1
|
Han Y, Sun Y, Peng S, Tang T, Zhang B, Yu R, Sun X, Guo S, Ma L, Li P, Yang P. PI3K/AKT pathway: A potential therapeutic target in cerebral ischemia-reperfusion injury. Eur J Pharmacol 2025; 998:177505. [PMID: 40118329 DOI: 10.1016/j.ejphar.2025.177505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
Cerebral ischemia is a prevalent cerebrovascular disorder, with the restoration of blocked blood vessels serving as the current standard clinical treatment. However, reperfusion can exacerbate neuronal damage and neurological dysfunction, resulting in cerebral ischemia-reperfusion (I/R) injury. Presently, clinical treatment strategies for cerebral I/R injury are limited, creating an urgent need to identify new effective therapeutic targets. The PI3K/AKT signaling pathway, a pro-survival pathway associated with cerebral I/R injury, has garnered significant attention. We conducted a comprehensive review of the literature on the PI3K/AKT pathway in the context of cerebral I/R. Our findings indicate that activation of the PI3K/AKT signaling pathway following cerebral I/R can alleviate oxidative stress, reduce endoplasmic reticulum stress (ERS), inhibit inflammatory responses, decrease neuronal apoptosis, autophagy, and pyroptosis, mitigate blood-brain barrier (BBB) damage, and promote neurological function recovery. Consequently, this pathway ultimately reduces neuronal death, alleviates brain tissue damage, decreases the volume of cerebral infarction, and improves behavioral impairments. These results suggest that the PI3K/AKT signaling pathway is a promising therapeutic target for further research and drug development, holding significant potential for the treatment of cerebral I/R injury.
Collapse
Affiliation(s)
- Yiming Han
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yu Sun
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shiyu Peng
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Tingting Tang
- First Clinical College, Xinxiang Medical University, Xinxiang, China
| | - Beibei Zhang
- First Clinical College, Xinxiang Medical University, Xinxiang, China
| | - Ruonan Yu
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Xiaoyan Sun
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shanshan Guo
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China; Staff Hospital of Henan Fifth Construction Group Co., Ltd, Zhengzhou, Henan, China
| | - Lijuan Ma
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Pengfei Yang
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| |
Collapse
|
2
|
Li X, Guan L, Liu Z, Du Z, Yuan Q, Zhou F, Yang X, Lv M, Lv L. Ubiquitination of ATAD3A by TRIM25 exacerbates cerebral ischemia-reperfusion injury via regulating PINK1/Parkin signaling pathway-mediated mitophagy. Free Radic Biol Med 2024; 224:757-769. [PMID: 39307194 DOI: 10.1016/j.freeradbiomed.2024.09.029] [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: 07/18/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND Cerebral ischemia-reperfusion injury (CI/RI) is a complex process leading to neuronal damage and death, with mitophagy implicated in its pathogenesis. However, the significance of mitophagy in CI/RI remains debated. HYPOTHESIS We hypothesized that TRIM25 reduces ATAD3A expression by ubiquitinating ATAD3A, promoting mitophagy via the PINK1/Parkin pathway, and aggravating CI/RI. STUDY DESIGN Rat middle cerebral artery occlusion (MCAO) followed by reperfusion and oxygen-glucose deprivation and reoxygenation (OGD/R) in PC12 cells were used as animal and cell models, respectively. METHODS To evaluate the success of the CI/R modeling, TTC and HE staining were employed. The determination of serum biochemical indexes was carried out using relative assay kits. The Western Blot analysis was employed to assess the expression of ATAD3A, TRIM25, as well as mitophagy-related proteins (PINK1, Parkin, P62, and LC3II/LC3I). The mRNA levels were detected using QRT-PCR. Mitochondrial membrane potential was assessed through JC-1 staining. Mitosox Red Assay Kit was utilized to measure mitochondrial reactive oxygen species levels in PC12 cells. Additionally, characterization of the mitophagy structure was performed using transmission electron microscopy (TEM). RESULTS Our findings showed down-regulation of ATAD3A and up-regulation of TRIM25 in both in vivo and in vitro CI/RI models. Various experimental techniques such as Western Blot, JC-1 staining, Mitosox assay, Immunofluorescence assay, and TEM observation supported the occurrence of PINK1/Parkin signaling pathway-mediated mitophagy in both models. ATAD3A suppressed mitophagy, while TRIM25 promoted it during CI/RI injury. Additionally, the results indicated that TRIM25 interacted with and ubiquitinated ATAD3A via the proteasome pathway, affecting ATAD3A protein stability and expression. CONCLUSION TRIM25 promoted Pink1/Parkin-dependent excessive mitophagy by destabilizing ATAD3A, exacerbating CI/RI. Targeting TRIM25 and ATAD3A may offer therapeutic strategies for mitigating CI/RI and associated neurological damage.
Collapse
Affiliation(s)
- Xin Li
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China
| | - Liyang Guan
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China
| | - Zhi'en Liu
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China; Inner Mongolia Autonomous Region People's Hospital, Inner Mongolia Autonomous Region, 010017, PR China
| | - Zaixing Du
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China
| | - Qianhui Yuan
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China
| | - Fuxin Zhou
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China
| | - Xiaobo Yang
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China.
| | - Mei Lv
- The ENT & Head and Neck Surgery Department, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, PR China.
| | - Li Lv
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, 116044, PR China.
| |
Collapse
|
3
|
Ren XS, He J, Li S, Hu H, Kyle M, Kohsaka S, Zhao LR. Hematopoietic Growth Factors Regulate the Entry of Monocytes into the Adult Brain via Chemokine Receptor CCR5. Int J Mol Sci 2024; 25:8898. [PMID: 39201584 PMCID: PMC11354986 DOI: 10.3390/ijms25168898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Monocytes are circulating macrophage precursors generated from bone marrow hematopoietic stem cells. In adults, monocytes continuously replenish cerebral border-associated macrophages under physiological conditions. Monocytes also rapidly infiltrate the brain in pathological settings. The mechanisms of recruiting monocyte-derived macrophages into the brain under pathological conditions have been extensively studied. However, it remains unclear how monocytes enter the brain to renew border-associated macrophages under physiological conditions. Using both in vitro and in vivo approaches, this study reveals that a combination of two hematopoietic growth factors, stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF), complementarily and synergistically enhances the adhesion of monocytes to cerebral endothelial cells in a dose-dependent manner. Cysteine-cysteine chemokine receptor 5 (CCR5) in brain endothelial cells, but not the cell adhesion molecules mediating neuroinflammation-related infiltration of monocyte-derived macrophages, modulates SCF+G-CSF-enhanced monocyte-endothelial cell adhesion. Blocking CCR5 or genetically deleting CCR5 reduces monocyte-endothelial cell adhesion induced by SCF+G-CSF. The SCF+G-CSF-enhanced recruitment of bone marrow-derived monocytes/macrophages into the cerebral perivascular space is also reduced in adult CCR5 knockout mice. This study demonstrates the role of SCF and G-CSF in regulating the entry of monocytes into the adult brain to replenish perivascular macrophages.
Collapse
Affiliation(s)
- Xuefang Sophie Ren
- Department of Neurology, Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Junchi He
- Department of Neurosurgery, The State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Songruo Li
- Department of Neurosurgery, The State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Heng Hu
- Department of Neurology, Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Michele Kyle
- Department of Neurosurgery, The State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Shinichi Kohsaka
- National Institute of Neuroscience, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8502, Japan
| | - Li-Ru Zhao
- Department of Neurology, Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
- Department of Neurosurgery, The State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
4
|
Ren X, He J, Hu H, Kohsaka S, Zhao LR. Hematopoietic growth factors Regulate Entry of Monocytes into the Adult Brain via Chemokine Receptor CCR5. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594359. [PMID: 38798506 PMCID: PMC11118552 DOI: 10.1101/2024.05.15.594359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Monocytes are circulating macrophage precursors and are generated from bone marrow hematopoietic stem cells. In the adults, monocytes continuously replenish cerebral border-associated macrophages under a physiological condition. Monocytes also rapidly infiltrate into the brain in the settings of pathological conditions. The mechanisms of recruiting monocyte-derived macrophages into the brain under pathological conditions have been extensively studied. However, it remains unclear how monocytes enter the brain for renewal of border-associated macrophages under the physiological condition. Using both in vitro and in vivo approaches, this study reveals that the combination of two hematopoietic growth factors, stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF), complementarily and synergistically enhances adhesion of monocytes to cerebral endothelial cells in a dose dependent manner. Cysteine-cysteine chemokine receptor 5 (CCR5) in brain endothelial cells, but not cell adhesion molecules mediating neuroinflammation-related infiltration of monocyte-derived macrophages, modulates the SCF+G-CSF-enhanced monocyte-endothelial cell adhesion. Blocking CCR5 or genetically deleting CCR5 reduces monocyte-endothelial cell adhesion induced by SCF+G-CSF. SCF+G-CSF-enhanced recruitment of bone marrow-derived monocytes/macrophages in cerebral perivascular space is also reduced in adult CCR5 knockout mice. This study demonstrates the contribution of SCF and G-CSF in regulating the entry of monocytes into the adult brain to replenish perivascular macrophages.
Collapse
|
5
|
Zhang LM, Liang XL, Xiong GF, Xing XL, Zhang QJ, Zhang BR, Liu MW. Analysis and identification of oxidative stress-ferroptosis related biomarkers in ischemic stroke. Sci Rep 2024; 14:3803. [PMID: 38360841 PMCID: PMC10869843 DOI: 10.1038/s41598-024-54555-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/14/2024] [Indexed: 02/17/2024] Open
Abstract
Studies have shown that a series of molecular events caused by oxidative stress is associated with ferroptosis and oxidation after ischemic stroke (IS). Differential analysis was performed to identify differentially expressed mRNA (DEmRNAs) between IS and control groups. Critical module genes were identified using weighted gene co-expression network analysis (WGCNA). DEmRNAs, critical module genes, oxidative stress-related genes (ORGs), and ferroptosis-related genes (FRGs) were crossed to screen for intersection mRNAs. Candidate mRNAs were screened based on the protein-protein interaction (PPI) network and the MCODE plug-in. Biomarkers were identified based on two types of machine learning algorithms, and the intersection was obtained. Functional items and related pathways of the biomarkers were identified using gene set enrichment analysis (GSEA). Finally, single-sample GSEA (ssGSEA) and Wilcoxon tests were used to identify differential immune cells. An miRNA-mRNA-TF network was created. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to verify the expression levels of biomarkers in the IS and control groups. There were 8287 DE mRNAs between the IS and control groups. The genes in the turquoise module were selected as critical module genes for IS. Thirty intersecting mRNAs were screened for overlaps. Seventeen candidate mRNAs were also identified. Four biomarkers (CDKN1A, GPX4, PRDX1, and PRDX6) were identified using two types of machine-learning algorithms. GSEA results indicated that the biomarkers were associated with steroid biosynthesis. Nine types of immune cells (activated B cells and neutrophils) were markedly different between the IS and control groups. We identified 3747 miRNA-mRNA-TF regulatory pairs in the miRNA-mRNA-TF regulatory network, including hsa-miR-4469-CDKN1A-BACH2 and hsa-miR-188-3p-GPX4-ATF2. CDKN1A, PRDX1, and PRDX6 were upregulated in IS samples compared with control samples. This study suggests that four biomarkers (CDKN1A, GPX4, PRDX1, and PRDX6) are significantly associated with IS. This study provides a new reference for the diagnosis and treatment of IS.
Collapse
Affiliation(s)
- Lin-Ming Zhang
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Xing-Ling Liang
- Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Gui-Fei Xiong
- Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Xuan-Lin Xing
- Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Qiu-Juan Zhang
- Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Bing-Ran Zhang
- Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Ming-Wei Liu
- Department of Emergency, People's Hospital of Dali Bai Autonomous Prefecture, No. 35 Renmin South Road, Xiaguan Street, Dalí, 671000, Yunnan, China.
| |
Collapse
|
6
|
Tai SH, Chao LC, Huang SY, Lin HW, Lee AH, Chen YY, Lee EJ. Nicotinamide Deteriorates Post-Stroke Immunodepression Following Cerebral Ischemia-Reperfusion Injury in Mice. Biomedicines 2023; 11:2145. [PMID: 37626642 PMCID: PMC10452067 DOI: 10.3390/biomedicines11082145] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Inducing experimental stroke leads to biphasic immune responses, where the early activation of immune functions is followed by severe immunosuppression accompanied by spleen and thymus atrophy. Nicotinamide, a water-soluble B-group vitamin, is a known neuroprotectant against brain ischemia in animal models. We examined the effect of nicotinamide on the central and peripheral immune response in experimental stroke models. (2) Methods: Nicotinamide (500 mg/kg) or saline was intravenously administered to C57BL/6 mice during reperfusion after transiently occluding the middle cerebral artery or after LPS injection. On day 3, the animals were examined for behavioral performance and were then sacrificed to assess brain infarction, blood-brain barrier (BBB) integrity, and the composition of immune cells in the brain, thymus, spleen, and blood using flow cytometry. (3) Results: Nicotinamide reduced brain infarction and microglia/macrophage activation following MCAo (p < 0.05). Similarly, in LPS-injected mice, microglia/macrophage activation was decreased upon treatment with nicotinamide (p < 0.05), suggesting a direct inhibitory effect of nicotinamide on microglia/macrophage activation. Nicotinamide decreased the infiltration of neutrophils into the brain parenchyma and ameliorated Evans blue leakage (p < 0.05), suggesting that a decreased infiltration of neutrophils could, at least partially, be the result of a more integrated BBB structure following nicotinamide treatment. Our studies also revealed that administering nicotinamide led to retarded B-cell maturation in the spleen and subsequently decreased circulating B cells in the thymus and bloodstream (p < 0.05). (4) Conclusions: Cumulatively, nicotinamide decreased brain inflammation caused by ischemia-reperfusion injury, which was mediated by a direct anti-inflammatory effect of nicotinamide and an indirect protective effect on BBB integrity. Administering nicotinamide following brain ischemia resulted in a decrease in circulating B cells. This warrants attention with respect to future clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - E-Jian Lee
- Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| |
Collapse
|
7
|
Li J, Qiu Y, Zhang C, Wang H, Bi R, Wei Y, Li Y, Hu B. The role of protein glycosylation in the occurrence and outcome of acute ischemic stroke. Pharmacol Res 2023; 191:106726. [PMID: 36907285 DOI: 10.1016/j.phrs.2023.106726] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 03/12/2023]
Abstract
Acute ischemic stroke (AIS) is a serious and life-threatening disease worldwide. Despite thrombolysis or endovascular thrombectomy, a sizeable fraction of patients with AIS have adverse clinical outcomes. In addition, existing secondary prevention strategies with antiplatelet and anticoagulant drugs therapy are not able to adequately decrease the risk of ischemic stroke recurrence. Thus, exploring novel mechanisms for doing so represents an urgent need for the prevention and treatment of AIS. Recent studies have discovered that protein glycosylation plays a critical role in the occurrence and outcome of AIS. As a common co- and post-translational modification, protein glycosylation participates in a wide variety of physiological and pathological processes by regulating the activity and function of proteins or enzymes. Protein glycosylation is involved in two causes of cerebral emboli in ischemic stroke: atherosclerosis and atrial fibrillation. Following ischemic stroke, the level of brain protein glycosylation becomes dynamically regulated, which significantly affects stroke outcome through influencing inflammatory response, excitotoxicity, neuronal apoptosis, and blood-brain barrier disruption. Drugs targeting glycosylation in the occurrence and progression of stroke may represent a novel therapeutic idea. In this review, we focus on possible perspectives about how glycosylation affects the occurrence and outcome of AIS. We then propose the potential of glycosylation as a therapeutic drug target and prognostic marker for AIS patients in the future.
Collapse
Affiliation(s)
- Jianzhuang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanmei Qiu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunlin Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hailing Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rentang Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanhao Wei
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanan 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
|
8
|
Lee RD, Chen YJ, Singh L, Nguyen HM, Wulff H. Immunocytoprotection after reperfusion with Kv1.3 inhibitors has an extended treatment window for ischemic stroke. Front Pharmacol 2023; 14:1190476. [PMID: 37180699 PMCID: PMC10166874 DOI: 10.3389/fphar.2023.1190476] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/17/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction: Mechanical thrombectomy has improved treatment options and outcomes for acute ischemic stroke with large artery occlusion. However, as the time window of endovascular thrombectomy is extended there is an increasing need to develop immunocytoprotective therapies that can reduce inflammation in the penumbra and prevent reperfusion injury. We previously demonstrated, that by reducing neuroinflammation, KV1.3 inhibitors can improve outcomes not only in young male rodents but also in female and aged animals. To further explore the therapeutic potential of KV1.3 inhibitors for stroke therapy, we here directly compared a peptidic and a small molecule KV1.3 blocker and asked whether KV1.3 inhibition would still be beneficial when started at 72 hours after reperfusion. Methods: Transient middle cerebral artery occlusion (tMCAO, 90-min) was induced in male Wistar rats and neurological deficit assessed daily. On day-8 infarction was determined by T2-weighted MRI and inflammatory marker expression in the brain by quantitative PCR. Potential interactions with tissue plasminogen activator (tPA) were evaluated in-vitro with a chromogenic assay. Results: In a direct comparison with administration started at 2 hours after reperfusion, the small molecule PAP-1 significantly improved outcomes on day-8, while the peptide ShK-223 failed to reduce infarction and neurological deficits despite reducing inflammatory marker expression. PAP-1 still provided benefits when started 72 hours after reperfusion. PAP-1 does not reduce the proteolytic activity of tPA. Discussion: Our studies suggest that KV1.3 inhibition for immunocytoprotection after ischemic stroke has a wide therapeutic window for salvaging the inflammatory penumbra and requires brain-penetrant small molecules.
Collapse
Affiliation(s)
- Ruth D. Lee
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Yi-Je Chen
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
- Animal Models Core, Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Latika Singh
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Hai M. Nguyen
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
9
|
Selective ischemic-hemisphere targeting Ginkgolide B liposomes with improved solubility and therapeutic efficacy for cerebral ischemia-reperfusion injury. Asian J Pharm Sci 2023; 18:100783. [PMID: 36891470 PMCID: PMC9986716 DOI: 10.1016/j.ajps.2023.100783] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/23/2022] [Accepted: 01/24/2023] [Indexed: 02/13/2023] Open
Abstract
Cerebral ischemia-reperfusion injury (CI/RI) remains the main cause of disability and death in stroke patients due to lack of effective therapeutic strategies. One of the main issues related to CI/RI treatment is the presence of the blood-brain barrier (BBB), which affects the intracerebral delivery of drugs. Ginkgolide B (GB), a major bioactive component in commercially available products of Ginkgo biloba, has been shown significance in CI/RI treatment by regulating inflammatory pathways, oxidative damage, and metabolic disturbance, and seems to be a candidate for stroke recovery. However, limited by its poor hydrophilicity and lipophilicity, the development of GB preparations with good solubility, stability, and the ability to cross the BBB remains a challenge. Herein, we propose a combinatorial strategy by conjugating GB with highly lipophilic docosahexaenoic acid (DHA) to obtain a covalent complex GB-DHA, which can not only enhance the pharmacological effect of GB, but can also be encapsulated in liposomes stably. The amount of finally constructed Lipo@GB-DHA targeting to ischemic hemisphere was validated 2.2 times that of free solution in middle cerebral artery occlusion (MCAO) rats. Compared to the marketed ginkgolide injection, Lipo@GB-DHA significantly reduced infarct volume with better neurobehavioral recovery in MCAO rats after being intravenously administered both at 2 h and 6 h post-reperfusion. Low levels of reactive oxygen species (ROS) and high neuron survival in vitro was maintained via Lipo@GB-DHA treatment, while microglia in the ischemic brain were polarized from the pro-inflammatory M1 phenotype to the tissue-repairing M2 phenotype, which modulate neuroinflammatory and angiogenesis. In addition, Lipo@GB-DHA inhibited neuronal apoptosis via regulating the apoptotic pathway and maintained homeostasis by activating the autophagy pathway. Thus, transforming GB into a lipophilic complex and loading it into liposomes provides a promising nanomedicine strategy with excellent CI/RI therapeutic efficacy and industrialization prospects.
Collapse
|
10
|
Offner H, Lockwood D, Meza-Romero R, Vandenbark AA. PD-L1 is required for estrogen-induced protection against severe EAE in IL-10 deficient mice 1. Metab Brain Dis 2023; 38:589-599. [PMID: 36454506 PMCID: PMC9976593 DOI: 10.1007/s11011-022-01129-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND IL-10 knockout (KO) mice can be protected against experimental autoimmune encephalomyelitis (EAE) with low-dose estrogen (E2) treatment similar to wild type (WT) mice, indicating that IL-10 is not required for E2-induced EAE protection. Our previous study demonstrated that E2 treatment induced an increase in programmed death ligands 1 (PD-L1) and 2 (PD-L2) on monocytes and macrophages in the periphery and within the CNS. In this study, we selectively inhibited the function of PD-L1 and PD-L2 to evaluate their critical role in maintaining E2-induced protection against EAE in IL-10-KO mice. METHODS This study used female IL-10 KO mice pre-treated with either E2 or sham pellets seven days prior to induction of EAE and subsequently treated with Vehicle or antibodies to PD-L1, PD-L2 or respective isotype controls. Mice were scored daily for EAE severity over 21 days post-EAE induction. Cells from the spleen and brain were evaluated by flow cytometry. RESULTS Differences in EAE severity were assessed in E2 and sham pre-treated IL-10-KO mice treated with α-PD-L1 or α-PD-L2 antibodies over the course of disease compared to treatment with Vehicle or isotype control antibodies. The results revealed real-time development of severe EAE in E2-pre-treated IL-10-KO mice treated with α-PD-L1 but not α-PD-L2 antibodies, mediated in part by increased percentages of activated CD74+CD11b+ myeloid cells in spleen and brain as well as splenic B-cells, T-cells and CD73+ cells. CONCLUSION These results demonstrate unequivocally that PD-L1 but not PD-L2 was required to retain the inhibitory effects of E2 on clinical EAE scores in female IL-10-KO mice and further implicate the emergence of the MIF/CD74 axis as a contributing pathogenic mechanism.
Collapse
Affiliation(s)
- Halina Offner
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA.
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA.
- Veterans Affairs Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., 97239, Portland, OR, USA.
| | - Denesa Lockwood
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA
| | - Roberto Meza-Romero
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
11
|
Bai Y, He Z, Duan W, Gu H, Wu K, Yuan W, Liu W, Huang H, Li Y. Sodium formononetin-3'-sulphonate alleviates cerebral ischemia-reperfusion injury in rats via suppressing endoplasmic reticulum stress-mediated apoptosis. BMC Neurosci 2022; 23:74. [PMID: 36482320 PMCID: PMC9733209 DOI: 10.1186/s12868-022-00762-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Sodium formononetin-3'-sulphonate (Sul-F) may alleviate I/R injury in vivo with uncertain mechanism. Endoplasmic reticulum (ER) stress-mediated apoptosis participates in the process of cerebral ischemia-reperfusion (I/R) injury. Our aim is to figure out the effect of Sul-F on cerebral I/R injury and to verify whether it works through suppressing ER stress-mediated apoptosis. RESULTS The cerebral lesions of middle cerebral artery occlusion (MCAO) model in SD rats were aggravated after 24 h of reperfusion, including impaired neurological function, increased infarct volume, intensified inflammatory response and poor cell morphology. After intervention, the edaravone (EDA, 3 mg/kg) group and Sul-F high-dose (Sul-F-H, 80 mg/kg) group significantly alleviated I/R injury via decreasing neurological score, infarct volume and the serum levels of inflammatory factors (TNF-α, IL-1β and IL-6), as well as alleviating pathological injury. Furthermore, the ER stress level and apoptosis rate were elevated in the ischemic penumbra of MCAO group, and were significantly blocked by EDA and Sul-F-H. In addition, EDA and Sul-F-H significantly down-regulated the ER stress related PERK/eIF2α/ATF4 and IRE1 signal pathways, which led to reduced cell apoptosis rate compared with the MCAO group. Furthermore, there was no difference between the EDA and Sul-F-H group in terms of therapeutic effect on cerebral I/R injury, indicating a therapeutic potential of Sul-F for ischemic stroke. CONCLUSIONS Sul-F-H can significantly protects against cerebral I/R injury through inhibiting ER stress-mediated apoptosis in the ischemic penumbra, which might be a novel therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Yue Bai
- grid.256883.20000 0004 1760 8442Department of Internal Medicine, Shijiazhuang Pingan Hospital, Hebei Medical University, Shijiazhuang, 050000 Hebei China
| | - Zhiwei He
- grid.256883.20000 0004 1760 8442Department of Internal Medicine, Shijiazhuang Pingan Hospital, Hebei Medical University, Shijiazhuang, 050000 Hebei China
| | - Weisong Duan
- grid.452702.60000 0004 1804 3009Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei China
| | - He Gu
- grid.256883.20000 0004 1760 8442Department of Internal Medicine, Shijiazhuang Pingan Hospital, Hebei Medical University, Shijiazhuang, 050000 Hebei China
| | - Kefeng Wu
- grid.256883.20000 0004 1760 8442Department of Internal Medicine, Shijiazhuang Pingan Hospital, Hebei Medical University, Shijiazhuang, 050000 Hebei China
| | - Wei Yuan
- grid.256883.20000 0004 1760 8442Department of Internal Medicine, Shijiazhuang Pingan Hospital, Hebei Medical University, Shijiazhuang, 050000 Hebei China
| | - Wenkang Liu
- grid.256883.20000 0004 1760 8442Department of Internal Medicine, Shijiazhuang Pingan Hospital, Hebei Medical University, Shijiazhuang, 050000 Hebei China
| | - Huaipeng Huang
- grid.256883.20000 0004 1760 8442Department of Internal Medicine, Shijiazhuang Pingan Hospital, Hebei Medical University, Shijiazhuang, 050000 Hebei China
| | - Yanan Li
- grid.256883.20000 0004 1760 8442Department of Clinical Laboratory Diagnosis, Shijiazhuang Pingan Hospital, Hebei Medical University, Shijiazhuang, 050000 Hebei China
| |
Collapse
|
12
|
Zhu T, Wang L, Wang LP, Wan Q. Therapeutic targets of neuroprotection and neurorestoration in ischemic stroke: Applications for natural compounds from medicinal herbs. Biomed Pharmacother 2022; 148:112719. [DOI: 10.1016/j.biopha.2022.112719] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/13/2022] Open
|
13
|
Amu S, Malone K. Flow cytometry and stroke: from current methodology to future applications. Neural Regen Res 2022; 17:1748-1750. [PMID: 35017430 PMCID: PMC8820698 DOI: 10.4103/1673-5374.332138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Sylvie Amu
- Cancer Research , Ireland.,UCC, University College Cork, Cork, Ireland
| | - Kyle Malone
- Department of Pharmacology and Therapeutics, Western Gateway Building; School of Pharmacy, University College Cork, Cork, Ireland
| |
Collapse
|
14
|
Fu K, Zhang D, Song Y, Xu M, Wu R, Xiong X, Liu X, Wu L, Guo Y, Zhou Y, Li X, Wang Z. Tibetan Medicine Qishiwei Zhenzhu Pills Can Reduce Cerebral Ischemia-Reperfusion Injury by Regulating Gut Microbiota and Inhibiting Inflammation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:2251679. [PMID: 34804175 PMCID: PMC8601817 DOI: 10.1155/2021/2251679] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/19/2021] [Accepted: 10/27/2021] [Indexed: 01/11/2023]
Abstract
Cerebral ischemia is a series of harmful reactions, such as acute necrosis of tissue, inflammation, apoptosis, autophagy, and blood-brain barrier injury, due to the insufficient blood supply to the brain. Inflammatory response and gut microbiota imbalance are important concomitant factors of cerebral ischemia and may increase the severity of cerebral ischemia through the gut-brain axis. Qishiwei Zhenzhu pills (QSW) contain more than 70 kinds of medicinal materials, which have the effects of anti-cerebral infarction, anti-convulsion, anti-dementia, and so on. It is a treasure of Tibetan medicine commonly used in the treatment of cerebral ischemia in Tibetan areas. In this study, we gave rats QSW (66.68 mg/kg) once by gavage in advance and then immediately established the rat middle cerebral artery occlusion (MCAO) model. After 24 hours of treatment, the neuroprotection, intestinal pathology, and gut microbiota were examined. The results showed that QSW could significantly reduce the neurobehavioral abnormalities and cerebral infarction rate in MCAO rats. Furthermore, qPCR, western blot, and immunohistochemistry results showed that QSW could effectively inhibit IL-6, IL-1β, and other inflammatory factors so as to effectively reduce the inflammatory response of MCAO rats. Furthermore, QSW could improve intestinal integrity and reduce intestinal injury. 16S rRNA sequencing showed that QSW could significantly improve the gut microbiota disorder of MCAO rats. Specifically, at the phylum level, it can regulate the abundance of Firmicutes and Proteobacteria in the gut microbiota of rats with MCAO. At the genus level, it can adjust the abundance of Escherichia and Shigella. At the species level, it can adjust the abundance of Lactobacillus johnsonii and Lactobacillus reuteri. All in all, this study is the first to show that QSW can reduce the severity of cerebral ischemia-reperfusion injury by regulating gut microbiota and inhibiting the inflammatory response.
Collapse
Affiliation(s)
- Ke Fu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dewei Zhang
- Wanzhou Institute for Drug and Food Control, Chongqing 404000, China
| | - Yinglian Song
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Min Xu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ruixia Wu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xueqing Xiong
- Wanzhou Institute for Drug and Food Control, Chongqing 404000, China
| | - Xianwu Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lei Wu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ya Guo
- Wanzhou Institute for Drug and Food Control, Chongqing 404000, China
| | - You Zhou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaoli Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhang Wang
- College of Ethnomedicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
15
|
Wang Y, Wang Y, Li S, Cui Y, Liang X, Shan J, Gu W, Qiu J, Li Y, Wang G. Functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke. J Nanobiotechnology 2021; 19:331. [PMID: 34674712 PMCID: PMC8529766 DOI: 10.1186/s12951-021-01067-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/29/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Ischemic stroke is an acute and severe neurological disease, and reperfusion is an effective way to reverse brain damage after stroke. However, reperfusion causes secondary tissue damage induced by inflammatory responses, called ischemia/reperfusion (I/R) injury. Current therapeutic strategies that control inflammation to treat I/R are less than satisfactory. RESULTS We report a kind of shield and sword nano-soldier functionalized nanoparticles (monocyte membranes-coated rapamycin nanoparticles, McM/RNPs) that can reduce inflammation and relieve I/R injury by blocking monocyte infiltration and inhibiting microglia proliferation. The fabricated McM/RNPs can actively target and bind to inflammatory endothelial cells, which inhibit the adhesion of monocytes to the endothelium, thus acting as a shield. Subsequently, McM/RNPs can penetrate the endothelium to reach the injury site, similar to a sword, and release the RAP drug to inhibit the proliferation of inflammatory cells. In a rat I/R injury model, McM/RNPs exhibited improved active homing to I/R injury areas and greatly ameliorated neuroscores and infarct volume. Importantly, in vivo animal studies revealed good safety for McM/RNPs treatment. CONCLUSION The results demonstrated that the developed McM/RNPs may serve as an effective and safe nanovehicles for I/R injury therapy.
Collapse
Affiliation(s)
- Yanyun Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Shuyu Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yuliang Cui
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Xiping Liang
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Juanjuan Shan
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Wei Gu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
| | - Yiliang Li
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, Guangdong, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
16
|
García-Belda P, Prima-García H, Aliena-Valero A, Castelló-Ruiz M, Ulloa-Navas MJ, Ten-Esteve A, Martí-Bonmatí L, Salom JB, García-Verdugo JM, Gil-Perotín S. Intravenous SPION-labeled adipocyte-derived stem cells targeted to the brain by magnetic attraction in a rat stroke model: An ultrastructural insight into cell fate within the brain. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 39:102464. [PMID: 34583057 DOI: 10.1016/j.nano.2021.102464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 07/09/2021] [Accepted: 08/06/2021] [Indexed: 12/19/2022]
Abstract
Mesenchymal stem cell therapy after stroke is a promising option investigated in animal models and clinical trials. The intravenous route is commonly used in clinical settings guaranteeing an adequate safety profile although low yields of engraftment. In this report, rats subjected to ischemic stroke were injected with adipose-derived stem cells (ADSCs) labeled with superparamagnetic iron oxide nanoparticles (SPIONs) applying an external magnetic field in the skull to retain the cells. Although most published studies demonstrate viability of ADSCs, only a few have used ultrastructural techniques. In our study, the application of a local magnetic force resulted in a tendency for higher yields of SPION-ADSCs targeting the brain. However, grafted cells displayed morphological signs of death, one day after administration, and correlative microscopy showed active microglia and astrocytes associated in the process of scavenging. Thus, we conclude that, although successfully targeted within the brain, SPION-ADSCs viability was rapidly compromised.
Collapse
Affiliation(s)
- Paula García-Belda
- Laboratory of Comparative Neurobiology, Institute Cavanilles, University of Valencia, Valencia, Spain
| | - Helena Prima-García
- Instituto de Ciencia Molecular (ICMol), Universitat de València, Paterna, Spain
| | - Alicia Aliena-Valero
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe-Universidad de Valencia, Valencia, Spain
| | - María Castelló-Ruiz
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe-Universidad de Valencia, Valencia, Spain; Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Burjassot, Spain
| | - María José Ulloa-Navas
- Laboratory of Comparative Neurobiology, Institute Cavanilles, University of Valencia, Valencia, Spain
| | - Amadeo Ten-Esteve
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute, Valencia, Spain
| | - Luis Martí-Bonmatí
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute, Valencia, Spain
| | - Juan B Salom
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe-Universidad de Valencia, Valencia, Spain; Departamento de Fisiología, Universidad de Valencia, Valencia, Spain.
| | | | - Sara Gil-Perotín
- Laboratory of Central Neuroimmunology, IIS Hospital La Fe, Valencia, Spain.
| |
Collapse
|
17
|
The Need for New Biomarkers to Assist with Stroke Prevention and Prediction of Post-Stroke Therapy Based on Plasma-Derived Extracellular Vesicles. Biomedicines 2021; 9:biomedicines9091226. [PMID: 34572411 PMCID: PMC8466486 DOI: 10.3390/biomedicines9091226] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/07/2021] [Accepted: 09/13/2021] [Indexed: 12/24/2022] Open
Abstract
The risk of having a stroke event doubles each decade after the age of 55. Therefore, it is of great interest to develop neurorestorative therapies of stroke which occurs mostly in elderly people. However, to date, patients at risk for these sequels of stroke are not duly diagnosed and treated due to the lack of reliable biomarkers. Extracellular vesicles (EVs) are lipid bilayer-delimited particles that are shed by the brain cells and are able to cross the blood–brain barrier and enter the blood stream; thus, they may be used to interrogate molecular and cellular events in the brain damaged area. In this review, we summarize the major molecular and cellular responses of astroglia and neurons to cerebral ischemia and assess their impact on post-stroke recovery and rehabilitation. In particular, we ask if EVs secreted by brain cells are responses to cerebral ischemia, and they may shed new light on the interplay between exosomes-mediated interactions between brain cells and the question of how to exploit it in order to predict the individual course of the disease and to introduce specific preventive or therapeutic strategies. Given these findings, we are left with two options: either to (i) transplant neuronal precursors into the damaged cortical area or (ii) to covert abundantly present proliferating astrocytes in the perilesional area into neurons by using recently developed genetic technologies. However, given the complexity of molecular and cellular responses to cerebral ischemia and our limited capabilities to restore brain structure and function, we are left with only one realistic aim: to invest more in prevention.
Collapse
|
18
|
Zhang X, Zhang X, Qiu C, Shen H, Zhang H, He Z, Song Z, Zhou W. The imbalance of Th17/Treg via STAT3 activation modulates cognitive impairment in P. gingivalis LPS-induced periodontitis mice. J Leukoc Biol 2021; 110:511-524. [PMID: 34342041 DOI: 10.1002/jlb.3ma0521-742rrr] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 01/05/2023] Open
Abstract
Periodontitis is one of the most common oral diseases worldwide, and it is associated with various systemic diseases, including cognitive diseases. STAT3 regulates the inflammatory cascade and influences adaptive immunity by modulating Th17/Treg cell differentiation. In this study, we aimed to explore the effect of adaptive immunity inside and outside the brain on the association between periodontitis and cognitive impairment and understand the role of the STAT3 signaling pathway. We established Porphyromonas gingivalis LPS-induced periodontitis mice models by injecting P. gingivalis LPS into the gingival sulcus of mice. Behavioral tests showed that learning and memory abilities were impaired. The flow cytometry data showed an imbalance in the Th17/Treg ratio in the blood and brain samples of the mice. The expression of Th17-related cytokines (IL-1β, IL-17A, IL-21, and IL-22) increased, whereas that of Treg-related cytokines (IL-2 and IL-10) decreased in both the blood and the brain. The level of LPS increased and the STAT3 signaling pathway was activated during this process. These effects were reversed by C188-9, a STAT3 inhibitor. In conclusion, P. gingivalis LPS-induced periodontitis may promote the occurrence and progression of cognitive impairment by modulating the Th17/Treg balance inside and outside the brain. The STAT3 signaling pathway may have immunoregulatory effects on the mouth-to-brain axis.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China.,Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Xuan Zhang
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Che Qiu
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hui Shen
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Huanyu Zhang
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhiyan He
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhongchen Song
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Wei Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Research Institute of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong Univerisity; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
19
|
Hernández J, Francos-Quijorna I, Redondo-Castro E, López-Vales R, Navarro X. Microglia Stimulation by Protein Extract of Injured Rat Spinal Cord. A Novel In vitro Model for Studying Activated Microglia. Front Mol Neurosci 2021; 14:582497. [PMID: 34093123 PMCID: PMC8176957 DOI: 10.3389/fnmol.2021.582497] [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] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 04/14/2021] [Indexed: 01/07/2023] Open
Abstract
Research on microglia has established the differentiation between the so-called M1 and M2 phenotypes. However, new frameworks have been proposed attempting to discern between meaningful microglia profiles. We have set up an in vitro microglial activation model by adding an injured spinal cord (SCI) lysate to microglial cultures, obtained from postnatal rats, in order to mimic the environment of the spinal cord after injury. We found that under the presence of the SCI lysate microglial cells changed their phenotype, developing less ramified but longer processes, and proliferated. The SCI lysate also led to upregulation of pro-inflammatory cytokines, such as IL-1β, IL-6, and TNF-α, downregulation of the anti-inflammatory cytokines IL-10 and IL-4, and a biphasic profile of iNOS. In addition, a latex beads phagocytosis assay revealed the SCI lysate stimulated the phagocytic capacity of microglia. Flow cytometry analysis indicated that microglial cells showed a pro-inflammatory profile in the presence of SCI lysate. Finally, characterization of the microglial activation in the spinal cord on day 7 after contusion injury, we showed that these cells have a pro-inflammatory phenotype. Overall, these results indicate that the use of SCI lysates could be a useful tool to skew microglia towards a closer phenotype to that observed after the spinal cord contusion injury than the use of LPS or IFNγ.
Collapse
Affiliation(s)
- Joaquim Hernández
- Group of Neuroplasticity and Regeneration, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Red de Terapia Celular (TerCel), Bellaterra, Spain
| | - Isaac Francos-Quijorna
- Group of Neuroplasticity and Regeneration, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Red de Terapia Celular (TerCel), Bellaterra, Spain
| | - Elena Redondo-Castro
- Group of Neuroplasticity and Regeneration, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Red de Terapia Celular (TerCel), Bellaterra, Spain
| | - Rubén López-Vales
- Group of Neuroplasticity and Regeneration, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Red de Terapia Celular (TerCel), Bellaterra, Spain
| | - Xavier Navarro
- Group of Neuroplasticity and Regeneration, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Red de Terapia Celular (TerCel), Bellaterra, Spain
| |
Collapse
|
20
|
Anyaegbu CC, Mao Y, McGonigle T, Raja S, Clarke T, Black AMB, Solomon T, Fuller K, Fitzgerald M. Simultaneous flow cytometric characterization of multiple cell types and metabolic states in the rat brain after repeated mild traumatic brain injury. J Neurosci Methods 2021; 359:109223. [PMID: 34004202 DOI: 10.1016/j.jneumeth.2021.109223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Cellular responses at the sub-acute phase of mild traumatic brain injury (mTBI), and their contribution to ongoing damage, are unclear, complex and require simultaneous assessment of multiple cells to elucidate. NEW METHOD An 11-colour flow cytometry method for analysing brain cells was evaluated in a weight-drop rat model of repeated mTBI. Animals received sham, one, two or three mTBI delivered at 24 h intervals (n = 6/group). Cerebrum homogenates were prepared 11 days after first mTBI, in two cohorts of n = 3/group to enable same-day staining of fresh tissue. Percentages of neurons, astrocytes, microglia, mature oligodendrocytes and NeuN + CC1+ cells, neutrophils, macrophages and non-myeloid leukocytes, and their immunoreactivity for cell damage indicators (inducible nitric oxide synthase; iNOS, proliferating cell nuclear antigen; PCNA, 8-Oxo-2'-deoxyguanosine; 8OHDG and 4-hydroxynonenal; HNE), were assessed. RESULTS Median fluorescence intensity (MFI) of iNOS in activated microglia increased following two, but not one or three, mTBI (p = 0.04). However, there were differences between processing cohorts in terms of percentages and MFI of some PCNA+, iNOS+, 8OHDG + and HNE + cell populations. COMPARISON WITH EXISTING METHODS Previous applications of flow cytometry for rat brain analysis were typically limited to three or four markers. This method uses 11 markers to identify nine cell populations and evaluate their immunoreactivity to four metabolic indicators of cell damage. CONCLUSIONS Flow cytometry can be useful for discerning injury-related changes in multiple rat brain cells. However, markers sensitive to subtle changes in experimental conditions must be identified in pilot experiments and subsequently analysed in the same tissue-processing cohort.
Collapse
Affiliation(s)
- Chidozie C Anyaegbu
- Curtin Health Innovation Research Institute, Curtin University, Belmont, Western Australia, Australia.
| | - Yilin Mao
- Curtin Health Innovation Research Institute, Curtin University, Belmont, Western Australia, Australia
| | - Terry McGonigle
- Curtin Health Innovation Research Institute, Curtin University, Belmont, Western Australia, Australia
| | - Sushmitha Raja
- Curtin Health Innovation Research Institute, Curtin University, Belmont, Western Australia, Australia
| | - Thomas Clarke
- Curtin Health Innovation Research Institute, Curtin University, Belmont, Western Australia, Australia
| | - Anna M B Black
- Curtin Health Innovation Research Institute, Curtin University, Belmont, Western Australia, Australia
| | - Tanya Solomon
- Curtin Health Innovation Research Institute, Curtin University, Belmont, Western Australia, Australia
| | - Kathy Fuller
- Division of Cancer Biology, School of Biomedical Sciences, The University of Western Australia, 35 Stirling Hwy, Nedlands 6009 Western Australia, Australia
| | - Melinda Fitzgerald
- Curtin Health Innovation Research Institute, Curtin University, Belmont, Western Australia, Australia; Perron Institute for Neurological and Translational Science, Sarich Neuroscience Research Institute Building, 8 Verdun St, Nedlands 6009 Western Australia, Australia
| |
Collapse
|
21
|
Chang CF, Goods BA, Askenase MH, Beatty HE, Osherov A, DeLong JH, Hammond MD, Massey J, Landreneau M, Love JC, Sansing LH. Divergent Functions of Tissue-Resident and Blood-Derived Macrophages in the Hemorrhagic Brain. Stroke 2021; 52:1798-1808. [PMID: 33840225 DOI: 10.1161/strokeaha.120.032196] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Che-Feng Chang
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei (C.-F.C.).,Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Brittany A Goods
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Cambridge (B.A.G., J.C.L.)
| | - Michael H Askenase
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT.,Immunobiology (M.H.A., H.E.B., J.H.D., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Hannah E Beatty
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT.,Immunobiology (M.H.A., H.E.B., J.H.D., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Artem Osherov
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Jonathan H DeLong
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT.,Immunobiology (M.H.A., H.E.B., J.H.D., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Matthew D Hammond
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Jordan Massey
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Margaret Landreneau
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - J Christopher Love
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Cambridge (B.A.G., J.C.L.)
| | - Lauren H Sansing
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT.,Immunobiology (M.H.A., H.E.B., J.H.D., L.H.S.), Yale University School of Medicine, New Haven, CT
| |
Collapse
|
22
|
Ye X, Peng X, Song Q, Zeng T, Xiong X, Huang Y, Cai X, Zhang C, Wang C, Wang B. Borneol-modified tanshinone IIA liposome improves cerebral ischemia reperfusion injury by suppressing NF-κB and ICAM-1 expression. Drug Dev Ind Pharm 2021; 47:609-617. [PMID: 33834937 DOI: 10.1080/03639045.2021.1908331] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To investigate the metabolism and brain tissue distribution of borneol-modified tanshinone IIA liposome (BO-TA-Lip) and its effect on NF-κB and ICAM-1 in cerebral ischemia reperfusion rats, thereby exploring the ameliorative mechanism of BO-TA-Lip on ischemic encephalopathy. METHODS Particle size, entrapment efficiency, drug loading were measured to evaluate the preparation comprehensively. Metabolism and brain tissue distributions in vivo were measured by HPLC, and the pharmacokinetic parameters were calculated. In addition, 24 SD rats were randomly divided into sham, model, STS (sodium tanshinone IIA sulfonate, 30 mg/kg) and BO-TA-Lip groups (44 mg/kg). The middle cerebral artery occlusion (MCAO) rats were constructed with thread embolism method. Neurological deficits were scored using Zea Longa scoring standard. TTC and HE staining were used for the cerebral infarction and histopathological examination, respectively. The protein expression was examined by immunohistochemistry and Western blot. RESULTS The average particle size, encapsulation efficiency and drug loading of BO-TA-Lip were (135.33 ± 7.25) nm, (85.95 ± 3.20)% and (4.06 ± 0.31)%, respectively. Both in the pharmacokinetic analysis of plasma and brain tissue, in BO-TA-Lip group, the peak concentration and the area under the curve increased, and the clearance rate decreased. The neurological deficit scores and infarct area of the BO-TA-Lip group were significantly lower than that of the model and STS groups. Besides, BO-TA-Lip reduced the protein expression of NF-κB, ICAM-1, IL-1β, TNF-α and IL-6 in the brain tissue. CONCLUSION BO-TA-Lip had higher bioavailability and better absorption in brain tissue, and could improve cerebral ischemia reperfusion injury, which might be related to the inhibitory effect of BO-TA-Lip in expression of NF-κB and ICAM-1.
Collapse
Affiliation(s)
- Xiaoli Ye
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xueying Peng
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qing Song
- Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Taohui Zeng
- First Affiliated Hospital, Gannan Medical College, Ganzhou, China
| | | | - Yuye Huang
- The Affiliated Cangnan Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinjun Cai
- Zhejiang Chinese Medicine and Western Medicine Integrated Hospital, Hangzhou, Zhejiang, China
| | - Chao Zhang
- Hangzhou Lin'an district People's Hospital, Hangzhou, Zhejiang, China
| | - Congyao Wang
- The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Binhui Wang
- Municipal Hospital Affiliated to Medical School of Taizhou University, Taizhou, Zhejiang, China
| |
Collapse
|
23
|
Bekhbat M, Mukhara D, Dozmorov MG, Stansfield JC, Benusa SD, Hyer MM, Rowson SA, Kelly SD, Qin Z, Dupree JL, Tharp GK, Tansey MG, Neigh GN. Adolescent stress sensitizes the adult neuroimmune transcriptome and leads to sex-specific microglial and behavioral phenotypes. Neuropsychopharmacology 2021; 46:949-958. [PMID: 33558677 PMCID: PMC8115118 DOI: 10.1038/s41386-021-00970-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 01/30/2023]
Abstract
Adolescent exposure to chronic stress, a risk factor for mood disorders in adulthood, sensitizes the neuroinflammatory response to a subsequent immune challenge. We previously showed that chronic adolescent stress (CAS) in rats led to distinct patterns of neuroimmune priming in adult male and female rats. However, sex differences in the neuroimmune consequences of CAS and their underlying mechanisms are not fully understood. Here we hypothesized that biological sex would dictate differential induction of inflammation-related transcriptomic pathways and immune cell involvement (microglia activation and leukocyte presence) in the hippocampus of male and female rats with a history of CAS. Adolescent rats underwent CAS (six restraint and six social defeat episodes during postnatal days 38-49), and behavioral assessments were conducted in adolescence and adulthood. Neuroimmune measures were obtained following vehicle or a systemic lipopolysaccharide (LPS) challenge in adulthood. CAS led to increased time in the corners of the open field in adolescence. In males, CAS also increased social avoidance. As adults, CAS rats displayed an exaggerated enrichment of the nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) pathway and chemokine induction following LPS challenge, and increased number of perivascular CD45+ cells in the hippocampus. However, CAS females, but not males, showed exaggerated glucocorticoid receptor (GR) pathway enrichment and increased microglial complexity. These results provide further insight to the mechanisms by which peripheral immune events may influence neuroimmune responses differentially among males and females and further demonstrate the importance of adolescent stress in shaping adult responses.
Collapse
Affiliation(s)
- Mandakh Bekhbat
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA
| | - Deepika Mukhara
- grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Mikhail G. Dozmorov
- grid.417264.20000 0001 2194 2791Center for Clinical and Translational Research, Virginia Commonwealth University Medical Center, Richmond, VA 23298 USA
| | - John C. Stansfield
- grid.417264.20000 0001 2194 2791Center for Clinical and Translational Research, Virginia Commonwealth University Medical Center, Richmond, VA 23298 USA
| | - Savannah D. Benusa
- grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Molly M. Hyer
- grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Sydney A. Rowson
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA
| | - Sean D. Kelly
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA
| | - Zhaohui Qin
- grid.189967.80000 0001 0941 6502Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA USA
| | - Jeffrey L. Dupree
- grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Gregory K. Tharp
- grid.189967.80000 0001 0941 6502Division of Developmental & Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA 30322 USA
| | - Malú G. Tansey
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA
| | - Gretchen N. Neigh
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA ,grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA ,grid.189967.80000 0001 0941 6502Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA 30322 USA
| |
Collapse
|
24
|
Cutibacterium acnes Central Nervous System Catheter Infection Induces Long-Term Changes in the Cerebrospinal Fluid Proteome. Infect Immun 2021; 89:IAI.00531-20. [PMID: 33288649 PMCID: PMC8090941 DOI: 10.1128/iai.00531-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/24/2020] [Indexed: 11/20/2022] Open
Abstract
Cutibacterium acnes is the third most common cause of cerebrospinal fluid (CSF) shunt infection and is likely underdiagnosed due to the difficulty in culturing this pathogen. Shunt infections lead to grave neurologic morbidity for patients especially when there is a delay in diagnosis. Currently, the gold standard for identifying CSF shunt infections is microbiologic culture. However, C. acnes infection often results in falsely negative cultures; therefore, new diagnostic methods are needed. To investigate potential CSF biomarkers of C. acnes CSF shunt infection we adapted a rat model of CSF catheter infection to C. acnes. We found elevated levels of interleukin-1β (IL-1β), IL-6, chemokine ligand 2, and IL-10 in the CSF and brain tissues of animals implanted with C. acnes-infected catheters compared to sterile controls at day 1 postinfection. This coincided with modest increases in neutrophils in the CSF and, to a greater extent, in the brain tissues of animals with C. acnes infection, which closely mirrors the clinical findings in patients with C. acnes shunt infection. Mass spectrometry revealed that the CSF proteome is altered during C. acnes shunt infection and changes over the course of disease, typified at day 1 postinfection by an acute-phase and pathogen neutralization response evolving to a response consistent with wound resolution at day 28 compared to a sterile catheter placement. Collectively, these results demonstrate that it is possible to distinguish C. acnes infection from sterile postoperative inflammation and that CSF proteins could be useful in a diagnostic strategy for this pathogen that is difficult to diagnose.
Collapse
|
25
|
Deng Z, Ou H, Ren F, Guan Y, Huan Y, Cai H, Sun B. LncRNA SNHG14 promotes OGD/R-induced neuron injury by inducing excessive mitophagy via miR-182-5p/BINP3 axis in HT22 mouse hippocampal neuronal cells. Biol Res 2020; 53:38. [PMID: 32912324 PMCID: PMC7488096 DOI: 10.1186/s40659-020-00304-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Long non-coding RNA (lncRNA) small nucleolar RNA host gene 14 (SNHG14) is associated with cerebral ischemia-reperfusion (CI/R) injury. This work aims to explore the role of SNHG14 in CI/R injury. METHODS HT22 (mouse hippocampal neuronal cells) cell model was established by oxygen-glucose deprivation/reoxygenation (OGD/R) treatment. The interaction among SNHG14, miR-182-5p and BNIP3 was verified by luciferase reporter assay. Flow cytometry, western blot and quantitative real-time PCR were performed to examine apoptosis, the expression of genes and proteins. RESULTS SNHG14 and BNIP3 were highly expressed, and miR-182-5p was down-regulated in the OGD/R-induced HT22 cells. OGD/R-induced HT22 cells exhibited an increase in apoptosis. SNHG14 overexpression promoted apoptosis and the expression of cleaved-caspase-3 and cleaved-caspase-9 in the OGD/R-induced HT22 cells. Moreover, SNHG14 up-regulation enhanced the expression of BNIP3, Beclin-1, and LC3II/LC3I in the OGD/R-induced HT22 cells. Furthermore, SNHG14 regulated BNIP3 expression by sponging miR-182-5p. MiR-182-5p overexpression or BNIP3 knockdown repressed apoptosis in OGD/R-induced HT22 cells, which was abolished by SNHG14 up-regulation. CONCLUSION Our study demonstrates that lncRNA SNHG14 promotes OGD/R-induced neuron injury by inducing excessive mitophagy via miR-182-5p/BINP3 axis in HT22 mouse hippocampal neuronal cells. Thus, SNHG14/miR-182-5p/BINP3 axis may be a valuable target for CI/R injury therapies.
Collapse
Affiliation(s)
- Zexiang Deng
- Department of Anesthesiology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Hao Ou
- Department of Emergency and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Fei Ren
- Department of Anesthesiology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yujiao Guan
- Department of Anesthesiology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Ye Huan
- Department of Anesthesiology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Hongwei Cai
- Department of Anesthesiology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China.
| | - Bei Sun
- Department of Anesthesiology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
26
|
Hsia CH, Jayakumar T, Sheu JR, Hsia CW, Huang WC, Velusamy M, Lien LM. Synthetic Ruthenium Complex TQ-6 Potently Recovers Cerebral Ischemic Stroke: Attenuation of Microglia and Platelet Activation. J Clin Med 2020; 9:jcm9040996. [PMID: 32252398 PMCID: PMC7230480 DOI: 10.3390/jcm9040996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022] Open
Abstract
Activated microglia are crucial in the regulation of neuronal homeostasis and neuroinflammation. They also contribute to neuropathological processes after ischemic stroke. Thus, finding new approaches for reducing neuroinflammation has gained considerable attention. The metal ruthenium has gained notable attention because of its ability to form new complexes that can be used in disease treatment. [Ru(η6-cymene)2-(1H-benzoimidazol-2-yl)-quinoline Cl]BF4 (TQ-6), a potent ruthenium (II)-derived compound, was used in this study to investigate its neuroprotective action against microglia activation, middle cerebral artery occlusion (MCAO)-induced embolic stroke, and platelet activation, respectively. TQ-6 (2 μM) potently diminished inflammatory mediators (nitric oxide/inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2)) expression, nuclear factor kappa B (NF-κB) p65 phosphorylation, nuclear translocation, and hydroxyl radical (OH•) formation in LPS-stimulated microglia. Conversely, TQ-6 increased the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). Moreover, it significantly reduced brain infarct volume and edema in MCAO mice. Additionally, it drastically inhibited platelet aggregation and OH• production in mice platelets. This study confirmed that TQ-6 exerts an anti-neuroinflammatory effect on microglia activation through neuroprotection, antiplatelet activation, and free radical scavenging. The authors propose that TQ-6 might mitigate neurodegenerative pathology by inhibiting the NF-κB-mediated downstream pathway (iNOS and COX-2) and enhancing Nrf2/HO-1 signaling molecules in microglia.
Collapse
Affiliation(s)
- Chih-Hsuan Hsia
- Translational Medicine Center, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan;
- Graduate Institute of Medical Sciences and Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.J.); (J.-R.S.); (C.-W.H.); (W.-C.H.)
| | - Thanasekaran Jayakumar
- Graduate Institute of Medical Sciences and Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.J.); (J.-R.S.); (C.-W.H.); (W.-C.H.)
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences and Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.J.); (J.-R.S.); (C.-W.H.); (W.-C.H.)
| | - Chih-Wei Hsia
- Graduate Institute of Medical Sciences and Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.J.); (J.-R.S.); (C.-W.H.); (W.-C.H.)
| | - Wei-Chieh Huang
- Graduate Institute of Medical Sciences and Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.J.); (J.-R.S.); (C.-W.H.); (W.-C.H.)
| | - Marappan Velusamy
- Department of Chemistry, North Eastern Hill University, Shillong 793022, India;
| | - Li-Ming Lien
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan
- Correspondence: ; Tel.: +886-2-283-322-11 (ext. 2071)
| |
Collapse
|
27
|
Zera KA, Buckwalter MS. The Local and Peripheral Immune Responses to Stroke: Implications for Therapeutic Development. Neurotherapeutics 2020; 17:414-435. [PMID: 32193840 PMCID: PMC7283378 DOI: 10.1007/s13311-020-00844-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The immune response to stroke is an exciting target for future stroke therapies. Stroke is a leading cause of morbidity and mortality worldwide, and clot removal (mechanical or pharmacological) to achieve tissue reperfusion is the only therapy currently approved for patient use. Due to a short therapeutic window and incomplete effectiveness, however, many patients are left with infarcted tissue that stimulates inflammation. Although this is critical to promote repair, it can also damage surrounding healthy brain tissue. In addition, acute immunodepression and subsequent infections are common and are associated with worse patient outcomes. Thus, the acute immune response is a major focus of researchers attempting to identify ways to amplify its benefits and suppress its negative effects to improve short-term recovery of patients. Here we review what is known about this powerful process. This includes the role of brain resident cells such as microglia, peripherally activated cells such as macrophages and neutrophils, and activated endothelium. The role of systemic immune activation and subsequent immunodepression in the days after stroke is also discussed, as is the chronic immune responses and its effects on cognitive function. The biphasic role of inflammation, as well as complex timelines of cell production, differentiation, and trafficking, suggests that the relationship between the acute and chronic phases of stroke recovery is complex. Gaining a more complete understanding of this intricate process by which inflammation is initiated, propagated, and terminated may potentially lead to therapeutics that can treat a larger population of stroke patients than what is currently available. The immune response plays a critical role in patient recovery in both the acute and chronic phases after stroke. In patients, the immune response can be beneficial by promoting repair and recovery, and also detrimental by propagating a pro-inflammatory microenvironment. Thus, it is critical to understand the mechanisms of immune activation following stroke in order to successfully design therapeutics.
Collapse
Affiliation(s)
- Kristy A Zera
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Department of Neurosurgery, Stanford Univeristy School of Medicine, Stanford, CA, USA.
| |
Collapse
|
28
|
El-Missiry MA, Othman AI, Amer MA, Sedki M, Ali SM, El-Sherbiny IM. Nanoformulated ellagic acid ameliorates pentylenetetrazol-induced experimental epileptic seizures by modulating oxidative stress, inflammatory cytokines and apoptosis in the brains of male mice. Metab Brain Dis 2020; 35:385-399. [PMID: 31728888 DOI: 10.1007/s11011-019-00502-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 09/30/2019] [Indexed: 12/11/2022]
Abstract
The present study evaluated the neuroprotective and antiepileptic efficacy of ellagic acid (EA) encapsulated in calcium-alginate nanoparticles (Ca2+-ALG NPs) in pentylenetetrazol (PTZ)-induced seizures in male mice. EA was encapsulated in ALG NPs using a nanospray drying method followed by ionotropic crosslinking with Ca2+. Characterization of the developed Ca2+-crosslinked EA-ALG NPs showed spherical, high stability NPs; successful loading of EA within crosslinked ALG NPs; and sustained release of EA. Male Swiss albino mice were divided into ten groups as follows; Group I- (control), Group II (50 mg EA /kg) - (EA), Group III polyethylene glycol (PEG), Group IV EA NPs (50 mg/kg) - (EA NP), Group (50 mg/kg alginate) V void V NPs - (void NPs), Group VI: (37.5 PTZ mg/kg) -(PTZ), Group VII: PTZ and EA - (PTZ-EA). Group VIII: animals received PTZ and PEG concurrently (PTZ-PEG). Group IX; animals received PTZ and void NPs concurrently - (PTZ-void). Group X: animals received PTZ and EA NPs concurrently (PTZ-EA NPs). PTZ was used to induce experimental epilepsy. Ca2+-ALG NPs prevented seizures throughout the experimental period and had a more prominent effect than free EA did. Ca2+-ALG NPs prevented increased glutamate, decreased GABA concentrations and ameliorated increased amyloid-β and homocysteine levels in the serum and brain. Ca2+-EA-ALG NPs were superior to free EA in improving increased IL-6 and TNF-α. Ca2+-ALG NPs ameliorated PTZ-induced oxidative stress, as evidenced by decreased 4HNE levels and enhanced GSH, GR and GPx levels in the brain. These changes were accompanied by amelioration of apoptosis and its regulating proteins, including Cytochrome C, P53, Bax, Bcl2 and caspase-3 and caspase-9, and protected against DNA damage. Histological examination of the hippocampus confirmed that the neuroprotective effect of Ca2+-EA-ALG NPs was superior and more effective than that of free EA.
Collapse
Affiliation(s)
| | - Azza I Othman
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Maher A Amer
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Mohammed Sedki
- Nanomaterials Laboratory, Center for Materials Science, Zewail City of Science and Technology, 6th of October City, Giza, 12578, Egypt
| | - Sara M Ali
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Ibrahim M El-Sherbiny
- Nanomaterials Laboratory, Center for Materials Science, Zewail City of Science and Technology, 6th of October City, Giza, 12578, Egypt.
| |
Collapse
|
29
|
Calvo B, Rubio F, Fernández M, Tranque P. Dissociation of neonatal and adult mice brain for simultaneous analysis of microglia, astrocytes and infiltrating lymphocytes by flow cytometry. IBRO Rep 2020; 8:36-47. [PMID: 32215337 PMCID: PMC7090101 DOI: 10.1016/j.ibror.2019.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 12/31/2019] [Indexed: 12/29/2022] Open
Abstract
Recovery of neural cells is higher with 30 % Percoll gradient than 30–70 %. Papain enhances combined extraction of microglia, astrocytes and lymphocytes. Dispase II potentiates papain action only in adult brain. Mechanical dissociation isolates neonatal and adult astrocytes better than enzymes. Papain + dispase II alows cell cytometry quantification of glial activation by LPS.
The technical difficulty to isolate microglia, astrocytes and infiltrating immune cells from mouse brain is nowadays a limiting factor in the study of neuroinflammation. Brain isolation requirements are cell-type and animal-age dependent, but current brain dissociation procedures are poorly standardized. This lack of comprehensive studies hampers the selection of optimized methodologies. Thus, we present here a comparative analysis of dissociation methods and Percoll-based separation to identify the most efficient procedure for the combined isolation of healthy microglia, astrocytes and infiltrated leukocytes; distinguishing neonatal and adult mouse brain. Gentle mechanical dissociation and DNase I incubation was supplemented with papain or collagenase II. Dispase II digestion was also used alone or in combination. In addition, cell separation efficiency of 30 % and 30–70 % Percoll gradients was compared. In these experiments, cell yield and integrity of freshly dissociated cells was measured by flow cytometry. We found that papain digestion in combination with dispase II followed by 30 % Percoll separation is the most balanced method to obtain a mixture of microglia, astrocytes and infiltrated immune cells; while addition of dispase II was not an advantage for neonatal brain. These dissociation conditions allowed flow cytometry detection of a slight glial activation triggered by sublethal LPS injection. In conclusion, the enzymes and Percoll density gradients tested here affected differently resting microglia, activated microglia/macrophages, astrocytes and infiltrated lymphocytes. Also, newborn and adult brain showed contrasting reactions to digestion. Our study highlights the strength of flow cytometry for the simultaneous analysis of neuroimmune cell populations once extraction is optimized.
Collapse
Key Words
- ANOVA, one-way analysis of variance
- Astrocytes
- CNS, Central Nervous System
- CaCl2, calcium chloride
- EBSS, Earle's Balanced Salt Solution
- EDTA, ethylenediaminetetraacetic acid
- FACS, Fluorescence-activated cell sorter
- FSC, forward-scattered light
- Flow cytometry
- Glia reactivity
- HBSS, Hank's Balanced Salt Solution
- LD, lethal dose
- LPS, lipopolysaccharide
- Lymphocytes
- MgCl2, magnesium chloride
- MgSO4, magnesium sulfate
- Microglia
- Neuroimmunity
- PBS, phosphate-buffered saline
- RT, room temperature
- SIP, stock solution of isotonic Percoll
- SSC, side-scattered light
- i.p, intraperitoneal injection
Collapse
Affiliation(s)
- Belén Calvo
- Neuroglia Laboratory, Research Institute for Neurological Disorders (IDINE), Medical School, University of Castilla-La Mancha (UCLM), Albacete, Spain
| | - Felipe Rubio
- Neuroglia Laboratory, Research Institute for Neurological Disorders (IDINE), Medical School, University of Castilla-La Mancha (UCLM), Albacete, Spain
| | - Miriam Fernández
- Neuroglia Laboratory, Research Institute for Neurological Disorders (IDINE), Medical School, University of Castilla-La Mancha (UCLM), Albacete, Spain
| | - Pedro Tranque
- Neuroglia Laboratory, Research Institute for Neurological Disorders (IDINE), Medical School, University of Castilla-La Mancha (UCLM), Albacete, Spain
| |
Collapse
|
30
|
Costa IM, Lima FOV, Fernandes LCB, Norrara B, Neta FI, Alves RD, Cavalcanti JRLP, Lucena EES, Cavalcante JS, Rego ACM, Filho IA, Queiroz DB, Freire MAM, Guzen FP. Astragaloside IV Supplementation Promotes A Neuroprotective Effect in Experimental Models of Neurological Disorders: A Systematic Review. Curr Neuropharmacol 2020; 17:648-665. [PMID: 30207235 PMCID: PMC6712289 DOI: 10.2174/1570159x16666180911123341] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 08/10/2018] [Accepted: 08/28/2018] [Indexed: 01/22/2023] Open
Abstract
Background: Neurological disorders constitute a growing worldwide concern due to the progressive aging of the population and the risky behavior they represent. Herbal medicines have scientific relevance in the treatment of these pathol-ogies. One of these substances, Astragaloside IV (AS-IV), is the main active compound present in the root of Astragalus membranaceus (Fisch.) Bge, a Chinese medicinal herb with neuroprotective properties. Objective: In the present study we performed a systematic review that sought to comprehend the neuroprotective effect pre-sented by AS-IV in experimental models of neurological disorders. Method: This study is a systematic review, where an electronic search in United States National Library of Medicine (Pub-Med), Science Direct, Cochrane Library, Scientific Electronic Library Online (SciELO), Scopus, Web of Science, Medline via Proquest and Periodicos Capes databases covering the years between 2007 and 2017, using “Astragaloside IV” and “Neurodegenerative diseases”; “Astragaloside IV” and “ Neurological disorders” as reference terms was made. Results: A total of 16 articles were identified, in which the efficacy of AS-IV was described in experimental models of Par-kinson’s disease, Alzheimer’s disease, cerebral ischemia and autoimmune encephalomyelitis, by improving motor deficits and/or neurochemical activity, especially antioxidant systems, reducing inflammation and oxidative stress. Conclusion: The findings of the present study indicate that the administration of AS-IV can improve behavioral and neuro-chemical deficits largely due to its antioxidant, antiapoptotic and anti-inflammatory properties, emerging as an alternative therapeutic approach for the treatment of neurological disorders.
Collapse
Affiliation(s)
- Ianara M Costa
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte (UERN), Mossoro/RN, Brazil
| | - Francisca O V Lima
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte (UERN), Mossoro/RN, Brazil
| | - Luciana C B Fernandes
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte (UERN), Mossoro/RN, Brazil
| | - Bianca Norrara
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte (UERN), Mossoro/RN, Brazil
| | - Francisca I Neta
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte (UERN), Mossoro/RN, Brazil
| | - Rodrigo D Alves
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte (UERN), Mossoro/RN, Brazil
| | - José R L P Cavalcanti
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte (UERN), Mossoro/RN, Brazil
| | - Eudes E S Lucena
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte (UERN), Mossoro/RN, Brazil
| | - Jeferson S Cavalcante
- Laboratory of Neurochemical Studies, Center of Biological Sciences, Federal University of Rio Grande do Norte (UFRN), Natal/RN, Brazil
| | - Amalia C M Rego
- Post Graduation Program in Biotechnology, Health School, Potiguar University (UnP), Natal/RN, Brazil
| | - Irami A Filho
- Post Graduation Program in Biotechnology, Health School, Potiguar University (UnP), Natal/RN, Brazil
| | - Dinalva B Queiroz
- Post Graduation Program in Biotechnology, Health School, Potiguar University (UnP), Natal/RN, Brazil
| | - Marco A M Freire
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte (UERN), Mossoro/RN, Brazil
| | - Fausto P Guzen
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Faculty of Health Sciences, University of the State of Rio Grande do Norte (UERN), Mossoro/RN, Brazil.,Post Graduation Program in Biotechnology, Health School, Potiguar University (UnP), Natal/RN, Brazil
| |
Collapse
|
31
|
Yang C, Hou X, Feng Q, Li Y, Wang X, Qin L, Yang P. Lupus serum IgG induces microglia activation through Fc fragment dependent way and modulated by B-cell activating factor. J Transl Med 2019; 17:426. [PMID: 31864410 PMCID: PMC6925475 DOI: 10.1186/s12967-019-02175-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 12/10/2019] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Neuropsychiatric manifestations are frequent in patients with systemic lupus erythematosus (SLE), yet the etiology and pathogenesis of brain damage in SLE remains unclear. Because the production of autoantibodies, formation and deposition of immunocomplexes are major serological characteristics of SLE, the elevated level of serum immunoglobulin may contribute to brain tissue injury of SLE. To testify this, in this study, we examined whether immunoglobulin G (IgG) in the serum of SLE patients affects the cellular functions in central nervous system and the potential mechanism. METHODS In vivo intracerebral injection of SLE-serum in mouse was used to activate microglia and the production of pro-inflammatory cytokine was assessed by ELISA. Sera was divided into IgG and IgG depleted fractions, while IgG was further divided into Fc and Fab fragments to examine which part has an effect on microglia. Flow cytometry, immunofluorescence and quantitative PCR (qPCR) were used to verify the synergistic effect of B-cell activating factor (BAFF) on IgG stimulation of microglia. RESULTS We found that IgG in lupus sera can induce M1 activation of brain microglia following intraventricular injection into normal mice, and BAFF facilitates this process. In vitro, we identified that IgG bound to microglia through Fc rather than Fab fragments, and BAFF up-regulated the expression of Fc receptors (FcγR) on the surface of microglia, consequently, promote IgG binding to microglia. CONCLUSION Our results suggest that lupus serum IgG causes inflammatory responses of microglia by involving the Fc signaling pathway and the activity could be up-regulated by BAFF. Accordingly, disruption of the FcγR-mediated signaling pathway and blockade of microglia activation may be a therapeutic target in patients with neuropsychiatric lupus erythematosus.
Collapse
Affiliation(s)
- Chunshu Yang
- Department of 1st Cancer Institute, First Affiliated Hospital, China Medical University, Shenyang, 110001 People’s Republic of China
| | - Xiaoyu Hou
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, 110001 People’s Republic of China
| | - Qianhui Feng
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, 110001 People’s Republic of China
| | - Yingzhuo Li
- Department of Physiology, School of Life Science, China Medical University, Shenyang, 110122 People’s Republic of China
| | - Xuejiao Wang
- Department of Physiology, School of Life Science, China Medical University, Shenyang, 110122 People’s Republic of China
| | - Ling Qin
- Department of Physiology, School of Life Science, China Medical University, Shenyang, 110122 People’s Republic of China
| | - Pingting Yang
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, 110001 People’s Republic of China
| |
Collapse
|
32
|
Seifert HA, Gerstner G, Kent G, Vandenbark AA, Offner H. Estrogen-induced compensatory mechanisms protect IL-10-deficient mice from developing EAE. J Neuroinflammation 2019; 16:195. [PMID: 31665042 PMCID: PMC6821034 DOI: 10.1186/s12974-019-1588-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/12/2019] [Indexed: 02/08/2023] Open
Abstract
Background IL-10 knockout (KO) mice are protected from experimental autoimmune encephalomyelitis (EAE) with low-dose estrogen (E2) treatment similar to wild-type (WT) mice. Previous studies have demonstrated a decrease in tumor necrosis factor in all E2-treated groups, which led to the protection of the mice. Methods This study used IL-10 KO mice and WT mice treated either with E2 or sham pellets 7 days prior to induction of EAE. Mice were observed for 21 days post-immunization. The spleen, inguinal lymph nodes, and brain were evaluated by flow cytometry. Spinal cords were evaluated using a cytokine/chemokine array, RT-PCR, and histology. Results This study demonstrates that E2 treatment induced three heightened regulatory mechanisms that potentially protect IL-10 KO mice from EAE: (1) an increase in programmed death-ligands 1 and 2 on monocytes and macrophages in the periphery and within the CNS; (2) an increase in CD73 in the inflamed CNS, which can increase the production of the anti-inflammatory molecule adenosine; and (3) a decrease in CD4+CD25+FoxP3+ regulatory T cells in the spleen. Together, these factors comprise an alternative compensatory mechanism that significantly downregulates key pro-inflammatory cytokine, chemokine, and chemokine receptor genes which are enhanced in the spinal cord of IL-10 KO mice. This group of E2-treated mice remained asymptomatic after EAE challenge similar to E2-treated WT mice, despite their having more T and B lymphocytes in the brain, and modestly increased demyelination in the spinal cord. Conclusion These results indicate that previously unrecognized compensatory mechanisms of EAE protection are stimulated by E2 in the absence of IL-10, which can provide disease protection comparable to the IL-10-dependent effects induced by E2 in WT mice.
Collapse
Affiliation(s)
- Hilary A Seifert
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Neuroimmunology Research, VA Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., Portland, OR, 97239, USA
| | - Grant Gerstner
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Neuroimmunology Research, VA Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., Portland, OR, 97239, USA
| | - Gail Kent
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Neuroimmunology Research, VA Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., Portland, OR, 97239, USA
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.,Neuroimmunology Research, VA Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., Portland, OR, 97239, USA.,Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Halina Offner
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA. .,Neuroimmunology Research, VA Portland Health Care System, R&D-31, 3710 SW US Veterans Hospital Rd., Portland, OR, 97239, USA. .,Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
33
|
Identification of Potential Cerebrospinal Fluid Biomarkers To Discriminate between Infection and Sterile Inflammation in a Rat Model of Staphylococcus epidermidis Catheter Infection. Infect Immun 2019; 87:IAI.00311-19. [PMID: 31262978 PMCID: PMC6704599 DOI: 10.1128/iai.00311-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 06/18/2019] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus epidermidis cerebrospinal fluid (CSF) shunt infection is a common complication of hydrocephalus treatment, creating grave neurological consequences for patients, especially when diagnosis is delayed. The current method of diagnosis relies on microbiological culture; however, awaiting culture results may cause treatment delays, or culture may fail to identify infection altogether, so newer methods are needed. Staphylococcus epidermidis cerebrospinal fluid (CSF) shunt infection is a common complication of hydrocephalus treatment, creating grave neurological consequences for patients, especially when diagnosis is delayed. The current method of diagnosis relies on microbiological culture; however, awaiting culture results may cause treatment delays, or culture may fail to identify infection altogether, so newer methods are needed. To investigate potential CSF biomarkers of S. epidermidis shunt infection, we developed a rat model allowing for serial CSF sampling. We found elevated levels of interleukin-10 (IL-10), IL-1β, chemokine ligand 2 (CCL2), and CCL3 in the CSF of animals implanted with S. epidermidis-infected catheters compared to sterile controls at day 1 postinfection. Along with increased chemokine and cytokine expression early in infection, neutrophil influx was significantly increased in the CSF of animals with infected catheters, suggesting that coupling leukocyte counts with inflammatory mediators may differentiate infection from sterile inflammation. Mass spectrometry analysis revealed that the CSF proteome in sterile animals was similar to that in infected animals at day 1; however, by day 5 postinfection, there was an increase in the number of differently expressed proteins in the CSF of infected compared to sterile groups. The expansion of the proteome at day 5 postinfection was interesting, as bacterial burdens began to decline by this point, yet the CSF proteome data indicated that the host response remained active, especially with regard to the complement cascade. Collectively, these results provide potential biomarkers to distinguish S. epidermidis infection from sterile postoperative inflammation.
Collapse
|
34
|
Contribution of synovial macrophages to rat advanced osteoarthritis pain resistant to cyclooxygenase inhibitors. Pain 2019; 160:895-907. [PMID: 30585984 DOI: 10.1097/j.pain.0000000000001466] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Most advanced knee osteoarthritis (OA) patients experience chronic pain resistant to cyclooxygenase (COX) inhibitors. However, the cells and molecules involved in this advanced OA pain remain poorly understood. In this study, we developed a rat model of advanced knee OA by modification of the monoiodoacetate-induced OA pain model and examined involvement of synovial macrophages in advanced OA pain. Cyclooxygenase inhibitors, such as celecoxib and naproxen, and a steroid were ineffective, but an opioid and anti-nerve growth factor (NGF) antibody was effective for pain management in the advanced OA model. Similar to advanced OA patients, histological analysis indicated severe bone marrow damages, synovitis, and cartilage damage and an increase of macrophages with high expression of interleukin-1β, NGF, nitric oxide synthase (NOS) 1, NOS2, and COX-2 in the knee joint of the advanced OA model. Intravenous injection of clodronate liposomes depleted synovial macrophages, which decreased the level of not only proinflammatory mediator interleukin-1β but also NGF in the knee joint, leading to pain suppression in the advanced OA model. These data suggest the involvement of synovial macrophages in advanced knee OA pain resistant to COX inhibitors by increasing proinflammatory mediators, and that drugs targeting synovial macrophages might have potent analgesic effects.
Collapse
|
35
|
Edwards DN, Bix GJ. The Inflammatory Response After Ischemic Stroke: Targeting β 2 and β 1 Integrins. Front Neurosci 2019; 13:540. [PMID: 31191232 PMCID: PMC6546847 DOI: 10.3389/fnins.2019.00540] [Citation(s) in RCA: 24] [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: 03/07/2019] [Accepted: 05/09/2019] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke is a leading cause of death and disability with limited therapeutic options. Resulting inflammatory mechanisms after reperfusion (removal of the thrombus) result in cytokine activation, calcium influx, and leukocytic infiltration to the area of ischemia. In particular, leukocytes migrate toward areas of inflammation by use of integrins, particularly integrins β1 and β2. Integrins have been shown to be necessary for leukocyte adhesion and migration, and thus are of immediate interest in many inflammatory diseases, including ischemic stroke. In this review, we identify the main integrins involved in leukocytic migration following stroke (α L β2, αDβ2, α4β1, and α5β1) and targeted clinical therapeutic interventions.
Collapse
Affiliation(s)
- Danielle N. Edwards
- Sanders–Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Gregory J. Bix
- Department of Neurology, University of Kentucky, Lexington, KY, United States
- Department of Neurosurgery, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
36
|
Tuttolomondo A, Di Raimondo D, Pecoraro R, Casuccio A, Di Bona D, Aiello A, Accardi G, Arnao V, Clemente G, Corte VD, Maida C, Simonetta I, Caruso C, Squatrito R, Pinto A. HLA and killer cell immunoglobulin-like receptor (KIRs) genotyping in patients with acute ischemic stroke. J Neuroinflammation 2019; 16:88. [PMID: 30995924 PMCID: PMC6471781 DOI: 10.1186/s12974-019-1469-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 03/27/2019] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION In humans, a major component of natural killer (NK) and T cell target recognition depends on the surveillance of human leukocyte antigen (HLA) class I molecules by killer immunoglobulin-like receptors (KIRs). AIMS To implement the knowledge about the immunological genetic background of acute ischemic stroke susceptibility in relation to the frequency of the KIR genes and HLA alleles. METHODS Subjects with acute ischemic stroke and subjects without stroke were genotyped for the presence of KIR genes and of the three major KIR ligand groups, HLA-C1, HLA-C2, and HLA-Bw4, both HLA-B and HLA-A loci. RESULTS Between November 2013 and February 2016, consecutive patients with acute ischemic stroke were recruited. As healthy controls, we enrolled subjects without acute ischemic stroke. Subjects with acute ischemic stroke in comparison with controls showed a higher frequency of 2DL3, 2DL5B, 2DS2, and 2DS4 KIR genes and a lower frequency of HLA-B-Bw4I alleles. Subjects without acute ischemic stroke showed a higher frequency of interaction between KIR 2DS2 and HLAC2. We also observed a higher frequency of 2DL3 and 2 DL4 KIR genes in subjects with atherosclerotic (LAAS) subtype. Multiple logistic regression analysis showed a protective effect towards stroke of HLA-B-Bw4I and interaction between KIR 2DL2 and HLAC1 and 2DS2-HLAC2 and a detrimental effect of 2DL2-HLA-C1_A interactions. CONCLUSION Our findings of a higher frequency of activating KIR genes seem to be consistent with findings previously reported patients with coronary syndrome. This higher frequency of "proinflammatory" genes in subjects with ischemic stroke could also explain the immunoinflammatory activation of the acute phase of stroke.
Collapse
Affiliation(s)
- Antonino Tuttolomondo
- Internal Medicine and Stroke Care Ward, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, P.zza delle Cliniche n.2, 90127 Palermo, Italy
| | - Domenico Di Raimondo
- Internal Medicine and Stroke Care Ward, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, P.zza delle Cliniche n.2, 90127 Palermo, Italy
| | - Rosaria Pecoraro
- Pronto Soccorso Unit, Giuseppe Giglio Hospital, Cefalù, Italy
- PhD Programme in Clinical Medicine and Behavioural Sciences, University of Palermo, Palermo, PA 90133 Italy
| | - Alessandra Casuccio
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, P.zza delle Cliniche n.2, 90127 Palermo, Italy
| | - Danilo Di Bona
- School and Chair of Allergology, Dipartimento delle Emergenze e Trapianti d’Organo, University of Bari, Bari, Italy
| | - Anna Aiello
- Dipartimento di Biopatologia e Biotecnologie Mediche, Universita’ degli Studi di Palermo, Palermo, Italy
| | - Giulia Accardi
- Dipartimento di Biopatologia e Biotecnologie Mediche, Universita’ degli Studi di Palermo, Palermo, Italy
| | - Valentina Arnao
- Dipartimento di BioMedicina Sperimentale e Neuroscienze Cliniche, Università degli Studi di Palermo, Palermo, Italy
| | - Giuseppe Clemente
- Internal Medicine and Stroke Care Ward, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, P.zza delle Cliniche n.2, 90127 Palermo, Italy
| | - Vittoriano Della Corte
- PhD Programme in Molecular and Clinical Medicine, University of Palermo, Palermo, PA 90133 Italy
| | - Carlo Maida
- Internal Medicine and Stroke Care Ward, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, P.zza delle Cliniche n.2, 90127 Palermo, Italy
| | - Irene Simonetta
- Internal Medicine and Stroke Care Ward, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, P.zza delle Cliniche n.2, 90127 Palermo, Italy
| | - Calogero Caruso
- Dipartimento di Biopatologia e Biotecnologie Mediche, Universita’ degli Studi di Palermo, Palermo, Italy
| | | | - Antonio Pinto
- Internal Medicine and Stroke Care Ward, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, P.zza delle Cliniche n.2, 90127 Palermo, Italy
| |
Collapse
|
37
|
Vandenbark AA, Meza-Romero R, Benedek G, Offner H. A novel neurotherapeutic for multiple sclerosis, ischemic injury, methamphetamine addiction, and traumatic brain injury. J Neuroinflammation 2019; 16:14. [PMID: 30683115 PMCID: PMC6346590 DOI: 10.1186/s12974-018-1393-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 12/27/2018] [Indexed: 02/08/2023] Open
Abstract
Neurovascular, autoimmune, and traumatic injuries of the central nervous system (CNS) all have in common an initial acute inflammatory response mediated by influx across the blood-brain barrier of activated mononuclear cells followed by chronic and often progressive disability. Although some anti-inflammatory therapies can reduce cellular infiltration into the initial lesions, there are essentially no effective treatments for the progressive phase. We here review the successful treatment of animal models for four separate neuroinflammatory and neurodegenerative CNS conditions using a single partial MHC class II construct called DRa1-hMOG-35-55 or its newest iteration, DRa1(L50Q)-hMOG-35-55 (DRhQ) that can be administered without a need for class II tissue type matching due to the conserved DRα1 moiety of the drug. These constructs antagonize the cognate TCR and bind with high affinity to their cell-bound CD74 receptor on macrophages and dendritic cells, thereby competitively inhibiting downstream signaling and pro-inflammatory effects of macrophage migration inhibitory factor (MIF) and its homolog, d-dopachrome tautomerase (D-DT=MIF-2) that bind to identical residues of CD74 leading to progressive disease. These effects suggest the existence of a common pathogenic mechanism involving a chemokine-driven influx of activated monocytes into the CNS tissue that can be reversed by parenteral injection of the DRa1-MOG-35-55 constructs that also induce anti-inflammatory macrophages and microglia within the CNS. Due to their ability to block this common pathway, these novel drugs appear to be prime candidates for therapy of a wide range of neuroinflammatory and neurodegenerative CNS conditions.
Collapse
Affiliation(s)
- Arthur A Vandenbark
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA. .,Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA. .,Department of Molecular Microbiology & Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| | - Roberto Meza-Romero
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA.,Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Gil Benedek
- Present Address: Tissue Typing and Immunogenetics Laboratory, Hadassah Medical Center, Jerusalem, Israel
| | - Halina Offner
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA.,Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.,Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| |
Collapse
|
38
|
Arya AK, Hu B. Brain-gut axis after stroke. Brain Circ 2018; 4:165-173. [PMID: 30693343 PMCID: PMC6329216 DOI: 10.4103/bc.bc_32_18] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 11/25/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022] Open
Abstract
Stroke leads to inflammatory and immune response in the brain and immune organs. The gut or gastrointestinal tract is a major immune organ equipped with the largest pool of immune cells representing more than 70% of the entire immune system and the largest population of macrophages in the human body. The bidirectional communication between the brain and the gut is commonly known as brain–gut or gut–brain axis. Stroke often leads to gut dysmotility, gut microbiota dysbiosis, “leaky” gut, gut hemorrhage, and even gut-origin sepsis, which is often associated with poor prognosis. Emerging evidence suggests that gut inflammatory and immune response plays a key role in the pathophysiology of stroke and may become a key therapeutic target for its treatment. Ischemic brain tissue produces damage-associated molecular patterns to initiate innate and adaptive immune response both locally and systemically through the specialized pattern-recognition receptors (e.g., toll-like receptors). After stroke, innate immune cells including neutrophils, microglia or macrophages, mast cells, innate lymphocytes (IL-17 secreting γδ T-cell), and natural killer T-cell respond within hours, followed by the adaptive immune response through activation of T and B lymphocytes. Subpopulations of T-cells can help or worsen ischemic brain injury. Pro-inflammatory Th1, Th17, and γδ T-cells are often associated with increased inflammatory damage, whereas regulatory T-cells are known to suppress postischemic inflammation by increasing the secretion of anti-inflammatory cytokine IL-10. Although known to play a key role, research in the gut inflammatory and immune response after stroke is still in its initial stage. A better understanding of the gut inflammatory and immune response after stroke may be important for the development of effective stroke therapies. The present review will discuss recent advances in the studies of the brain–gut axis after stroke, the key issues to be solved, and the future directions.
Collapse
Affiliation(s)
- Awadhesh K Arya
- Department of Neurology and Anesthesiology, Shock Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bingren Hu
- Department of Neurology and Anesthesiology, Shock Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA.,Veterans Affairs Maryland Health Center System, Baltimore, MD, USA
| |
Collapse
|
39
|
García-Culebras A, Durán-Laforet V, Peña-Martínez C, Ballesteros I, Pradillo JM, Díaz-Guzmán J, Lizasoain I, Moro MA. Myeloid cells as therapeutic targets in neuroinflammation after stroke: Specific roles of neutrophils and neutrophil-platelet interactions. J Cereb Blood Flow Metab 2018; 38:2150-2164. [PMID: 30129391 PMCID: PMC6282223 DOI: 10.1177/0271678x18795789] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Ischemic brain injury causes a local inflammatory response, involving the activation of resident brain cells such as microglia and the recruitment of infiltrating immune cells. Increasing evidence supports that plasticity of the myeloid cell lineage is determinant for the specific role of these cells on stroke outcome, from initiation and maintenance to resolution of post-ischemic inflammation. The aim of this review is to summarize some of the key characteristics of these cells and the mechanisms for their recruitment into the injured brain through interactions with platelets, endothelial cells and other leukocytes. Also, we discuss the existence of different leukocyte subsets in the ischemic tissue and, specifically, the impact of different myeloid phenotypes on stroke outcome, with special emphasis on neutrophils and their interplay with platelets. Knowledge of these cellular phenotypes and interactions may pave the way to new therapies able to promote protective immune responses and tissue repair after cerebral ischemia.
Collapse
Affiliation(s)
- Alicia García-Culebras
- 1 Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.,2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,3 Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Madrid, Spain
| | - Violeta Durán-Laforet
- 1 Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.,2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,3 Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Madrid, Spain
| | - Carolina Peña-Martínez
- 1 Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.,2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,3 Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Madrid, Spain
| | - Iván Ballesteros
- 4 Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jesús M Pradillo
- 1 Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.,2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,3 Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Madrid, Spain
| | - Jaime Díaz-Guzmán
- 2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,5 Servicio de Neurología, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - Ignacio Lizasoain
- 1 Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.,2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,3 Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Madrid, Spain
| | - María A Moro
- 1 Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.,2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,3 Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Madrid, Spain
| |
Collapse
|
40
|
Yu H, Wang X, Kang F, Chen Z, Meng Y, Dai M. Propofol attenuates inflammatory damage on neurons following cerebral infarction by inhibiting excessive activation of microglia. Int J Mol Med 2018; 43:452-460. [PMID: 30431058 DOI: 10.3892/ijmm.2018.3974] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 02/02/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Hang Yu
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Xiaozhi Wang
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Fuxin Kang
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Zhile Chen
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Yunxia Meng
- Intensive Care Unit, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Mingming Dai
- Department of Internal Neurology, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570311, P.R. China
| |
Collapse
|
41
|
Brockmann C, Kociok N, Dege S, Davids AM, Brockmann T, Miller KR, Joussen AM. Local partial depletion of CD11b + cells and their influence on choroidal neovascularization using the CD11b-HSVTK mouse model. Acta Ophthalmol 2018. [PMID: 29536657 DOI: 10.1111/aos.13716] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE To assess the influence of retinal macrophages and microglia on the formation of choroidal neovascularization (CNV). Therefore, we used a transgenic mouse (CD11b-HSVTK) in which the application of ganciclovir (GCV) results in a depletion of CD11b+ cells. METHODS We first investigated if a local depletion of CD11b+ macrophages and microglia in the retina is feasible. In a second step, the influence of CD11b+ cell depletion on CNV formation was analysed. One eye of each CD11b-HSVTK mouse was injected with GCV, and the fellow eye received sodium chloride solution (NaCl). Cell counting was performed at day 3 and 7 (one injection) or at day 14 and 21 (two injections). Choroidal neovascularization (CNV) was induced by argon laser and analysed at day 14. RESULTS The most effective CD11b+ cell depletion was achieved 7 days after a single injection and 14 days after two injections of GCV. After two injections of GCV, we found a significant reduction of CD11b+ cells in central (52 ± 23.9 cells/mm2 ) and peripheral retina (53 ± 20.6 cells/mm2 ); compared to eyes received NaCl (216 ± 49.0 and 210 ± 50.5 cells/mm2 , p < 0.001, respectively). Regarding CNV areas, no statistical significance was found between the groups. CONCLUSION The CD11b-HSVTK mouse is a feasible model for a local depletion of CD11b+ cells in the retina. Nevertheless, only a partial depletion of CD11b+ cells could be achieved compared to baseline data without any intravitreal injections. Our results did not reveal a significant reduction in CNV areas. In the light of previous knowledge, the potential influence of systemic immune cells on CNV formation might be more relevant than expected.
Collapse
Affiliation(s)
- Claudia Brockmann
- Department of Ophthalmology; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health; Berlin Germany
- Berlin Institute of Health (BIH); Berlin Germany
| | - Norbert Kociok
- Department of Ophthalmology; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health; Berlin Germany
| | - Sabrina Dege
- Department of Ophthalmology; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health; Berlin Germany
| | - Anja-Maria Davids
- Department of Ophthalmology; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health; Berlin Germany
| | - Tobias Brockmann
- Department of Ophthalmology; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health; Berlin Germany
- Berlin Institute of Health (BIH); Berlin Germany
| | - Kelly R. Miller
- Department of Neuropathology; Charite - Universitätsmedizin Berlin; Corporate Member of Freie Universität Berlin; Humboldt-Universität Berlin, and Berlin Institute of Health; Berlin Germany
| | - Antonia M. Joussen
- Department of Ophthalmology; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health; Berlin Germany
| |
Collapse
|
42
|
El-Missiry MA, Othman AI, El-Sawy MR, Lebede MF. Neuroprotective effect of epigallocatechin-3-gallate (EGCG) on radiation-induced damage and apoptosis in the rat hippocampus. Int J Radiat Biol 2018; 94:798-808. [PMID: 29939076 DOI: 10.1080/09553002.2018.1492755] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
PURPOSE This study investigated the potential neuroprotective effect of epigallocatechin-3-gallate (EGCG) on radiation-induced cell death and damage in the hippocampus. MATERIALS AND METHODS Adult male Wister rats received oral treatment with EGCG at doses of 2.5 and 5 mg/kg/d for 3 d before 4 Gy γ irradiation. RESULTS The pretreatment of irradiated rats with EGCG significantly ameliorated the increased plasma levels of homocysteine, amyloid β, TNF-α and IL-6 levels and the decrease of dopamine and serotonin. Pretreatment with EGCG also significantly ameliorated the irradiation-induced increase in the 4-HNE and protein carbonyl levels and the decreased antioxidants including glutathione level, and the activities of glutathione peroxidase and glutathione reductase in the hippocampus. EGCG treatment prior to radiation exposure protected against DNA damage and apoptosis in the hippocampus. The increase in the levels of p53, Cytochrome-c, Bax and caspases 3 and 9 in the hippocampus were significantly ameliorated with a significant increase in Bcl-2. These changes were supported by marked protection of the dentate gyrus that exhibited a similar histological structure of the control animals. CONCLUSIONS EGCG can attenuate the severity of radiation-induced damage and cell death in hippocampus recommending polyphenols as successful option for protecting against radiation-induced hippocampal damage.
Collapse
Affiliation(s)
- Mohamed A El-Missiry
- a Zoology Department, Faculty of Science , Mansoura University , Mansoura , Egypt.,b Prince Sultan Military Collage of Health Sciences , Dhahran , KSA
| | - Azza I Othman
- a Zoology Department, Faculty of Science , Mansoura University , Mansoura , Egypt
| | - Mamdouh R El-Sawy
- a Zoology Department, Faculty of Science , Mansoura University , Mansoura , Egypt
| | - Mohamad F Lebede
- c Medical Laboratory Department, Faculty of Medical Technology , Tobruk University , Tobruk , Libya
| |
Collapse
|
43
|
El-Missiry MA, ElKomy MA, Othman AI, AbouEl-Ezz AM. Punicalagin ameliorates the elevation of plasma homocysteine, amyloid-β, TNF-α and apoptosis by advocating antioxidants and modulating apoptotic mediator proteins in brain. Biomed Pharmacother 2018; 102:472-480. [PMID: 29579708 DOI: 10.1016/j.biopha.2018.03.096] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 12/21/2022] Open
Abstract
The present study investigated the neuroprotective role of punicalagin, a major polyphenolic compound of pomegranate on methionine-induced brain injury. Hyperhomocysteinemia (HHcy) was induced in two months old male BALB c mice by methionine supplementation in drinking water (1 g/kg body weight) for 30 days. Punicalagin (1 mg/kg) was injected i.p every other day concurrently with methionine. Punicalagin significantly prevented the rise in the levels of homocysteine, amyloid-β and TNF-α. HHcy is associated with a decrease in the activities of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (PGx) and glutathione reductase (GR) and glutathione (GSH) levels in the brains of methionine-treated mice while these antioxidants are increased by punicalagin supplementation. The treatment with punicalagin significantly decreased oxidative stress as indicated by decreased malondialdehyde and protein carbonyl formation in the brain. Compared with methionine-treated animals, mice that treated with methionine and punicalagin remarkably displayed less apoptosis, indicated by the lower level of proapoptotic protein (Bax, caspases- 3, 9 and p53) and higher levels of antiapoptotic Bcl-2 protein than those in hyperhomocysteinemic mice. The potent bioactivity of punicalagin extends to protect neuronal DNA as evidenced by the inhibition of the increase of comet parameters compared to the methionine-treated mice. In conclusion, punicalagin protected from methionine-induced HHcy and brain damage with an ability to repress apoptosis by modulating apoptotic mediators and maintaining DNA integrity in the brain of mice.
Collapse
Affiliation(s)
- Mohammed A El-Missiry
- Zoology Department, Faculty of Science, Mansoura University, Egypt; Prince Sultan Military Collage of Health Science, Dhahran, Saudi Arabia.
| | - Magda A ElKomy
- Zoology Department, Faculty of Science, Mansoura University, Egypt
| | - Azza I Othman
- Zoology Department, Faculty of Science, Mansoura University, Egypt
| | - Ali M AbouEl-Ezz
- Zoology Department, Faculty of Science, Mansoura University, Egypt.
| |
Collapse
|
44
|
Jin Y, Wei F, Dai X, Qi M, Ma Y. Anti-inflammatory effect of 4-methylcyclopentadecanone in rats submitted to ischemic stroke. Fundam Clin Pharmacol 2018; 32:270-278. [PMID: 29344983 DOI: 10.1111/fcp.12348] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 12/31/2017] [Accepted: 01/11/2018] [Indexed: 12/22/2022]
Abstract
This study aimed to investigate the anti-inflammatory effect of 4-methylcyclopentadecanone (4-MCPC) in rats suffering from a cerebral ischemia/reperfusion (I/R) injury. In this study, the focal cerebral ischemia in rats was induced by middle cerebral artery occlusion (MCAO) for 2 h, and the rats were treated with 4-MCPC (8 mg/kg) just 0.5 h before reperfusion. The ischemic infarct volume was recorded 24 h after the MCAO. In addition, myeloperoxidase (MPO) activity and TNF-α and IL-1β levels in the ischemic cerebral cortex were determined by ELISA, while nuclear translocation of NF-κB p65 subunit and expression of p-IκBα were investigated by Western blotting. Our results showed that 4-MCPC treatment decreased infarct volume significantly, compared with I/R group (16.8%±7.5% vs. 39.7%±10.9%); it reduced MPO activity (0.43 ± 0.10 vs. 1.00 ± 0.51 U/g) and expression levels of TNF-α (18.90 ± 3.65 vs. 35.87 ± 4.87 ng/g) and IL-1β (1.68 ± 0.23 vs. 2.67 ± 0.38 ng/g) in ischemic brain tissues of rats. Further study revealed that 4-MCPC treatment markedly reduced nuclear translocation of NF-κB p65 subunit and expression of p-IκBα in ischemic cerebral cortex. Taken together, our results suggest that 4-MCPC protects against cerebral I/R injury and displays anti-inflammatory actions through inhibition of the NF-κB signal pathway.
Collapse
Affiliation(s)
- Yan Jin
- Shandong Provincial Key Laboratory of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, 250101, Jinan, China.,Shandong Freda Pharmaceutical Group Co., Ltd., 250101, Jinan, China
| | - Fang Wei
- Weifang People's Hospital, 261000, Weifang, China
| | - Xiaoli Dai
- Shandong Provincial Key Laboratory of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, 250101, Jinan, China
| | - Min Qi
- Shandong Provincial Key Laboratory of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, 250101, Jinan, China
| | - Yukui Ma
- Shandong Provincial Key Laboratory of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, 250101, Jinan, China
| |
Collapse
|
45
|
Usage of Multiparameter Flow Cytometry to Study Microglia and Macrophage Heterogeneity in the Central Nervous System During Neuroinflammation and Neurodegeneration. Methods Mol Biol 2018; 1745:167-177. [PMID: 29476469 DOI: 10.1007/978-1-4939-7680-5_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The resident macrophages of the central nervous system (CNS), also known as microglia, and blood-derived macrophages play an important role in the functional activity of the normal CNS, as well as in the development of neuroinflammation during various neurodegenerative disorders. Microglia and macrophages represent heterogeneous populations, which can modulate CNS environment and have different effects on neuronal regeneration. In this chapter, the main features of microglial and macrophage subsets and current methods for investigation of their heterogeneity will be discussed.
Collapse
|
46
|
Zrzavy T, Machado-Santos J, Christine S, Baumgartner C, Weiner HL, Butovsky O, Lassmann H. Dominant role of microglial and macrophage innate immune responses in human ischemic infarcts. Brain Pathol 2017; 28:791-805. [PMID: 29222823 PMCID: PMC6334527 DOI: 10.1111/bpa.12583] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 11/27/2017] [Indexed: 01/06/2023] Open
Abstract
Inflammatory mechanisms, involving granulocytes, T‐cells, B‐cells, macrophages and activated microglia, have been suggested to play a pathogenic role in experimental models of stroke and may be targets for therapeutic intervention. However, knowledge on the inflammatory response in human stroke lesions is limited. Here, we performed a quantitative study on the inflammatory reaction in human ischemic infarct lesions. We found increased numbers of T‐lymphocytes, mainly CD8+ cells, but not of B‐lymphocytes. Their number was very low in comparison to that seen in inflammatory diseases of the central nervous system and they did not show signs of activation. Polymorphonuclear leukocytes were present in meninges and less prominently in the perivascular space in early lesions, but their infiltration into the lesioned tissue was sparse with the exception of a single case. Microglia were lost in the necrotic core of fresh lesions, their number was increased in the surrounding penumbra, apparently due to proliferation. Using TMEM119 as a marker for the resident microglia pool, macrophages in lesions were in part derived from the original microglia pool, depending on the lesion stage. Most microglia and macrophages revealed a pro‐inflammatory activation pattern, expressing molecules involved in phagocytosis, oxidative injury, antigen presentation and iron metabolism and had partially lost the expression of P2RY12, an antigen expressed on homeostatic (“resting”) microglia in rodents. At later lesion stages, the majority of macrophages showed intermediate activation patterns, expressing pro‐inflammatory and anti‐inflammatory markers. Microglia in the normal white matter of controls and stroke patients were already partly activated toward a pro‐inflammatory phenotype. Our data suggest that the direct contribution of lymphocytes and granulocytes to active tissue injury in human ischemic infarct lesions is limited and that stroke therapy that targets pro‐inflammatory microglia and macrophage activation may be effective.
Collapse
Affiliation(s)
- Tobias Zrzavy
- Center for Brain Research, Medical University of Vienna, Austria
| | | | - Sheren Christine
- Center for Brain Research, Medical University of Vienna, Austria
| | - Christoph Baumgartner
- Department of Neurology, General Hospital Hietzing with Neurological Center Rosenhügel, Sigmund Freud University, Vienna, Austria
| | - Howard L Weiner
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Oleg Butovsky
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Austria
| |
Collapse
|
47
|
Chen YJ, Nguyen HM, Maezawa I, Jin LW, Wulff H. Inhibition of the potassium channel Kv1.3 reduces infarction and inflammation in ischemic stroke. Ann Clin Transl Neurol 2017; 5:147-161. [PMID: 29468176 PMCID: PMC5817832 DOI: 10.1002/acn3.513] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/10/2017] [Accepted: 11/20/2017] [Indexed: 12/26/2022] Open
Abstract
Objective Inhibitors of the voltage‐gated K+ channel Kv1.3 are currently in development as immunomodulators for the treatment of autoimmune diseases. As Kv1.3 is also expressed on microglia and has been shown to be specifically up‐regulated on “M1‐like” microglia, we here tested the therapeutic hypothesis that the brain‐penetrant small‐molecule Kv1.3‐inhibitor PAP‐1 reduces secondary inflammatory damage after ischemia/reperfusion. Methods We studied microglial Kv1.3 expression using electrophysiology and immunohistochemistry, and evaluated PAP‐1 in hypoxia‐exposed organotypic hippocampal slices and in middle cerebral artery occlusion (MCAO) with 8 days of reperfusion in both adult male C57BL/6J mice (60 min MCAO) and adult male Wistar rats (90 min MCAO). In both models, PAP‐1 administration was started 12 h after reperfusion. Results We observed Kv1.3 staining on activated microglia in ischemic infarcts in mice, rats, and humans and found higher Kv1.3 current densities in acutely isolated microglia from the infarcted hemisphere than in microglia isolated from the contralateral hemisphere of MCAO mice. PAP‐1 reduced microglia activation and increased neuronal survival in hypoxia‐exposed hippocampal slices as effectively as minocycline. In mouse MCAO, PAP‐1 dose‐dependently reduced infarct area, improved neurological deficit score, and reduced brain levels of IL‐1β and IFN‐γ without affecting IL‐10 and brain‐derived nerve growth factor (BDNF) levels or inhibiting ongoing phagocytosis. The beneficial effects on infarct area and neurological deficit score were reproduced in rats providing confirmation in a second species. Interpretation Our findings suggest that Kv1.3 constitutes a promising therapeutic target for preferentially inhibiting “M1‐like” inflammatory microglia/macrophage functions in ischemic stroke.
Collapse
Affiliation(s)
- Yi-Je Chen
- Department of Pharmacology University of California Davis 95616 California
| | - Hai M Nguyen
- Department of Pharmacology University of California Davis 95616 California
| | - Izumi Maezawa
- Department of Pathology and Laboratory Medicine University of California Davis, Sacramento 95817 California.,M.I.N.D. Institute University of California Davis 95817 California
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine University of California Davis, Sacramento 95817 California.,M.I.N.D. Institute University of California Davis 95817 California
| | - Heike Wulff
- Department of Pharmacology University of California Davis 95616 California
| |
Collapse
|
48
|
Brockmann C, Dege S, Crespo-Garcia S, Kociok N, Brockmann T, Strauß O, Joussen AM. Spatial distribution of CD115 + and CD11b + cells and their temporal activation during oxygen-induced retinopathy in mice. Graefes Arch Clin Exp Ophthalmol 2017; 256:313-323. [PMID: 29185100 DOI: 10.1007/s00417-017-3845-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/11/2017] [Accepted: 11/04/2017] [Indexed: 12/25/2022] Open
Abstract
PURPOSE The model of oxygen-induced retinopathy (OIR) is widely used to analyze pathomechanisms in retinal neovascularization. Previous studies have shown that macrophages (MP) play a key role in vessel formation in OIR, the influence of microglia (MG) having been discussed. The aim of our study was to analyze the spatial and temporal distribution and activation of MP/MG expressing CD115 and CD11b during the process of neovascularization in OIR. METHODS We used MacGreen mice expressing the green fluorescence protein (GFP) under the promoter for CD115. CD115+ cells were investigated in vivo by scanning laser ophthalmoscopy at postnatal days (P) 17 and 21 in MacGreen mice with OIR (75% oxygen from P7 to P12), and were compared to MacGreen room-air controls. In addition MP/MG were examined ex vivo using immunohistochemistry for CD11b+ detection on retinal flatmounts at P14, P17, and P21 of wild type mice with OIR. RESULTS In-vivo imaging revealed the highest density of activated MP/MG in tuft areas at P17 of MacGreen mice with OIR. Tufts and regions with a high density of CD115+ cells were detected close to veins, rather to arteries. In peripheral, fully vascularized areas, the distribution of CD115+ cells in MacGreen mice with OIR was similar to MacGreen room-air controls. Correspondingly, immunohistochemical analyses of retinal flatmounts from wild type mice with OIR induction revealed that the number of CD11b+ cells significantly varies between vascular, avascular, and tuft areas as well as between the retinal layers. Activated CD11b+ cells were almost exclusively found in avascular areas and tufts of wild type mice with OIR induction; here, the proportion of activated cells related to the total number of CD11b+ cells remained stable over the course of time. CONCLUSIONS Using two different approaches to monitor MP/MG cells, our findings demonstrated that MP/MG concentrate within pathologically vascularized areas during OIR. We were able to clarify that reactive changes of CD11b+ cell distribution to OIR primarily occur in the deep retinal layers. Furthermore, we found the highest proportion of activated CD11b+ cells in regions with pathologic neovascularization processes. Our findings support previous reports about activated MP/MG guiding revascularization in avascular areas and playing a key role in the formation and regression of neovascular tufts.
Collapse
Affiliation(s)
- Claudia Brockmann
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany.
- Berlin Institute of Health (BIH), Berlin, Germany.
| | - Sabrina Dege
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Sergio Crespo-Garcia
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Norbert Kociok
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Tobias Brockmann
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Olaf Strauß
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Antonia M Joussen
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany
| |
Collapse
|
49
|
Jin R, Xiao AY, Chen R, Granger DN, Li G. Inhibition of CD147 (Cluster of Differentiation 147) Ameliorates Acute Ischemic Stroke in Mice by Reducing Thromboinflammation. Stroke 2017; 48:3356-3365. [PMID: 29114092 DOI: 10.1161/strokeaha.117.018839] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/03/2017] [Accepted: 10/11/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND PURPOSE Inflammation and thrombosis currently are recognized as critical contributors to the pathogenesis of ischemic stroke. CD147 (cluster of differentiation 147), also known as extracellular matrix metalloproteinase inducer, can function as a key mediator of inflammatory and immune responses. CD147 expression is increased in the brain after cerebral ischemia, but its role in the pathogenesis of ischemic stroke remains unknown. In this study, we show that CD147 acts as a key player in ischemic stroke by driving thrombotic and inflammatory responses. METHODS Focal cerebral ischemia was induced in C57BL/6 mice by a 60-minute transient middle cerebral artery occlusion. Animals were treated with anti-CD147 function-blocking antibody (αCD147) or isotype control antibody. Blood-brain barrier permeability, thrombus formation, and microvascular patency were assessed 24 hours after ischemia. Infarct size, neurological deficits, and inflammatory cells invaded in the brain were assessed 72 hours after ischemia. RESULTS CD147 expression was rapidly increased in ischemic brain endothelium after transient middle cerebral artery occlusion. Inhibition of CD147 reduced infarct size and improved functional outcome on day 3 after transient middle cerebral artery occlusion. The neuroprotective effects were associated with (1) prevented blood-brain barrier damage, (2) decreased intravascular fibrin and platelet deposition, which in turn reduced thrombosis and increased cerebral perfusion, and (3) reduced brain inflammatory cell infiltration. The underlying mechanism may include reduced NF-κB (nuclear factor κB) activation, MMP-9 (matrix metalloproteinase-9) activity, and PAI-1 (plasminogen activator inhibitor-1) expression in brain microvascular endothelial cells. CONCLUSIONS Inhibition of CD147 ameliorates acute ischemic stroke by reducing thromboinflammation. CD147 might represent a novel and promising therapeutic target for ischemic stroke and possibly other thromboinflammatory disorders.
Collapse
Affiliation(s)
- Rong Jin
- From the Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey (R.J., G.L.); and Department of Neurosurgery (R.C., R.J., G.L.) and the Department of Molecular and Cellular Physiology (A.Y.X., G.L., D.N.G.), Louisiana State University Health Sciences Center, Shreveport
| | - Adam Y Xiao
- From the Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey (R.J., G.L.); and Department of Neurosurgery (R.C., R.J., G.L.) and the Department of Molecular and Cellular Physiology (A.Y.X., G.L., D.N.G.), Louisiana State University Health Sciences Center, Shreveport
| | - Rui Chen
- From the Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey (R.J., G.L.); and Department of Neurosurgery (R.C., R.J., G.L.) and the Department of Molecular and Cellular Physiology (A.Y.X., G.L., D.N.G.), Louisiana State University Health Sciences Center, Shreveport
| | - D Neil Granger
- From the Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey (R.J., G.L.); and Department of Neurosurgery (R.C., R.J., G.L.) and the Department of Molecular and Cellular Physiology (A.Y.X., G.L., D.N.G.), Louisiana State University Health Sciences Center, Shreveport
| | - Guohong Li
- From the Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey (R.J., G.L.); and Department of Neurosurgery (R.C., R.J., G.L.) and the Department of Molecular and Cellular Physiology (A.Y.X., G.L., D.N.G.), Louisiana State University Health Sciences Center, Shreveport.
| |
Collapse
|
50
|
Seifert HA, Benedek G, Nguyen H, Kent G, Vandenbark AA, Offner H. Estrogen protects both sexes against EAE by promoting common regulatory cell subtypes independent of endogenous estrogen. Metab Brain Dis 2017; 32:1747-1754. [PMID: 28689297 PMCID: PMC5650507 DOI: 10.1007/s11011-017-0063-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 06/22/2017] [Indexed: 10/19/2022]
Abstract
Autoimmune diseases including multiple sclerosis predominantly affect females. Although high levels of sex hormones, particularly estrogen (E2), can reduce proinflammatory immune responses, it remains unclear if a lack of endogenous sex hormones might affect treatment with exogenous sex hormones. Pretreatment with E2 almost completely prevents intact female and male mice from developing clinical and histological signs of experimental autoimmune encephalomyelitis (EAE) by promoting various regulatory immune cell phenotypes. To evaluate the effects of exogenous estrogen in the absence of endogenous sex hormones, the current study compared EAE severity and the emergence of different immunoregulatory cell populations after E2 pretreatment of ovariectomized (OVX) female versus male mice. We found that E2 equally protected both OVX females and males from EAE over a 21 day observation period concomitant with reduced total cell numbers in spleen and spinal cord (males only), but enhanced percentages of CD19+CD5+CD1dhi, CD19+CD138+CD44hi and CD19+Tim-1+ Breg cells, CD8+CD122+ Treg cells and CD11b+CD 206+ARG-1+ anti-inflammatory M2-like monocytes/macrophages in both groups. In contrast, E2 decreased the percentage of CD4+CD25+FoxP3+ Treg cells in OVX females but increased these Treg cells in males and intact female mice. These data suggest that with the exception of CD4+CD25+FoxP3+ Treg cells, E2 protection against EAE promotes highly overlapping immunoregulatory subsets in OVX females and males.
Collapse
Affiliation(s)
- Hilary A Seifert
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Gil Benedek
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Ha Nguyen
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Gail Kent
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
| | - Halina Offner
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA.
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA.
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA.
| |
Collapse
|