1
|
Xie F, Shen B, Luo Y, Zhou H, Xie Z, Zhu S, Wei X, Chang Z, Zhu Z, Ding C, Jin K, Yang C, Batzu L, Chaudhuri KR, Chan LL, Tan EK, Wang Q. Repetitive transcranial magnetic stimulation alleviates motor impairment in Parkinson's disease: association with peripheral inflammatory regulatory T-cells and SYT6. Mol Neurodegener 2024; 19:80. [PMID: 39456006 DOI: 10.1186/s13024-024-00770-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) has been used to treat various neurological disorders. However, the molecular mechanism underlying the therapeutic effect of rTMS on Parkinson's disease (PD) has not been fully elucidated. Neuroinflammation like regulatory T-cells (Tregs) appears to be a key modulator of disease progression in PD. If rTMS affects the peripheral Tregs in PD remains unknown. METHODS Here, we conducted a prospective clinical study (Chinese ClinicalTrials. gov: ChiCTR 2100051140) involving 54 PD patients who received 10-day rTMS (10 Hz) stimulation on the primary motor cortex (M1) region or sham treatment. Clinical and function assessment as well as flow cytology study were undertaken in 54 PD patients who were consecutively recruited from the department of neurology at Zhujiang hospital between September 2021 and January 2022. Subsequently, we implemented flow cytometry analysis to examine the Tregs population in spleen of MPTP-induced PD mice that received rTMS or sham treatment, along with quantitative proteomic approach reveal novel molecular targets for Parkinson's disease, and finally, the RNA interference method verifies the role of these new molecular targets in the treatment of PD. RESULTS We demonstrated that a 10-day rTMS treatment on the M1 motor cortex significantly improved motor dysfunction in PD patients. The beneficial effects persisted for up to 40 days, and were associated with an increase in peripheral Tregs. There was a positive correlation between Tregs and motor improvements in PD cases. Similarly, a 10-day rTMS treatment on the brains of MPTP-induced PD mice significantly ameliorated motor symptoms. rTMS reversed the downregulation of circulating Tregs and tyrosine hydroxylase neurons in these mice. It also increased anti-inflammatory mediators, deactivated microglia, and decreased inflammatory cytokines. These effects were blocked by administration of a Treg inhibitor anti-CD25 antibody in MPTP-induced PD mice. Quantitative proteomic analysis identified TLR4, TH, Slc6a3 and especially Syt6 as the hub node proteins related to Tregs and rTMS therapy. Lastly, we validated the role of Treg and rTMS-related protein syt6 in MPTP mice using the virus interference method. CONCLUSIONS Our clinical and experimental studies suggest that rTMS improves motor function by modulating the function of Tregs and suppressing toxic neuroinflammation. Hub node proteins (especially Syt6) may be potential therapeutic targets. TRIAL REGISTRATION Chinese ClinicalTrials, ChiCTR2100051140. Registered 15 December 2021, https://www.chictr.org.cn/bin/project/edit?pid=133691.
Collapse
Affiliation(s)
- Fen Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Bibiao Shen
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Yuqi Luo
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Hang Zhou
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Zhenchao Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Xiaobo Wei
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Zihan Chang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Zhaohua Zhu
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Changhai Ding
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Chengwu Yang
- Division of Biostatistics and Health Services Research, Department of Population and Quantitative Health Sciences, T. H. Chan School of Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Lucia Batzu
- Parkinson Foundation International Centre of Excellence at King's College Hospital, and Kings College, Denmark Hill, London, SE5 9RS, UK
| | - K Ray Chaudhuri
- Parkinson Foundation International Centre of Excellence at King's College Hospital, and Kings College, Denmark Hill, London, SE5 9RS, UK
| | - Ling-Ling Chan
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
- 7Singapore General Hospital, Singapore; Duke-NUS Medical School, Singapore, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore.
- 7Singapore General Hospital, Singapore; Duke-NUS Medical School, Singapore, Singapore.
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China.
| |
Collapse
|
2
|
He Y, Jin W, Wan H, Zhang L, Yu L. Research progress on immune-related therapeutic targets of brain injury caused by cerebral ischemia. Cytokine 2024; 180:156651. [PMID: 38761715 DOI: 10.1016/j.cyto.2024.156651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
Stroke is the second leading cause of death worldwide and a leading cause of disability. The innate immune response occurs immediately after cerebral ischemia, resulting in adaptive immunity. More and more experimental evidence has proved that the immune response caused by cerebral ischemia plays an important role in early brain injury and later the recovery of brain injury. Innate immune cells and adaptive cells promote the occurrence of cerebral ischemic injury but also protect brain cells. A large number of studies have shown that cytokines and immune-related substances also have dual functions of promoting injury, reducing injury, or promoting injury recovery in the later stage of cerebral ischemia. They can be an important target for treating cerebral ischemic recovery. Therefore, this study discussed the immune cells, cytokines, and immune-related substances with dual roles in cerebral ischemia and summarized the therapeutic targets of cerebral ischemia. To explore more effective methods to treat cerebral ischemia, promote the recovery of brain function, and improve the prognosis of patients.
Collapse
Affiliation(s)
- Yuejia He
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Weifeng Jin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Haitong Wan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Lijiang Zhang
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Li Yu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Lee S, Kim J, You JS, Hyun YM, Kim JY, Lee JE. Ischemic stroke outcome after promoting CD4+CD25+ Treg cell migration through CCR4 overexpression in a tMCAO animal model. Sci Rep 2024; 14:10201. [PMID: 38702399 PMCID: PMC11068779 DOI: 10.1038/s41598-024-60358-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
The importance of neuroinflammation during the ischemic stroke has been extensively studied. The role of CD4+CD25+ regulatory T (Treg) cells during the recovery phase have shown infarct size reduction and functional improvement, possibly through the mitigation of inflammatory immune responses. We aimed to investigate the molecular factors involved in microglia-Treg cell communication that result in Treg trafficking. First, we observed the migration patterns of CD8+ (cytotoxic) T cells and Treg cells and then searched for chemokines released by activated microglia in an oxygen-glucose deprivation (OGD) model. The transwell migration assay showed increased migration into OGD media for both cell types, in agreement with the increase in chemokines involved in immune cell trafficking from the mouse chemokine profiling array. MSCV retrovirus was transduced to overexpress CCR4 in Treg cells. CCR4-overexpressed Treg cells were injected into the mouse transient middle cerebral artery occlusion (tMCAO) model to evaluate the therapeutic potential via the tetrazolium chloride (TTC) assay and behavioral tests. A general improvement in the prognosis of animals after tMCAO was observed. Our results suggest the increased mobility of CCR4-overexpressed Treg cells in response to microglia-derived chemokines in vitro and the therapeutic potential of Treg cells with increased mobility in cellular therapy.
Collapse
Affiliation(s)
- Seowoo Lee
- Department of Anatomy, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jiwon Kim
- Department of Anatomy, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Je Sung You
- Department of Emergency Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul, 06273, Republic of Korea
| | - Young-Min Hyun
- Department of Anatomy, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Luo J, Ma J. Correlation between thrombus composition and regulatory T cell counts with clinical outcomes of acute ischemic stroke patients with thrombectomy. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:160-167. [PMID: 38650441 PMCID: PMC11057995 DOI: 10.3724/zdxbyxb-2023-0424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/29/2024] [Indexed: 04/25/2024]
Abstract
OBJECTIVES To analyze the relationship of thrombus composition and regulatory T cell expression with clinical outcome in acute ischemic stroke (AIS) patients with thrombectomy. METHODS A total of 44 AIS patients who underwent thrombectomy in the Department of Neurology of Shaoxing Hospital from June 2021 to October 2022 were enrolled. All thrombus specimens were subjected to hematoxylin-eosin staining and immunohistochemistry. Semi-quantitative analysis was performed to determine the content of red blood cells, fibrinogen/platelets, and regulatory T (CD4+CD25+) cells. Clinical data, vascular recanalization status, and neurologic outcomes at 3 months after thrombectomy were collected. A modified Rankin Scale score of 0-2 was defined as a favorable outcome. RESULTS Among 44 patients with complete thrombus data there were 15 cases of red cell type, 11 cases of mixed type and 18 cases of fibrin/platelet type. There was a significant difference in trial of ORG 10172 in acute stroke treatment (TOAST) etiological classification among the three groups (P<0.01), while no significant differences were found in other general clinical and surgical data (all P>0.05). According to the TOAST etiology, 28 cases were classified as large atherosclerosis type and 16 cases as cardioembolic type. The proportion of red blood cells in thrombus was significantly higher in patients with large atherosclerosis type than that in those with cardioembolic type [58.0% (44.2%, 72.5%) vs. 24.5% (12.7%, 48.0%), P<0.01]. The ratio of fibrin to platelet in patients with cardiogenic embolism was significantly higher than that in patients with large atherosclerosis [73.0% (49.2%, 84.5%) vs. 40.0% (25.2%, 54.5%), P<0.01). Among the 44 patients, 19 had good while 25 had poor neurological outcomes. Univariate binary logistic regression analysis showed that age, operation time, CD4+CD25+T cell number were correlated with the functional outcomes of the patients (all P<0.05). Multivariate binary logistic regression analysis showed that thrombus CD4+CD25+T cell count was an independent factor affecting the functional outcome of patients (OR=1.369, 95%CI: 1.101-1.701, P<0.01). CONCLUSIONS There is no significant correlation of erythrocyte and fibrin/platelet components in thrombus with functional outcome in AIS patients, but an increased count of regulatory T cells associates with good functional outcome.
Collapse
Affiliation(s)
- Jia Luo
- Department of Neurology, Shaoxing Hospital Affiliated to China Medical University, Shaoxing 312030, Zhejiang Province, China.
| | - Jun Ma
- Department of Neurology, Shaoxing Hospital Affiliated to China Medical University, Shaoxing 312030, Zhejiang Province, China.
| |
Collapse
|
5
|
Liu Y, Dong J, Zhang Z, Liu Y, Wang Y. Regulatory T cells: A suppressor arm in post-stroke immune homeostasis. Neurobiol Dis 2023; 189:106350. [PMID: 37952680 DOI: 10.1016/j.nbd.2023.106350] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/09/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023] Open
Abstract
The activation of the immune system and the onset of pro- and anti-inflammatory responses play crucial roles in the pathophysiological processes of ischaemic stroke (IS). CD4+ regulatory T (Treg) cells is the main immunosuppressive cell population that is studied in the context of peripheral tolerance, autoimmunity, and the development of chronic inflammatory diseases. In recent years, more studies have focused on immune modulation after IS, and Treg cells have been demonstrated to be essential in the remission of inflammation, nerve regeneration, and behavioural recovery. However, the exact effects of Treg cells in the context of IS remain controversial, with some studies suggesting a negative correlation with stroke outcomes. In this review, we aim to provide a comprehensive overview of the current understanding of Treg cell involvement in post-stroke homeostasis. We summarized the literature focusing on the temporal changes in Treg cell populations after IS, the mechanisms of Treg cell-mediated immunomodulation in the brain, and the potential of Treg cell-based therapies for treatment. The purposes of the current article are to address the importance of Treg cells and inspire more studies to help physicians, as well as scientists, understand the whole map of immune responses during IS.
Collapse
Affiliation(s)
- Yiqi Liu
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jing Dong
- Department of Medical Engineering, Tsinghua University Yuquan Hospital, Beijing 100049, China
| | - Ziqing Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yunpeng Liu
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yang Wang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
6
|
Lukasik M, Telec M, Kazmierski R, Wojtasz I, Andrzejewska-Gorczyńska N, Kociemba W, Dworacki G, Kozubski WP, Frydrychowicz M. Temporal changes in regulatory T cell subsets defined by the transcription factor Helios in stroke and their potential role in stroke-associated infection: a prospective case-control study. J Neuroinflammation 2023; 20:275. [PMID: 37996909 PMCID: PMC10666369 DOI: 10.1186/s12974-023-02957-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) are involved in the systemic immune response after ischemic stroke. However, their role remains unclear, and the effect appears to be both neuroprotective and detrimental. Treg suppressor function may result in immunodepression and promote stroke-associated infection (SAI). Thus we assume that the bidirectional effects of Tregs may be in part attributed to the intracellular transcription factor Helios. Tregs with Helios expression (H+ Tregs) constitute 70-90% of all Treg cells and more frequently than Helios-negative Tregs (H- Tregs) express molecules recognized as markers of Tregs with suppressor abilities. METHODS AND RESULTS We prospectively assessed the circulating Treg population with flow cytometry in 52 subjects on days 1, 3, 10 and 90 after ischemic stroke and we compared the results with those obtained in concurrent age-, sex- and vascular risk factor-matched controls. At all studied time points the percentage of H+ Tregs decreased in stroke subjects-D1: 69.1% p < 0.0001; D3: 62.5% (49.6-76.6), p < 0.0001; D10: 60.9% (56.5-72.9), p < 0.0001; D90: 79.2% (50.2-91.7), p = 0.014 vs. controls: 92.7% (81.9-97.0) and the percentage of H- Tregs increased accordingly. In patients with SAI the percentage of pro-suppressor H+ Tregs on post-stroke day 3 was higher than in those without infection (p = 0.03). After adjustment for confounders, the percentage of H+ Tregs on day 3 independently correlated with SAI [OR 1.29; CI 95%: 1.08-1.27); p = 0.02]. Although the percentage of H+ Tregs on day 3 correlated positively with NIHSS score on day 90 (rS = 0.62; p < 0.01) and the infarct volume at day 90 (rS = 0.58; p < 0.05), in regression analysis it was not an independent risk factor. CONCLUSIONS On the first day after stroke the proportion of H+ vs. H- Tregs changes in favor of pro-inflammatory H- Tregs, and this shift continues toward normalization when assessed on day 90. A higher percentage of pro-suppressive H+ Tregs on day 3 independently correlates with SAI and is associated positively with NIHSS score, but it does not independently affect the outcome and stroke area in the convalescent phase of stroke.
Collapse
Affiliation(s)
- Maria Lukasik
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland.
| | - Magdalena Telec
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Radoslaw Kazmierski
- Department of Neurology, Collegium Medicum, University of Zielona Gora, Zielona Gora, Poland
| | | | | | | | - Grzegorz Dworacki
- Department of Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Wojciech P Kozubski
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | | |
Collapse
|
7
|
Cui W, Xu L, Huang L, Tian Y, Yang Y, Li Y, Yu Q. Changes of gut microbiota in patients at different phases of stroke. CNS Neurosci Ther 2023; 29:3416-3429. [PMID: 37309276 PMCID: PMC10580337 DOI: 10.1111/cns.14271] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 06/14/2023] Open
Abstract
AIMS Gut dysbiosis appears rapidly after acute stroke and may affect the prognosis, whereas changes in gut microbiota with gradual recovery from stroke are unknown and rarely studied. The purpose of this study is to explore the characteristics of gut microbiota changes over time after stroke. METHODS Stroke patients and healthy subjects were selected to compare the clinical data and gut microbiota of the patient group in two phases with that of healthy subjects and 16S rRNA gene sequencing was used to search the differences of gut microbiota in subjects. RESULTS Compared with the healthy subjects, the subacute patients mainly decreased the abundance of some gut microbial communities, while the decreased communities reduced and more communities increased the abundance in the convalescent patients. The abundance of Lactobacillaceae increased in both phases in patient group, while Butyricimona, Peptostreptococaceae and Romboutsia decreased in both phases. Correlation analysis found that the MMSE scores of patients in the two phases had the greatest correlation with the gut microbiota. CONCLUSION Gut dysbiosis still existed in patients in the subacute phase and convalescent phase, and gradually improved with the recovery of stroke. Gut microbiota may affect the prognosis of stroke by affecting BMI and/or related indicators, and there is a strong correlation between gut microbiota and cognitive function after stroke.
Collapse
Affiliation(s)
- Wei Cui
- Department of Rehabilitation MedicineSichuan Provincial People's Hospital, University of Electronic Science and Technology of ChinaChengduChina
| | - Li Xu
- Department of Rehabilitation MedicineSichuan Provincial People's Hospital, University of Electronic Science and Technology of ChinaChengduChina
| | - Lin Huang
- Department of Rehabilitation MedicineSichuan Provincial People's Hospital, University of Electronic Science and Technology of ChinaChengduChina
| | - Yang Tian
- Department of Rehabilitation MedicineSichuan Provincial People's Hospital, University of Electronic Science and Technology of ChinaChengduChina
| | - Yan Yang
- Department of Rehabilitation MedicineSichuan Provincial People's Hospital, University of Electronic Science and Technology of ChinaChengduChina
| | - Yamei Li
- Department of Rehabilitation MedicineSichuan Provincial People's Hospital, University of Electronic Science and Technology of ChinaChengduChina
| | - Qian Yu
- Department of Rehabilitation MedicineSichuan Provincial People's Hospital, University of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
8
|
Lin FH, Yang YX, Wang YJ, Subbiah SK, Wu XY. Amniotic membrane mesenchymal stromal cell-derived secretome in the treatment of acute ischemic stroke: A case report. World J Clin Cases 2023; 11:6543-6550. [PMID: 37900223 PMCID: PMC10601006 DOI: 10.12998/wjcc.v11.i27.6543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/30/2023] [Accepted: 08/18/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Stroke is the second and third leading cause of death and disability, respectively. To date, no definitive treatment can repair lost brain function. Recently, various preclinical studies have been reported on mesenchymal stromal cells (MSCs) and their derivatives and their potential as alternative therapies for stroke. CASE SUMMARY A 45-year-old female suffered an acute stroke, which led to paralysis in the left upper and lower limbs. The amniotic membrane MSC-derived secretome (MSC-secretome) was intravenously transplanted once a week for 4 wk. MSC-secretome-regulated regulatory T cells were investigated for the beneficial effects. The clinical improvement of this patient was accompanied by an increased frequency of regulatory T cells after transplantation. CONCLUSION Intravenous administration of MSC-secretome can potentially treat patients who suffer from acute ischemic stroke.
Collapse
Affiliation(s)
- Fu-Hong Lin
- Department of Neurology, Affiliated Hospital of Chifeng College, Chifeng 024000, Inner Mongolia Autonomous Region, China
| | - Yu-Xiao Yang
- Department of Technology, Beijing Protercell Biotechnology Co. Ltd., Beijing 102600, China
- Department of Technology, Inner Mongolia Protercell Biotechnology Co. Ltd., Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Yu-Jun Wang
- Department of Technology, Beijing Protercell Biotechnology Co. Ltd., Beijing 102600, China
- Department of Technology, Inner Mongolia Protercell Biotechnology Co. Ltd., Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Suresh Kumar Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai 600126, India
| | - Xiao-Yun Wu
- Department of Technology, Beijing Protercell Biotechnology Co. Ltd., Beijing 102600, China
- Department of Technology, Inner Mongolia Protercell Biotechnology Co. Ltd., Hohhot 010000, Inner Mongolia Autonomous Region, China
- Department of Interventional, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| |
Collapse
|
9
|
Nguyen JN, Chauhan A. Bystanders or not? Microglia and lymphocytes in aging and stroke. Neural Regen Res 2023; 18:1397-1403. [PMID: 36571333 PMCID: PMC10075112 DOI: 10.4103/1673-5374.360345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
As the average age of the world population increases, more people will face debilitating aging-associated conditions, including dementia and stroke. Not only does the incidence of these conditions increase with age, but the recovery afterward is often worse in older patients. Researchers and health professionals must unveil and understand the factors behind age-associated diseases to develop a therapy for older patients. Aging causes profound changes in the immune system including the activation of microglia in the brain. Activated microglia promote T lymphocyte transmigration leading to an increase in neuroinflammation, white matter damage, and cognitive impairment in both older humans and rodents. The presence of T and B lymphocytes is observed in the aged brain and correlates with worse stroke outcomes. Preclinical strategies in stroke target either microglia or the lymphocytes or the communications between them to promote functional recovery in aged subjects. In this review, we examine the role of the microglia and T and B lymphocytes in aging and how they contribute to cognitive impairment. Additionally, we provide an important update on the contribution of these cells and their interactions in preclinical aged stroke.
Collapse
Affiliation(s)
- Justin N. Nguyen
- University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Anjali Chauhan
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| |
Collapse
|
10
|
Wang M, Thomson AW, Yu F, Hazra R, Junagade A, Hu X. Regulatory T lymphocytes as a therapy for ischemic stroke. Semin Immunopathol 2023; 45:329-346. [PMID: 36469056 PMCID: PMC10239790 DOI: 10.1007/s00281-022-00975-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/17/2022] [Indexed: 12/09/2022]
Abstract
Unrestrained excessive inflammatory responses exacerbate ischemic brain injury and impede post-stroke brain recovery. CD4+CD25+Foxp3+ regulatory T (Treg) cells play important immunosuppressive roles to curtail inflammatory responses and regain immune homeostasis after stroke. Accumulating evidence confirms that Treg cells are neuroprotective at the acute stage after stroke and promote brain repair at the chronic phases. The beneficial effects of Treg cells are mediated by diverse mechanisms involving cell-cell interactions and soluble factor release. Multiple types of cells, including both immune cells and non-immune CNS cells, have been identified to be cellular targets of Treg cells. In this review, we summarize recent findings regarding the function of Treg cells in ischemic stroke and the underlying cellular and molecular mechanisms. The protective and reparative properties of Treg cells endorse them as good candidates for immune therapy. Strategies that boost the numbers and functions of Treg cells have been actively developing in the fields of transplantation and autoimmune diseases. We discuss the approaches for Treg cell expansion that have been tested in stroke models. The application of these approaches to stroke patients may bring new hope for stroke treatments.
Collapse
Affiliation(s)
- Miao Wang
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, SBST, Pittsburgh, PA, 15213, USA
| | - Angus W Thomson
- Department of Surgery and Department of Immunology, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Fang Yu
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, SBST, Pittsburgh, PA, 15213, USA
| | - Rimi Hazra
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Aditi Junagade
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, SBST, Pittsburgh, PA, 15213, USA
| | - Xiaoming Hu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, SBST, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
11
|
Jian L, Hu Y, Gao M, Shu L. Promoted Generation of T Helper 1-Like Regulatory T Cells After Transient Middle Cerebral Artery Occlusion in Type-2 Diabetic Mice. Immunol Invest 2023:1-17. [PMID: 37076315 DOI: 10.1080/08820139.2023.2197009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
BACKGROUND Regulatory T cells (Tregs) play a remarkable role in modulating post-ischemic neuroinflammation. However, the characteristics of Tregs in diabetic ischemic stroke remain unknown. METHODS Transient middle cerebral artery occlusion (MCAO) was conducted on leptin receptor-mutated db/db mice and db/+ mice. The number, cytokine production, and signaling features of Tregs in peripheral blood and ipsilateral hemispheres were evaluated by flow cytometry. Treg plasticity was assessed by the adoptive transfer of splenic Tregs into mice. The effect of ipsilateral macrophages/microglia on Treg plasticity was determined by in vitro co-culture analysis. RESULTS db/db mice had more infiltrating Tregs in their ipsilateral hemispheres than db/+ mice. Infiltrating Tregs in db/db mice expressed higher transforming growth factor-β (TGF-β), interleukin-10 (IL-10), forkhead box P3 (Foxp3), interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and T-box expressed in T cells (T-bet) in comparison to infiltrating Tregs in db/+ mice, suggesting promoted generation of T helper 1 (Th1)-like Tregs in the brains of db/db mice after stroke. The post-ischemic brain microenvironment of db/db mice significantly up-regulated IFN-γ, TNF-α, T-bet, IL-10, and TGF-β in infiltrating Tregs. Moreover, ipsilateral macrophages/microglia remarkably enhanced the expression of IFN-γ, TNF-α, and T-bet but not IL-10 and TGF-β in Tregs. db/db macrophages/microglia were more potent in up-regulating IFN-γ, TNF-α, and T-bet than db/+ macrophages/microglia. Interleukin-12 (IL-12) blockage partially abolished the modulatory effect of macrophages/microglia on Tregs. CONCLUSION The generation of Th1-like Tregs was promoted in the brains of type 2 diabetic mice after stroke. Our study reveals significant Treg plasticity in diabetic stroke.Abbreviations: Foxp3: forkhead box P3; IFN-γ: interferon-γ; IL-10: interleukin-10; IL-12: interleukin-12; MCAO: middle cerebral artery occlusion; PBS: phosphate-buffered saline; STAT1: Signal transducer and activator of transcription 1; STAT5: Signal transducer and activator of transcription 1; T-bet: T-box expressed in T cells; TGF-β: transforming growth factor-β; Th1: T helper 1; TNF-α: tumor necrosis factor-α; Tregs: regulatory T cells. Foxp3: forkhead box P3; IFN-γ: interferon-γ; IL-10: interleukin-10; IL-12: interleukin-12; MCAO: middle cerebral artery occlusion; PBS: phosphate-buffered saline; STAT1: Signal transducer and activator of transcription 1; STAT5: Signal transducer and activator of transcription 1; T-bet: T-box expressed in T cells; TGF-β: transforming growth factor-β; Th1: T helper 1; TNF-α: tumor necrosis factor-α; Tregs: regulatory T cells.
Collapse
Affiliation(s)
- Lei Jian
- The department of Endocrinology, Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province, China
| | - Yanqi Hu
- The department of Endocrinology, Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province, China
| | - Mingjie Gao
- The Department of Neurology, Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province, China
| | - Long Shu
- The Department of Neurology, Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province, China
| |
Collapse
|
12
|
Olson KE, Mosley RL, Gendelman HE. The potential for treg-enhancing therapies in nervous system pathologies. Clin Exp Immunol 2023; 211:108-121. [PMID: 36041453 PMCID: PMC10019130 DOI: 10.1093/cei/uxac084] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/28/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
While inflammation may not be the cause of disease, it is well known that it contributes to disease pathogenesis across a multitude of peripheral and central nervous system disorders. Chronic and overactive inflammation due to an effector T-cell-mediated aberrant immune response ultimately leads to tissue damage and neuronal cell death. To counteract peripheral and neuroinflammatory responses, research is being focused on regulatory T cell enhancement as a therapeutic target. Regulatory T cells are an immunosuppressive subpopulation of CD4+ T helper cells essential for maintaining immune homeostasis. The cells play pivotal roles in suppressing immune responses to maintain immune tolerance. In so doing, they control T cell proliferation and pro-inflammatory cytokine production curtailing autoimmunity and inflammation. For nervous system pathologies, Treg are known to affect the onset and tempo of neural injuries. To this end, we review recent findings supporting Treg's role in disease, as well as serving as a therapeutic agent in multiple sclerosis, myasthenia gravis, Guillain-Barre syndrome, Parkinson's and Alzheimer's diseases, and amyotrophic lateral sclerosis. An ever-broader role for Treg in the control of neurologic disease has been shown for traumatic brain injury, stroke, neurotrophic pain, epilepsy, and psychiatric disorders. To such ends, this review serves to examine the role played by Tregs in nervous system diseases with a focus on harnessing their functional therapeutic role(s).
Collapse
Affiliation(s)
- Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - R L Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| |
Collapse
|
13
|
Tuohy MC, Hillman EMC, Marshall R, Agalliu D. The age-dependent immune response to ischemic stroke. Curr Opin Neurobiol 2023; 78:102670. [PMID: 36586305 PMCID: PMC9845177 DOI: 10.1016/j.conb.2022.102670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/31/2022]
Abstract
Stroke is a devastating cause of global morbidity and mortality. Ischemic brain injury triggers a profound local and systemic immune response that participates in stroke pathophysiology. In turn, this immune response has emerged as a potential therapeutic target. In order to maximize its therapeutic potential, it is critical to understand how the immune response to ischemic brain injury is affected by age - the strongest non-modifiable risk factor for stroke. The development of multi-omics and single-cell technologies has provided a more comprehensive characterization of transcriptional and cellular changes that occur during aging. In this review, we summarize recent advances in our understanding of how age-related immune alterations shape differential stroke outcomes in older versus younger organisms, highlighting studies in both experimental mouse models and patient cohorts. Wherever possible, we emphasize outstanding questions that present important avenues for future investigation with therapeutic value for the aging population.
Collapse
Affiliation(s)
- Mary Claire Tuohy
- Doctoral Program in Neurobiology and Behavior, Columbia University Irving Medical Center, New York, NY, 10032, USA; Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA; Zuckerman Mind Brain Behavior Institute and Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Elizabeth M C Hillman
- Zuckerman Mind Brain Behavior Institute and Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA. https://twitter.com/HillmanLab
| | - Randolph Marshall
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Dritan Agalliu
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
14
|
Immunotherapy as a Treatment for Stroke: Utilizing Regulatory T Cells. BRAIN HEMORRHAGES 2023. [DOI: 10.1016/j.hest.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
|
15
|
Ren P, Wang JY, Chen HL, Lin XW, Zhao YQ, Guo WZ, Zeng ZR, Li YF. Diagnostic model constructed by nine inflammation-related genes for diagnosing ischemic stroke and reflecting the condition of immune-related cells. Front Immunol 2022; 13:1046966. [PMID: 36582228 PMCID: PMC9792959 DOI: 10.3389/fimmu.2022.1046966] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
Background Ischemic cerebral infarction is the most common type of stroke with high rates of mortality, disability, and recurrence. However, the known diagnostic biomarkers and therapeutic targets for ischemic stroke (IS) are limited. In the current study, we aimed to identify novel inflammation-related biomarkers for IS using machine learning analysis and to explore their relationship with the levels of immune-related cells in whole blood samples. Methods Gene expression profiles of healthy controls and patients with IS were download from the Gene Expression Omnibus. Analysis of differentially expressed genes (DEGs) was performed in healthy controls and patients with IS. Single-sample gene set enrichment analysis was performed to calculate inflammation scores, and weighted gene co-expression network analysis was used to analyze genes in significant modules associated with inflammation scores. Key DEGs in significant modules were then analyzed using LASSO regression analysis for constructing a diagnostic model. The effectiveness and specificity of the diagnostic model was verified in healthy controls and patients with IS and with cerebral hemorrhage (CH) using qRT-PCR. The relationship between diagnostic score and the levels of immune-related cells in whole blood were analyzed using Pearson correlations. Results A total of 831 DEGs were identified. Both chronic and acute inflammation scores were higher in patients with IS, while 54 DEGs were also clustered in the gene modules associated with chronic and acute inflammation scores. Among them, a total of 9 genes were selected to construct a diagnostic model. Interestingly, RT-qPCR showed that the diagnostic model had better diagnostic value for IS but not for CH. The levels of lymphocytes were lower in blood of patients with IS, while the levels of monocytes and neutrophils were increased. The diagnostic score of the model was negatively associated with the levels of lymphocytes and positively associated with levels of monocytes and neutrophils. Conclusions Taken together, the diagnostic model constructed using the inflammation-related genes TNFSF10, ID1, PAQR8, OSR2, PDK4, PEX11B, TNIP1, FFAR2, and JUN exhibited high and specific diagnostic value for IS and reflected the condition of lymphocytes, monocytes, and neutrophils in the blood. The diagnostic model may contribute to the diagnosis of IS.
Collapse
Affiliation(s)
- Peng Ren
- Beijing Institute of Basic Medical Sciences, Beijing, China,Department of Anesthesiology, Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jing-Ya Wang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hong-Lei Chen
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiao-Wan Lin
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yong-Qi Zhao
- Beijing Institute of Basic Medical Sciences, Beijing, China,*Correspondence: Yun-Feng Li, ; Zhi-Rui Zeng, ; Wen-Zhi Guo, ; Yong-Qi Zhao,
| | - Wen-Zhi Guo
- Department of Anesthesiology, Seventh Medical Center of Chinese PLA General Hospital, Beijing, China,*Correspondence: Yun-Feng Li, ; Zhi-Rui Zeng, ; Wen-Zhi Guo, ; Yong-Qi Zhao,
| | - Zhi-Rui Zeng
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou, China,*Correspondence: Yun-Feng Li, ; Zhi-Rui Zeng, ; Wen-Zhi Guo, ; Yong-Qi Zhao,
| | - Yun-Feng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China,Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China,*Correspondence: Yun-Feng Li, ; Zhi-Rui Zeng, ; Wen-Zhi Guo, ; Yong-Qi Zhao,
| |
Collapse
|
16
|
Rizzo PA, Bellavia S, Scala I, Colò F, Broccolini A, Antonica R, Vitali F, Angeloni BM, Brunetti V, Di Iorio R, Monforte M, Della Marca G, Calabresi P, Luigetti M, Frisullo G. COVID-19 Vaccination Is Associated with a Better Outcome in Acute Ischemic Stroke Patients: A Retrospective Observational Study. J Clin Med 2022; 11:jcm11236878. [PMID: 36498464 PMCID: PMC9737827 DOI: 10.3390/jcm11236878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Background: It is unclear whether and how COVID-19 vaccination may affect the outcome of patients with acute ischemic stroke (AIS). We investigated this potential association in a retrospective study by comparing previously vaccinated (VAX) versus unvaccinated (NoVAX) stroke patients. Methods: We collected clinical reports for all consecutive AIS patients admitted to our hospital and evaluated the outcome predictors in VAX and NoVAX groups. Adjustments were made for possible confounders in multivariable logistic regression analysis, and adjusted hazard ratios were calculated. Results: A total of 466 AIS patients (287 VAX and 179 NoVAX) were included in this study. The NIHSS score at discharge and mRS score at a 3-month follow-up visit were significantly lower in VAX patients compared to NoVAX patients (p < 0.001). Good outcomes (mRS 0−2) were significantly associated with COVID-19 vaccination before AIS (adjusted hazard ratio, 0.400 [95% CI = 0.216−0.741]). Conclusions: The observation that COVID-19 vaccination can influence the outcome of AIS provides support for further studies investigating the role of immunity in ischemic brain damage.
Collapse
Affiliation(s)
- Pier Andrea Rizzo
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Simone Bellavia
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Irene Scala
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Francesca Colò
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Aldobrando Broccolini
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Riccardo Antonica
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Francesca Vitali
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | | | - Valerio Brunetti
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Riccardo Di Iorio
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Mauro Monforte
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Giacomo Della Marca
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Paolo Calabresi
- UOC Neurology, Catholic University of Sacred Heart, 00168 Rome, Italy
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Marco Luigetti
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
- Correspondence: ; Tel.: +39-06-3015-4435
| | - Giovanni Frisullo
- UOC Neurology, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| |
Collapse
|
17
|
The Role of Extracellular Vesicles in Ischemic Stroke Severity. BIOLOGY 2022; 11:biology11101489. [PMID: 36290393 PMCID: PMC9598264 DOI: 10.3390/biology11101489] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 11/13/2022]
Abstract
The possibility of characterizing the extracellular vesicles (EVs) based on parental cell surface markers and their content makes them a new attractive prognostic biomarker. Thus, our study aims to verify the role of EVs as relevant prognostic factors for acute and mid-term outcomes in ischemic stroke. Forty-seven patients with acute ischemic stroke were evaluated at admission (T0), immediately after recanalization treatment or after 2 h in non-treated patients (T1) and after one week (Tw) using the National Institutes of Health Stroke Scale (NIHSS), and after 3 months using the Modified Rankin Scale (mRS). Total count and characterization of EVs were assessed by Nanosight analysis and flow cytometry. The relationships between stroke outcomes and EV count were assessed through multivariable negative binomial regression models. We found that the amount of platelet-derived EVs at admission was positively associated with the severity of ischemic stroke at the onset as well as with the severity of mid-term outcome. Moreover, our study revealed that T-cell-derived EVs at admission were positively related to both early and mid-term ischemic stroke outcomes. Finally, T-cell-derived EVs at T1 were positively related to mid-term ischemic stroke outcome. The present study suggests that specific EV subtypes are associated with stroke severity and both short- and long-term outcomes. EVs could represent a valid tool to improve risk stratification in patients with ischemic stroke and post-recanalization treatment monitoring.
Collapse
|
18
|
Wu F, Liu Z, Zhou L, Ye D, Zhu Y, Huang K, Weng Y, Xiong X, Zhan R, Shen J. Systemic immune responses after ischemic stroke: From the center to the periphery. Front Immunol 2022; 13:911661. [PMID: 36211352 PMCID: PMC9533176 DOI: 10.3389/fimmu.2022.911661] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 07/18/2022] [Indexed: 12/01/2022] Open
Abstract
Ischemic stroke is a leading cause of disability and death. It imposes a heavy economic burden on individuals, families and society. The mortality rate of ischemic stroke has decreased with the help of thrombolytic drug therapy and intravascular intervention. However, the nerve damage caused by ischemia-reperfusion is long-lasting and followed by multiple organ dysfunction. In this process, the immune responses manifested by systemic inflammatory responses play an important role. It begins with neuroinflammation following ischemic stroke. The large number of inflammatory cells released after activation of immune cells in the lesion area, along with the deactivated neuroendocrine and autonomic nervous systems, link the center with the periphery. With the activation of systemic immunity and the emergence of immunosuppression, peripheral organs become the second “battlefield” of the immune response after ischemic stroke and gradually become dysfunctional and lead to an adverse prognosis. The purpose of this review was to describe the systemic immune responses after ischemic stroke. We hope to provide new ideas for future research and clinical treatments to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Fan Wu
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zongchi Liu
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lihui Zhou
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Di Ye
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yu Zhu
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Kaiyuan Huang
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yuxiang Weng
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoxing Xiong
- Department of Clinical Laboratory, Renmin Hospital, Faculty of Medical Sciences, Wuhan University, Wuhan, China
| | - Renya Zhan
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Jian Shen, ; Renya Zhan,
| | - Jian Shen
- Department of Neurosurgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Jian Shen, ; Renya Zhan,
| |
Collapse
|
19
|
Choi J, Kim BR, Akuzum B, Chang L, Lee JY, Kwon HK. TREGking From Gut to Brain: The Control of Regulatory T Cells Along the Gut-Brain Axis. Front Immunol 2022; 13:916066. [PMID: 35844606 PMCID: PMC9279871 DOI: 10.3389/fimmu.2022.916066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022] Open
Abstract
The human gastrointestinal tract has an enormous and diverse microbial community, termed microbiota, that is necessary for the development of the immune system and tissue homeostasis. In contrast, microbial dysbiosis is associated with various inflammatory and autoimmune diseases as well as neurological disorders in humans by affecting not only the immune system in the gastrointestinal tract but also other distal organs. FOXP3+ regulatory T cells (Tregs) are a subset of CD4+ helper T cell lineages that function as a gatekeeper for immune activation and are essential for peripheral autoimmunity prevention. Tregs are crucial to the maintenance of immunological homeostasis and tolerance at barrier regions. Tregs reside in both lymphoid and non-lymphoid tissues, and tissue-resident Tregs have unique tissue-specific phenotype and distinct function. The gut microbiota has an impact on Tregs development, accumulation, and function in periphery. Tregs, in turn, modulate antigen-specific responses aimed towards gut microbes, which supports the host–microbiota symbiotic interaction in the gut. Recent studies have indicated that Tregs interact with a variety of resident cells in central nervous system (CNS) to limit the progression of neurological illnesses such as ischemic stroke, Alzheimer’s disease, and Parkinson’s disease. The gastrointestinal tract and CNS are functionally connected, and current findings provide insights that Tregs function along the gut-brain axis by interacting with immune, epithelial, and neuronal cells. The purpose of this study is to explain our current knowledge of the biological role of tissue-resident Tregs, as well as the interaction along the gut-brain axis.
Collapse
Affiliation(s)
- Juli Choi
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - Bo-Ram Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea
| | - Begum Akuzum
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea
| | - Leechung Chang
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - June-Yong Lee
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: June-Yong Lee, ; Ho-Keun Kwon,
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: June-Yong Lee, ; Ho-Keun Kwon,
| |
Collapse
|
20
|
DeLong JH, Ohashi SN, O'Connor KC, Sansing LH. Inflammatory Responses After Ischemic Stroke. Semin Immunopathol 2022; 44:625-648. [PMID: 35767089 DOI: 10.1007/s00281-022-00943-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/20/2022] [Indexed: 12/25/2022]
Abstract
Ischemic stroke generates an immune response that contributes to neuronal loss as well as tissue repair. This is a complex process involving a range of cell types and effector molecules and impacts tissues outside of the CNS. Recent reviews address specific aspects of this response, but several years have passed and important advances have been made since a high-level review has summarized the overall state of the field. The present review examines the initiation of the inflammatory response after ischemic stroke, the complex impacts of leukocytes on patient outcome, and the potential of basic science discoveries to impact the development of therapeutics. The information summarized here is derived from broad PubMed searches and aims to reflect recent research advances in an unbiased manner. We highlight valuable recent discoveries and identify gaps in knowledge that have the potential to advance our understanding of this disease and therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Jonathan Howard DeLong
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Sarah Naomi Ohashi
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kevin Charles O'Connor
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Lauren Hachmann Sansing
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
21
|
Liu Y, Dong J, Zhang Z, Liu Y, Wang Y. How Brain Infarction Links With the Microbiota-Gut-Brain Axis: Hints From Studies Focusing on the Risk Factors for Ischemic Stroke. Front Neurosci 2022; 16:877937. [PMID: 35685776 PMCID: PMC9170980 DOI: 10.3389/fnins.2022.877937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/14/2022] [Indexed: 11/23/2022] Open
Abstract
Ischemic stroke (IS) is among the top prevalent neurologic disorders globally today. Risk factors such as hypertension, diabetes, and aging, contribute to the development of IS, and patients with these risk factors face heavier therapeutic burden and worse prognosis. Microbiota–gut–brain axis describes the crosstalk between the gut flora, intestine, and center nervous system, which conduct homeostatic effects through the bacterial metabolites, the regulation of immune activity, also the contact with enteric nerve ends and vagus nerve. Nowadays, more studies have paid attention to the important roles that microbiota–gut–brain axis played in the risk factors of IS. In the current article, we will review the recent works focusing on the bi-directional impacts of gut dysbiosis and the pathogenic process of IS-related risk factors, for the purpose to summarize some novel findings in this area, and try to understand how probiotics could limit the development of IS via different strategies.
Collapse
Affiliation(s)
- Yunpeng Liu
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jing Dong
- Department of Medical Engineering, Tsinghua University Yuquan Hospital, Beijing, China
| | - Ziqing Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yiqi Liu
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yang Wang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Salminen A. Clinical perspectives on the age-related increase of immunosuppressive activity. J Mol Med (Berl) 2022; 100:697-712. [PMID: 35384505 PMCID: PMC8985067 DOI: 10.1007/s00109-022-02193-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/10/2022] [Accepted: 03/28/2022] [Indexed: 11/10/2022]
Abstract
The aging process is associated with a remodeling of the immune system involving chronic low-grade inflammation and a gradual decline in the function of the immune system. These processes are also called inflammaging and immunosenescence. The age-related immune remodeling is associated with many clinical changes, e.g., risk for cancers and chronic infections increases, whereas the efficiency of vaccination and immunotherapy declines with aging. On the other hand, there is convincing evidence that chronic inflammatory states promote the premature aging process. The inflammation associated with aging or chronic inflammatory conditions stimulates a counteracting immunosuppression which protects tissues from excessive inflammatory injuries but promotes immunosenescence. Immunosuppression is a driving force in tumors and chronic infections and it also induces the tolerance to vaccination and immunotherapies. Immunosuppressive cells, e.g., myeloid-derived suppressor cells (MDSC), regulatory T cells (Treg), and type M2 macrophages, have a crucial role in tumorigenesis and chronic infections as well as in the tolerance to vaccination and immunotherapies. Interestingly, there is substantial evidence that inflammaging is also associated with an increased immunosuppressive activity, e.g., upregulation of immunosuppressive cells and anti-inflammatory cytokines. Given that both the aging and chronic inflammatory states involve the activation of immunosuppression and immunosenescence, this might explain why aging is a risk factor for tumorigenesis and chronic inflammatory states and conversely, chronic inflammatory insults promote the premature aging process in humans.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
23
|
Qiu Z, Guo T, Sheng X, Tang Y, Du H. Prognostic Value of the Neutrophil-to-Lymphocyte Ratio in Patients with Chronic Internal Carotid Artery Occlusion Complicated by Cerebral Infarction. Neuropsychiatr Dis Treat 2022; 18:2265-2271. [PMID: 36268267 PMCID: PMC9578499 DOI: 10.2147/ndt.s384512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/08/2022] [Indexed: 12/03/2022] Open
Abstract
PURPOSE This study aims to investigate the prognostic value of the peripheral neutrophil-to-lymphocyte ratio (NLR) in patients with chronic internal carotid artery occlusion (CICAO) complicated by cerebral infarction. PATIENTS AND METHODS The clinical data of 99 CICAO patients complicated by cerebral infarction were retrospectively analyzed. The modified Rankin Scale (mRS) was used to assess their 3-month prognosis, and a multivariate logistic regression model was established to explore risk factors for poor prognosis. RESULTS Multivariate logistic regression analysis demonstrated that NLR (OR=2.114; 95% CI: 1.129-3.959) and baseline National Institute of Health Stroke Scale (NIHSS; OR=1.288, 95% CI: 1.053-1.574) score were risk factors of poor prognosis. The area under the receiver operator characteristic (ROC) curve of NLR in predicting the 3-month outcome after onset was 0.717 (95% CI: 0.606-0.828, P<0.000). The optimal cut-off value was 3.22, with a sensitivity of 0.743 and a specificity of 0.791. CONCLUSION NLR is an independent risk factor for the poor prognosis of CICAO patients complicated by cerebral infarction and can serve as an indicator for clinical prognosis.
Collapse
Affiliation(s)
- Zhuoyin Qiu
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou, People's Republic of China.,Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| | - Tingting Guo
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou, People's Republic of China.,Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| | - Xihua Sheng
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou, People's Republic of China.,Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| | - Ying Tang
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou, People's Republic of China.,Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| | - Huaping Du
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou, People's Republic of China.,Department of Neurology, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
24
|
Harkins AL, Kopec AL, Keeler AM. Regulatory T Cell Therapeutics for Neuroinflammatory Disorders. Crit Rev Immunol 2022; 42:1-27. [PMID: 37017285 PMCID: PMC11465901 DOI: 10.1615/critrevimmunol.2022045080] [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] [Indexed: 11/13/2022]
Abstract
A delicate balance of immune regulation exists in the central nervous system (CNS) that is often dysreg-ulated in neurological diseases, making them complicated to treat. With altered immune surveillance in the diseased or injured CNS, signals that are beneficial in the homeostatic CNS can be disrupted and lead to neuroinflammation. Recent advances in niche immune cell subsets have provided insight into the complicated cross-talk between the nervous system and the immune system. Regulatory T cells (Tregs) are a subset of T cells that are capable of suppressing effector T-cell activation and regulating immune tolerance, and play an important role in neuroprotection. Tregs have been shown to be effective therapies in a variety of immune-related disorders including, graft-versus-host disease (GVHD), type 1 diabetes (T1D), and inflammatory bowel disease (IBD), as well as within the CNS. Recently, significant advancements in engineering T cells, such as chimeric antigen receptor (CAR) T cells, have led to several approved therapies suggesting the safety and efficacy for similar engineered Treg therapies. Further, as understanding of the immune system's role in neuroinflammation has progressed, Tregs have recently become a potential therapeutic in the neurology space. In this review, we discuss Tregs and their evolving role as therapies for neuroinflammatory related disorders.
Collapse
Affiliation(s)
- Ashley L. Harkins
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences
- Horae Gene Therapy Center
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA
| | | | - Allison M. Keeler
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences
- Horae Gene Therapy Center
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
25
|
Yoshimura A, Ohyagi M, Ito M. T cells in the brain inflammation. Adv Immunol 2022; 157:29-58. [PMID: 37061287 DOI: 10.1016/bs.ai.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The immune system is deeply involved in autoimmune diseases of the central nervous system (CNS), such as multiple sclerosis, N-methyl-d-aspartate (NMDA) receptor encephalitis, and narcolepsy. Additionally, the immune system is involved in various brain diseases including cerebral infarction and neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). In particular, reports related to T cells are increasing. T cells may also play important roles in brain deterioration and dementia that occur with aging. Our understanding of the role of immune cells in the context of the brain has been greatly improved by the use of acute ischemic brain injury models. Additionally, similar neural damage and repair events are shown to occur in more chronic brain neurodegenerative brain diseases. In this review, we focus on the role of T cells, including CD4+ T cells, CD8+ T cells and regulatory T cells (Tregs) in cerebral infarction and neurodegenerative diseases.
Collapse
|
26
|
Roth W, Mohamadzadeh M. Vitamin B12 and gut-brain homeostasis in the pathophysiology of ischemic stroke. EBioMedicine 2021; 73:103676. [PMID: 34749301 PMCID: PMC8586745 DOI: 10.1016/j.ebiom.2021.103676] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 10/14/2021] [Accepted: 10/22/2021] [Indexed: 12/31/2022] Open
Abstract
Stroke is a leading cause of morbidity and mortality worldwide. It inflicts immeasurable suffering on patients and their loved ones and carries an immense social cost. Efforts to mitigate the impact of stroke have focused on identifying therapeutic targets for the prevention and treatment. The gut microbiome represents one such potential target given its multifaceted effects on conditions known to cause and worsen the severity of stroke. Vitamin B12 (VB12) serves as a cofactor for two enzymes, methylmalonyl-CoA synthase and methionine synthase, vital for methionine and nucleotide biosynthesis. VB12 deficiency results in a buildup of metabolic substrates, such as homocysteine, that alter immune homeostasis and contribute to atherosclerotic disorders, including ischemic stroke. In addition to its support of cellular function, VB12 serves as a metabolic cofactor for gut microbes. By shaping microbial communities, VB12 further impacts local and peripheral immunity. Growing evidence suggests that gut dysbiosis-related immune dysfunction induced by VB12 deficiency may potentially contributes to stroke pathogenesis, its severity, and patient outcomes. In this review, we discuss the complex interactions of VB12, gut microbes and the associated metabolites, and immune homeostasis throughout the natural history of ischemic stroke.
Collapse
Affiliation(s)
- William Roth
- Department of Neurology, University of Florida, Gainesville, FL 32608, USA.
| | - Mansour Mohamadzadeh
- Division of Gastroenterology & Nutrition, Department of Medicine, College of Medicine, University of Texas Health, San Antonio, TX, USA.
| |
Collapse
|
27
|
Wang H, Wang Z, Wu Q, Yuan Y, Cao W, Zhang X. Regulatory T cells in ischemic stroke. CNS Neurosci Ther 2021; 27:643-651. [PMID: 33470530 PMCID: PMC8111493 DOI: 10.1111/cns.13611] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
The pathophysiological mechanisms of neuroinflammation, angiogenesis, and neuroplasticity are currently the hotspots of researches in ischemic stroke. Regulatory T cells (Tregs), a subset of T cells that control inflammatory and immune responses in the body, are closely related to the pathogenesis of ischemic stroke. They participate in the inflammatory response and neuroplasticity process of ischemic stroke by various mechanisms, such as secretion of anti‐inflammatory factors, inhibition of pro‐inflammatory factors, induction of cell lysis, production of the factors that promote neural regeneration, and modulation of microglial and macrophage polarization. However, it remains unclear whether Tregs play a beneficial or deleterious role in ischemic stroke and the effect of Tregs in different stages of ischemic stroke. Here, we discuss the dynamic changes of Tregs at various stages of experimental and clinical stroke, the potential mechanisms under Tregs in regulating stroke and the preclinical studies of Tregs‐related treatments, in order to provide a reference for clinical treatment.
Collapse
Affiliation(s)
- Huan Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Zhao Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Qianqian Wu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Yujia Yuan
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Wen Cao
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei, PR China.,Hebei Vascular Homeostasis Key Laboratory, Shijiazhuang, Hebei, PR China
| |
Collapse
|
28
|
The combination of C C chemokine receptor type 5(CCR5) and Treg cells predicts prognosis in patients with ischemic stroke. J Neuroimmunol 2020; 349:577404. [DOI: 10.1016/j.jneuroim.2020.577404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/27/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022]
|
29
|
Zhao H, Li F, Huang Y, Zhang S, Li L, Yang Z, Wang R, Tao Z, Han Z, Fan J, Zheng Y, Ma Q, Luo Y. Prognostic significance of plasma IL-2 and sIL-2Rα in patients with first-ever ischaemic stroke. J Neuroinflammation 2020; 17:237. [PMID: 32795376 PMCID: PMC7427726 DOI: 10.1186/s12974-020-01920-3] [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: 05/11/2020] [Accepted: 08/05/2020] [Indexed: 11/30/2022] Open
Abstract
Background An imbalance between circulating neuroprotective and neurotoxic T cell subsets leads to poor prognosis in acute ischaemic stroke (AIS). Preclinical studies have indicated that the soluble form of the interleukin-2 receptor α (sIL-2Rα)-IL-2 complex regulates T cell differentiation. However, the association between sIL-2Rα levels and AIS remains unclear. Methods A total of 201 first-ever AIS patients within 24 h after stroke onset and 76 control subjects were recruited. The National Institutes of Health Stroke Scale (NIHSS) score and 3-month functional outcome (modified Rankin Scale [mRS] score) at admission were assessed. Plasma sIL-2Rα and IL-2 levels at admission were measured. Prognostic significance was identified by using univariate and multivariate logistic regression analyses. Results Patients with poor functional outcomes at 3 months had significantly higher levels of sIL-2Rα and lower levels of IL-2 than patients with good outcomes. Moreover, sIL-2Rα levels showed a strong positive correlation with NIHSS and mRS scores (p < 0.0001), whereas IL-2 levels were negatively correlated with mRS scores (p < 0.01). Univariate analyses showed that higher sIL-2Rα and IL-2 levels were associated with an increased and reduced risk of unfavourable outcomes, respectively. After adjusting for confounding variables, the sIL-2Rα level remained independently associated with an increased risk of an unfavourable outcome, and adding sIL-2Rα levels to the conventional risk factor model significantly improved risk reclassification (net reclassification improvement 17.56%, p = 0.003; integrated discrimination improvement 5.78%, p = 0.0003). Conclusions sIL-2Rα levels represent a novel, independent prognostic marker that can improve the currently used risk stratification of AIS patients. Our findings also highlight that elevated plasma sIL-2Rα and IL-2 levels manifested opposite correlations with functional outcome, underlining the importance of IL-2/IL-2R autocrine loops in AIS.
Collapse
Affiliation(s)
- Haiping Zhao
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Fangfang Li
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Yuyou Huang
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Sijia Zhang
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Lingzhi Li
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Zhenhong Yang
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Rongliang Wang
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Zhen Tao
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Ziping Han
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Junfen Fan
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Yangmin Zheng
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Qingfeng Ma
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China. .,Beijing Institute for Brain Disorders, Beijing, China.
| | - Yumin Luo
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, China. .,National Clinical Research Center for Geriatric Disorders, Beijing, China. .,Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
30
|
Nagy EE, Frigy A, Szász JA, Horváth E. Neuroinflammation and microglia/macrophage phenotype modulate the molecular background of post-stroke depression: A literature review. Exp Ther Med 2020; 20:2510-2523. [PMID: 32765743 PMCID: PMC7401670 DOI: 10.3892/etm.2020.8933] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022] Open
Abstract
Increasing evidence hints to the central role of neuroinflammation in the development of post-stroke depression. Danger signals released in the acute phase of ischemia trigger microglial activation, along with the infiltration of neutrophils and macrophages. The increased secretion of proinflammatory cytokines interleukin (IL)-1β, IL-6, IL-8, and tumor necrosis factor α (TNFα) provokes neuronal degeneration and apoptosis, whereas IL-6, interferon γ (IFNγ), and TNFα induce aberrant tryptophane degradation with the accumulation of the end-product quinolinic acid in resident glial cells. This promotes glutamate excitotoxicity via hyperexcitation of N-methyl-D-aspartate receptors and antagonizes 5-hydroxy-tryptamine, reducing synaptic plasticity and neuronal survival, thus favoring depression. In the post-stroke period, CX3CL1 and the CD200-CD200R interaction mediates the activation of glial cells, whereas CCL-2 attracts infiltrating macrophages. CD206 positive cells grant the removal of excessive danger signals; the high number of regulatory T cells, IL-4, IL-10, transforming growth factor β (TGFβ), and intracellular signaling via cAMP response element-binding protein (CREB) support the M2 type differentiation. In favorable conditions, these cells may exert efficient clearance, mediate tissue repair, and might be essential players in the downregulation of molecular pathways that promote post-stroke depression.
Collapse
Affiliation(s)
- Előd Ernő Nagy
- Department of Biochemistry and Environmental Chemistry, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Attila Frigy
- Department of Internal Medicine IV, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - József Attila Szász
- Neurology Clinic II, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540136 Targu Mures, Romania
| | - Emőke Horváth
- Department of Pathology, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| |
Collapse
|