1
|
Dunbar H, Hawthorne IJ, Tunstead C, McNamee EN, Weiss DJ, Armstrong ME, Donnelly SC, English K. Mesenchymal stromal cells dampen trained immunity in house dust mite-primed macrophages expressing human macrophage migration inhibitory factor polymorphism. Cytotherapy 2024; 26:1245-1251. [PMID: 38819366 DOI: 10.1016/j.jcyt.2024.05.010] [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: 02/07/2024] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Trained immunity results in long-term immunological memory, provoking a faster and greater immune response when innate immune cells encounter a secondary, often heterologous, stimulus. We have previously shown that house dust mite (HDM)-induced innate training is amplified in mice expressing the human macrophage migration inhibitory factor (MIF) CATT7 functional polymorphism. AIM This study investigated the ability of mesenchymal stromal cells (MSCs) to modulate MIF-driven trained immunity both in vitro and in vivo. METHODS Compared with wild-type mice, in vivo HDM-primed bone marrow-derived macrophages (BMDMs) from CATT7 mice expressed significantly higher levels of M1-associated genes following lipopolysaccharide stimulation ex vivo. Co-cultures of CATT7 BMDMs with MSCs suppressed this HDM-primed effect, with tumor necrosis factor alpha (TNF-α) being significantly decreased in a cyclooxygenase 2 (COX-2)-dependent manner. Interestingly, interleukin 6 (IL-6) was suppressed by MSCs independently of COX-2. In an in vitro training assay, MSCs significantly abrogated the enhanced production of pro-inflammatory cytokines by HDM-trained CATT7 BMDMs when co-cultured at the time of HDM stimulus on day 0, displaying their therapeutic efficacy in modulating an overzealous human MIF-dependent immune response. Utilizing an in vivo model of HDM-induced trained immunity, MSCs administered systemically on day 10 and day 11 suppressed this trained phenomenon by significantly reducing TNF-α and reducing IL-6 and C-C motif chemokine ligand 17 (CCL17) production. CONCLUSIONS This novel study elucidates how MSCs can attenuate an MIF-driven, HDM-trained response in CATT7 mice in a model of allergic airway inflammation.
Collapse
Affiliation(s)
- Hazel Dunbar
- Department of Biology, Maynooth University, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Ian J Hawthorne
- Department of Biology, Maynooth University, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Courteney Tunstead
- Department of Biology, Maynooth University, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Eóin N McNamee
- Department of Biology, Maynooth University, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Daniel J Weiss
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Michelle E Armstrong
- Department of Medicine, Trinity College Dublin and Tallaght University Hospital, Dublin, Ireland
| | - Seamas C Donnelly
- Department of Medicine, Trinity College Dublin and Tallaght University Hospital, Dublin, Ireland
| | - Karen English
- Department of Biology, Maynooth University, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland.
| |
Collapse
|
2
|
Qin M, Hu J, Li X, Liu J, Jiang R, Shi Y, Wang Z, Zhang L, Zhao Y, Gao H, Zhang Q, Zhao H, Li M, Huang C. Exosomal membrane proteins analysis using a silicon nanowire field effect transistor biosensor. Talanta 2024; 278:126534. [PMID: 39002259 DOI: 10.1016/j.talanta.2024.126534] [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: 05/17/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Exosomes are of great significance in clinical diagnosis, due to their high homology with parental generation, which can reflect the pathophysiological status. However, the quantitative and classification detection of exosomes is still faced with the challenges of low sensitivity and complex operation. In this study, we develop an electrical and label-free method to directly detect exosomes with high sensitivity based on a Silicon nanowire field effect transistor biosensor (Si-NW Bio-FET). First, the impact of Debye length on Si-NW Bio-FET detection was investigated through simulation. The simulation results demonstrated that as the Debye length increased, the electrical response to Si-NW produced by charged particle at a certain distance from the surface of Si-NW was greater. A Si-NW Bio-FET modified with specific antibody CD81 on the nanowire was fabricated then used for detection of cell line-derived exosomes, which achieved a low limit of detection (LOD) of 1078 particles/mL in 0.01 × PBS. Furthermore, the Si-NW Bio-FETs modified with specific antibody CD9, CD81 and CD63 respectively, were employed to distinguish exosomes derived from human promyelocytic leukemia (HL-60) cell line in three different states (control group, lipopolysaccharide (LPS) inflammation group, and LPS + Romidepsin (FK228) drug treatment group), which was consistent with nano-flow cytometry. This study provides a highly sensitive method of directly quantifying exosomes without labeling, indicating its potential as a tool for disease surveillance and medication instruction.
Collapse
Affiliation(s)
- Meiyan Qin
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Jiawei Hu
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; North China University of Technology, Beijing 100144, China
| | - Xue Li
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Jinlong Liu
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Rui Jiang
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yimin Shi
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Zizhen Wang
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Lingqian Zhang
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China
| | - Yang Zhao
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China
| | - Hang Gao
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China
| | - Qingzhu Zhang
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China
| | - Haiping Zhao
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Mingxiao Li
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China.
| | - Chengjun Huang
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
3
|
Araghi MME, Abdolmaleki A, Ghaleh HEG, Kondori BJ, Alvanegh AG, Moghaddam MM, Anbaran SJHN. Transplantation of human umbilical cord mesenchymal stem cells optimized with IFN-γ is a potential procedure for modification of motor impairment in multiple sclerosis cases: a preclinical systematic review and meta-analysis study. Anat Cell Biol 2024; 57:333-345. [PMID: 38978508 PMCID: PMC11424570 DOI: 10.5115/acb.24.082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 07/10/2024] Open
Abstract
Stem cells transplantation (SCT) is known as a newfound strategy for multiple sclerosis (MS) treatment. Human umbilical cord mesenchymal stem cells (hUCMSCs) contain various regenerative features. Experimental autoimmune encephalomyelitis (EAE) is a laboratory model of MS. This meta-analysis study was conducted to assess the overall therapeutic effects of hUCMSCs on reduction of clinical score (CS) and restoration of active movement in EAE-induced animals. For comprehensive searching (in various English and Persian databases until May 1, 2024), the main keywords of "Experimental Autoimmune Encephalomyelitis", "Multiple Sclerosis", "Human", "Umbilical Cord", "Mesenchymal", and "Stem Cell" were hired. Collected data were transferred to the citation manager software (EndNote x8) and duplicate papers were merged. Primary and secondary screenings were applied (according to the inclusion and exclusion criteria) and eligible studies were prepared for data collection. CS of two phases of peak and recovery of EAE were extracted as the difference in means and various analyses including heterogeneity, publication bias, funnel plot, and sensitivity index were reported. Meta-analysis was applied by CMA software (v.2), P<0.05 was considered a significant level, and the confidence interval (CI) was determined 95% (95% CI). Six eligible high-quality (approved by ARRIVE checklist) papers were gathered. The difference in means of peak and recovery phases were -0.775 (-1.325 to -0.225; P=0.006; I2=90.417%) and -1.230 (-1.759 to -0.700; P<0.001; I2=93.402%), respectively. The overall therapeutic effects of SCT of hUCMSCs on the EAE cases was -1.011 (95% CI=-1.392 to -0.629; P=0.001). hUCMSCs transplantation through the intravenous route to the animal MS model (EAE) seems a considerably effective procedure for the alleviation of motor defects in both phases of peak and recovery.
Collapse
Affiliation(s)
| | - Amir Abdolmaleki
- Department of Operating Room, Nahavand School of Allied Medical Sciences, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hadi Esmaeili Gouvarchin Ghaleh
- Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Bahman Jalali Kondori
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
- Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | | | | |
Collapse
|
4
|
Chen M, Kang X, Zhang Y, Liu Y. Trained immunity: A link between risk factors and cardiovascular disease. Br J Pharmacol 2024. [PMID: 38824960 DOI: 10.1111/bph.16472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/01/2024] [Accepted: 05/04/2024] [Indexed: 06/04/2024] Open
Abstract
Cardiovascular diseases are significant contributors to human mortality, closely associated with inflammation. With the changing living conditions and the extension of human lifespan, greater attention has been directed towards understanding the impact of early, long-term events on the development of cardiovascular events. Lifestyle factors such as stress, unhealthy diet and physical inactivity can increase the risk of cardiovascular diseases. Interestingly, even if the risk factors are addressed later, their influence may persist. Recently, the concept of trained innate immunity (TRIM), defined as sustained alterations in the function of innate immunocyte that promote a more robust response to downstream stimuli, has been proposed to be involved in cardiovascular diseases. It is hypothesized that TRIM may serve as a mediator bridging the impacts of aforementioned risk factors. This review aims to elucidate the role of TRIM in cardiovascular diseases and highlight its significance in uncovering new mechanisms and therapeutic targets.
Collapse
Affiliation(s)
- Mingqi Chen
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xuya Kang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yan Zhang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yahan Liu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
5
|
Pratiwi DIN, Alhajlah S, Alawadi A, Hjazi A, Alawsi T, Almalki SG, Alsalamy A, Kumar A. Mesenchymal stem cells and their extracellular vesicles as emerging therapeutic tools in the treatment of ischemic stroke. Tissue Cell 2024; 87:102320. [PMID: 38342071 DOI: 10.1016/j.tice.2024.102320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/13/2024]
Abstract
Ischemic stroke (IS) is a neurological condition characterized by severe long-term consequences and an unfavorable prognosis for numerous patients. Despite advancements in stroke treatment, existing therapeutic approaches possess certain limitations. However, accumulating evidence suggests that mesenchymal stem/stromal cells (MSCs) hold promise as a potential therapy for various neurological disorders, including IS, owing to their advantageous properties, such as immunomodulation and tissue regeneration. Additionally, MSCs primarily exert their therapeutic effects through the release of extracellular vesicles (EVs), highlighting the significance of their paracrine activities. These EVs are small double-layered phospholipid membrane vesicles, carrying a diverse cargo of proteins, lipids, and miRNAs that enable effective cell-to-cell communication. Notably, EVs have emerged as attractive substitutes for stem cell therapy due to their reduced immunogenicity, lower tumorigenic potential, and ease of administration and handling. Hence, this review summarizes the current preclinical and clinical studies performed to investigate the safety and therapeutic potential of MSCs and their EVs derived from different sources, including bone marrow, adipose tissue, umbilical cord blood, and Wharton's jelly in IS.
Collapse
Affiliation(s)
| | - Sharif Alhajlah
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Ahmed Alawadi
- College of technical engineering, the Islamic University, Najaf, Iraq; College of technical engineering, the Islamic University of Al Diwaniyah, Iraq; College of technical engineering, the Islamic University of Babylon, Iraq
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Taif Alawsi
- Department of Laser and Optoelectronics Engineering, University of Technology, Baghdad, Iraq
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Ali Alsalamy
- College of technical engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after the First President of Russia Boris Yeltsin, Ekaterinburg 620002, Russia
| |
Collapse
|
6
|
Saleh RO, Majeed AA, Margiana R, Alkadir OKA, Almalki SG, Ghildiyal P, Samusenkov V, Jabber NK, Mustafa YF, Elawady A. Therapeutic gene delivery by mesenchymal stem cell for brain ischemia damage: Focus on molecular mechanisms in ischemic stroke. Cell Biochem Funct 2024; 42:e3957. [PMID: 38468129 DOI: 10.1002/cbf.3957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 03/13/2024]
Abstract
Cerebral ischemic damage is prevalent and the second highest cause of death globally across patient populations; it is as a substantial reason of morbidity and mortality. Mesenchymal stromal cells (MSCs) have garnered significant interest as a potential treatment for cerebral ischemic damage, as shown in ischemic stroke, because of their potent intrinsic features, which include self-regeneration, immunomodulation, and multi-potency. Additionally, MSCs are easily obtained, isolated, and cultured. Despite this, there are a number of obstacles that hinder the effectiveness of MSC-based treatment, such as adverse microenvironmental conditions both in vivo and in vitro. To overcome these obstacles, the naïve MSC has undergone a number of modification processes to enhance its innate therapeutic qualities. Genetic modification and preconditioning modification (with medications, growth factors, and other substances) are the two main categories into which these modification techniques can be separated. This field has advanced significantly and is still attracting attention and innovation. We examine these cutting-edge methods for preserving and even improving the natural biological functions and therapeutic potential of MSCs in relation to adhesion, migration, homing to the target site, survival, and delayed premature senescence. We address the use of genetically altered MSC in stroke-induced damage. Future strategies for improving the therapeutic result and addressing the difficulties associated with MSC modification are also discussed.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | - Ali A Majeed
- Department of Pathological Analyses, Faculty of Science, University of Kufa, Najaf, Iraq
| | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Ola Kamal A Alkadir
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudi Arabia
| | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Vadim Samusenkov
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Ahmed Elawady
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
7
|
Zhang S, Zhang Y, Liu H, Wu F, Wang Z, Li L, Huang H, Qiu S, Li Y. Enriched environment remodels the central immune environment and improves the prognosis of acute ischemic stroke in elderly mice with chronic ischemia. Front Immunol 2023; 14:1114596. [PMID: 36969204 PMCID: PMC10033834 DOI: 10.3389/fimmu.2023.1114596] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
With the aging of many populations, cognitive and motor dysfunction caused by ischemic stroke (IS) secondary to long-term chronic cerebral ischemia presents a global problem. Enriched environment (EE), a classic paradigm of environment response and genetic interaction, has shown tremendous influence on the brain. This research aimed to investigate the potential effect of EE on cognitive and motor function in mice with chronic cerebral ischemia and secondary IS. In the chronic cerebral hypoperfusion (CCH) phase, EE treatment improved behavior performance by alleviating neuronal loss and white matter myelin damage, promoting the expression of brain-derived neurotrophic factor (BDNF) and phosphor-cAMP response element binding protein (p-CREB). Furthermore, infiltration of microglia/macrophages and astrocytes was inhibited, and the levels of IL-1β and TNFα were decreased. In the IS phase, EE altered the neuronal outcome on day 21 but not on day one after IS. In addition, EE inhibited IS-induced infiltration of microglia/macrophages and astrocytes, mediated the polarization of microglia/macrophages, and reduced pro-inflammatory factors. Importantly, EE improved IS-induced cognitive and motor deficits on day 21. Collectively, our work demonstrates that EE protects mice from cognitive and motor dysfunction and inhibits neuroinflammation caused by CCH and IS.
Collapse
Affiliation(s)
- Shehong Zhang
- Department of Rehabilitation Medicine, Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
- Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuromodulation, Huzhou, China
| | - Yonggang Zhang
- Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuromodulation, Huzhou, China
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - He Liu
- Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuromodulation, Huzhou, China
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Fengfeng Wu
- Department of Rehabilitation Medicine, Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Zhihong Wang
- Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuromodulation, Huzhou, China
| | - Liqin Li
- Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuromodulation, Huzhou, China
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Huilian Huang
- Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuromodulation, Huzhou, China
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Sheng Qiu
- Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuromodulation, Huzhou, China
- *Correspondence: Sheng Qiu, ; Yuntao Li,
| | - Yuntao Li
- Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuromodulation, Huzhou, China
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
- *Correspondence: Sheng Qiu, ; Yuntao Li,
| |
Collapse
|
8
|
Hao L, Yang Y, Xu X, Guo X, Zhan Q. Modulatory effects of mesenchymal stem cells on microglia in ischemic stroke. Front Neurol 2023; 13:1073958. [PMID: 36742051 PMCID: PMC9889551 DOI: 10.3389/fneur.2022.1073958] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/28/2022] [Indexed: 01/20/2023] Open
Abstract
Ischemic stroke accounts for 70-80% of all stroke cases. Immunity plays an important role in the pathophysiology of ischemic stroke. Microglia are the first line of defense in the central nervous system. Microglial functions are largely dependent on their pro-inflammatory (M1-like) or anti-inflammatory (M2-like) phenotype. Modulating neuroinflammation via targeting microglia polarization toward anti-inflammatory phenotype might be a novel treatment for ischemic stroke. Mesenchymal stem cells (MSC) and MSC-derived extracellular vesicles (MSC-EVs) have been demonstrated to modulate microglia activation and phenotype polarization. In this review, we summarize the physiological characteristics and functions of microglia in the healthy brain, the activation and polarization of microglia in stroke brain, the effects of MSC/MSC-EVs on the activation of MSC in vitro and in vivo, and possible underlying mechanisms, providing evidence for a possible novel therapeutics for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Lei Hao
- Department of Neurology, The First Branch of The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Yongtao Yang
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Xiaoli Xu
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Xiuming Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Xiuming Guo ✉
| | - Qunling Zhan
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, China,Qunling Zhan ✉
| |
Collapse
|
9
|
Tan N, Xin W, Huang M, Mao Y. Mesenchymal stem cell therapy for ischemic stroke: Novel insight into the crosstalk with immune cells. Front Neurol 2022; 13:1048113. [PMID: 36425795 PMCID: PMC9679024 DOI: 10.3389/fneur.2022.1048113] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/17/2022] [Indexed: 09/29/2023] Open
Abstract
Stroke, a cerebrovascular accident, is prevalent and the second highest cause of death globally across patient populations; it is as a significant cause of morbidity and mortality. Mesenchymal stem cell (MSC) transplantation is emerging as a promising treatment for alleviating neurological deficits, as indicated by a great number of animal and clinical studies. The potential of regulating the immune system is currently being explored as a therapeutic target after ischemic stroke. This study will discuss recent evidence that MSCs can harness the immune system by interacting with immune cells to boost neurologic recovery effectively. Moreover, a notion will be given to MSCs participating in multiple pathological processes, such as increasing cell survival angiogenesis and suppressing cell apoptosis and autophagy in several phases of ischemic stroke, consequently promoting neurological function recovery. We will conclude the review by highlighting the clinical opportunities for MSCs by reviewing the safety, feasibility, and efficacy of MSCs therapy.
Collapse
Affiliation(s)
- Nana Tan
- Department of Health Management, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenqiang Xin
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Min Huang
- Department of Health Management, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuling Mao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
10
|
Wang J, Huang D, Ren H, Shang L. Biomimic Trained Immunity-MSCs Delivery Microcarriers for Acute Liver Failure Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200858. [PMID: 35411651 DOI: 10.1002/smll.202200858] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/03/2022] [Indexed: 06/14/2023]
Abstract
Mesenchymal stem cells (MSCs) have a demonstrated value for acute liver failure (ALF) regeneration, while their delivery stratagems with long-term biological functions, low immune response, and high biocompatibility are still a challenge. Here, a lipopolysaccharide (LPS)-loaded photoresponsive cryogel porous microcarrier (CPM) for MSCs delivery and colonization is presented to promote defect liver regeneration. The CPMs are fabricated with graphene oxide, poly(N-isopropylacrylamide), and gelatin methacrylate (GelMA) via droplet microfluidic technology and a gradient-cooling procedure. Benefitting from the biocompatible GelMA component and the porous microstructure of the CPMs, MSCs can be nondestructively captured and abundantly delivered. Because the LPS can be released from the CPMs under NIR irradiation, the delivered MSCs are imparted with the feature of "trained immunity." Thus, when the MSCs-laden CPMs are tailored into the liver matched patches by bioprinting and applied in ALF rats, they display superior anti-inflammatory and more significant liver regeneration properties than the untrained MSCs. These features make the CPMs an excellent MSCs delivery system for clinical applications in tissue repair.
Collapse
Affiliation(s)
- Jinglin Wang
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Hepatobiliary Institute of Nanjing University, Nanjing, 210008, China
| | - Danqing Huang
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Hepatobiliary Institute of Nanjing University, Nanjing, 210008, China
| | - Haozhen Ren
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Hepatobiliary Institute of Nanjing University, Nanjing, 210008, China
| | - Luoran Shang
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Hepatobiliary Institute of Nanjing University, Nanjing, 210008, China
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| |
Collapse
|
11
|
Funes SC, Rios M, Fernández-Fierro A, Di Genaro MS, Kalergis AM. Trained Immunity Contribution to Autoimmune and Inflammatory Disorders. Front Immunol 2022; 13:868343. [PMID: 35464438 PMCID: PMC9028757 DOI: 10.3389/fimmu.2022.868343] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
A dysregulated immune response toward self-antigens characterizes autoimmune and autoinflammatory (AIF) disorders. Autoantibodies or autoreactive T cells contribute to autoimmune diseases, while autoinflammation results from a hyper-functional innate immune system. Aside from their differences, many studies suggest that monocytes and macrophages (Mo/Ma) significantly contribute to the development of both types of disease. Mo/Ma are innate immune cells that promote an immune-modulatory, pro-inflammatory, or repair response depending on the microenvironment. However, understanding the contribution of these cells to different immune disorders has been difficult due to their high functional and phenotypic plasticity. Several factors can influence the function of Mo/Ma under the landscape of autoimmune/autoinflammatory diseases, such as genetic predisposition, epigenetic changes, or infections. For instance, some vaccines and microorganisms can induce epigenetic changes in Mo/Ma, modifying their functional responses. This phenomenon is known as trained immunity. Trained immunity can be mediated by Mo/Ma and NK cells independently of T and B cell function. It is defined as the altered innate immune response to the same or different microorganisms during a second encounter. The improvement in cell function is related to epigenetic and metabolic changes that modify gene expression. Although the benefits of immune training have been highlighted in a vaccination context, the effects of this type of immune response on autoimmunity and chronic inflammation still remain controversial. Induction of trained immunity reprograms cellular metabolism in hematopoietic stem cells (HSCs), transmitting a memory-like phenotype to the cells. Thus, trained Mo/Ma derived from HSCs typically present a metabolic shift toward glycolysis, which leads to the modification of the chromatin architecture. During trained immunity, the epigenetic changes facilitate the specific gene expression after secondary challenge with other stimuli. Consequently, the enhanced pro-inflammatory response could contribute to developing or maintaining autoimmune/autoinflammatory diseases. However, the prediction of the outcome is not simple, and other studies propose that trained immunity can induce a beneficial response both in AIF and autoimmune conditions by inducing anti-inflammatory responses. This article describes the metabolic and epigenetic mechanisms involved in trained immunity that affect Mo/Ma, contraposing the controversial evidence on how it may impact autoimmune/autoinflammation conditions.
Collapse
Affiliation(s)
- Samanta C. Funes
- Instituto Multidisciplinario de Investigaciones Biológicas-San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de San Luis (UNSL), San Luis, Argentina
| | - Mariana Rios
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ayleen Fernández-Fierro
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María S. Di Genaro
- Instituto Multidisciplinario de Investigaciones Biológicas-San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de San Luis (UNSL), San Luis, Argentina
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Alexis M. Kalergis,
| |
Collapse
|
12
|
Carloni E, Ramos A, Hayes LN. Developmental Stressors Induce Innate Immune Memory in Microglia and Contribute to Disease Risk. Int J Mol Sci 2021; 22:13035. [PMID: 34884841 PMCID: PMC8657756 DOI: 10.3390/ijms222313035] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/26/2022] Open
Abstract
Many types of stressors have an impact on brain development, function, and disease susceptibility including immune stressors, psychosocial stressors, and exposure to drugs of abuse. We propose that these diverse developmental stressors may utilize a common mechanism that underlies impaired cognitive function and neurodevelopmental disorders such as schizophrenia, autism, and mood disorders that can develop in later life as a result of developmental stressors. While these stressors are directed at critical developmental windows, their impacts are long-lasting. Immune activation is a shared pathophysiology across several different developmental stressors and may thus be a targetable treatment to mitigate the later behavioral deficits. In this review, we explore different types of prenatal and perinatal stressors and their contribution to disease risk and underlying molecular mechanisms. We highlight the impact of developmental stressors on microglia biology because of their early infiltration into the brain, their critical role in brain development and function, and their long-lived status in the brain throughout life. Furthermore, we introduce innate immune memory as a potential underlying mechanism for developmental stressors' impact on disease. Finally, we highlight the molecular and epigenetic reprogramming that is known to underlie innate immune memory and explain how similar molecular mechanisms may be at work for cells to retain a long-term perturbation after exposure to developmental stressors.
Collapse
Affiliation(s)
- Elisa Carloni
- Department of Molecular and Cellular Biology, Dartmouth College, Hanover, NH 03755, USA;
| | - Adriana Ramos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA;
| | - Lindsay N. Hayes
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
13
|
Lajqi T, Braun M, Kranig SA, Frommhold D, Pöschl J, Hudalla H. LPS Induces Opposing Memory-like Inflammatory Responses in Mouse Bone Marrow Neutrophils. Int J Mol Sci 2021; 22:ijms22189803. [PMID: 34575963 PMCID: PMC8469609 DOI: 10.3390/ijms22189803] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/23/2021] [Accepted: 09/07/2021] [Indexed: 01/03/2023] Open
Abstract
A growing body of evidence suggests that innate immune cells can respond in a memory-like (adaptive) fashion, which is referred to as trained immunity. Only few in vivo studies have shown training effects in neutrophils; however, no in vitro setup has been established to study the induction of trained immunity or tolerance in neutrophils by microbial agents. In light of their short lifespan (up to 48 h), we suggest to use the term trained sensitivity for neutrophils in an in vitro setting. Here, we firstly describe a feasible two-hit model, using different doses of lipopolysaccharide (LPS) in bone marrow neutrophils. We found that low doses (10 pg/mL) induce pro-inflammatory activation (trained sensitivity), whereas priming with high doses (100 ng/mL) leads to suppression of pro-inflammatory mediators such as TNF-α or IL-6 (tolerance) (p < 0.05). On a functional level, trained neutrophils displayed increased phagocytic activity and LFA-1 expression as well as migrational capacity and CD11a expression, whereas tolerant neutrophils show contrasting effects in vitro. Mechanistically, TLR4/MyD88/PI3Ks regulate the activation of p65, which controls memory-like responses in mouse bone marrow neutrophils (p < 0.05). Our results open a new window for further in vitro studies on memory-like inflammatory responses of short-lived innate immune cells such as neutrophils.
Collapse
Affiliation(s)
- Trim Lajqi
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (M.B.); (S.A.K.); (D.F.); (J.P.)
| | - Maylis Braun
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (M.B.); (S.A.K.); (D.F.); (J.P.)
| | - Simon Alexander Kranig
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (M.B.); (S.A.K.); (D.F.); (J.P.)
| | - David Frommhold
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (M.B.); (S.A.K.); (D.F.); (J.P.)
- Klinik für Kinderheilkunde und Jugendmedizin, D-87700 Memmingen, Germany
| | - Johannes Pöschl
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (M.B.); (S.A.K.); (D.F.); (J.P.)
| | - Hannes Hudalla
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (M.B.); (S.A.K.); (D.F.); (J.P.)
- Correspondence: ; Tel.: +49-6221-56-1983; Fax: +49-6221-56-5626
| |
Collapse
|
14
|
Xin WQ, Wei W, Pan YL, Cui BL, Yang XY, Bähr M, Doeppner TR. Modulating poststroke inflammatory mechanisms: Novel aspects of mesenchymal stem cells, extracellular vesicles and microglia. World J Stem Cells 2021; 13:1030-1048. [PMID: 34567423 PMCID: PMC8422926 DOI: 10.4252/wjsc.v13.i8.1030] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/25/2021] [Accepted: 08/06/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammation plays an important role in the pathological process of ischemic stroke, and systemic inflammation affects patient prognosis. As resident immune cells in the brain, microglia are significantly involved in immune defense and tissue repair under various pathological conditions, including cerebral ischemia. Although the differentiation of M1 and M2 microglia is certainly oversimplified, changing the activation state of microglia appears to be an intriguing therapeutic strategy for cerebral ischemia. Recent evidence indicates that both mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles (EVs) regulate inflammation and modify tissue repair under preclinical stroke conditions. However, the precise mechanisms of these signaling pathways, especially in the context of the mutual interaction between MSCs or MSC-derived EVs and resident microglia, have not been sufficiently unveiled. Hence, this review summarizes the state-of-the-art knowledge on MSC- and MSC-EV-mediated regulation of microglial activity under ischemic stroke conditions with respect to various signaling pathways, including cytokines, neurotrophic factors, transcription factors, and microRNAs.
Collapse
Affiliation(s)
- Wen-Qiang Xin
- Department of Neurology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Wei Wei
- Department of Neurology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Yong-Li Pan
- Department of Neurology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Bao-Long Cui
- Department of Neurology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Xin-Yu Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Thorsten R Doeppner
- Department of Neurology, University Medical Center Göttingen, Göttingen 37075, Germany
| |
Collapse
|
15
|
Magadán S, Mikelez-Alonso I, Borrego F, González-Fernández Á. Nanoparticles and trained immunity: Glimpse into the future. Adv Drug Deliv Rev 2021; 175:113821. [PMID: 34087325 DOI: 10.1016/j.addr.2021.05.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/27/2021] [Accepted: 05/29/2021] [Indexed: 12/17/2022]
Abstract
Emerging evidences show that innate immune cells can display changes in their functional programs after infection or vaccination, which lead to immunomodulation (increased or reduced responsiveness) upon secondary activation to the same stimuli or even to a different one. Innate cells acquire features of immunological memory, nowadays using the new term of "trained immunity" or "innate immune memory", which is different from the specific memory immune response elicited by B and T lymphocytes. The review focused on the concept of trained immunity, mostly on myeloid cells. Special attention is dedicated to the pathogen recognition along the evolution (bacteria, plants, invertebrate and vertebrate animals), and to techniques used to study epigenetic reprogramming and metabolic rewiring. Nanomaterials can be recognized by immune cells offering a very promising way to learn about trained immunity. Nanomaterials could be modified in order to immunomodulate the responses ad hoc. Many therapeutic possibilities are opened, and they should be explored.
Collapse
|
16
|
Ahmed A, Rakshit S, Adiga V, Dias M, Dwarkanath P, D'Souza G, Vyakarnam A. A century of BCG: Impact on tuberculosis control and beyond. Immunol Rev 2021; 301:98-121. [PMID: 33955564 DOI: 10.1111/imr.12968] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/13/2021] [Accepted: 03/13/2021] [Indexed: 12/14/2022]
Abstract
BCG turns 100 this year and while it might not be the perfect vaccine, it has certainly contributed significantly towards eradication and prevention of spread of tuberculosis (TB). The search for newer and better vaccines for TB is an ongoing endeavor and latest results from trials of candidate TB vaccines such as M72AS01 look promising. However, recent encouraging data from BCG revaccination trials in adults combined with studies on mucosal and intravenous routes of BCG vaccination in non-human primate models have renewed interest in BCG for TB prevention. In addition, several well-demonstrated non-specific effects of BCG, for example, prevention of viral and respiratory infections, give BCG an added advantage. Also, BCG vaccination is currently being widely tested in human clinical trials to determine whether it protects against SARS-CoV-2 infection and/or death with detailed analyses and outcomes from several ongoing trials across the world awaited. Through this review, we attempt to bring together information on various aspects of the BCG-induced immune response, its efficacy in TB control, comparison with other candidate TB vaccines and strategies to improve its efficiency including revaccination and alternate routes of administration. Finally, we discuss the future relevance of BCG use especially in light of its several heterologous benefits.
Collapse
Affiliation(s)
- Asma Ahmed
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Srabanti Rakshit
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Vasista Adiga
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Mary Dias
- Division of Infectious Diseases, St John's Research Institute, Bangalore, India
| | | | - George D'Souza
- Division of Infectious Diseases, St John's Research Institute, Bangalore, India.,Department of Pulmonary Medicine, St John's Medical College, Bangalore, India
| | - Annapurna Vyakarnam
- Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India.,Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, Guy's Hospital, King's College London, London, UK
| |
Collapse
|
17
|
Feng Y, Guo M, Zhao H, Han S, Hao Y, Yuan Y, Shen W, Sun J, Dong Q, Cui M. Dl-3-n-Butylphthalide Alleviates Demyelination and Improves Cognitive Function by Promoting Mitochondrial Dynamics in White Matter Lesions. Front Aging Neurosci 2021; 13:632374. [PMID: 33762923 PMCID: PMC7982723 DOI: 10.3389/fnagi.2021.632374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/02/2021] [Indexed: 11/30/2022] Open
Abstract
White matter lesions (WMLs) are a type of cerebrovascular disorder accompanied by demyelination and cognitive decline. Dl-3-n-butylphthalide (D1-NBP) is a neuroprotective drug used for the treatment of ischemic cerebrovascular diseases, although the function of DI-NBP on WML is still not clear. This study aims to investigate whether DI-NBP affects cognitive function and ameliorates demyelination in a model of WML. The bilateral carotid artery stenosis (BCAS) mouse model and in vitro brain slice cultures with low glucose and low oxygen (LGLO) treatment were adopted. The Dl-NBP was administered intragastrically for 28 days after BCAS or added at a dose of 50 μm for 48 h after LGLO. Spatial learning and memory were evaluated by an eight-arm radial maze. Demyelination was detected using a TEM. Mitochondrial dynamics were assessed by time-lapse imaging in the cultured brain slices. The function of the synapse was evaluated by the patch clamp technique. In BCAS mice, obvious demyelination and cognitive decline were observed, while both were significantly relieved by a high-dose D1-NBP treatment (100 mg/kg). Along with demyelination, mitochondrial accumulation in the axons was significantly increased in the BCAS mice model, but with the treatment of a high-dose D1-NBP, mitochondrial accumulation was mitigated, and the anterograde/retrograde transport of mitochondria was increased. Following the improved anterograde/retrograde transport of mitochondria, the synapse activity was significantly upregulated while the reactive oxygen species (ROS) generation was remarkably decreased in the cultured brain slices. In addition, we identified syntaphilin (SNPH) as the downstream target of D1-NBP. The overexpression of SNPH mediated the effects of D1-NBP in mitigating axonal mitochondrial accumulation. In conclusion, the D1-NBP treatment significantly relieved demyelination and improved spatial learning and memory in the WML model by promoting mitochondrial dynamics. These neuroprotective effects of D1-NBP were mediated by inhibiting the mitochondrial arching protein, SNPH, which provided a potential therapeutic target for WML.
Collapse
Affiliation(s)
- Yiwei Feng
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Min Guo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongchen Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Sida Han
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yining Hao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiwen Yuan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Weiwei Shen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian Sun
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Liu H, Reiter S, Zhou X, Chen H, Ou Y, Lenahan C, He Y. Insight Into the Mechanisms and the Challenges on Stem Cell-Based Therapies for Cerebral Ischemic Stroke. Front Cell Neurosci 2021; 15:637210. [PMID: 33732111 PMCID: PMC7959708 DOI: 10.3389/fncel.2021.637210] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/03/2021] [Indexed: 01/01/2023] Open
Abstract
Strokes are the most common types of cerebrovascular disease and remain a major cause of death and disability worldwide. Cerebral ischemic stroke is caused by a reduction in blood flow to the brain. In this disease, two major zones of injury are identified: the lesion core, in which cells rapidly progress toward death, and the ischemic penumbra (surrounding the lesion core), which is defined as hypoperfusion tissue where cells may remain viable and can be repaired. Two methods that are approved by the Food and Drug Administration (FDA) include intravenous thrombolytic therapy and endovascular thrombectomy, however, the narrow therapeutic window poses a limitation, and therefore a low percentage of stroke patients actually receive these treatments. Developments in stem cell therapy have introduced renewed hope to patients with ischemic stroke due to its potential effect for reversing the neurological sequelae. Over the last few decades, animal tests and clinical trials have been used to treat ischemic stroke experimentally with various types of stem cells. However, several technical and ethical challenges must be overcome before stem cells can become a choice for the treatment of stroke. In this review, we summarize the mechanisms, processes, and challenges of using stem cells in stroke treatment. We also discuss new developing trends in this field.
Collapse
Affiliation(s)
- Huiyong Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sydney Reiter
- Department of Kinesiology, University of Texas at Austin, Austin, TX, United States
| | - Xiangyue Zhou
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hanmin Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yibo Ou
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cameron Lenahan
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| | - Yue He
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Hou K, Li G, Yu J, Xu K, Wu W. Receptors, Channel Proteins, and Enzymes Involved in Microglia-mediated Neuroinflammation and Treatments by Targeting Microglia in Ischemic Stroke. Neuroscience 2021; 460:167-180. [PMID: 33609636 DOI: 10.1016/j.neuroscience.2021.02.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 12/12/2022]
Abstract
Stroke is the largest contributor to global neurological disability-adjusted life-years, posing a huge economic and social burden to the world. Though pharmacological recanalization with recombinant tissue plasminogen activator and mechanical thrombectomy have greatly improved the prognosis of patients with ischemic stroke, clinically, there is still no effective treatment for the secondary injury caused by cerebral ischemia. In recent years, more and more evidences show that neuroinflammation plays a pivotal role in the pathogenesis and progression of ischemic cerebral injury. Microglia are brain resident innate immune cells and act the role peripheral macrophages. They play critical roles in mediating neuroinflammation after ischemic stroke. Microglia-mediated neuroinflammation is not an isolated process and has complex relationships with other pathophysiological processes as oxidative/nitrative stress, excitotoxicity, necrosis, apoptosis, pyroptosis, autophagy, and adaptive immune response. Upon activation, microglia differentially express various receptors, channel proteins, and enzymes involved in promoting or inhibiting the inflammatory processes, making them the targets of intervention for ischemic stroke. To inhibit microglia-related neuroinflammation and promote neurological recovery after ischemic stroke, numerous biochemical agents, cellular therapies, and physical methods have been demonstrated to have therapeutic potentials. Though accumulating experimental evidences have demonstrated that targeting microglia is a promising approach in the treatment of ischemic stroke, the clinical progress is slow. Till now, no clinical study could provide convincing evidence that any biochemical or physical therapies could exert neuroprotective effect by specifically targeting microglia following ischemic stroke.
Collapse
Affiliation(s)
- Kun Hou
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Guichen Li
- Department of Neurology, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Jinlu Yu
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Kan Xu
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Wei Wu
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| |
Collapse
|
20
|
Feng Y, Guo M, Zhao H, Han S, Dong Q, Cui M. Mesenchymal-Stem-Cell-Derived Extracellular Vesicles Mitigate Trained Immunity in the Brain. Front Bioeng Biotechnol 2020; 8:599058. [PMID: 33330429 PMCID: PMC7710545 DOI: 10.3389/fbioe.2020.599058] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
Trained immunity was recently discovered in innate immune cells and shown to facilitate the clearance of pathogens at the time of occurrence of the second insult. However, it exacerbates several aspects of neuropathologies, and proper therapy is needed to rectify this abnormal immune reaction. Mesenchymal-stem cells (MSCs) exhibit a distinct capability for brain repair but are associated with safety concerns. Extracellular vesicles derived from MSCs are a promising alternative therapy. In this study, we used lipopolysaccharides to activate trained immunity in the brain and examined the therapeutic potential of MSC-derived extracellular vesicles in mitigating the trained-immunity-induced exacerbated neuropathology. We found that MSC-derived extracellular vesicles showed comparable effects to those of MSCs in the mitigation of trained immunity in the brain. Moreover, the administration of MCS-derived extracellular vesicles mitigated the aggregated inflammatory responses in the acute stage of stroke and alleviated the trained-immunity-induced increased load of amyloid-β in APP/PS1 mice. We further investigated the molecular machinery of MSC-derived extracellular vesicles and found that IL-10 is important for the mediation of the therapeutic potential of MSC-derived extracellular vesicles toward the alleviation of trained immunity. Our study indicates that extracellular-vesicle-based regenerative strategies might be useful to mitigate trained immunity in the brain.
Collapse
Affiliation(s)
- Yiwei Feng
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Min Guo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongchen Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Sida Han
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|