere, but did not alter T or B cells numbers. The anti-inflammatory and anti-apoptotic effects of H2-relaxin when administered at 6 h post-cerebral ischaemia may provide a novel therapeutic option for patients following ischaemic stroke.
Collapse
Affiliation(s)
- Shirley H T Truong
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Benjamin Bonnici
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Samoda Rupasinghe
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Barbara K Kemp-Harper
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Chrishan S Samuel
- Fibrosis Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Brad R S Broughton
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
21
|
Involvement of T-bet and GATA3 transcription factors in Mesenchymal stem cells and royal jelly combination treatment in brain stroke. J Stroke Cerebrovasc Dis 2022; 31:106678. [PMID: 35882098 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Mesenchymal stem cells (MSC) therapy is a promising therapeutic strategy to overcome the brain stroke side effects. However, it may be associated with long-term complications, including induction of inflammation. This project was designed to examine the effects of MSC administration and its combination with royal jelly (RJ) on the differentiation of T helper subsets. MATERIAL AND METHODS In this project, the mice were divided to the six groups, including control (healthy without stroke), stroke (mice model of middle cerebral artery occlusion (MCAO)), treated with mouse MSC (mMSC), royal jelly (RJ), combination of mMSC and RJ (mMSC + RJ) and MSC conditioned medium (SUP). Thereafter, sticky test, brain mRNA levels of T-bet (transcription factor for Th1 subset), GATA3 (transcription factor for Th2 subset), and ROR-γ (transcription factor for Th17 subset) and percentage of myeloperoxidase (MPO) activities were explored in the groups. RESULTS Administration of mMSC and mMSC + RJ improved the sticky test times and decreased the MPO activities. Using mMSCs and RJ was associated with increased expression of T-bet and GATA3 transcription factors. Transplantation of mMSCs in combination with RJ reduced expression of T-bet in the infarcted tissue. CONCLUSION Using mMSC may be associated with Th1-related inflammation in the long term. RJ co-administration significantly reduced the risks, hence, to decrease the plausible side effects of MSCs, it can be proposed to use RJ in combination with MSC to reduce stroke complications.
Collapse
|
22
|
Tarkowska A, Furmaga-Jabłońska W, Bogucki J, Kocki J, Pluta R. Hypothermia after Perinatal Asphyxia Does Not Affect Genes Responsible for Amyloid Production in Neonatal Peripheral Lymphocytes. J Clin Med 2022; 11:3263. [PMID: 35743334 PMCID: PMC9225259 DOI: 10.3390/jcm11123263] [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: 03/28/2022] [Revised: 05/11/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022] Open
Abstract
In this study, the expression of the genes of the amyloid protein precursor, β-secretase, presenilin 1 and 2 by RT-PCR in the lymphocytes of newborns after perinatal asphyxia and perinatal asphyxia treated with hypothermia was analyzed at the age of 15-21 days. The relative quantification of Alzheimer's-disease-related genes was first performed by comparing the peripheral lymphocytes of non-asphyxia control versus those with asphyxia or asphyxia with hypothermia. In the newborns who had perinatal asphyxia, the peripheral lymphocytes presented a decreased expression of the amyloid protein precursor and β-secretase genes. On the other hand, the expression of the presenilin 1 and 2 genes increased in the studied group. The expression of the studied genes in the asphyxia group treated with hypothermia had an identical pattern of changes that were not statistically significant to the asphyxia group. This suggests that the expression of the genes involved in the metabolism of the amyloid protein precursor in the peripheral lymphocytes may be a biomarker of progressive pathological processes in the brain after asphyxia that are not affected by hypothermia. These are the first data in the world showing the role of hypothermia in the gene changes associated with Alzheimer's disease in the peripheral lymphocytes of newborns after asphyxia.
Collapse
Affiliation(s)
- Agata Tarkowska
- Department of Neonate and Infant Pathology, Medical University of Lublin, 20-093 Lublin, Poland; (A.T.); (W.F.-J.)
| | - Wanda Furmaga-Jabłońska
- Department of Neonate and Infant Pathology, Medical University of Lublin, 20-093 Lublin, Poland; (A.T.); (W.F.-J.)
| | - Jacek Bogucki
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, 20-080 Lublin, Poland;
| | - Ryszard Pluta
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| |
Collapse
|
23
|
Leone P, Mincheva G, Balzano T, Malaguarnera M, Felipo V, Llansola M. Rifaximin Improves Spatial Learning and Memory Impairment in Rats with Liver Damage-Associated Neuroinflammation. Biomedicines 2022; 10:1263. [PMID: 35740285 PMCID: PMC9219896 DOI: 10.3390/biomedicines10061263] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/04/2022] Open
Abstract
Patients with non-alcoholic fatty liver disease (NAFLD) may show mild cognitive impairment. Neuroinflammation in the hippocampus mediates cognitive impairment in rat models of minimal hepatic encephalopathy (MHE). Treatment with rifaximin reverses cognitive impairment in a large proportion of cirrhotic patients with MHE. However, the underlying mechanisms remain unclear. The aims of this work were to assess if rats with mild liver damage, as a model of NAFLD, show neuroinflammation in the hippocampus and impaired cognitive function, if treatment with rifaximin reverses it, and to study the underlying mechanisms. Mild liver damage was induced with carbon-tetrachloride. Infiltration of immune cells, glial activation, and cytokine expression, as well as glutamate receptors expression in the hippocampus and cognitive function were assessed. We assessed the effects of daily treatment with rifaximin on the alterations showed by these rats. Rats with mild liver damage showed hippocampal neuroinflammation, reduced membrane expression of glutamate N-methyl-D-aspartate (NMDA) receptor subunits, and impaired spatial memory. Increased C-C Motif Chemokine Ligand 2 (CCL2), infiltration of monocytes, microglia activation, and increased tumor necrosis factor α (TNFα) were reversed by rifaximin, that normalized NMDA receptor expression and improved spatial memory. Thus, rifaximin reduces neuroinflammation and improves cognitive function in rats with mild liver damage, being a promising therapy for patients with NAFLD showing mild cognitive impairment.
Collapse
Affiliation(s)
- Paola Leone
- Mar Lab Department of Neuroscience, NYU Grossman School of Medicine Science Building, New York, NY 10016, USA;
| | - Gergana Mincheva
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (G.M.); (M.L.)
| | - Tiziano Balzano
- Centro Integral de Neurociencias, A.C. HM Hospital Universitario Puerta del Sur CINAC, 28938 Madrid, Spain;
| | - Michele Malaguarnera
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, 46010 Valencia, Spain;
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (G.M.); (M.L.)
| | - Marta Llansola
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (G.M.); (M.L.)
| |
Collapse
|
24
|
Optimising the photothrombotic model of stroke in the C57BI/6 and FVB/N strains of mouse. Sci Rep 2022; 12:7598. [PMID: 35534531 PMCID: PMC9085761 DOI: 10.1038/s41598-022-11793-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
The photothrombotic stroke model relies on the interaction between photosensitive-dye and light for clot formation. Interestingly, the relationship between the length of light exposure and stroke-outcome has never been examined. This model has yet to be established in the FVB/N strain, even though stroke-outcomes are strain-specific. Therefore, this study aimed to examine the effect of different lengths of light exposure in two strains of mice on photothrombotic stroke. Male FVB/N and C57Bl/6 mice were subjected to stroke using 15, 18, or 20-min light exposure. Mice underwent functional testing for up to 7 days. Infarct volume was assessed with thionin staining, and cellular responses to injury analysed via immunofluorescence at 7-days post-stroke. Blood brain barrier (BBB) breakdown was assessed using Evans blue dye at 4.5-h post-stroke. Increasing light exposure from 15 to 20-min increased infarct volume but not functional deficit. Interestingly, there were strain-specific differences in functional outcomes, with FVB/N mice having less deficit on the hanging wire test than C57BI/6 after 15-min of light exposure. The opposite was seen in the adhesive removal test. There was no difference in the number of neurons, astrocytes, microglia, macrophages, and T cells between the strains, despite FVB/N mice demonstrating greater BBB breakdown and an enlarged spleen post-stroke. Increasing light exposure systematically increases infarct volume but does not worsen functional outcomes. FVB/N and C57Bl/6 mice exhibit subtle differences in functional outcomes post stroke, which highlights the need to choose tests which are appropriate for the mouse strain being used.
Collapse
|
25
|
Xie Y, Zou X, Han J, Zhang Z, Feng Z, Ouyang Q, Hua S, Liu Z, Li C, Cai Y, Zou Y, Tang Y, Jiang X. Indole-3-propionic acid alleviates ischemic brain injury in a mouse middle cerebral artery occlusion model. Exp Neurol 2022; 353:114081. [DOI: 10.1016/j.expneurol.2022.114081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/16/2022] [Accepted: 04/05/2022] [Indexed: 11/24/2022]
|
26
|
Li S, Huang Y, Liu Y, Rocha M, Li X, Wei P, Misilimu D, Luo Y, Zhao J, Gao Y. Change and predictive ability of circulating immunoregulatory lymphocytes in long-term outcomes of acute ischemic stroke. J Cereb Blood Flow Metab 2021; 41:2280-2294. [PMID: 33641517 PMCID: PMC8393304 DOI: 10.1177/0271678x21995694] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lymphocytes play an important role in the immune response after stroke. However, our knowledge of the circulating lymphocytes in ischemic stroke is limited. Herein, we collected the blood samples of clinical ischemic stroke patients to detect the change of lymphocytes from admission to 3 months after ischemic stroke by flow cytometry. A total of 87 healthy controls and 210 patients were enrolled, and the percentages of circulating T cells, CD4+ T cells, CD8+ T cells, double negative T cells (DNTs), CD4+ regulatory T cells (Tregs), CD8+ Tregs, B cells and regulatory B cells (Bregs) were measured. Among patients, B cells, Bregs and CD8+ Tregs increased significantly, while CD4+ Tregs dropped and soon reversed after ischemic stroke. CD4+ Tregs, CD8+ Tregs, and DNTs also showed high correlations with the infarct volume and neurological scores of patients. Moreover, these lymphocytes enhanced the predictive ability of long-term prognosis of neurological scores when added to basic clinical information. The percentage of CD4+ Tregs within lymphocytes showed high correlations with both acute and long-term neurological outcomes, which exhibited a great independent predictive ability. These findings suggest that CD4+ Tregs can be a biomarker to predict stroke outcomes and improve existing therapeutic strategies of immunoregulatory lymphocytes.
Collapse
Affiliation(s)
- Sicheng Li
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yichen Huang
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yang Liu
- Department of Neurology, Minhang Hospital Affiliated to Fudan University, Shanghai, China
| | - Marcelo Rocha
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiaofan Li
- Department of Neurology, Minhang Hospital Affiliated to Fudan University, Shanghai, China
| | - Pengju Wei
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Dilidaer Misilimu
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yunhe Luo
- Department of Neurology, Minhang Hospital Affiliated to Fudan University, Shanghai, China
| | - Jing Zhao
- Department of Neurology, Minhang Hospital Affiliated to Fudan University, Shanghai, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Li QQ, Li JY, Zhou M, Qin ZH, Sheng R. Targeting neuroinflammation to treat cerebral ischemia - The role of TIGAR/NADPH axis. Neurochem Int 2021; 148:105081. [PMID: 34082063 DOI: 10.1016/j.neuint.2021.105081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/24/2021] [Accepted: 05/22/2021] [Indexed: 01/30/2023]
Abstract
Cerebral ischemia is a disease of ischemic necrosis of brain tissue caused by intracranial artery stenosis or occlusion and cerebral artery embolization. Neuroinflammation plays an important role in the pathophysiology of cerebral ischemia. Microglia, astrocytes, leukocytes and other cells that release a variety of inflammatory factors involved in neuroinflammation may play a damaging or protective role during the process of cerebral ischemia. TP53-induced glycolysis and apoptotic regulators (TIGAR) may facilitate the production of nicotinamide adenine dinucleotide phosphoric acid (NADPH) via the pentose phosphate pathway (PPP) to inhibit oxidative stress and neuroinflammation. TIGAR can also directly inhibit NF-κB to inhibit neuroinflammation. TIGAR thus protect against cerebral ischemic injury. Exogenous NADPH can inhibit neuroinflammation by inhibiting oxidative stress and regulating a variety of signals. However, since NADPH oxidase (NOX) may use NADPH as a substrate to generate reactive oxygen species (ROS) to mediate neuroinflammation, the combination of NADPH and NOX inhibitors may produce more powerful anti-neuroinflammatory effects. Here, we review the cells and regulatory signals involved in neuroinflammation during cerebral ischemia, and discuss the possible mechanisms of targeting neuroinflammation in the treatment of cerebral ischemia with TIGAR/NADPH axis, so as to provide new ideas for the prevention and treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Qi-Qi Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Jia-Ying Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Ming Zhou
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China.
| |
Collapse
|
28
|
Yu F, Huang T, Ran Y, Li D, Ye L, Tian G, Xi J, Liu Z. New Insights Into the Roles of Microglial Regulation in Brain Plasticity-Dependent Stroke Recovery. Front Cell Neurosci 2021; 15:727899. [PMID: 34421544 PMCID: PMC8374071 DOI: 10.3389/fncel.2021.727899] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 07/13/2021] [Indexed: 01/07/2023] Open
Abstract
Stroke remains the leading cause of long-term disability worldwide with significant long-term sequelae. However, there is no highly effective treatment to enhance post-stroke recovery despite extensive efforts in exploring rehabilitative therapies. Neurorehabilitation is recognized as the cornerstone of functional restoration therapy in stroke, where treatments are focused on neuroplastic regulation to reverse neural structural disruption and improve neurofunctional networks. Post-stroke neuroplasticity changes begin within hours of symptom onset and reaches a plateau by 3 to 4 weeks within the global brain in animal studies. It plays a determining role in spontaneous stroke recovery. Microglia are immediately activated following cerebral ischemia, which has been found both proximal to the primary ischemic injury and at the remote brain regions which have functional connections to the primary injury area. Microglia exhibit different activation profiles based on the microenvironment and adaptively switch their phenotypes in a spatiotemporal manner in response to brain injuries. Microglial activation coincides with neuroplasticity after stroke, which provides the fundamental base for the microglia-mediated inflammatory responses involved in the entire neural network rewiring and brain repair. Microglial activation exerts important effects on spontaneous recovery after stroke, including structural and functional reestablishment of neurovascular networks, neurogenesis, axonal remodeling, and blood vessel regeneration. In this review, we focus on the crosstalk between microglial activation and endogenous neuroplasticity, with a special focus on the plastic alterations in the whole brain network and their implications for structural and functional restoration after stroke. We then summarize recent advances in the impacts of microglial phenotype polarization on brain plasticity, trying to discuss the potential efficacy of microglia-based extrinsic restorative interventions in promoting post-stroke recovery.
Collapse
Affiliation(s)
- Fang Yu
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Anesthesiology, Westchester Medical Center, New York Medical College, Valhalla, NY, United States
| | - Tingting Huang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Ran
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Da Li
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Lin Ye
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China
| | - Guiqin Tian
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Jianing Xi
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
29
|
Schächtle MA, Rosshart SP. The Microbiota-Gut-Brain Axis in Health and Disease and Its Implications for Translational Research. Front Cell Neurosci 2021; 15:698172. [PMID: 34335190 PMCID: PMC8321234 DOI: 10.3389/fncel.2021.698172] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/16/2021] [Indexed: 12/22/2022] Open
Abstract
Over the past decades, microbiome research has evolved rapidly and became a hot topic in basic, preclinical and clinical research, for the pharmaceutical industry and for the general public. With the help of new high-throughput sequencing technologies tremendous progress has been made in the characterization of host-microbiota interactions identifying the microbiome as a major factor shaping mammalian physiology. This development also led to the discovery of the gut-brain axis as the crucial connection between gut microbiota and the nervous system. Consequently, a rapidly growing body of evidence emerged suggesting that the commensal gut microbiota plays a vital role in brain physiology. Moreover, it became evident that the communication along this microbiota-gut-brain axis is bidirectional and primarily mediated by biologically active microbial molecules and metabolites. Further, intestinal dysbiosis leading to changes in the bidirectional relationship between gut microbiota and the nervous system was linked to the pathogenesis of several psychiatric and neurological disorders. Here, we discuss the impact of the gut microbiota on the brain in health and disease, specifically as regards to neuronal homeostasis, development and normal aging as well as their role in neurological diseases of the highest socioeconomic burden such as Alzheimer's disease and stroke. Subsequently, we utilize Alzheimer's disease and stroke to examine the translational research value of current mouse models in the spotlight of microbiome research. Finally, we propose future strategies on how we could conduct translational microbiome research in the field of neuroscience that may lead to the identification of novel treatments for human diseases.
Collapse
Affiliation(s)
- Melanie Anna Schächtle
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stephan Patrick Rosshart
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
30
|
Guo H, Lin J, Zheng L, Yang F. An effective fluorescent sensor for ClO - in aqueous media based on thiophene-cyanostilbene Schiff-base. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 256:119744. [PMID: 33819762 DOI: 10.1016/j.saa.2021.119744] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 06/12/2023]
Abstract
Although some reports on sensing ClO- had been presented, the ClO- sensor with high selectivity and sensitivity in aqueous media was still expected. Herein, an effective fluorescent sensor for ClO- in aqueous media was designed and synthesized by simple procedure based on cyanostilbene derivative (TCS). TCS exhibited strong fluorescence in aqueous media, which could be selectively quenched by ClO- among various species. The detection limit was as low as 3.2 × 10-8 M. The sensing mechanism of the oxidation of sulfur in thiophene unit was confirmed by the FT-IR spectrum, fluorescence Job's plot, 1H NMR spectrum and MS spectrum. This sensor was successfully applied on detecting ClO- in real sample and living-cell imaging, suggesting its potential application for sensing ClO- in both vitro assay and vivo environment.
Collapse
Affiliation(s)
- Hongyu Guo
- College of Chemistry and Materials, Fujian Normal University, Fuzhou 350007, PR China; Fujian Key Laboratory of Polymer Materials, Fuzhou 350007, PR China; Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fuzhou 350007, PR China
| | - Jianrong Lin
- College of Chemistry and Materials, Fujian Normal University, Fuzhou 350007, PR China
| | - Linlu Zheng
- Fujian Provincial Key Laboratory of Featured Biochemical and Chemical Materials, Ningde Normal University, Ningde 352106, PR China
| | - Fafu Yang
- College of Chemistry and Materials, Fujian Normal University, Fuzhou 350007, PR China; Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fuzhou 350007, PR China.
| |
Collapse
|
31
|
Qiu YM, Zhang CL, Chen AQ, Wang HL, Zhou YF, Li YN, Hu B. Immune Cells in the BBB Disruption After Acute Ischemic Stroke: Targets for Immune Therapy? Front Immunol 2021; 12:678744. [PMID: 34248961 PMCID: PMC8260997 DOI: 10.3389/fimmu.2021.678744] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/31/2021] [Indexed: 12/15/2022] Open
Abstract
Blood-Brain Barrier (BBB) disruption is an important pathophysiological process of acute ischemic stroke (AIS), resulting in devastating malignant brain edema and hemorrhagic transformation. The rapid activation of immune cells plays a critical role in BBB disruption after ischemic stroke. Infiltrating blood-borne immune cells (neutrophils, monocytes, and T lymphocytes) increase BBB permeability, as they cause microvascular disorder and secrete inflammation-associated molecules. In contrast, they promote BBB repair and angiogenesis in the latter phase of ischemic stroke. The profound immunological effects of cerebral immune cells (microglia, astrocytes, and pericytes) on BBB disruption have been underestimated in ischemic stroke. Post-stroke microglia and astrocytes can adopt both an M1/A1 or M2/A2 phenotype, which influence BBB integrity differently. However, whether pericytes acquire microglia phenotype and exert immunological effects on the BBB remains controversial. Thus, better understanding the inflammatory mechanism underlying BBB disruption can lead to the identification of more promising biological targets to develop treatments that minimize the onset of life-threatening complications and to improve existing treatments in patients. However, early attempts to inhibit the infiltration of circulating immune cells into the brain by blocking adhesion molecules, that were successful in experimental stroke failed in clinical trials. Therefore, new immunoregulatory therapeutic strategies for acute ischemic stroke are desperately warranted. Herein, we highlight the role of circulating and cerebral immune cells in BBB disruption and the crosstalk between them following acute ischemic stroke. Using a robust theoretical background, we discuss potential and effective immunotherapeutic targets to regulate BBB permeability after acute ischemic stroke.
Collapse
Affiliation(s)
| | | | | | | | | | - Ya-nan Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
32
|
Feng Y, Li Y, Zhang Y, Zhang BH, Zhao H, Zhao X, Shi FD, Jin WN, Zhang XA. miR-1224 contributes to ischemic stroke-mediated natural killer cell dysfunction by targeting Sp1 signaling. J Neuroinflammation 2021; 18:133. [PMID: 34118948 PMCID: PMC8196447 DOI: 10.1186/s12974-021-02181-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/21/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Brain ischemia compromises natural killer (NK) cell-mediated immune defenses by acting on neurogenic and intracellular pathways. Less is known about the posttranscriptional mechanisms that regulate NK cell activation and cytotoxicity after ischemic stroke. METHODS Using a NanoString nCounter® miRNA array panel, we explored the microRNA (miRNA) profile of splenic NK cells in mice subjected to middle cerebral artery occlusion. Differential gene expression and function/pathway analysis were applied to investigate the main functions of predicted miRNA target genes. miR-1224 inhibitor/mimics transfection and passive transfer of NK cells were performed to confirm the impact of miR-1224 in NK cells after brain ischemia. RESULTS We observed striking dysregulation of several miRNAs in response to ischemia. Among those miRNAs, miR-1224 markedly increased 3 days after ischemic stroke. Transfection of miR-1224 mimics into NK cells resulted in suppression of NK cell activity, while an miR-1224 inhibitor enhanced NK cell activity and cytotoxicity, especially in the periphery. Passive transfer of NK cells treated with an miR-1224 inhibitor prevented the accumulation of a bacterial burden in the lungs after ischemic stroke, suggesting an enhanced immune defense of NK cells. The transcription factor Sp1, which controls cytokine/chemokine release by NK cells at the transcriptional level, is a predicted target of miR-1224. The inhibitory effect of miR-1224 on NK cell activity was blocked in Sp1 knockout mice. CONCLUSIONS These findings indicate that miR-1224 may serve as a negative regulator of NK cell activation in an Sp1-dependent manner; this mechanism may be a novel target to prevent poststroke infection specifically in the periphery and preserve immune defense in the brain.
Collapse
Affiliation(s)
- Yan Feng
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Li
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Ying Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Bo-Hao Zhang
- The Third Affiliated Hospital of Zhengzhou University, No. 7 Kangfu front ST, Zhengzhou, Henan, China
| | - Hui Zhao
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Zhao
- The Third Affiliated Hospital of Zhengzhou University, No. 7 Kangfu front ST, Zhengzhou, Henan, China
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wei-Na Jin
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China.
- China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xiao-An Zhang
- The Third Affiliated Hospital of Zhengzhou University, No. 7 Kangfu front ST, Zhengzhou, Henan, China.
- School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, Henan, China.
| |
Collapse
|
33
|
Li Y, Gong M. Analysis of the neuroprotective effect of GLP-1 receptor agonist peptide on cerebral ischemia-reperfusion injury by Quantitative Proteomics Mass Spectrometry. Brain Behav 2021; 11:e02190. [PMID: 34018701 PMCID: PMC8213929 DOI: 10.1002/brb3.2190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE The pathological characteristics of cerebral ischemia-reperfusion injury (CIRI) are complex, and the mechanism involved remains unknown. The treatment for CIRI has become an increasingly important challenge in the clinic, prompting us to explore the mechanism of CIRI. It was reported that GLP-1 receptor agonist, Liraglutide, exhibited alleviating effects on CIRI. The previous findings suggested that the administration of Liraglutide in rodents results in the attenuation of the infarct volume following ischemia-reperfusion injury by mediating the reactive oxygen species, apoptotic and necroptotic pathways. METHODS Here, a proteomic study was performed aiming to clarify the physiological protection role of GLP-1 receptor agonist during the development of CIRI in MCAO mice. This proteomic investigations is contributed to reveal the mechanism associated with the treatment of GLP-1 receptor agonist in MCAO mice. RESULTS The results indicated that the occurrence of ischemia-reperfusion led to complex pathological processes, including inflammation, necroptosis and apoptosis. The treatment of Liraglutide significantly reduced the infract volume resulted from ischemia reperfusion injury. The proteomic data revealed that the administration of Liraglutide in MCAO mice induced the various effects on proteins expression level and phosphorylation. CONCLUSIONS The findings in this study was beneficial for identifying the novel therapeutic target for the treatment of ischemia reperfusion.
Collapse
Affiliation(s)
- Ying Li
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, China
| | - Min Gong
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, China.,Department of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
34
|
Alzheimer's Disease Associated Presenilin 1 and 2 Genes Dysregulation in Neonatal Lymphocytes Following Perinatal Asphyxia. Int J Mol Sci 2021; 22:ijms22105140. [PMID: 34067945 PMCID: PMC8152038 DOI: 10.3390/ijms22105140] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
Perinatal asphyxia is mainly a brain disease leading to the development of neurodegeneration, in which a number of peripheral lesions have been identified; however, little is known about the expression of key genes involved in amyloid production by peripheral cells, such as lymphocytes, during the development of hypoxic-ischemic encephalopathy. We analyzed the gene expression of the amyloid protein precursor, β-secretase, presenilin 1 and 2 and hypoxia-inducible factor 1-α by RT-PCR in the lymphocytes of post-asphyxia and control neonates. In all examined periods after asphyxia, decreased expression of the genes of the amyloid protein precursor, β-secretase and hypoxia-inducible factor 1-α was noted in lymphocytes. Conversely, expression of presenilin 1 and 2 genes decreased on days 1–7 and 8–14 but increased after survival for more than 15 days. We believe that the expression of presenilin genes in lymphocytes could be a potential biomarker to determine the severity of the post-asphyxia neurodegeneration or to identify the underlying factors for brain neurodegeneration and get information about the time they occurred. This appears to be the first worldwide data on the role of the presenilin 1 and 2 genes associated with Alzheimer’s disease in the dysregulation of neonatal lymphocytes after perinatal asphyxia.
Collapse
|
35
|
Wang G, Zhang H, Sun J, Zhang Y, He F, Zou J. Cyclosporin A impairs neurogenesis and cognitive abilities in brain development via the IFN-γ-Shh-BDNF pathway. Int Immunopharmacol 2021; 96:107744. [PMID: 33993101 DOI: 10.1016/j.intimp.2021.107744] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/15/2021] [Accepted: 04/29/2021] [Indexed: 01/07/2023]
Abstract
A wealth of evidence indicate that the peripheral immune activation alters brain development. However, it is still largely unclear whether and how peripheral immunosuppression affects neurodevelopment. Here, we found that the immunosuppressant cyclosporin A (CsA) decreased the number of BrdU+, BrdU+/DCX+, BrdU+/NeuN + cells in the hippocampus, impaired learning and memory and inhibited protein levels of the shh signaling pathway, including Shh, Smo and Gli1. However, the shh pathway receptor agonist SAG could block the impairment of cognitive ability and the decrease of hippocampal neurogenesis and brain-derived neurotrophic factor (BDNF) level induced by CsA. We also found that CsA decreased the level of interferon-gamma (IFN-γ), while up-regulation of IFN-γ altered the inhibitory effect of the shh signaling pathway and the decrease of BDNF induced by CsA. Collectively, these data indicate that peripheral CsA impairs neurogenesis and cognition in brain development through downregulating the IFN-γ-Shh-BDNF pathway. The present study guides us to correctly apply immunomodulatory drugs in early life and suggests that the IFN-γ-Shh-BDNF pathway may represent a novel protective target for neurodevelopment under the condition of immunosuppression.
Collapse
Affiliation(s)
- Ge Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200032 Shanghai, People's Republic of China
| | - Hongyang Zhang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jiancong Sun
- Department of Radiation Oncology, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yuwei Zhang
- Department of Anatomy, Wannan Medical College, Wuhu, Anhui, People's Republic of China
| | - Fen He
- Department of Radiation Oncology, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Juntao Zou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
36
|
Pluta R, Januszewski S, Czuczwar SJ. Neuroinflammation in Post-Ischemic Neurodegeneration of the Brain: Friend, Foe, or Both? Int J Mol Sci 2021; 22:4405. [PMID: 33922467 PMCID: PMC8122836 DOI: 10.3390/ijms22094405] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
One of the leading causes of neurological mortality, disability, and dementia worldwide is cerebral ischemia. Among the many pathological phenomena, the immune system plays an important role in the development of post-ischemic degeneration of the brain, leading to the development of neuroinflammatory changes in the brain. After cerebral ischemia, the developing neuroinflammation causes additional damage to the brain cells, but on the other hand it also plays a beneficial role in repair activities. Inflammatory mediators are sources of signals that stimulate cells in the brain and promote penetration, e.g., T lymphocytes, monocytes, platelets, macrophages, leukocytes, and neutrophils from systemic circulation to the brain ischemic area, and this phenomenon contributes to further irreversible ischemic brain damage. In this review, we focus on the issues related to the neuroinflammation that occurs in the brain tissue after ischemia, with particular emphasis on ischemic stroke and its potential treatment strategies.
Collapse
Affiliation(s)
- Ryszard Pluta
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, PL 02-106 Warsaw, Poland;
| | - Sławomir Januszewski
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, PL 02-106 Warsaw, Poland;
| | - Stanisław J. Czuczwar
- Department of Pathophysiology, Medical University of Lublin, PL 20-090 Lublin, Poland;
| |
Collapse
|
37
|
Liu B, Hu Y, Ma G, Xiao Y, Zhang B, Liang Y, Zhong P, Zeng X, Lin Z, Kong H, Wu G, Du Z, Fang Y, Huang M, Wang L, Yang X, Yu H. Reduced Retinal Microvascular Perfusion in Patients With Stroke Detected by Optical Coherence Tomography Angiography. Front Aging Neurosci 2021; 13:628336. [PMID: 33927607 PMCID: PMC8078175 DOI: 10.3389/fnagi.2021.628336] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/16/2021] [Indexed: 12/20/2022] Open
Abstract
Currently there is a shortage of biomarkers for stroke, one of the leading causes of death and disability in aging populations. Retinal vessels offer a unique and accessible “window” to study the microvasculature in vivo. However, the relationship between the retinal microvasculature and stroke is not entirely clear. To investigate the retinal microvascular characteristics in stroke, we recruited patients with stroke and age-matched control subjects from a tertiary hospital in China. The macular vessel density (VD) in the superficial capillary plexus (SCP) and deep capillary plexus (DCP), foveal avascular zone (FAZ) metrics, and optical coherence tomography angiography (OCTA) measured optic disc VD were recorded for analysis. A total of 189 patients with stroke and 195 control subjects were included. After adjusting for sex, visual acuity, systolic and diastolic blood pressure, a history of smoking, levels of hemoglobulin (HbA1c), cholesterol, and high-density lipoprotein (HDL), the macular VD of SCP and DCP in all sectors was decreased in patients with stroke. In the stroke group, the VD around the FAZ and the VD of the optic disk were lower. Logistic regression found the parafovea-superior-hemi VD of DCP > 54.53% [odds ratio (OR): 0.169] as a protective factor of stroke. Using the integration of all OCTA parameters and traditional risk factors, the area under the receiver operating characteristic (AUC) curve of distinguishing patients with stroke was 0.962, with a sensitivity of 0.944 and a specificity of 0.871. Our study demonstrates that the retinal VD is decreased in patients with stroke independently of the traditional risk factors of stroke, which may shed light on the monitoring of stroke using the retinal microvascular parameters.
Collapse
Affiliation(s)
- Baoyi Liu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yijun Hu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Refractive Surgery Center, Aier Institute of Refractive Surgery, Guangzhou Aier Eye Hospital, Guangzhou, China.,Aier School of Ophthalmology, Central South University, Changsha, China
| | - Guixian Ma
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yu Xiao
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Bin Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yingying Liang
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Pingting Zhong
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xiaomin Zeng
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhanjie Lin
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Huiqian Kong
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Guanrong Wu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zijing Du
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ying Fang
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Manqing Huang
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Lijuan Wang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaohong Yang
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Honghua Yu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
38
|
Suzuki K, Shinohara M, Uno Y, Tashiro Y, Gheni G, Yamamoto M, Fukumori A, Shindo A, Mashimo T, Tomimoto H, Sato N. Deletion of B-cell translocation gene 2 (BTG2) alters the responses of glial cells in white matter to chronic cerebral hypoperfusion. J Neuroinflammation 2021; 18:86. [PMID: 33812385 PMCID: PMC8019185 DOI: 10.1186/s12974-021-02135-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/19/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Subcortical ischemic vascular dementia, one of the major subtypes of vascular dementia, is characterized by lacunar infarcts and white matter lesions caused by chronic cerebral hypoperfusion. In this study, we used a mouse model of bilateral common carotid artery stenosis (BCAS) to investigate the role of B-cell translocation gene 2 (BTG2), an antiproliferation gene, in the white matter glial response to chronic cerebral hypoperfusion. METHODS Btg2-/- mice and littermate wild-type control mice underwent BCAS or sham operation. Behavior phenotypes were assessed by open-field test and Morris water maze test. Brain tissues were analyzed for the degree of white matter lesions and glial changes. To further confirm the effects of Btg2 deletion on proliferation of glial cells in vitro, BrdU incorporation was investigated in mixed glial cells derived from wild-type and Btg2-/- mice. RESULTS Relative to wild-type mice with or without BCAS, BCAS-treated Btg2-/- mice exhibited elevated spontaneous locomotor activity and poorer spatial learning ability. Although the severities of white matter lesions did not significantly differ between wild-type and Btg2-/- mice after BCAS, the immunoreactivities of GFAP, a marker of astrocytes, and Mac2, a marker of activated microglia and macrophages, in the white matter of the optic tract were higher in BCAS-treated Btg2-/- mice than in BCAS-treated wild-type mice. The expression level of Gfap was also significantly elevated in BCAS-treated Btg2-/- mice. In vitro analysis showed that BrdU incorporation in mixed glial cells in response to inflammatory stimulation associated with cerebral hypoperfusion was higher in Btg2-/- mice than in wild-type mice. CONCLUSION BTG2 negatively regulates glial cell proliferation in response to cerebral hypoperfusion, resulting in behavioral changes.
Collapse
Affiliation(s)
- Kaoru Suzuki
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430, Morioka, Obu, Aichi, 474-8511, Japan
| | - Mitsuru Shinohara
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430, Morioka, Obu, Aichi, 474-8511, Japan
- Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshihiro Uno
- Institute of Experimental Animal Sciences, Osaka University Medical School, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshitaka Tashiro
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430, Morioka, Obu, Aichi, 474-8511, Japan
| | - Ghupurjan Gheni
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430, Morioka, Obu, Aichi, 474-8511, Japan
| | - Miho Yamamoto
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430, Morioka, Obu, Aichi, 474-8511, Japan
| | - Akio Fukumori
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430, Morioka, Obu, Aichi, 474-8511, Japan
- Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Akihiko Shindo
- Department of Neurology, Graduate School of Medicine, Mie University, 174, Edobashi 2-chome, Tsu, Mie, 514-8507, Japan
| | - Tomoji Mashimo
- Institute of Experimental Animal Sciences, Osaka University Medical School, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Graduate School of Medicine, Mie University, 174, Edobashi 2-chome, Tsu, Mie, 514-8507, Japan
| | - Naoyuki Sato
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430, Morioka, Obu, Aichi, 474-8511, Japan.
- Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
39
|
Beuker C, Strecker JK, Rawal R, Schmidt-Pogoda A, Ruck T, Wiendl H, Klotz L, Schäbitz WR, Sommer CJ, Minnerup H, Meuth SG, Minnerup J. Immune Cell Infiltration into the Brain After Ischemic Stroke in Humans Compared to Mice and Rats: a Systematic Review and Meta-Analysis. Transl Stroke Res 2021; 12:976-990. [PMID: 33496918 PMCID: PMC8557159 DOI: 10.1007/s12975-021-00887-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/14/2022]
Abstract
Although several studies have suggested that anti-inflammatory strategies reduce secondary infarct growth in animal stroke models, clinical studies have not yet demonstrated a clear benefit of immune modulation in patients. Potential reasons include systematic differences of post-ischemic neuroinflammation between humans and rodents. We here performed a systematic review and meta-analysis to summarize and compare the spatial and temporal distribution of immune cell infiltration in human and rodent stroke. Data on spatiotemporal distribution of immune cells (T cells, macrophages, and neutrophils) and infarct volume were extracted. Data from all rodent studies were pooled by means of a random-effect meta-analysis. Overall, 20 human and 188 rodent stroke studies were included in our analyses. In both patients and rodents, the infiltration of macrophages and neutrophils preceded the lymphocytic influx. Macrophages and neutrophils were the predominant immune cells within 72 h after infarction. Although highly heterogeneously across studies, the temporal profile of the poststroke immune response was comparable between patients and rodents. In rodent stroke, the extent of the immune cell infiltration depended on the duration and location of vessel occlusion and on the species. The density of infiltrating immune cells correlated with the infarct volume. In summary, we provide the first systematic analysis and comparison of human and rodent post-ischemic neuroinflammation. Our data suggest that the inflammatory response in rodent stroke models is comparable to that in patients with stroke. However, the overall heterogeneity of the post-ischemic immune response might contribute to the translational failure in stroke research.
Collapse
Affiliation(s)
- Carolin Beuker
- Department of Neurology with Institute of Translational Neurology, University of Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| | - Jan-Kolja Strecker
- Department of Neurology with Institute of Translational Neurology, University of Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| | - Rajesh Rawal
- Institute of Epidemiology and Social Medicine, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Antje Schmidt-Pogoda
- Department of Neurology with Institute of Translational Neurology, University of Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| | - Tobias Ruck
- Department of Neurology with Institute of Translational Neurology, University of Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University of Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University of Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| | | | - Clemens J Sommer
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Heike Minnerup
- Institute of Epidemiology and Social Medicine, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, University of Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany
| | - Jens Minnerup
- Department of Neurology with Institute of Translational Neurology, University of Münster, Albert-Schweitzer-Campus 1, Gebäude A1, 48149, Münster, Germany.
| |
Collapse
|
40
|
Cellular and Molecular Mechanisms of R/S-Roscovitine and CDKs Related Inhibition under Both Focal and Global Cerebral Ischemia: A Focus on Neurovascular Unit and Immune Cells. Cells 2021; 10:cells10010104. [PMID: 33429982 PMCID: PMC7827530 DOI: 10.3390/cells10010104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/29/2020] [Accepted: 01/05/2021] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke is the second leading cause of death worldwide. Following ischemic stroke, Neurovascular Unit (NVU) inflammation and peripheral leucocytes infiltration are major contributors to the extension of brain lesions. For a long time restricted to neurons, the 10 past years have shown the emergence of an increasing number of studies focusing on the role of Cyclin-Dependent Kinases (CDKs) on the other cells of NVU, as well as on the leucocytes. The most widely used CDKs inhibitor, (R)-roscovitine, and its (S) isomer both decreased brain lesions in models of global and focal cerebral ischemia. We previously showed that (S)-roscovitine acted, at least, by modulating NVU response to ischemia. Interestingly, roscovitine was shown to decrease leucocytes-mediated inflammation in several inflammatory models. Specific inhibition of roscovitine majors target CDK 1, 2, 5, 7, and 9 showed that these CDKs played key roles in inflammatory processes of NVU cells and leucocytes after brain lesions, including ischemic stroke. The data summarized here support the investigation of roscovitine as a potential therapeutic agent for the treatment of ischemic stroke, and provide an overview of CDK 1, 2, 5, 7, and 9 functions in brain cells and leucocytes during cerebral ischemia.
Collapse
|
41
|
Filling the gaps on stroke research: Focus on inflammation and immunity. Brain Behav Immun 2021; 91:649-667. [PMID: 33017613 PMCID: PMC7531595 DOI: 10.1016/j.bbi.2020.09.025] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/10/2020] [Accepted: 09/23/2020] [Indexed: 02/08/2023] Open
Abstract
For the last two decades, researchers have placed hopes in a new era in which a combination of reperfusion and neuroprotection would revolutionize the treatment of stroke. Nevertheless, despite the thousands of papers available in the literature showing positive results in preclinical stroke models, randomized clinical trials have failed to show efficacy. It seems clear now that the existing data obtained in preclinical research have depicted an incomplete picture of stroke pathophysiology. In order to ameliorate bench-to-bed translation, in this review we first describe the main actors on stroke inflammatory and immune responses based on the available preclinical data, highlighting the fact that the link between leukocyte infiltration, lesion volume and neurological outcome remains unclear. We then describe what is known on neuroinflammation and immune responses in stroke patients, and summarize the results of the clinical trials on immunomodulatory drugs. In order to understand the gap between clinical trials and preclinical results on stroke, we discuss in detail the experimental results that served as the basis for the summarized clinical trials on immunomodulatory drugs, focusing on (i) experimental stroke models, (ii) the timing and selection of outcome measuring, (iii) alternative entry routes for leukocytes into the ischemic region, and (iv) factors affecting stroke outcome such as gender differences, ageing, comorbidities like hypertension and diabetes, obesity, tobacco, alcohol consumption and previous infections like Covid-19. We can do better for stroke treatment, especially when targeting inflammation following stroke. We need to re-think the design of stroke experimental setups, notably by (i) using clinically relevant models of stroke, (ii) including both radiological and neurological outcomes, (iii) performing long-term follow-up studies, (iv) conducting large-scale preclinical stroke trials, and (v) including stroke comorbidities in preclinical research.
Collapse
|
42
|
Tarkanyi G, Karadi ZN, Szabo Z, Szegedi I, Csiba L, Szapary L. Relationship between leukocyte counts and large vessel occlusion in acute ischemic stroke. BMC Neurol 2020; 20:440. [PMID: 33272235 PMCID: PMC7716438 DOI: 10.1186/s12883-020-02017-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/30/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuroinflammation plays an important role in the pathogenesis of acute ischemic stroke (AIS) and peripheral leukocyte counts have proved to be independent predictors of stroke severity and outcomes. Clinical significance of large vessel occlusion (LVO) in AIS is increasing, as these patients are potential candidates for endovascular thrombectomy and likely to have worse outcomes if not treated urgently. The aim of our study was to assess the relationship between on admission leukocyte counts and the presence of LVO in the early phase of AIS. METHODS We have conducted a cross-sectional, observational study based on a registry of consecutive AIS patients admitted up to 4.5 h after stroke onset. Blood samples were taken at admission and leukocyte counts were measured immediately. The presence of LVO was verified based on the computed tomography angiography scan on admission. RESULTS Total white blood cell (WBC) and neutrophil counts were significantly higher in patients with LVO than those without LVO (P < 0.001 respectively). After adjustment for potential confounders total WBC counts (adjusted OR: 1.405 per 1 × 109/L increase, 95% CI: 1.209 to 1.632) and neutrophil counts (adjusted OR: 1.344 per 1 × 109/L increase, 95% CI: 1.155 to 1.564) were found to have the strongest associations with the presence of LVO. Total WBC and neutrophil counts had moderate ability to discriminate an LVO in AIS (AUC: 0.667 and 0.655 respectively). No differences were recorded in leukocyte counts according to the size of the occluded vessel and the status of collateral circulation in the anterior vascular territory. However, total WBC and neutrophil counts tended to be higher in patients with LVO in the posterior circulation (p = 0.005 and 0.010 respectively). CONCLUSION Higher admission total WBC and neutrophil counts are strongly associated with the presence of LVO and has moderate ability to discriminate an LVO in AIS. Detailed evaluation of stroke-evoked inflammatory mechanisms and changes according to the presence of LVO demands further investigation.
Collapse
Affiliation(s)
- Gabor Tarkanyi
- Department of Neurology, University of Pécs, Ifjúság u. 13, Pécs, 7624, Hungary
| | | | - Zsofia Szabo
- Department of Neurology, University of Debrecen, Debrecen, Hungary
| | - Istvan Szegedi
- Department of Neurology, University of Debrecen, Debrecen, Hungary
| | - Laszlo Csiba
- Department of Neurology, University of Debrecen, Debrecen, Hungary
| | - Laszlo Szapary
- Department of Neurology, University of Pécs, Ifjúság u. 13, Pécs, 7624, Hungary.
| |
Collapse
|
43
|
Bernstein DL, Zuluaga-Ramirez V, Gajghate S, Reichenbach NL, Polyak B, Persidsky Y, Rom S. miR-98 reduces endothelial dysfunction by protecting blood-brain barrier (BBB) and improves neurological outcomes in mouse ischemia/reperfusion stroke model. J Cereb Blood Flow Metab 2020; 40:1953-1965. [PMID: 31601141 PMCID: PMC7786850 DOI: 10.1177/0271678x19882264] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Most neurological diseases, including stroke, lead to some degree of blood-brain barrier (BBB) dysfunction. A significant portion of BBB injury is caused by inflammation, due to pro-inflammatory factors produced in the brain, and by leukocyte engagement of the brain endothelium. Recently, microRNAs (miRNAs) have appeared as major regulators of inflammation-induced changes to gene expression in the microvascular endothelial cells (BMVEC) that comprise the BBB. However, miRNAs' role during cerebral ischemia/reperfusion is still underexplored. Endothelial levels of miR-98 were significantly altered following ischemia/reperfusion insults, both in vivo and in vitro, transient middle cerebral artery occlusion (tMCAO), and oxygen-glucose deprivation (OGD), respectively. Overexpression of miR-98 reduced the mouse's infarct size after tMCAO. Further, miR-98 lessened infiltration of proinflammatory Ly6CHI leukocytes into the brain following stroke and diminished the prevalence of M1 (activated) microglia within the impacted area. miR-98 attenuated BBB permeability, as demonstrated by changes to fluorescently-labeled dextran penetration in vivo and improved transendothelial electrical resistance (TEER) in vitro. Treatment with miR-98 improved significantly the locomotor impairment. Our study provides identification and functional assessment of miRNAs in brain endothelium and lays the groundwork for improving therapeutic approaches for patients suffering from ischemic attacks.
Collapse
Affiliation(s)
- David L Bernstein
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, USA
| | | | - Sachin Gajghate
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, USA
| | - Nancy L Reichenbach
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, USA
| | - Boris Polyak
- Department of Surgery, Drexel University College of Medicine, PA, USA
| | - Yuri Persidsky
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, USA.,Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Slava Rom
- Department of Pathology and Laboratory Medicine, Temple University, Philadelphia, PA, USA.,Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
44
|
Liu F, Cheng X, Zhong S, Liu C, Jolkkonen J, Zhang X, Liang Y, Liu Z, Zhao C. Communications Between Peripheral and the Brain-Resident Immune System in Neuronal Regeneration After Stroke. Front Immunol 2020; 11:1931. [PMID: 33042113 PMCID: PMC7530165 DOI: 10.3389/fimmu.2020.01931] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
Cerebral ischemia may cause irreversible neural network damage and result in functional deficits. Targeting neuronal repair after stroke potentiates the formation of new connections, which can be translated into a better functional outcome. Innate and adaptive immune responses in the brain and the periphery triggered by ischemic damage participate in regulating neural repair after a stroke. Immune cells in the blood circulation and gut lymphatic tissues that have been shaped by immune components including gut microbiota and metabolites can infiltrate the ischemic brain and, once there, influence neuronal regeneration either directly or by modulating the properties of brain-resident immune cells. Immune-related signalings and metabolites from the gut microbiota can also directly alter the phenotypes of resident immune cells to promote neuronal regeneration. In this review, we discuss several potential mechanisms through which peripheral and brain-resident immune components can cooperate to promote first the resolution of neuroinflammation and subsequently to improved neural regeneration and a better functional recovery. We propose that new insights into discovery of regulators targeting pro-regenerative process in this complex neuro-immune network may lead to novel strategies for neuronal regeneration.
Collapse
Affiliation(s)
- Fangxi Liu
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Xi Cheng
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Shanshan Zhong
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Chang Liu
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Jukka Jolkkonen
- A.I. Virtanen Institute and Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Xiuchun Zhang
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Yifan Liang
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Zhouyang Liu
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Chuansheng Zhao
- Neurology, The First Hospital of China Medical University, Shenyang, China.,Stroke Center, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
45
|
Urolithin A Prevents Focal Cerebral Ischemic Injury via Attenuating Apoptosis and Neuroinflammation in Mice. Neuroscience 2020; 448:94-106. [PMID: 32946950 DOI: 10.1016/j.neuroscience.2020.09.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 12/31/2022]
Abstract
Neuroinflammation contributes to neuronal death in cerebral ischemia. Urolithin A (UA), a gut microbial metabolite of ellagic acid, has emerged as a potential anti-inflammatory agent. However, its roles and precise mechanisms in stroke remain unknown. Here we found that UA treatment ameliorated infarction, neurological deficit scores, and spatial memory deficits after cerebral ischemia. Furthermore, UA significantly reduced neuron loss and promoted neurogenesis after ischemic stroke. We also found that UA attenuated apoptosis by regulating apoptotic-related proteins. Meanwhile, UA treatment inhibited glial activation via affecting inflammatory signaling pathways, specifically by enhancing cerebral AMPK and IκBa activation while decreasing the activation of Akt, P65NFκB, ERK, JNK, and P38MAPK. Our findings reveal a key role of UA against ischemic stroke through modulating apoptosis and neuroinflammation in mice.
Collapse
|
46
|
Maida CD, Norrito RL, Daidone M, Tuttolomondo A, Pinto A. Neuroinflammatory Mechanisms in Ischemic Stroke: Focus on Cardioembolic Stroke, Background, and Therapeutic Approaches. Int J Mol Sci 2020; 21:E6454. [PMID: 32899616 PMCID: PMC7555650 DOI: 10.3390/ijms21186454] [Citation(s) in RCA: 349] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 08/29/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
One of the most important causes of neurological morbidity and mortality in the world is ischemic stroke. It can be a result of multiple events such as embolism with a cardiac origin, occlusion of small vessels in the brain, and atherosclerosis affecting the cerebral circulation. Increasing evidence shows the intricate function played by the immune system in the pathophysiological variations that take place after cerebral ischemic injury. Following the ischemic cerebral harm, we can observe consequent neuroinflammation that causes additional damage provoking the death of the cells; on the other hand, it also plays a beneficial role in stimulating remedial action. Immune mediators are the origin of signals with a proinflammatory position that can boost the cells in the brain and promote the penetration of numerous inflammatory cytotypes (various subtypes of T cells, monocytes/macrophages, neutrophils, and different inflammatory cells) within the area affected by ischemia; this process is responsible for further ischemic damage of the brain. This inflammatory process seems to involve both the cerebral tissue and the whole organism in cardioembolic stroke, the stroke subtype that is associated with more severe brain damage and a consequent worse outcome (more disability, higher mortality). In this review, the authors want to present an overview of the present learning of the mechanisms of inflammation that takes place in the cerebral tissue and the role of the immune system involved in ischemic stroke, focusing on cardioembolic stroke and its potential treatment strategies.
Collapse
Affiliation(s)
- Carlo Domenico Maida
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (R.L.N.); (M.D.); (A.T.); (A.P.)
- Molecular and Clinical Medicine PhD Programme, University of Palermo, 90127 Palermo, Italy
| | - Rosario Luca Norrito
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (R.L.N.); (M.D.); (A.T.); (A.P.)
| | - Mario Daidone
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (R.L.N.); (M.D.); (A.T.); (A.P.)
| | - Antonino Tuttolomondo
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (R.L.N.); (M.D.); (A.T.); (A.P.)
| | - Antonio Pinto
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (R.L.N.); (M.D.); (A.T.); (A.P.)
| |
Collapse
|
47
|
Wang D, Liu F, Zhu L, Lin P, Han F, Wang X, Tan X, Lin L, Xiong Y. FGF21 alleviates neuroinflammation following ischemic stroke by modulating the temporal and spatial dynamics of microglia/macrophages. J Neuroinflammation 2020; 17:257. [PMID: 32867781 PMCID: PMC7457364 DOI: 10.1186/s12974-020-01921-2] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 08/10/2020] [Indexed: 12/26/2022] Open
Abstract
Background Resident microglia and macrophages are the predominant contributors to neuroinflammation and immune reactions, which play a critical role in the pathogenesis of ischemic brain injury. Controlling inflammatory responses is considered a promising therapeutic approach for stroke. Recombinant human fibroblast growth factor 21 (rhFGF21) presents anti-inflammatory properties by modulating microglia and macrophages; however, our knowledge of the inflammatory modulation of rhFGF21 in focal cerebral ischemia is lacking. Therefore, we investigated whether rhFGF21 improves ischemic outcomes in experimental stroke by targeting microglia and macrophages. Methods C57BL/6 mice were subjected to middle cerebral artery occlusion (MCAO) and randomly divided into groups that received intraperitoneal rhFGF21 or vehicle daily starting at 6 h after reperfusion. Behavior assessments were monitored for 14 days after MCAO, and the gene expression levels of inflammatory cytokines were analyzed via qRT-PCR. The phenotypic variation of microglia/macrophages and the presence of infiltrated immune cells were examined by flow cytometry and immunostaining. Additionally, magnetic cell sorting (MACS) in combination with fluorescence-activated cell sorting (FACS) was used to purify microglia and macrophages. Results rhFGF21 administration ameliorated neurological deficits in behavioral tests by regulating the secretion of pro-inflammatory and anti-inflammatory cytokines. rhFGF21 also attenuated the polarization of microglia/macrophages toward the M1 phenotype and the accumulation of peripheral immune cells after stroke, accompanied by a temporal evolution of the phenotype of microglia/macrophages and infiltration of peripheral immune cells. Furthermore, rhFGF21 treatment inhibited M1 polarization of microglia and pro-inflammatory cytokine expression through its actions on FGF receptor 1 (FGFR1) by suppressing nuclear factor-kappa B (NF-κB) and upregulating peroxisome proliferator-activated receptor-γ (PPAR-γ). Conclusions rhFGF21 treatment promoted functional recovery in experimental stroke by modulating microglia/macrophage-mediated neuroinflammation via the NF-κB and PPAR-γ signaling pathways, making it a potential anti-inflammatory agent for stroke treatment.
Collapse
Affiliation(s)
- Dongxue Wang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fei Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Liyun Zhu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ping Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fanyi Han
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xianxi Tan
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Li Lin
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China. .,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Ye Xiong
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
48
|
Adaptive Immune Cells Link Microbial Metabolites to Stroke Recovery. J Neurosci 2020; 40:5344-5346. [PMID: 32641452 DOI: 10.1523/jneurosci.0634-20.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/15/2020] [Accepted: 06/04/2020] [Indexed: 11/21/2022] Open
|
49
|
Ahnstedt H, Patrizz A, Chauhan A, Roy-O’Reilly M, Furr JW, Spychala MS, D’Aigle J, Blixt FW, Zhu L, Alegria JB, McCullough LD. Sex differences in T cell immune responses, gut permeability and outcome after ischemic stroke in aged mice. Brain Behav Immun 2020; 87:556-567. [PMID: 32058038 PMCID: PMC7590503 DOI: 10.1016/j.bbi.2020.02.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/15/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Stroke is a disease that presents with well-known sex differences. While women account for more stroke deaths, recent data show that after adjusting for age and pre-stroke functional status, mortality is higher in men. Immune responses are key determinants of stroke outcome and may differ by sex. This study examined sex differences in central and peripheral T cell immune responses, systemic effects on gut permeability and microbiota diversity and behavioral outcomes after stroke in aged mice. We hypothesized that there are sex differences in the immune response to stroke in aged animals. METHODS C57BL/6CR mice (20-22 months) were subjected to 60 min middle cerebral artery occlusion, or sham surgery. T cells were quantified in brain and blood at 3, 7 and 15 days (d) post-stroke by flow cytometry. Peripheral effects on gut permeability and microbiota diversity, as well as neurological function were assessed up to 14 d, and at 21 d (cognitive function) post-stroke. Brain glial fibrillary acidic protein (GFAP) expression was evaluated at 42 d post-stroke. RESULTS AND DISCUSSION Mortality (50% vs 14%, p < 0.05) and hemorrhagic transformation (44% vs 0%) were significantly higher in males than in females. No difference in infarct size at 3d were observed. Peripherally, stroke induced greater gut permeability of FITC-dextran in males at d3 (p < 0.05), and non-reversible alterations in microbiota diversity in males. Following the sub-acute phase, both sexes demonstrated a time-dependent increase of CD4+ and CD8+ T cells in the brain, with significantly higher levels of CD8+ T cells and Regulatory T cells in males at d15 (p < 0.01). Aged males demonstrated greater neurological deficits up to d5 and impaired sensorimotor function up to d15 when assessed by the corner asymmetry test (p < 0.001 and p < 0.01, respectively). A trend in greater cognitive decline was observed at d21 in males. Increased GFAP expression in the ischemic hemisphere, indicating astroglial activation and gliosis, was demonstrated in both males and females 42d post-stroke. Our findings indicate that despite a similar initial ischemic brain injury, aged male mice experience greater peripheral effects on the gut and ongoing central neuroinflammation past the sub-acute phase after stroke.
Collapse
Affiliation(s)
- Hilda Ahnstedt
- BRAINS Research Laboratory, Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bernstein DL, Gajghate S, Reichenbach NL, Winfield M, Persidsky Y, Heldt NA, Rom S. let-7g counteracts endothelial dysfunction and ameliorating neurological functions in mouse ischemia/reperfusion stroke model. Brain Behav Immun 2020; 87:543-555. [PMID: 32017988 PMCID: PMC7316629 DOI: 10.1016/j.bbi.2020.01.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/22/2020] [Accepted: 01/31/2020] [Indexed: 12/15/2022] Open
Abstract
Stroke is a debilitating disease, accounting for almost 20% of all hospital visits, and 8% of all fatalities in the United States in 2017. Following an ischemic attack, inflammatory processes originating from endothelial cells within the brain microvasculature can induce many toxic effects into the impacted area, from both sides of the blood brain barrier (BBB). In addition to increased BBB permeability, impacted brain microvascular endothelial cells can recruit macrophages and other immune cells from the periphery and can also trigger the activation of microglia and astrocytes within the brain. We have identified a key microRNA, let-7g, which levels were drastically diminished as consequence of transient middle cerebral artery occlusion (tMCAO) in vivo and oxygen-glucose deprivation (OGD) in vitro ischemia/reperfusion conditions, respectively. We have observed that let-7g* liposome-based delivery is capable of attenuating inflammation after stroke, reducing BBB permeability, limiting brain infiltration by CD3+CD4+ T-cells and Ly6G+ neutrophils, lessening microglia activation and neuronal death. These effects consequently improved clinical outcomes, shown by mitigating post-stroke gait asymmetry and extremity motor function. Due to the role of the endothelium in propagating the effects of stroke and other inflammation, treatments which can reduce endothelial inflammation and limit ischemic damage and improving recovery after a stroke are required. Our findings demonstrate a critical link between the CNS inflammation and the immune system reaction and lay important groundwork for future stroke pharmacotherapies.
Collapse
Affiliation(s)
- David L Bernstein
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Sachin Gajghate
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Nancy L Reichenbach
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Malika Winfield
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Yuri Persidsky
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Nathan A Heldt
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Slava Rom
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|