1
|
Lu M, Wang Y, Ren H, Yin X, Li H. Research progress on the mechanism of action and clinical application of remote ischemic post-conditioning for acute ischemic stroke. Clin Neurol Neurosurg 2024; 244:108397. [PMID: 38968813 DOI: 10.1016/j.clineuro.2024.108397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/07/2024]
Abstract
Remote ischemic post-conditioning (RIPostC) can reduce cerebral ischemia reperfusion injury (IRI) by inducing endogenous protective effects, the distal limb ischemia post-treatment and in situ ischemia post-treatment were classified according to the site of intervention. And in the process of clinical application distal limb ischemia post-treatment is more widely used and more conducive to clinical translation. Therefore, in this paper, we review the mechanism of action and clinical application of RIPostC in cerebral ischemia, hoping to provide reference help for future experimental directions and clinical translation.
Collapse
Affiliation(s)
- Meng Lu
- Department of Nursing, The First Hospital of Jilin University, Changchun, China
| | - Yujiao Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Hui Ren
- Department of Nursing, The First Hospital of Jilin University, Changchun, China
| | - Xin Yin
- Department of Nursing, The First Hospital of Jilin University, Changchun, China.
| | - Hongyan Li
- Department of Nursing, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
2
|
Duan H, Cheng Z, Geng X, Rajah GB, Gao J, Guo Y, Cai L, Tong Y, Li F, Jiang Q, Han Z, Ding Y. Prospective randomized controlled trial on the safety and neuroprotective efficacy of remote administration of hypothermia over spleen during acute ischemic stroke with mechanical thrombectomy: rationale, design, and protocol. Front Neurol 2024; 15:1382365. [PMID: 39081338 PMCID: PMC11286455 DOI: 10.3389/fneur.2024.1382365] [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: 02/05/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Background Brain inflammation plays a key role in ischemia/reperfusion (I/R) injury and is the main cause of "ineffective or futile recanalization" after successful mechanical thrombectomy (MT) in acute ischemic stroke (AIS). One of the primary sources of inflammatory cells after AIS are derived from the spleen. As an innovative and potential neuroprotective strategy after stroke, Remote Administration of Hypothermia (RAH) temporarily suppresses immune activities in the spleen, reduces the release of inflammatory cells and cytokines into blood, and thus reversibly diminishes inflammatory injury in the brain. Methods This single-center, prospective, randomized controlled study (RCT) is proposed for AIS patients with anterior circulation large vessel occlusion (LVO). Subjects will be randomly assigned to either the control or intervention groups in a 1:1 ratio (n = 40). Participants allocated to the intervention group will receive RAH on the abdomen above the spleen prior to recanalization until 6 h after thrombectomy. All enrolled patients will receive standard stroke Guideline care. The main adverse events associated with RAH are focal cold intolerance and abdominal pain. The primary outcome will assess safety as it pertains to RAH application. The secondary outcomes include the efficacy of RAH on spleen, determined by spleen volumes, blood inflammatory factor (cells and cytokines), and on brain injury, determined by infarction volumes and poststroke functional outcomes. Discussion This study aims to examine the safety and preliminary effectiveness of RAH over the spleen during endovascular therapy in AIS patients. The results of this study are expected to facilitate larger randomized clinical trials and hopefully prove RAH administration confers adjuvant neuroprotective properties in AIS treated with MT. Clinical trial registration https://www.chictr.org.cn/. Identifier ChiCTR 2300077052.
Collapse
Affiliation(s)
- Honglian Duan
- Department of Neurology and Stroke Center, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
| | - Zhe Cheng
- Department of Neurology and Stroke Center, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- Department of Neurology and Stroke Center, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Gary B. Rajah
- Department of Neurosurgery, Munson Medical Center, Traverse City, MI, United States
| | - Jie Gao
- Department of Neurology and Stroke Center, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yang Guo
- Department of Neurology and Stroke Center, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Lipeng Cai
- Department of Neurology and Stroke Center, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yanna Tong
- Department of Neurology and Stroke Center, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Fengwu Li
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
| | - Qian Jiang
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
| | - Zhenzhen Han
- Department of Neurology and Stroke Center, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
3
|
Xie L, He M, Ying C, Chu H. Mechanisms of inflammation after ischemic stroke in brain-peripheral crosstalk. Front Mol Neurosci 2024; 17:1400808. [PMID: 38932932 PMCID: PMC11199882 DOI: 10.3389/fnmol.2024.1400808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Stroke is a devastating disease with high morbidity, disability, and mortality, among which ischemic stroke is more common. However, there is still a lack of effective methods to improve the prognosis and reduce the incidence of its complications. At present, there is evidence that peripheral organs are involved in the inflammatory response after stroke. Moreover, the interaction between central and peripheral inflammation includes the activation of resident and peripheral immune cells, as well as the activation of inflammation-related signaling pathways, which all play an important role in the pathophysiology of stroke. In this review, we discuss the mechanisms of inflammatory response after ischemic stroke, as well as the interactions through circulatory pathways between peripheral organs (such as the gut, heart, lung and spleen) and the brain to mediate and regulate inflammation after ischemic stroke. We also propose the potential role of meningeal lymphatic vessels (MLVs)-cervical lymph nodes (CLNs) as a brain-peripheral crosstalk lymphatic pathway in ischemic stroke. In addition, we also summarize the mechanisms of anti-inflammatory drugs in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ling Xie
- Department of Critical Medicine, First People's Hospital of Linping District, Hangzhou, China
| | - Ming He
- Department of Critical Medicine, First People's Hospital of Linping District, Hangzhou, China
| | - Caidi Ying
- Department of Hepatobiliary and Pancreatic Surgery, The Traditional Chinese Medicine Hospital of Ningbo, Ningbo, China
| | - Haifeng Chu
- Department of Neurosurgery, The Traditional Chinese Medicine Hospital of Linping District, Hangzhou, China
| |
Collapse
|
4
|
Chin SP, Saffery NS, Then KY, Cheong SK. Preclinical assessments of safety and tumorigenicity of very high doses of allogeneic human umbilical cord mesenchymal stem cells. In Vitro Cell Dev Biol Anim 2024; 60:307-319. [PMID: 38421574 PMCID: PMC11014873 DOI: 10.1007/s11626-024-00852-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/08/2024] [Indexed: 03/02/2024]
Abstract
Human umbilical cord-mesenchymal stem cells (hUC-MSCs) have been widely investigated as a new therapeutic agent to treat injuries and inflammatory-mediated and autoimmune diseases. Previous studies have reported on the safety of low-dose infusion of hUC-MSCs, but information on the cell behaviour at higher doses and frequency of injection of the cells remains uncertain. The aim of the present study was to demonstrate the safety and efficacy of hUC-MSCs by Cytopeutics® (Selangor, Malaysia) from low to an extremely high dose in different monitoring periods in healthy BALB/c mice as well as assessing the tumorigenicity of the cells in B-NDG SCID immunocompromised mice. Umbilical cord from two healthy human newborns was obtained and the isolation of the hUC-MSCs was performed based on previous established method. Assessment of the cells at different doses of single or multiple administrations was performed on healthy BALB/c mice in dose range finding, sub-acute (7 d and 28 d) and sub-chronic periods (90 d). Tumorigenicity potential of Cytopeutics® hUC-MSCs was also evaluated on B-NDG immunocompromised mice for 26 wk. Single or multiple administrations of Cytopeutics® hUC-MSCs up to 40 × 106 cells per kilogramme of body weight (kg BW) were found to have no adverse effect in terms of clinical symptoms, haematology and other laboratory parameters, and histology examination in healthy BALB/c mice. hUC-MSCs were also found to reduce pro-inflammatory cytokines (IL-6 and TNF-α) in a dose-dependent manner. No sign of tumor formation was observed in B-NDG mice in the 26-wk tumorigenicity assessment. Single or multiple administration of allogenic Cytopeutics® hUC-MSCs was safe even at very high doses, is non-tumorigenic and did not cause adverse effects in mice throughout the evaluation periods. In addition, Cytopeutics® hUC-MSCs exhibited immunomodulatory effect in a dose-dependent manner.
Collapse
Affiliation(s)
- Sze-Piaw Chin
- Cytopeutics Sdn Bhd, Bio-X Centre, Persiaran Cyberpoint Selatan, Suite 2-3, 2nd Floor, Cyber 8, 63000, Cyberjaya, Selangor, Malaysia.
- CMH Specialist Hospital, Jalan Tun Dr. Ismail, 70200, Seremban, Negeri Sembilan, Malaysia.
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman (UTAR), Bandar Sungai Long, 43000, Kajang, Selangor, Malaysia.
| | - Nik Syazana Saffery
- Cytopeutics Sdn Bhd, Bio-X Centre, Persiaran Cyberpoint Selatan, Suite 2-3, 2nd Floor, Cyber 8, 63000, Cyberjaya, Selangor, Malaysia
| | - Kong-Yong Then
- Cytopeutics Sdn Bhd, Bio-X Centre, Persiaran Cyberpoint Selatan, Suite 2-3, 2nd Floor, Cyber 8, 63000, Cyberjaya, Selangor, Malaysia
- Cryocord Sdn Bhd, Cyber 8, 63000, Cyberjaya, Selangor, Malaysia
| | - Soon-Keng Cheong
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman (UTAR), Bandar Sungai Long, 43000, Kajang, Selangor, Malaysia
| |
Collapse
|
5
|
Gajghate S, Li H, Rom S. GPR55 Inactivation Diminishes Splenic Responses and Improves Neurological Outcomes in the Mouse Ischemia/Reperfusion Stroke Model. Cells 2024; 13:280. [PMID: 38334672 PMCID: PMC10855118 DOI: 10.3390/cells13030280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
Although strokes are frequent and severe, treatment options are scarce. Plasminogen activators, the only FDA-approved agents for clot treatment (tissue plasminogen activators (tPAs)), are used in a limited patient group. Moreover, there are few approaches for handling the brain's inflammatory reactions to a stroke. The orphan G protein-coupled receptor 55 (GPR55)'s connection to inflammatory processes has been recently reported; however, its role in stroke remains to be discovered. Post-stroke neuroinflammation involves the central nervous system (CNS)'s resident microglia activation and the infiltration of leukocytes from circulation into the brain. Additionally, splenic responses have been shown to be detrimental to stroke recovery. While lymphocytes enter the brain in small numbers, they regularly emerge as a very influential leukocyte subset that causes secondary inflammatory cerebral damage. However, an understanding of how this limited lymphocyte presence profoundly impacts stroke outcomes remains largely unclear. In this study, a mouse model for transient middle cerebral artery occlusion (tMCAO) was used to mimic ischemia followed by a reperfusion (IS/R) stroke. GPR55 inactivation, with a potent GPR55-specific antagonist, ML-193, starting 6 h after tMCAO or the absence of the GPR55 in mice (GPR55 knock out (GPR55ko)) resulted in a reduced infarction volume, improved neurological outcomes, and decreased splenic responses. The inhibition of GPR55 with ML-193 diminished CD4+T-cell spleen egress and attenuated CD4+T-cell brain infiltration. Additionally, ML-193 treatment resulted in an augmented number of regulatory T cells (Tregs) in the brain post-tMCAO. Our report offers documentation and the functional evaluation of GPR55 in the brain-spleen axis and lays the foundation for refining therapeutics for patients after ischemic attacks.
Collapse
Affiliation(s)
- Sachin Gajghate
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Hongbo Li
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Slava Rom
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| |
Collapse
|
6
|
Cherkashova E, Namestnikova D, Leonov G, Gubskiy I, Sukhinich K, Melnikov P, Chekhonin V, Yarygin K, Goldshtein D, Salikhova D. Comparative study of the efficacy of intra-arterial and intravenous transplantation of human induced pluripotent stem cells-derived neural progenitor cells in experimental stroke. PeerJ 2023; 11:e16358. [PMID: 38025691 PMCID: PMC10640846 DOI: 10.7717/peerj.16358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/05/2023] [Indexed: 12/01/2023] Open
Abstract
Background Cell therapy using neural progenitor cells (NPCs) is a promising approach for ischemic stroke treatment according to the results of multiple preclinical studies in animal stroke models. In the vast majority of conducted animal studies, the therapeutic efficacy of NPCs was estimated after intracerebral transplantation, while the information of the effectiveness of systemic administration is limited. Nowadays, several clinical trials aimed to estimate the safety and efficacy of NPCs transplantation in stroke patients were also conducted. In these studies, NPCs were transplanted intracerebrally in the subacute/chronic phase of stroke. The results of clinical trials confirmed the safety of the approach, however, the degree of functional improvement (the primary efficacy endpoint) was not sufficient in the majority of the studies. Therefore, more studies are needed in order to investigate the optimal transplantation parameters, especially the timing of cell transplantation after the stroke onset. This study aimed to evaluate the therapeutic effects of intra-arterial (IA) and intravenous (IV) administration of NPCs derived from induced pluripotent stem cells (iNPCs) in the acute phase of experimental stroke in rats. Induced pluripotent stem cells were chosen as the source of NPCs as this technology is perspective, has no ethical concerns and provides the access to personalized medicine. Methods Human iNPCs were transplanted IA or IV into male Wistar rats 24 h after the middle cerebral artery occlusion stroke modeling. Therapeutic efficacy was monitored for 14 days and evaluated in comparison with the cell transplantation-free control group. Additionally, cell distribution in the brain was assessed. Results The obtained results show that both routes of systemic transplantation (IV and IA) significantly reduced the mortality and improved the neurological deficit of experimental animals compared to the control group. At the same time, according to the MRI data, only IA administration led to faster and prominent reduction of the stroke volume. After IA administration, iNPCs transiently trapped in the brain and were not detected on day 7 after the transplantation. In case of IV injection, transplanted cells were not visualized in the brain. The obtained data demonstrated that the systemic transplantation of human iNPCs in the acute phase of ischemic stroke can be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Elvira Cherkashova
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency of Russian Federation, Moscow, Russian Federation
| | - Daria Namestnikova
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency of Russian Federation, Moscow, Russian Federation
| | - Georgiy Leonov
- Orekhovich Research Institute of Biomedical Chemistry of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Ilya Gubskiy
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency of Russian Federation, Moscow, Russian Federation
| | - Kirill Sukhinich
- Orekhovich Research Institute of Biomedical Chemistry of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Pavel Melnikov
- Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
| | - Vladimir Chekhonin
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
| | - Konstantin Yarygin
- Orekhovich Research Institute of Biomedical Chemistry of the Russian Academy of Sciences, Moscow, Russian Federation
- Russian Medical Academy of Continuous Professional Education of the Ministry of Healthcare of the Russian Federation, Moscow, Russian Federation
| | | | - Diana Salikhova
- Institute of Molecular and Cellular Medicine, Medical Institute, RUDN University, Moscow, Russian Federation
| |
Collapse
|
7
|
Alsbrook DL, Di Napoli M, Bhatia K, Biller J, Andalib S, Hinduja A, Rodrigues R, Rodriguez M, Sabbagh SY, Selim M, Farahabadi MH, Jafarli A, Divani AA. Neuroinflammation in Acute Ischemic and Hemorrhagic Stroke. Curr Neurol Neurosci Rep 2023; 23:407-431. [PMID: 37395873 PMCID: PMC10544736 DOI: 10.1007/s11910-023-01282-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2023] [Indexed: 07/04/2023]
Abstract
PURPOSE OF REVIEW This review aims to provide an overview of neuroinflammation in ischemic and hemorrhagic stroke, including recent findings on the mechanisms and cellular players involved in the inflammatory response to brain injury. RECENT FINDINGS Neuroinflammation is a crucial process following acute ischemic stroke (AIS) and hemorrhagic stroke (HS). In AIS, neuroinflammation is initiated within minutes of the ischemia onset and continues for several days. In HS, neuroinflammation is initiated by blood byproducts in the subarachnoid space and/or brain parenchyma. In both cases, neuroinflammation is characterized by the activation of resident immune cells, such as microglia and astrocytes, and infiltration of peripheral immune cells, leading to the release of pro-inflammatory cytokines, chemokines, and reactive oxygen species. These inflammatory mediators contribute to blood-brain barrier disruption, neuronal damage, and cerebral edema, promoting neuronal apoptosis and impairing neuroplasticity, ultimately exacerbating the neurologic deficit. However, neuroinflammation can also have beneficial effects by clearing cellular debris and promoting tissue repair. The role of neuroinflammation in AIS and ICH is complex and multifaceted, and further research is necessary to develop effective therapies that target this process. Intracerebral hemorrhage (ICH) will be the HS subtype addressed in this review. Neuroinflammation is a significant contributor to brain tissue damage following AIS and HS. Understanding the mechanisms and cellular players involved in neuroinflammation is essential for developing effective therapies to reduce secondary injury and improve stroke outcomes. Recent findings have provided new insights into the pathophysiology of neuroinflammation, highlighting the potential for targeting specific cytokines, chemokines, and glial cells as therapeutic strategies.
Collapse
Affiliation(s)
- Diana L Alsbrook
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mario Di Napoli
- Neurological Service, SS Annunziata Hospital, Sulmona, L'Aquila, Italy
| | - Kunal Bhatia
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA
| | - José Biller
- Department of Neurology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Sasan Andalib
- Research Unit of Neurology, Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Archana Hinduja
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Roysten Rodrigues
- Department of Neurology, University of Louisville, Louisville, KY, USA
| | - Miguel Rodriguez
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sara Y Sabbagh
- Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Magdy Selim
- Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Alibay Jafarli
- Department of Neurology, Tufts Medical Center, Boston, MA, USA
| | - Afshin A Divani
- Department of Neurology, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
8
|
Li H, Xiao G, Tan X, Liu G, Xu Y, Gu S. Human umbilical cord blood mononuclear cells ameliorate ischemic brain injury via promoting microglia/macrophages M2 polarization in MCAO Rats. Exp Brain Res 2023; 241:1585-1598. [PMID: 37142782 DOI: 10.1007/s00221-023-06600-1] [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] [Received: 02/03/2023] [Accepted: 03/13/2023] [Indexed: 05/06/2023]
Abstract
Cerebral infarction is one of the most prevalent cerebrovascular disorders. Microglia and infiltrating macrophages play a key role in regulating the inflammatory response after ischemic stroke. Regulation of microglia/macrophages polarization contributes to the recovery of neurological function in cerebral infarction. In recent decades, human umbilical cord blood mononuclear cells (hUCBMNCs) have been considered a potential therapeutic alternative. However, the mechanism of action is yet unclear. Our study aimed to explore whether hUCBMNCs treatment for cerebral infarction is via regulation of microglia/macrophages polarization. Adult male Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) and were treated by intravenous routine with or without hUCBMNCs at 24 h following MCAO. We evaluated the therapeutic effects of hUCBMNCs on cerebral infarction by measuring animal behavior and infarct volume, and further explored the possible mechanisms of hUCBMNCs for cerebral infarction by measuring inflammatory factors and microglia/macrophages markers using Elisa and immunofluorescence staining, respectively. We found that administration with hUCBMNCs improved behavioral functions and reduced infarct volume. Rats treated with hUCBMNCs showed a significant reduction in the level of IL-6, and TNF-α and increased the level of IL-4 and IL-10 compared to those treated without hUCBMNCs. Furthermore, hUCBMNCs inhibited M1 polarization and promoted M2 polarization of microglia/macrophages after MCAO. We conclude that hUCBMNCs could ameliorate cerebral brain injury by promoting microglia/macrophages M2 polarization in MCAO Rats. This experiment provides evidence that hUCBMNCs represent a promising therapeutic option for ischemic stroke.
Collapse
Affiliation(s)
- Hongmei Li
- Department of Neurology, the Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Gai Xiao
- Department of Neurology, the Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xiao Tan
- Department of Neurology, the Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Guojun Liu
- Shandong Cord Blood Bank, Jinan, Shangdong, China
| | - Yangzhou Xu
- Department of Neurology, the Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Shaojuan Gu
- Department of Neurology, the Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
9
|
Deng J, Chen C, Xue S, Su D, Poon WS, Hou H, Wang J. Microglia-mediated inflammatory destruction of neuro-cardiovascular dysfunction after stroke. Front Cell Neurosci 2023; 17:1117218. [PMID: 37025698 PMCID: PMC10070726 DOI: 10.3389/fncel.2023.1117218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/23/2023] [Indexed: 04/08/2023] Open
Abstract
Stroke, a serious systemic inflammatory disease, features neurological deficits and cardiovascular dysfunction. Neuroinflammation is characterized by the activation of microglia after stroke, which disrupts the cardiovascular-related neural network and the blood-brain barrier. Neural networks activate the autonomic nervous system to regulate the cardiac and blood vessels. Increased permeability of the blood-brain barrier and the lymphatic pathways promote the transfer of the central immune components to the peripheral immune organs and the recruitment of specific immune cells or cytokines, produced by the peripheral immune system, and thus modulate microglia in the brain. In addition, the spleen will also be stimulated by central inflammation to further mobilize the peripheral immune system. Both NK cells and Treg cells will be generated to enter the central nervous system to suppress further inflammation, while activated monocytes infiltrate the myocardium and cause cardiovascular dysfunction. In this review, we will focus on microglia-mediated inflammation in neural networks that result in cardiovascular dysfunction. Furthermore, we will discuss neuroimmune regulation in the central-peripheral crosstalk, in which the spleen is a vital part. Hopefully, this will benefit in anchoring another therapeutic target for neuro-cardiovascular dysfunction.
Collapse
Affiliation(s)
- Jiahong Deng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Chenghan Chen
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Shuaishuai Xue
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Daoqing Su
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Wai Sang Poon
- Neuro-Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Wai Sang Poon
| | - Honghao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, China
- Honghao Hou
| | - Jun Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
- *Correspondence: Jun Wang
| |
Collapse
|
10
|
Wang ZY, Wen ZJ, Xu HM, Zhang Y, Zhang YF. Exosomal noncoding RNAs in central nervous system diseases: biological functions and potential clinical applications. Front Mol Neurosci 2022; 15:1004221. [PMID: 36438184 PMCID: PMC9681831 DOI: 10.3389/fnmol.2022.1004221] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/18/2022] [Indexed: 09/26/2023] Open
Abstract
Central nervous system (CNS) disease is a general term for a series of complex and diverse diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), CNS tumors, stroke, epilepsy, and amyotrophic lateral sclerosis (ALS). Interneuron and neuron-glia cells communicate with each other through their homeostatic microenvironment. Exosomes in the microenvironment have crucial impacts on interneuron and neuron-glia cells by transferring their contents, such as proteins, lipids, and ncRNAs, constituting a novel form of cell-to-cell interaction and communication. Exosomal noncoding RNAs (ncRNAs), including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), circular RNAs (circRNAs), and PIWI-interacting RNAs (piRNAs), regulate physiological functions and maintain CNS homeostasis. Exosomes are regarded as extracellular messengers that transfer ncRNAs between neurons and body fluids due to their ability to cross the blood-brain barrier. This review aims to summarize the current understanding of exosomal ncRNAs in CNS diseases, including prospective diagnostic biomarkers, pathological regulators, therapeutic strategies and clinical applications. We also provide an all-sided discussion of the comparison with some similar CNS diseases and the main limitations and challenges for exosomal ncRNAs in clinical applications.
Collapse
Affiliation(s)
- Zhong-Yu Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Zeng-Jin Wen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Hai-Ming Xu
- Department of Occupational and Environmental Medicine, School of Public Health and Management, Ningxia Medical University, Yinchuan, China
- The Key Laboratory of Environmental Factors and Chronic Disease Control of Ningxia, Ningxia Medical University, Yinchuan, China
| | - Yu Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
11
|
Chen H, Liu F, Sun D, Zhang J, Luo S, Liao Q, Tian F. The potential risk factors of early-onset post-stroke depression from immuno-inflammatory perspective. Front Immunol 2022; 13:1000631. [PMID: 36225923 PMCID: PMC9549963 DOI: 10.3389/fimmu.2022.1000631] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/23/2022] [Indexed: 01/08/2023] Open
Abstract
Background Mounting evidence strongly uncovered that peripheral immuno-inflammatory response induced by acute stroke is associated with the appearance of post-stroke depression (PSD), but the mechanism remains unclear. Methods 103 stroke patients were assessed at 2 weeks after onset using Diagnostic and Statistical Manual of Mental Disorders, 5th edition and then divided into PSD and non-PSD groups. Polymorphisms of inflammatory molecules (interleukin [IL]-1β, IL-6, IL-10, IL-18, tumor necrosis factor-α [TNF-α], interferon-γ [IFN-γ] and C-reactive protein [CRP]), complete blood count parameters, splenic attenuation (SA) and splenic volume (SV) on unenhanced chest computed tomography, demographic and other clinical characteristics were obtained. Binary logistic regression model was used to analyze the associations between inflammation-related factors and the occurrence of PSD at 2 weeks after stroke. Results 49 patients were diagnosed with PSD at 2 weeks after onset (early-onset PSD). The C/T genotypes of CRP rs2794520 and rs1205 were less in PSD group than non-PSD group (both adjusted odds ratio = 3.364; 95%CI: 1.039-10.898; p = 0.043). For CRP rs3091244, the frequency of G allele was higher (80.61% vs. 13.89%) while the frequency of A allele was lower (6.12% vs. 71.30%) in PSD patients than non-PSD patients (χ2 = 104.380; p<0.001). SA of PSD patients was lower than that of non-PSD patients in the presence of CRP rs2794520 C/T genotype and rs1205 C/T genotype (both t = 2.122; p = 0.039). Peripheral monocyte count was less in PSD group than non-PSD group (adjusted odds ratio = 0.057; 95%CI: 0.005-0.686; p = 0.024). Conclusions CRP polymorphisms, SA based on CRP genotype, and peripheral monocytes are associated with the risk of early-onset PSD, suggesting peripheral immuno-inflammatory activities elicited by stroke in its aetiology.
Collapse
|
12
|
Methamphetamine Induces Systemic Inflammation and Anxiety: The Role of the Gut–Immune–Brain Axis. Int J Mol Sci 2022; 23:ijms231911224. [PMID: 36232524 PMCID: PMC9569811 DOI: 10.3390/ijms231911224] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 12/21/2022] Open
Abstract
Methamphetamine (METH) is a highly addictive drug abused by millions of users worldwide, thus becoming a global health concern with limited management options. The inefficiency of existing treatment methods has driven research into understanding the mechanisms underlying METH-induced disorders and finding effective treatments. This study aims to understand the complex interactions of the gastrointestinal–immune–nervous systems following an acute METH dose administration as one of the potential underlying molecular mechanisms concentrating on the impact of METH abuse on gut permeability. Findings showed a decreased expression of tight junction proteins ZO-1 and EpCAm in intestinal tissue and the presence of FABP-1 in sera of METH treated mice suggests intestinal wall disruption. The increased presence of CD45+ immune cells in the intestinal wall further confirms gut wall inflammation/disruption. In the brain, the expression of inflammatory markers Ccl2, Cxcl1, IL-1β, TMEM119, and the presence of albumin were higher in METH mice compared to shams, suggesting METH-induced blood–brain barrier disruption. In the spleen, cellular and gene changes are also noted. In addition, mice treated with an acute dose of METH showed anxious behavior in dark and light, open field, and elevated maze tests compared to sham controls. The findings on METH-induced inflammation and anxiety may provide opportunities to develop effective treatments for METH addiction in the future.
Collapse
|
13
|
Zhu L, Huang L, Le A, Wang TJ, Zhang J, Chen X, Wang J, Wang J, Jiang C. Interactions between the Autonomic Nervous System and the Immune System after Stroke. Compr Physiol 2022; 12:3665-3704. [PMID: 35766834 DOI: 10.1002/cphy.c210047] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acute stroke is one of the leading causes of morbidity and mortality worldwide. Stroke-induced immune-inflammatory response occurs in the perilesion areas and the periphery. Although stroke-induced immunosuppression may alleviate brain injury, it hinders brain repair as the immune-inflammatory response plays a bidirectional role after acute stroke. Furthermore, suppression of the systemic immune-inflammatory response increases the risk of life-threatening systemic bacterial infections after acute stroke. Therefore, it is essential to explore the mechanisms that underlie the stroke-induced immune-inflammatory response. Autonomic nervous system (ANS) activation is critical for regulating the local and systemic immune-inflammatory responses and may influence the prognosis of acute stroke. We review the changes in the sympathetic and parasympathetic nervous systems and their influence on the immune-inflammatory response after stroke. Importantly, this article summarizes the mechanisms on how ANS regulates the immune-inflammatory response through neurotransmitters and their receptors in immunocytes and immune organs after stroke. To facilitate translational research, we also discuss the promising therapeutic approaches modulating the activation of the ANS or the immune-inflammatory response to promote neurologic recovery after stroke. © 2022 American Physiological Society. Compr Physiol 12:3665-3704, 2022.
Collapse
Affiliation(s)
- Li Zhu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Anh Le
- Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Tom J Wang
- Winston Churchill High School, Potomac, Maryland, USA
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Xuemei Chen
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Junmin Wang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Jian Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.,Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
14
|
Wang J, Zhang J, Ye Y, Xu Q, Li Y, Feng S, Xiong X, Jian Z, Gu L. Peripheral Organ Injury After Stroke. Front Immunol 2022; 13:901209. [PMID: 35720359 PMCID: PMC9200619 DOI: 10.3389/fimmu.2022.901209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/21/2022] [Indexed: 01/08/2023] Open
Abstract
Stroke is a disease with high incidence, mortality and disability rates. It is also the main cause of adult disability in developed countries. Stroke is often caused by small emboli on the inner wall of the blood vessels supplying the brain, which can lead to arterial embolism, and can also be caused by cerebrovascular or thrombotic bleeding. With the exception of recombinant tissue plasminogen activator (rt-PA), which is a thrombolytic drug used to recanalize the occluded artery, most treatments have been demonstrated to be ineffective. Stroke can also induce peripheral organ damage. Most stroke patients have different degrees of injury to one or more organs, including the lung, heart, kidney, spleen, gastrointestinal tract and so on. In the acute phase of stroke, severe inflammation occurs in the brain, but there is strong immunosuppression in the peripheral organs, which greatly increases the risk of peripheral organ infection and aggravates organ damage. Nonneurological complications of stroke can affect treatment and prognosis, may cause serious short-term and long-term consequences and are associated with prolonged hospitalization and increased mortality. Many of these complications are preventable, and their adverse effects can be effectively mitigated by early detection and appropriate treatment with various medical measures. This article reviews the pathophysiological mechanism, clinical manifestations and treatment of peripheral organ injury after stroke.
Collapse
Affiliation(s)
- Jin Wang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiehua Zhang
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingxue Xu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yina Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shi Feng
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Abstract
Stroke remains a significant unmet clinical need with few treatment options that have a very narrow therapeutic window, thereby causing massive mortality and morbidity in the United States and around the world. Accordingly, finding safe and effective novel treatments with a wider therapeutic window stands as an urgent need in stroke. The progressive inflammation that occurs centrally and peripherally after stroke serves as a unique therapeutic target to retard and even halt the secondary cell death. Stem cell therapy represents a potent approach that can diminish inflammation in both the stroke brain and periphery (eg, spleen), advancing a paradigm shift from a traditionally brain-focused therapy to treating stroke as a neurological disorder with a significant peripheral pathology. The purpose of this review article is to highlight the inflammation-mediated secondary cell death that plagues both brain and spleen in stroke and to evaluate the therapeutic potential of stem cell therapy in dampening these inflammatory responses.
Collapse
Affiliation(s)
- Stefan Anthony
- Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL 34211, USA
| | - Dorothy Cabantan
- Michigan State University College of Osteopathic Medicine, 965 Wilson Rd, East Lansing, MI 48824, USA
| | - Molly Monsour
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Cesario V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| |
Collapse
|
16
|
Mello TG, Rosado-de-Castro PH, Vasques JF, Pinhão C, Santos TM, de Lima RR, Foerster BU, Paiva FF, Mendez-Otero R, Pimentel-Coelho PM. Hyperacute transplantation of umbilical cord mesenchymal stromal cells in a model of severe intracerebral hemorrhage. Future Sci OA 2022; 8:FSO793. [PMID: 35369279 PMCID: PMC8965815 DOI: 10.2144/fsoa-2021-0121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 02/24/2022] [Indexed: 11/23/2022] Open
Abstract
Aim: Intracerebral hemorrhage (ICH) has limited therapeutic options. We have shown that an intravenous injection of human umbilical cord-derived mesenchymal stromal cells (hUC-MSC) 24 h after an ICH in rats reduced the residual hematoma volume after a moderate hemorrhage but was inefficient in severe ICH. Here, we investigated whether a treatment in the hyperacute phase would be more effective in severe ICH. Materials & methods: Wistar rats were randomly selected to receive an intravenous injection of hUC-MSC or the vehicle 1 h after a severe ICH. Results: The hyperacute treatment with hUC-MSC did not affect the 22-day survival rate, the motor function or the residual hematoma volume. Conclusion: These results indicate the need for optimization of hUC-MSC-based therapies for severe ICH. Hemorrhagic stroke, caused by the leakage of blood from blood vessels to the brain, is a life-threatening condition that reduces the quality of life of a large number of patients worldwide without effective treatments. Here, we induced a severe hemorrhagic stroke in rats to study the effects of a treatment using mesenchymal stromal cells, stem cells obtained from the umbilical cord tissue capable of producing protective molecules for the brain. The treatment; however, did not improve some aspects of the disease, such as the motor ability and the size of the brain lesion, indicating that further studies are still necessary.
Collapse
Affiliation(s)
- Tanira Giara Mello
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
- Instituto de Engenharia Nuclear, Comissão Nacional de Energia Nuclear, Rio de Janeiro, RJ, 21941-614, Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Paulo Henrique Rosado-de-Castro
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, RJ, 21941-902, Brazil
- Departamento de Radiologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Juliana Ferreira Vasques
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, RJ, 21941-902, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Carolina Pinhão
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Tayná Monteiro Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Renata Rodrigues de Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Bernd Uwe Foerster
- Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, SP, 13566-590, Brazil
| | | | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Pedro Moreno Pimentel-Coelho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, RJ, 21941-902, Brazil
| |
Collapse
|
17
|
Xue J, Gao J, Gu Y, Wang A, Yu S, Li B, Yin Y, Wang J, Su W, Zhang H, Ren W, Gu W, Lv Z, Mu Y, Cheng Y. Human umbilical cord-derived mesenchymal stem cells alleviate insulin resistance in diet-induced obese mice via an interaction with splenocytes. Stem Cell Res Ther 2022; 13:109. [PMID: 35313972 PMCID: PMC8935757 DOI: 10.1186/s13287-022-02791-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/28/2022] [Indexed: 11/22/2022] Open
Abstract
Background Previous research has demonstrated that the spleen plays an important role in mesenchymal stem cell (MSC)-mediated alleviation of acute inflammation, as MSC infusion increases the spleen-derived anti-inflammatory cytokine interleukin 10 (IL-10) levels. However, studies on splenic involvement in MSC-induced protection against chronic inflammatory diseases are limited. Obesity is characterized by chronic low-grade inflammation, a key driver of insulin resistance. This study aims to evaluate the effects of MSCs on obesity-related insulin resistance and explore the underlying mechanism, particularly regarding splenic involvement.
Methods We induced obesity in mice by feeding them high-fat diets for 20 weeks. Human umbilical cord-derived MSCs (UC-MSCs) were systemically infused into the obese mice once per week for 6 weeks. Systemic glucose metabolic homeostasis and insulin sensitivity in epididymal adipose tissue (EAT) were evaluated. Then, we conducted in vivo blockade of IL-10 during UC-MSC infusion by intraperitoneally administrating an IL-10-neutralizing antibody twice per week. We also investigated the therapeutic effects of UC-MSCs on obese mice after removal of the spleen by splenectomy. Results UC-MSC infusions improved systemic metabolic homeostasis and alleviated insulin resistance in EAT but elicited no change in weight. Despite rare engraftment of UC-MSCs in EAT, UC-MSC infusions attenuated insulin resistance in EAT by polarizing macrophages into the M2 phenotype, coupled with elevated serum IL-10 levels. In vivo blockade of IL-10 blunted the effects of UC-MSCs on obese mice. Furthermore, UC-MSCs overwhelmingly homed to the spleen, and the ability of UC-MSCs to elevate serum IL-10 levels and alleviate insulin resistance was impaired in the absence of the spleen. Further in vivo and in vitro studies revealed that UC-MSCs promoted the capacity of regulatory T cells (Treg cells) to produce IL-10 in the spleen. Conclusions Our results demonstrated that UC-MSCs elevated serum IL-10 levels and subsequently promoted macrophage polarization, leading to alleviation of insulin resistance in EAT. The underlying mechanism was that UC-MSCs improved the capacity of Treg cells to produce IL-10 in the spleen. Our findings indicated that the spleen played a critical role in amplifying MSC-mediated immunomodulatory effects, which may contribute to maximizing MSC efficacy in clinical applications in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02791-6.
Collapse
Affiliation(s)
- Jing Xue
- Medical School of Chinese PLA, Beijing, China.,Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China.,Department of Endocrinology, Diabetes Center of People's Liberation Army (PLA), PLA Strategic Support Force Characteristic Medical Center (The 306th Hospital of PLA), Beijing, China
| | - Jieqing Gao
- Department of Endocrinology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Yulin Gu
- Medical School of Chinese PLA, Beijing, China.,Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Aihong Wang
- Department of Endocrinology, Diabetes Center of People's Liberation Army (PLA), PLA Strategic Support Force Characteristic Medical Center (The 306th Hospital of PLA), Beijing, China
| | - Songyan Yu
- Department of Endocrinology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bing Li
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yaqi Yin
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jie Wang
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Wanlu Su
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Haixia Zhang
- Medical School of Chinese PLA, Beijing, China.,Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Weizheng Ren
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Weijun Gu
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhaohui Lv
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yiming Mu
- Medical School of Chinese PLA, Beijing, China. .,Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Yu Cheng
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
18
|
Fu C, Zhou Y, Wang L. The Effect of Bone Marrow Mesenchymal Stem Cells (BMSCs) on Brain Injury Repair and Synapse Regeneration in Mice Under Different Conditions of Intrauterine Ischemia and Hypoxia Through Wnt Pathway. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) can be differentiated into a variety of cells and repair damaged cells. We explore whether BMSCs can repair brain damage and synapses regeneration in mice under intrauterine ischemia and hypoxia. Twenty-five pregnant mice were assigned into
control group, 6% hypoxic injury group, 8% hypoxic injury group, 6% treatment group, 8% treatment group followed by analysis of the expression of MBP, MAG, CSPGs, IGF-1, NCAN, COLIV, SynD1G1, GFAP, GSK-3β, and β-actin by RT-PCR and Western blot. Our results showed that
the expression of MBP, MAG, COL IV, SynD1G1, IGF-1 in the treatment group were significantly higher than those in hypoxic injury group with significant differences between the 8% treatment group and 6% treatment group (P < 0.05). In conclusion, BMSCs can repair brain damage and synapse
regeneration in mice under different intrauterine ischemia and hypoxia conditions which might be through Wnt signaling pathway.
Collapse
Affiliation(s)
- Changtao Fu
- Department of Neurosurgery, Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, 443000, China
| | - Youdong Zhou
- Department of Neurosurgery, Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, 443000, China
| | - Lei Wang
- Department of Neurosurgery, Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, 443000, China
| |
Collapse
|
19
|
Yu H, Cai Y, Zhong A, Zhang Y, Zhang J, Xu S. The "Dialogue" Between Central and Peripheral Immunity After Ischemic Stroke: Focus on Spleen. Front Immunol 2022; 12:792522. [PMID: 34975893 PMCID: PMC8717871 DOI: 10.3389/fimmu.2021.792522] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022] Open
Abstract
The immune response generated by the body after the incidence of ischemic stroke, runs through the comprehensive process of aftermath. During this process of ischemic stroke, the central neuroinflammation and peripheral immune response seriously affect the prognosis of patients, which has been the focus of research in recent years. As this research scenario progressed, the "dialogue" between central nervous inflammation and peripheral immune response after ischemic stroke has become more closely related. It's worth noting that the spleen, as an important peripheral immune organ, plays a pivotal role in this dialogue. Multiple mechanisms have previously been reported for brain-spleen crosstalk after ischemic stroke. Further, neuroinflammation in the brain can affect the peripheral immune state by activating/inhibiting spleen function. However, the activation of the peripheral immune inflammatory response can work reversibly in the spleen. It further affects intracerebral neuroinflammation through the injured blood-brain barrier. Therefore, paying close attention to the role of spleen as the pivot between central and peripheral immunity in ischemic stroke may help to provide a new target for immune intervention in the treatment of ischemic stroke. In the present review, we reviewed the important role of spleen in central neuroinflammation and peripheral immune response after ischemic stroke. We summarized the relevant studies and reports on spleen as the target of immune intervention which can provide new ideas for the clinical treatment of ischemic stroke.
Collapse
Affiliation(s)
- Hongchen Yu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yichen Cai
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Aiqin Zhong
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese, Tianjin, China
| | - Junping Zhang
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| |
Collapse
|
20
|
Brain Research Bulletin Special Issue: Brain–body communication in health and diseases Brain–spleen axis in health and diseases: a review and future perspective. Brain Res Bull 2022; 182:130-140. [DOI: 10.1016/j.brainresbull.2022.02.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 02/06/2023]
|
21
|
Molecular Mechanisms of Neuroimmune Crosstalk in the Pathogenesis of Stroke. Int J Mol Sci 2021; 22:ijms22179486. [PMID: 34502395 PMCID: PMC8431165 DOI: 10.3390/ijms22179486] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 12/21/2022] Open
Abstract
Stroke disrupts the homeostatic balance within the brain and is associated with a significant accumulation of necrotic cellular debris, fluid, and peripheral immune cells in the central nervous system (CNS). Additionally, cells, antigens, and other factors exit the brain into the periphery via damaged blood–brain barrier cells, glymphatic transport mechanisms, and lymphatic vessels, which dramatically influence the systemic immune response and lead to complex neuroimmune communication. As a result, the immunological response after stroke is a highly dynamic event that involves communication between multiple organ systems and cell types, with significant consequences on not only the initial stroke tissue injury but long-term recovery in the CNS. In this review, we discuss the complex immunological and physiological interactions that occur after stroke with a focus on how the peripheral immune system and CNS communicate to regulate post-stroke brain homeostasis. First, we discuss the post-stroke immune cascade across different contexts as well as homeostatic regulation within the brain. Then, we focus on the lymphatic vessels surrounding the brain and their ability to coordinate both immune response and fluid homeostasis within the brain after stroke. Finally, we discuss how therapeutic manipulation of peripheral systems may provide new mechanisms to treat stroke injury.
Collapse
|
22
|
Juli C, Heryaman H, Nazir A, Ang ET, Defi IR, Gamayani U, Atik N. The Lymphocyte Depletion in Patients with Acute Ischemic Stroke Associated with Poor Neurologic Outcome. Int J Gen Med 2021; 14:1843-1851. [PMID: 34017192 PMCID: PMC8131088 DOI: 10.2147/ijgm.s308325] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/15/2021] [Indexed: 01/01/2023] Open
Abstract
Purpose Inflammation plays an important role and is involved in all stages of acute ischemic stroke. One of these stages involves the recruitment of leukocytes from the peripheral circulation into the ischemic tissue. Lymphocytes as a subtype of leukocytes are important mediators and can become a predictor of neurological outcome. Several studies have been conducted regarding the correlation between differential lymphocyte counts and acute ischemic stroke. Most of these studies analyzed lymphocyte ratio to other leukocyte subtypes such as neutrophils and monocytes. This study specifically observed the role of lymphocytes as an indicator of the inflammatory response in patients with acute ischemic stroke. This study aimed to observe the correlation among risk factors, infarct location, leukocyte counts, lymphocyte value and neurologic output in acute ischemic stroke patients. Patients and Methods We observed and analyzed 193 patients’ data from medical record which met the inclusion and exclusion criteria with a diagnosis of acute ischemic stroke at the Department of Neurology of Dr. Hasan Sadikin General Bandung. Data were then analysed using appropriate statistical tests. Results Most patients have more than one risk factor with a leukocyte count of less than 10,000 cell/mm3. Infarct was mostly located in subcortical area (basal ganglia), with moderate average NIHSS values at admission and at discharge. The number of lymphocytes decreased in the subject group with more than 10,000 cell/mm3 leukocytes. Subsequently, data were analyzed using Spearman’s test and there was a correlation between NIHSS on admission and lymphocyte depletion. Conclusion The lymphocyte depletion in patients with leukocytosis is a predictor of poor NIHSS.
Collapse
Affiliation(s)
- Cep Juli
- Doctoral Program, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia.,Department of Neurology, Dr. Hasan Sadikin General Hospital, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - Henhen Heryaman
- Doctoral Program, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - Arnengsih Nazir
- Doctoral Program, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - Eng-Tat Ang
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Irma Ruslina Defi
- Department of Physical Medicine and Rehabilitation, Dr. Hasan Sadikin General Hospital, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - Uni Gamayani
- Department of Neurology, Dr. Hasan Sadikin General Hospital, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - Nur Atik
- Department of Biomedical Sciences, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| |
Collapse
|
23
|
Smith JN, Dawson DM, Christo KF, Jogasuria AP, Cameron MJ, Antczak MI, Ready JM, Gerson SL, Markowitz SD, Desai AB. 15-PGDH inhibition activates the splenic niche to promote hematopoietic regeneration. JCI Insight 2021; 6:143658. [PMID: 33600377 PMCID: PMC8026178 DOI: 10.1172/jci.insight.143658] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 02/17/2021] [Indexed: 01/08/2023] Open
Abstract
The splenic microenvironment regulates hematopoietic stem and progenitor cell (HSPC) function, particularly during demand-adapted hematopoiesis; however, practical strategies to enhance splenic support of transplanted HSPCs have proved elusive. We have previously demonstrated that inhibiting 15-hydroxyprostaglandin dehydrogenase (15-PGDH), using the small molecule (+)SW033291 (PGDHi), increases BM prostaglandin E2 (PGE2) levels, expands HSPC numbers, and accelerates hematologic reconstitution after BM transplantation (BMT) in mice. Here we demonstrate that the splenic microenvironment, specifically 15-PGDH high-expressing macrophages, megakaryocytes (MKs), and mast cells (MCs), regulates steady-state hematopoiesis and potentiates recovery after BMT. Notably, PGDHi-induced neutrophil, platelet, and HSPC recovery were highly attenuated in splenectomized mice. PGDHi induced nonpathologic splenic extramedullary hematopoiesis at steady state, and pretransplant PGDHi enhanced the homing of transplanted cells to the spleen. 15-PGDH enzymatic activity localized specifically to macrophages, MK lineage cells, and MCs, identifying these cell types as likely coordinating the impact of PGDHi on splenic HSPCs. These findings suggest that 15-PGDH expression marks HSC niche cell types that regulate hematopoietic regeneration. Therefore, PGDHi provides a well-tolerated strategy to therapeutically target multiple HSC niches, promote hematopoietic regeneration, and improve clinical outcomes of BMT.
Collapse
Affiliation(s)
- Julianne Np Smith
- Department of Medicine and Case Comprehensive Cancer Center Case Western Reserve University, Cleveland, Ohio, USA
| | - Dawn M Dawson
- Department of Medicine and Case Comprehensive Cancer Center Case Western Reserve University, Cleveland, Ohio, USA
| | - Kelsey F Christo
- Department of Medicine and Case Comprehensive Cancer Center Case Western Reserve University, Cleveland, Ohio, USA
| | - Alvin P Jogasuria
- Department of Medicine and Case Comprehensive Cancer Center Case Western Reserve University, Cleveland, Ohio, USA
| | - Mark J Cameron
- Department of Medicine and Case Comprehensive Cancer Center Case Western Reserve University, Cleveland, Ohio, USA
| | - Monika I Antczak
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Stanton L Gerson
- Department of Medicine and Case Comprehensive Cancer Center Case Western Reserve University, Cleveland, Ohio, USA.,University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| | - Sanford D Markowitz
- Department of Medicine and Case Comprehensive Cancer Center Case Western Reserve University, Cleveland, Ohio, USA.,University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| | - Amar B Desai
- Department of Medicine and Case Comprehensive Cancer Center Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
24
|
Jafarzadeh-Esfehani R, Soudyab M, Parizadeh SM, Jaripoor ME, Nejad PS, Shariati M, Nabavi AS. Circulating Exosomes and Their Role in Stroke. Curr Drug Targets 2021; 21:89-95. [PMID: 31433753 DOI: 10.2174/1389450120666190821153557] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/29/2019] [Accepted: 08/07/2019] [Indexed: 12/12/2022]
Abstract
Stroke is an acute neurologic disorder which can be life-threatening if left untreated or diagnosed late. Various detecting techniques including neurologic imaging of the brain by computed tomography or magnetic resonance imaging can facilitate diagnosis of stroke. However, according to the recent advances in molecular detection techniques, new diagnostic and prognostic markers have emerged. Exosomes as an extra cellar particle are one of these markers which can have useful diagnostic, prognostic, and even therapeutic impact after stroke. We have previously discussed the role of exosomes in cardiovascular disease and in the present review we focus on the most common cerebrovascular disease. The aim of the present review is summarizing the recent diagnostic role of exosomes which are specifically secreted during a stroke and can guide clinicians to better diagnosis of stroke.
Collapse
Affiliation(s)
- Reza Jafarzadeh-Esfehani
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soudyab
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | - Payam Sasan Nejad
- Department of neurology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Shariati
- Department of neurology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ariane Sadr Nabavi
- Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
25
|
Malone K, Diaz Diaz AC, Shearer JA, Moore AC, Waeber C. The effect of fingolimod on regulatory T cells in a mouse model of brain ischaemia. J Neuroinflammation 2021; 18:37. [PMID: 33516262 PMCID: PMC7847573 DOI: 10.1186/s12974-021-02083-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/14/2021] [Indexed: 11/30/2022] Open
Abstract
Background The role of the immune system in stroke is well-recognised. Fingolimod, an immunomodulatory agent licensed for the management of relapsing-remitting multiple sclerosis, has been shown to provide benefit in rodent models of stroke. Its mechanism of action, however, remains unclear. We hypothesised fingolimod increases the number and/or function of regulatory T cells (Treg), a lymphocyte population which promotes stroke recovery. The primary aim of this study was to rigorously investigate the effect of fingolimod on Tregs in a mouse model of brain ischaemia. The effect of fingolimod in mice with common stroke-related comorbidities (ageing and hypercholesteremia) was also investigated. Methods Young (15–17 weeks), aged C57BL/6 mice (72–73 weeks), and ApoE−/− mice fed a high-fat diet (20–21 weeks) underwent permanent electrocoagulation of the left middle cerebral artery. Mice received either saline or fingolimod (0.5 mg/kg or 1 mg/kg) at 2, 24, and 48 h post-ischaemia via intraperitoneal injection. Another cohort of young mice (8–9, 17–19 weeks) received short-term (5 days) or long-term (10 days) fingolimod (0.5 mg/kg) treatment. Flow cytometry was used to quantify Tregs in blood, spleen, and lymph nodes. Immunohistochemistry was used to quantify FoxP3+ cell infiltration into the ischaemic brain. Results Fingolimod significantly increased the frequency of Tregs within the CD4+ T cell population in blood and spleen post-ischaemia in all three mouse cohorts compared to untreated ischemic mice. The highest splenic Treg frequency in fingolimod-treated mice was observed in ApoE−/− mice (9.32 ± 1.73% vs. 7.8 ± 3.01% in young, 6.09 ± 1.64% in aged mice). The highest circulating Treg frequency was also noted in ApoE−/− mice (8.39 ± 3.26% vs. 5.43 ± 2.74% in young, 4.56 ± 1.60% in aged mice). Fingolimod significantly increased the number of FoxP3+ cells in the infarct core of all mice. The most pronounced effects were seen when mice were treated for 10 days post-ischaemia. Conclusions Fingolimod increases Treg frequency in spleen and blood post-ischaemia and enhances the number of FoxP3+ cells in the ischaemic brain. The effect of fingolimod on this regulatory cell population may underlie its neuroprotective activity and could be exploited as part of future stroke therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02083-5.
Collapse
Affiliation(s)
- Kyle Malone
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Andrea C Diaz Diaz
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Jennifer A Shearer
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Anne C Moore
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Christian Waeber
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland. .,School of Pharmacy, University College Cork, Cork, Ireland.
| |
Collapse
|
26
|
Gaidhani N, Kem WR, Uteshev VV. Spleen is not required for therapeutic effects of 4OH-GTS-21, a selective α7 nAChR agonist, in the sub-acute phase of ischemic stroke in rats. Brain Res 2020; 1751:147196. [PMID: 33159972 DOI: 10.1016/j.brainres.2020.147196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 11/15/2022]
Abstract
Acute ischemic stroke (AIS) causes both central and peripheral inflammation, while activation of α7 nicotinic acetylcholine receptors (nAChRs) provides both central and peripheral anti-inflammatory and anti-apoptotic effects. Here, we provide evidence that 4OH-GTS-21, a selective α7 agonist, produces its therapeutic effects via primarily central sites of action because 4OH-GTS-21 was found equally effective in splenectomized and non-spenectomized rats in the sub-acute phase of ischemic stroke (≤1 week). However, the spleen may boost the therapeutic efficacy of 4OH-GTS-21 in certain behavioral tasks as our data also indicated. In our tests, AIS was modeled by transient middle cerebral artery occlusion (tMCAO). Splenectomy was done 2 weeks before tMCAO. We determined that: 1) Daily 4OH-GTS-21 treatments for 7 days after tMCAO significantly reduced neurological deficits and brain injury in both splenectomized and non-spelenectomized rats demonstrating that the spleen is not required for therapeutic benefits of 4OH-GTS-21; 2) The effects of 4OH-GTS-21 in the adhesive sticker removal test were significantly weaker in splenectomized animals suggesting that the spleen boosts the efficacy of 4OH-GTS-21 in the first week after tMCAO; and 3) Ischemic brain injury was not significantly affected by splenectomy in both vehicle-treated and 4OH-GTS-21-treated animals. These data support the hypothesis that the therapeutic efficacy of sub-chronic (≤1 week) 4OH-GTS-21 primarily originates from central sites of action. These results validate brain availability as a critical factor for developing novel α7 ligands for AIS.
Collapse
Affiliation(s)
- Nikhil Gaidhani
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States
| | - William R Kem
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, 1200 Newell Drive, Gainesville, FL 32610, United States
| | - Victor V Uteshev
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, United States.
| |
Collapse
|
27
|
Clinical Trials of Stem Cell Therapy for Cerebral Ischemic Stroke. Int J Mol Sci 2020; 21:ijms21197380. [PMID: 33036265 PMCID: PMC7582939 DOI: 10.3390/ijms21197380] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/24/2020] [Accepted: 10/02/2020] [Indexed: 12/17/2022] Open
Abstract
Despite recent developments in innovative treatment strategies, stroke remains one of the leading causes of death and disability worldwide. Stem cell therapy is currently attracting much attention due to its potential for exerting significant therapeutic effects on stroke patients. Various types of cells, including bone marrow mononuclear cells, bone marrow/adipose-derived stem/stromal cells, umbilical cord blood cells, neural stem cells, and olfactory ensheathing cells have enhanced neurological outcomes in animal stroke models. These stem cells have also been tested via clinical trials involving stroke patients. In this article, the authors review potential molecular mechanisms underlying neural recovery associated with stem cell treatment, as well as recent advances in stem cell therapy, with particular reference to clinical trials and future prospects for such therapy in treating stroke.
Collapse
|
28
|
Suda S, Nito C, Yokobori S, Sakamoto Y, Nakajima M, Sowa K, Obinata H, Sasaki K, Savitz SI, Kimura K. Recent Advances in Cell-Based Therapies for Ischemic Stroke. Int J Mol Sci 2020; 21:ijms21186718. [PMID: 32937754 PMCID: PMC7555943 DOI: 10.3390/ijms21186718] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022] Open
Abstract
Stroke is the most prevalent cardiovascular disease worldwide, and is still one of the leading causes of death and disability. Stem cell-based therapy is actively being investigated as a new potential treatment for certain neurological disorders, including stroke. Various types of cells, including bone marrow mononuclear cells, bone marrow mesenchymal stem cells, dental pulp stem cells, neural stem cells, inducible pluripotent stem cells, and genetically modified stem cells have been found to improve neurological outcomes in animal models of stroke, and there are some ongoing clinical trials assessing their efficacy in humans. In this review, we aim to summarize the recent advances in cell-based therapies to treat stroke.
Collapse
Affiliation(s)
- Satoshi Suda
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
- Correspondence: ; Tel.: +81-3-3822-2131; Fax: +81-3-3822-4865
| | - Chikako Nito
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| | - Shoji Yokobori
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan; (S.Y.); (H.O.); (K.S.)
| | - Yuki Sakamoto
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| | - Masataka Nakajima
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| | - Kota Sowa
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| | - Hirofumi Obinata
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan; (S.Y.); (H.O.); (K.S.)
| | - Kazuma Sasaki
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan; (S.Y.); (H.O.); (K.S.)
| | - Sean I. Savitz
- Institute for Stroke and Cerebrovascular Disease, UTHealth, Houston, TX 77030, USA;
| | - Kazumi Kimura
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| |
Collapse
|
29
|
Potential of stem cell therapy in intracerebral hemorrhage. Mol Biol Rep 2020; 47:4671-4680. [PMID: 32415506 DOI: 10.1007/s11033-020-05457-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/11/2020] [Indexed: 01/01/2023]
Abstract
Spontaneous intracerebral hemorrhage (ICH) is a common disease associated with high mortality and morbidity. The treatment of patients with ICH includes medical and surgical interventions. New areas of surgical intervention have been focused on the evacuation of hematoma through minimally invasive neurosurgery. In contrast, there have been no significant advances in the development of medical interventions for functional recovery after ICH. Stem cells exert multiple therapeutic functions and have emerged as a promising treatment strategy. Herein, we summarized the pathophysiology of ICH and its treatment targets, and we introduced the therapeutic mechanisms of stem cells (e.g. neutrotrophy and neuroregeneration). Moreover, we reviewed and summarized the experimental designs of the preclinical studies, including the types of cells and the timing and routes of stem cell administration. We further listed and reviewed the completed/published and ongoing clinical trials supporting the safety and efficacy of stem cell therapy in ICH. The limitations of translating preclinical studies into clinical trials and the objectives of future studies were discussed. In conclusion, current literatures showed that stem cell therapy is a promising treatment in ICH and further translation research on judiciously selected group of patients is warranted before it can be extensively applied in clinical practice.
Collapse
|
30
|
Brown J, Kingsbury C, Lee J, Vandenbark AA, Meza‐Romero R, Offner H, Borlongan CV. Spleen participation in partial MHC class II construct neuroprotection in stroke. CNS Neurosci Ther 2020; 26:663-669. [PMID: 32237074 PMCID: PMC7298973 DOI: 10.1111/cns.13369] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
Pathological progression of stroke in the peripheral and central nervous systems (PNS and CNS) is characterized by multiple converging signalling pathways that exacerbate neuroinflammation-mediated secondary cell death. This creates a need for a novel type of immunotherapy capable of simultaneously lowering the synergistic inflammatory responses in the PNS and CNS, specifically the spleen and brain. Previously, we demonstrated that partial major histocompatibility complex (MHC) class II constructs can be administered subcutaneously to promote histological and behavioural effects that alleviate common symptoms found in a murine model of transient stroke. This MHC class II manipulates T cell cytokine expression in both PNS and CNS, resulting in dampened inflammation. In our long-standing efforts towards translational research, we recently demonstrated that a potent next generation mouse-based partial MHC class II construct named DRmQ (DRa1L50Q -mMOG-35-55) similarly induces neuroprotection in stroke rats, replicating the therapeutic effects of the human homolog as DRhQ (DRa1L50Q -human (h)MOG-35-55) in stroke mice. Our preclinical studies showed that DRmQ reduces motor deficits, infarct volume and peri-infarct cell loss by targeting inflammation in this second species. Moreover, we provided mechanistic support in both animal studies that partial MHC class II constructs effectively modulate the spleen, an organ which plays a critical role in modulating secondary cell death. Together, these preclinical studies satisfy testing the constructs in two stroke models, which is a major criterion of the Stroke Therapy Academic Industry Roundtable (STAIR) criteria and a key step in effectively translating this drug to the clinic. Additional translational studies, including dose-response and larger animal models may be warranted to bring MHC class II constructs closer to the clinic.
Collapse
Affiliation(s)
- John Brown
- Department of Neurosurgery and Brain RepairCenter of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFLUSA
| | - Chase Kingsbury
- Department of Neurosurgery and Brain RepairCenter of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFLUSA
| | - Jea‐Young Lee
- Department of Neurosurgery and Brain RepairCenter of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFLUSA
| | - Arthur A. Vandenbark
- Neuroimmunology Research R&D‐31VA Portland Health Care SystemPortlandORUSA,Department of Neurology and Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Roberto Meza‐Romero
- Neuroimmunology Research R&D‐31VA Portland Health Care SystemPortlandORUSA,Department of Neurology and Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Halina Offner
- Neuroimmunology Research R&D‐31VA Portland Health Care SystemPortlandORUSA,Department of Neurology and Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Cesar V. Borlongan
- Department of Neurosurgery and Brain RepairCenter of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFLUSA
| |
Collapse
|
31
|
Liu W, Bai X, Zhang A, Huang J, Xu S, Zhang J. Role of Exosomes in Central Nervous System Diseases. Front Mol Neurosci 2019; 12:240. [PMID: 31636538 PMCID: PMC6787718 DOI: 10.3389/fnmol.2019.00240] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 09/19/2019] [Indexed: 12/18/2022] Open
Abstract
There are many types of intercellular communication, and extracellular vesicles are one of the important forms of this. They are released by a variety of cell types, are heterogeneous, and can roughly be divided into microvesicles and exosomes according to their occurrence and function. Of course, exosomes do not just play a role in cell-to-cell communication. In the nervous system, exosomes can participate in intercellular communication, maintain the myelin sheath, and eliminate waste. Similarly, exosomes in the brain can play a role in central nervous system diseases, such as stroke, Alzheimer's disease (AD), Parkinson's disease (PD), prion disease, and traumatic encephalopathy (CTE), with both positive and negative effects (such as the transfer of misfolded proteins). Exosomes contain a variety of key bioactive substances and can therefore be considered as a snapshot of the intracellular environment. Studies have shown that exosomes from the central nervous system can be found in cerebrospinal fluid and peripheral body fluids, and that their contents will change with disease occurrence. Because exosomes can penetrate the blood brain barrier (BBB) and are highly stable in peripheral circulation, they can protect disease-related molecules well and therefore, using exosomes as a biomarker of central nervous system diseases is an attractive prospect as they can be used to monitor disease development and enable early diagnosis and treatment optimization. In this review, we discuss the current state of knowledge of exosomes, and introduce their pathophysiological roles in different diseases of the central nervous system as well as their roles and applications as a viable pathological biomarker.
Collapse
Affiliation(s)
- Wanying Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaodan Bai
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ao Zhang
- Epidemiology, College of Global Public Health, New York University, New York, NY, United States
| | - Juanjuan Huang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shixin Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|