1
|
Planas AM. Role of microglia in stroke. Glia 2024; 72:1016-1053. [PMID: 38173414 DOI: 10.1002/glia.24501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024]
Abstract
Microglia play key roles in the post-ischemic inflammatory response and damaged tissue removal reacting rapidly to the disturbances caused by ischemia and working to restore the lost homeostasis. However, the modified environment, encompassing ionic imbalances, disruption of crucial neuron-microglia interactions, spreading depolarization, and generation of danger signals from necrotic neurons, induce morphological and phenotypic shifts in microglia. This leads them to adopt a proinflammatory profile and heighten their phagocytic activity. From day three post-ischemia, macrophages infiltrate the necrotic core while microglia amass at the periphery. Further, inflammation prompts a metabolic shift favoring glycolysis, the pentose-phosphate shunt, and lipid synthesis. These shifts, combined with phagocytic lipid intake, drive lipid droplet biogenesis, fuel anabolism, and enable microglia proliferation. Proliferating microglia release trophic factors contributing to protection and repair. However, some microglia accumulate lipids persistently and transform into dysfunctional and potentially harmful foam cells. Studies also showed microglia that either display impaired apoptotic cell clearance, or eliminate synapses, viable neurons, or endothelial cells. Yet, it will be essential to elucidate the viability of engulfed cells, the features of the local environment, the extent of tissue damage, and the temporal sequence. Ischemia provides a rich variety of region- and injury-dependent stimuli for microglia, evolving with time and generating distinct microglia phenotypes including those exhibiting proinflammatory or dysfunctional traits and others showing pro-repair features. Accurate profiling of microglia phenotypes, alongside with a more precise understanding of the associated post-ischemic tissue conditions, is a necessary step to serve as the potential foundation for focused interventions in human stroke.
Collapse
Affiliation(s)
- Anna M Planas
- Cerebrovascular Research Laboratory, Department of Neuroscience and Experimental Therapeutics, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
- Cerebrovascular Diseases, Area of Clinical and Experimental Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clínic, Barcelona, Spain
| |
Collapse
|
2
|
Dou X, Ji W, Dai M, Sun S, Chen R, Yang J, Long J, Ge Y, Lin Y. Spatial and temporal mapping of neuron-microglia interaction modes in acute ischemic stroke. Biochem Pharmacol 2023; 216:115772. [PMID: 37659736 DOI: 10.1016/j.bcp.2023.115772] [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: 06/07/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/04/2023]
Abstract
Ischemic stroke (IS) is a major cause of morbidity and mortality worldwide, accounting for 75-80% of all strokes. Under conditions of ischemia and hypoxia, neurons suffer damage or death, leading to a series of secondary immune reactions. Microglia, the earliest activated immune cells, can exert neurotoxic or neuroprotective effects on neurons through secretion of factors. There exists a complex interaction between neurons and microglia during this process. Moreover, the interaction between them becomes even more complex due to differences in the infarct area and reperfusion time. This review first elaborates on the differences in neuronal death modes between the ischemic core and penumbra, and then introduces the differences in microglial markers across different infarct areas with varying reperfusion time, indicating distinct functions. Finally, we focus on exploring the interaction modes between neurons and microglia in order to precisely target beneficial interactions and inhibit harmful ones, thus providing new therapeutic strategies for the treatment of IS.
Collapse
Affiliation(s)
- Xiaoke Dou
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Wei Ji
- Department of Anesthesiology, Yantai Affiliated Hospital of BinZhou Medical College, Yantai 264000, China
| | - Maosha Dai
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Shujun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China; Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Rui Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Juexi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Junhao Long
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Yangyang Ge
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China.
| | - Yun Lin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China.
| |
Collapse
|
3
|
Liang Z, Lou Y, Hao Y, Li H, Feng J, Liu S. The Relationship of Astrocytes and Microglia with Different Stages of Ischemic Stroke. Curr Neuropharmacol 2023; 21:2465-2480. [PMID: 37464832 PMCID: PMC10616922 DOI: 10.2174/1570159x21666230718104634] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 07/20/2023] Open
Abstract
Ischemic stroke is the predominant cause of severe morbidity and mortality worldwide. Post-stroke neuroinflammation has recently received increasing attention with the aim of providing a new effective treatment strategy for ischemic stroke. Microglia and astrocytes are major components of the innate immune system of the central nervous system. They can be involved in all phases of ischemic stroke, from the early stage, contributing to the first wave of neuronal cell death, to the late stage involving phagocytosis and repair. In the early stage of ischemic stroke, a vicious cycle exists between the activation of microglia and astrocytes (through astrocytic connexin 43 hemichannels), aggravating neuroinflammatory injury post-stroke. However, in the late stage of ischemic stroke, repeatedly activated microglia can induce the formation of glial scars by triggering reactive astrogliosis in the peri-infarct regions, which may limit the movement of activated microglia in reverse and restrict the diffusion of inflammation to healthy brain tissues, alleviating the neuroinflammatory injury poststroke. In this review, we elucidated the various roles of astrocytes and microglia and summarized their relationship with neuroinflammation. We also examined how astrocytes and microglia influence each other at different stages of ischemic stroke. Several potential therapeutic approaches targeting astrocytes and microglia in ischemic stroke have been reviewed. Understanding the details of astrocytemicroglia interaction processes will contribute to a better understanding of the mechanisms underlying ischemic stroke, contributing to the identification of new therapeutic interventions.
Collapse
Affiliation(s)
- Zhen Liang
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yingyue Lou
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Hui Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Songyan Liu
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
4
|
Kleinwort KJH, Degroote RL, Hirmer S, Korbonits L, Lorenz L, Scholz AM, Hauck SM, Deeg CA. Bovine Peripheral Blood Derived Lymphocyte Proteome and Secretome Show Divergent Reaction of Bovine Immune Phenotypes after Stimulation with Pokeweed Mitogen. Proteomes 2022; 10:proteomes10010007. [PMID: 35225986 PMCID: PMC8883952 DOI: 10.3390/proteomes10010007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/23/2022] [Accepted: 01/25/2022] [Indexed: 12/15/2022] Open
Abstract
We recently identified a deviant bovine immune phenotype characterized by hyperproliferation of lymphocytes after polyclonal stimulation. This phenotype was first discovered in dams that responded to PregSure BVD vaccination by producing pathological antibodies, triggering the fatal disease “bovine neonatal pancytopenia” in calves. The aim of the study was to gain deeper insights into molecular processes occurring in lymphocytes of immune phenotypes and the effect on their secretome after immune stimulation. Two discovery proteomic experiments were performed with unstimulated and Pokeweed Mitogen (PWM) stimulated lymphocytes, using label-free LC-MS/MS. In lymphocytes, 2447 proteins were quantified, and 1204 proteins were quantified in the secretome. Quantitative proteome analysis of immune deviant and control samples after PWM stimulation revealed clear differences. The increase in abundance of IL17A, IL17F, IL8, CCL5, LRRC59, and CLIC4 was higher in controls through mitogenic stimulation. In contrast, the abundance of IFNγ, IL2, IL2RA, CD83, and CD200 increased significantly more in immune deviant lymphocytes. Additional pathway enrichment analysis of differentially secreted proteins also yielded fundamental differences between the immune phenotypes. Our study provides a comprehensive dataset, which gives novel insights into proteome changes of lymphocytes from different bovine immune phenotypes. These differences point to the development of diverse immune responses of bovine immune phenotypes after immune stimulation.
Collapse
Affiliation(s)
- Kristina J. H. Kleinwort
- Department of Veterinary Sciences, LMU Munich, D-82152 Planegg, Germany; (K.J.H.K.); (R.L.D.); (S.H.); (L.K.); (L.L.)
| | - Roxane L. Degroote
- Department of Veterinary Sciences, LMU Munich, D-82152 Planegg, Germany; (K.J.H.K.); (R.L.D.); (S.H.); (L.K.); (L.L.)
| | - Sieglinde Hirmer
- Department of Veterinary Sciences, LMU Munich, D-82152 Planegg, Germany; (K.J.H.K.); (R.L.D.); (S.H.); (L.K.); (L.L.)
| | - Lucia Korbonits
- Department of Veterinary Sciences, LMU Munich, D-82152 Planegg, Germany; (K.J.H.K.); (R.L.D.); (S.H.); (L.K.); (L.L.)
| | - Lea Lorenz
- Department of Veterinary Sciences, LMU Munich, D-82152 Planegg, Germany; (K.J.H.K.); (R.L.D.); (S.H.); (L.K.); (L.L.)
| | - Armin M. Scholz
- Livestock Center of the Faculty of Veterinary Medicine, LMU Munich, D-85764 Oberschleißheim, Germany;
| | - Stefanie M. Hauck
- Research Unit Protein Science, Helmholtz Center Munich, German Research Center for Environmental Health, D-80939 Munich, Germany;
| | - Cornelia A. Deeg
- Department of Veterinary Sciences, LMU Munich, D-82152 Planegg, Germany; (K.J.H.K.); (R.L.D.); (S.H.); (L.K.); (L.L.)
- Correspondence:
| |
Collapse
|
5
|
Garcia-Bonilla L, Iadecola C, Anrather J. Inflammation and Immune Response. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
6
|
Haidar MA, Ibeh S, Shakkour Z, Reslan MA, Nwaiwu J, Moqidem YA, Sader G, Nickles RG, Babale I, Jaffa AA, Salama M, Shaito A, Kobeissy F. Crosstalk between Microglia and Neurons in Neurotrauma: An Overview of the Underlying Mechanisms. Curr Neuropharmacol 2022; 20:2050-2065. [PMID: 34856905 PMCID: PMC9886840 DOI: 10.2174/1570159x19666211202123322] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/22/2022] Open
Abstract
Microglia are the resident immune cells of the brain and play a crucial role in housekeeping and maintaining homeostasis of the brain microenvironment. Upon injury or disease, microglial cells become activated, at least partly, via signals initiated by injured neurons. Activated microglia, thereby, contribute to both neuroprotection and neuroinflammation. However, sustained microglial activation initiates a chronic neuroinflammatory response which can disturb neuronal health and disrupt communications between neurons and microglia. Thus, microglia-neuron crosstalk is critical in a healthy brain as well as during states of injury or disease. As most studies focus on how neurons and microglia act in isolation during neurotrauma, there is a need to understand the interplay between these cells in brain pathophysiology. This review highlights how neurons and microglia reciprocally communicate under physiological conditions and during brain injury and disease. Furthermore, the modes of microglia-neuron communication are exposed, focusing on cell-contact dependent signaling and communication by the secretion of soluble factors like cytokines and growth factors. In addition, it has been discussed that how microglia-neuron interactions could exert either beneficial neurotrophic effects or pathologic proinflammatory responses. We further explore how aberrations in microglia-neuron crosstalk may be involved in central nervous system (CNS) anomalies, namely traumatic brain injury (TBI), neurodegeneration, and ischemic stroke. A clear understanding of how the microglia-neuron crosstalk contributes to the pathogenesis of brain pathologies may offer novel therapeutic avenues of brain trauma treatment.
Collapse
Affiliation(s)
- Muhammad Ali Haidar
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Stanley Ibeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Zaynab Shakkour
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohammad Amine Reslan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Judith Nwaiwu
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yomna Adel Moqidem
- Biotechnology Program, School of Science and Engineering, The American University in Cairo, Cairo, Egypt
| | - Georgio Sader
- Faculty of Medicine, University of Balamand, Balamand, Lebanon
| | - Rachel G. Nickles
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Ismail Babale
- Department of Biomedical Engineering, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Aneese A. Jaffa
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| | - Mohamed Salama
- Institute of Global Health and Human Ecology (I-GHHE), The American University in Cairo, New Cairo 11835, Egypt
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
| | - Abdullah Shaito
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biomedical Engineering, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
7
|
Alia C, Cangi D, Massa V, Salluzzo M, Vignozzi L, Caleo M, Spalletti C. Cell-to-Cell Interactions Mediating Functional Recovery after Stroke. Cells 2021; 10:3050. [PMID: 34831273 PMCID: PMC8623942 DOI: 10.3390/cells10113050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022] Open
Abstract
Ischemic damage in brain tissue triggers a cascade of molecular and structural plastic changes, thus influencing a wide range of cell-to-cell interactions. Understanding and manipulating this scenario of intercellular connections is the Holy Grail for post-stroke neurorehabilitation. Here, we discuss the main findings in the literature related to post-stroke alterations in cell-to-cell interactions, which may be either detrimental or supportive for functional recovery. We consider both neural and non-neural cells, starting from astrocytes and reactive astrogliosis and moving to the roles of the oligodendrocytes in the support of vulnerable neurons and sprouting inhibition. We discuss the controversial role of microglia in neural inflammation after injury and we conclude with the description of post-stroke alterations in pyramidal and GABAergic cells interactions. For all of these sections, we review not only the spontaneous evolution in cellular interactions after ischemic injury, but also the experimental strategies which have targeted these interactions and that are inspiring novel therapeutic strategies for clinical application.
Collapse
Affiliation(s)
- Claudia Alia
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
| | - Daniele Cangi
- Department of Neurosciences, Psychology, Drugs and Child Health Area, School of Psychology, University of Florence, 50121 Florence, Italy;
| | - Verediana Massa
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
| | - Marco Salluzzo
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
- Department of Neurosciences, Psychology, Drugs and Child Health Area, School of Psychology, University of Florence, 50121 Florence, Italy;
| | - Livia Vignozzi
- Department of Biomedical Sciences, University of Padua, Viale G. Colombo 3, 35121 Padua, Italy;
| | - Matteo Caleo
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
- Department of Biomedical Sciences, University of Padua, Viale G. Colombo 3, 35121 Padua, Italy;
| | - Cristina Spalletti
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
| |
Collapse
|
8
|
Li Z, Zhang J, Halbgebauer S, Chandrasekar A, Rehman R, Ludolph A, Boeckers T, Huber-Lang M, Otto M, Roselli F, Heuvel FO. Differential effect of ethanol intoxication on peripheral markers of cerebral injury in murine blunt traumatic brain injury. BURNS & TRAUMA 2021; 9:tkab027. [PMID: 34604393 PMCID: PMC8484207 DOI: 10.1093/burnst/tkab027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 05/28/2021] [Indexed: 11/29/2022]
Abstract
Background Blood-based biomarkers have proven to be a reliable measure of the severity and outcome of traumatic brain injury (TBI) in both murine models and patients. In particular, neuron-specific enolase (NSE), neurofilament light (NFL) and S100 beta (S100B) have been investigated in the clinical setting post-injury. Ethanol intoxication (EI) remains a significant comorbidity in TBI, with 30–40% of patients having a positive blood alcohol concentration post-TBI. The effect of ethanol on blood-based biomarkers for the prognosis and diagnosis of TBI remains unclear. In this study, we investigated the effect of EI on NSE, NFL and S100B and their correlation with blood–brain barrier integrity in a murine model of TBI. Methods We used ultra-sensitive single-molecule array technology and enzyme-linked immunosorbent assay methods to measure NFL, NSE, S100B and claudin-5 concentrations in plasma 3 hours post-TBI. Results We showed that NFL, NSE and S100B were increased at 3 hours post-TBI. Interestingly, ethanol blood concentrations showed an inverse correlation with NSE but not with NFL or S100B. Claudin-5 levels were increased post-injury but no difference was detected compared to ethanol pretreatment. The increase in claudin-5 post-TBI was correlated with NFL but not with NSE or S100B. Conclusions Ethanol induces an effect on biomarker release in the bloodstream that is different from TBI not influenced by alcohol. This could be the basis of investigations into humans.
Collapse
Affiliation(s)
- Zhenghui Li
- Department of Neurology, Ulm University, ZBMF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Jin Zhang
- Department of Neurology, Ulm University, ZBMF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Steffen Halbgebauer
- Department of Neurology, Ulm University, ZBMF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Akila Chandrasekar
- Department of Neurology, Ulm University, ZBMF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Rida Rehman
- Department of Neurology, Ulm University, ZBMF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Albert Ludolph
- Department of Neurology, Ulm University, ZBMF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Tobias Boeckers
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital, ZBMF - Helmhotzstrasse 8/1, 89081 Ulm, Germany
| | - Markus Otto
- Department of Neurology, Ulm University, ZBMF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Francesco Roselli
- Department of Neurology, Ulm University, ZBMF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Florian Olde Heuvel
- Department of Neurology, Ulm University, ZBMF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| |
Collapse
|
9
|
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: 6.0] [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
|
10
|
Al Mamun A, Ngwa C, Qi S, Honarpisheh P, Datar S, Sharmeen R, Xu Y, McCullough LD, Liu F. Neuronal CD200 Signaling Is Protective in the Acute Phase of Ischemic Stroke. Stroke 2021; 52:3362-3373. [PMID: 34353112 DOI: 10.1161/strokeaha.120.032374] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE CD200 (cluster of differentiation 200), a highly glycosylated protein primarily expressed on neurons in the central nervous system, binds with its receptor CD200R to form an endogenous inhibitory signal against immune responses. However, little is known about the effect of neuronal CD200 signaling in cerebral ischemia. The aim of this study was to investigate how neuronal CD200 signaling impacts poststroke inflammation and the ischemic injury. METHODS CD200 tma1lf/fl:Thy1CreER mice were treated with tamoxifen to induce conditional gene knockout (ICKO) of neuronal CD200. The mice were subjected to a 60-minute transient middle cerebral artery occlusion. Stroke outcomes, apoptotic cell death, immune cell infiltration, microglia activation, and other inflammatory profiles were evaluated at 3 and 7 days after stroke. RESULTS Infarct volumes were significantly larger, and behavioral deficits more severe in ICKO versus control mice at 3 days after middle cerebral artery occlusion. Terminal deoxynucleotidyl transferase dUTP nick end labeling assay also revealed a significant increase in apoptotic neuronal death in CD200 ICKO mice. An enhancement in lymphocytic infiltration and microglial proinflammatory responses were revealed by flow cytometry at 3 and 7 days after stroke in ICKO mice, accompanied by an increased microglial phagocytosis activity. Plasma proinflammatory cytokine (TNFα [tumor necrosis factor alpha] and IL [interleukin]-1β) levels significantly increased at 3 days, and IL-1β/IL-6 levels increased at 7 days in ICKO versus control animals. ICKO led to significantly lower baseline level of CD200 both in brain and plasma. CONCLUSIONS Neuronal CD200 inhibits proinflammatory responses and is protective against stroke injury.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Conelius Ngwa
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Shaohua Qi
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Pedram Honarpisheh
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Saumil Datar
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Romana Sharmeen
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Yan Xu
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Louise D McCullough
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Fudong Liu
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| |
Collapse
|
11
|
Semenova E, Grudniak MP, Machaj EK, Bocian K, Chroscinska-Krawczyk M, Trochonowicz M, Stepaniec IM, Murzyn M, Zagorska KE, Boruczkowski D, Kolanowski TJ, Oldak T, Rozwadowska N. Mesenchymal Stromal Cells from Different Parts of Umbilical Cord: Approach to Comparison & Characteristics. Stem Cell Rev Rep 2021; 17:1780-1795. [PMID: 33860454 PMCID: PMC8553697 DOI: 10.1007/s12015-021-10157-3] [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] [Accepted: 03/17/2021] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) are a unique population of cells that play an important role in the regeneration potential of the body. MSCs exhibit a characteristic phenotype and are capable of modulating the immune response. MSCs can be isolated from various tissues such as: bone marrow, adipose tissue, placenta, umbilical cord and others. The umbilical cord as a source of MSCs, has strong advantages, such as no-risk procedure of tissue retrieval after birth and easiness of the MSCs isolation. As the umbilical cord (UC) is a complex organ and we decided to evaluate, whether the cells derived from different regions of umbilical cord show similar or distinct properties. In this study we characterized and compared MSCs from three regions of the umbilical cord: Wharton's Jelly (WJ), the perivascular space (PRV) and the umbilical membrane (UCM). The analysis was carried out in terms of morphology, phenotype, immunomodulation potential and secretome. Based on the obtained results, we were able to conclude, that MSCs derived from distinct UC regions differ in their properties. According to our result WJ-MSCs have high and stabile proliferation potential and phenotype, when compare with other MSCs and can be treated as a preferable source of cells for medical application.
Collapse
Affiliation(s)
- Ekaterina Semenova
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Mariusz P Grudniak
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Eugeniusz K Machaj
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Katarzyna Bocian
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland.,Faculty of Biology, Department of Immunology, University of Warsaw, Warsaw, Poland
| | | | - Marzena Trochonowicz
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Igor M Stepaniec
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Magdalena Murzyn
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland.,Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Karolina E Zagorska
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Dariusz Boruczkowski
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Tomasz J Kolanowski
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland.,Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Tomasz Oldak
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland.
| | | |
Collapse
|
12
|
Rabaneda-Lombarte N, Serratosa J, Bové J, Vila M, Saura J, Solà C. The CD200R1 microglial inhibitory receptor as a therapeutic target in the MPTP model of Parkinson's disease. J Neuroinflammation 2021; 18:88. [PMID: 33823877 PMCID: PMC8025338 DOI: 10.1186/s12974-021-02132-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 03/15/2021] [Indexed: 01/22/2023] Open
Abstract
Background It is suggested that neuroinflammation, in which activated microglial cells play a relevant role, contributes to the development of Parkinson’s disease (PD). Consequently, the modulation of microglial activation is a potential therapeutic target to be taken into account to act against the dopaminergic neurodegeneration occurring in this neurological disorder. Several soluble and membrane-associated inhibitory mechanisms contribute to maintaining microglial cells in a quiescent/surveillant phenotype in physiological conditions. However, the presence of activated microglial cells in the brain in PD patients suggests that these mechanisms have been somehow overloaded. We focused our interest on one of the membrane-associated mechanisms, the CD200-CD200R1 ligand-receptor pair. Methods The acute MPTP experimental mouse model of PD was used to study the temporal pattern of mRNA expression of CD200 and CD200R1 in the context of MPTP-induced dopaminergic neurodegeneration and neuroinflammation. Dopaminergic damage was assessed by tyrosine hydroxylase (TH) immunoreactivity, and neuroinflammation was evaluated by the mRNA expression of inflammatory markers and IBA1 and GFAP immunohistochemistry. The effect of the modulation of the CD200-CD200R1 system on MPTP-induced damage was determined by using a CD200R1 agonist or CD200 KO mice. Results MPTP administration resulted in a progressive decrease in TH-positive fibres in the striatum and TH-positive neurons in the substantia nigra pars compacta, which were accompanied by transient astrogliosis, microgliosis and expression of pro- and anti-inflammatory markers. CD200 mRNA levels rapidly decreased in the ventral midbrain after MPTP treatment, while a transient decrease of CD200R1 mRNA expression was repeatedly observed in this brain area at earlier and later phases. By contrast, a transient increase in CD200R1 expression was observed in striatum. The administration of a CD200R1 agonist resulted in the inhibition of MPTP-induced dopaminergic neurodegeneration, while microglial cells showed signs of earlier activation in CD200-deficient mice. Conclusions Collectively, these findings provide evidence for a correlation between CD200-CD200R1 alterations, glial activation and neuronal loss. CD200R1 stimulation reduces MPTP-induced loss of dopaminergic neurons, and CD200 deficiency results in earlier microglial activation, suggesting that the potentiation of CD200R1 signalling is a possible approach to controlling neuroinflammation and neuronal death in PD.
Collapse
Affiliation(s)
- Neus Rabaneda-Lombarte
- Department of Cerebral Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August-Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Biochemistry and Molecular Biology Unit, School of Medicine, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Joan Serratosa
- Department of Cerebral Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August-Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Bové
- Vall d'Hebrón Research Institute-CIBERNED, Barcelona, Spain
| | - Miquel Vila
- Vall d'Hebrón Research Institute-CIBERNED, Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Josep Saura
- Biochemistry and Molecular Biology Unit, School of Medicine, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Carme Solà
- Department of Cerebral Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August-Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
13
|
Lee TK, Shin MC, Ahn JH, Kim DW, Kim B, Sim H, Lee JC, Cho JH, Park JH, Kim YM, Won MH, Lee CH. CD200 Change Is Involved in Neuronal Death in Gerbil Hippocampal CA1 Field Following Transient Forebrain Ischemia and Postischemic Treatment with Risperidone Displays Neuroprotection without CD200 Change. Int J Mol Sci 2021; 22:ijms22031116. [PMID: 33498705 PMCID: PMC7865463 DOI: 10.3390/ijms22031116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 11/16/2022] Open
Abstract
It has been reported that CD200 (Cluster of Differentiation 200), expressed in neurons, regulates microglial activation in the central nervous system, and a decrease in CD200 expression causes an increase in microglial activation and neuronal loss. The aim of this study was to investigate time-dependent changes in CD200 expression in the hippocampus proper (CA1, 2, and 3 fields) after transient forebrain ischemia for 5 min in gerbils. In this study, 5-min ischemia evoked neuronal death (loss) of pyramidal neurons in the CA1 field, but not in the CA2/3 fields, at 5 days postischemia. In the sham group, CD200 expression was found in pyramidal neurons of the CA1 field, and the immunoreactivity in the group with ischemia was decreased at 6 h postischemia, dramatically increased at 12 h postischemia, decreased (to level found at 6 h postischemia) at 1 and 2 days postischemia, and significantly increased again at 5 days postischemia. At 5 days postischemia, CD200 immunoreactivity was strongly expressed in microglia and GABAergic neurons. However, in the CA3 field, the change in CD200 immunoreactivity in pyramidal neurons was markedly weaker than that in the CA1 field, showing there was no expression of CD 200 in microglia and GABAergic neurons. In addition, treatment of 10 mg/kg risperidone (an atypical antipsychotic drug) after the ischemia hardly changed CD200 immunoreactivity in the CA1 field, showing that CA1 pyramidal neurons were protected from the ischemic injury. These results indicate that the transient ischemia-induced change in CD200 expression may be associated with specific and selective neuronal death in the hippocampal CA1 field following transient forebrain ischemia.
Collapse
Affiliation(s)
- Tae-Kyeong Lee
- Department of Biomedical Science and Research, Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Gangwon, Korea;
| | - Myoung Cheol Shin
- Department of Emergency Medicine, Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 24289, Gangwon, Korea; (M.C.S.); (J.H.C.)
| | - Ji Hyeon Ahn
- Department of Physical Therapy, College of Health Science, Youngsan University, Yangsan 50510, Gyeongnam, Korea;
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology and Research Institute of Oral Sciences, College of Dentistry, Gangnung-Wonju National University, Gangneung 25457, Gangwon, Korea;
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Hyejin Sim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Jun Hwi Cho
- Department of Emergency Medicine, Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 24289, Gangwon, Korea; (M.C.S.); (J.H.C.)
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju 38066, Gyeongbuk, Korea;
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea;
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (B.K.); (H.S.); (J.-C.L.)
- Correspondence: (M.-H.W.); (C.-H.L.); Tel.: +82-33-250-8891 (M.-H.W.); +82-41-550-1441 (C.-H.L.); Fax: +82-33-256-1614 (M.-H.W.); +82-41-559-7899 (C.-H.L.)
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Chungnam, Korea
- Correspondence: (M.-H.W.); (C.-H.L.); Tel.: +82-33-250-8891 (M.-H.W.); +82-41-550-1441 (C.-H.L.); Fax: +82-33-256-1614 (M.-H.W.); +82-41-559-7899 (C.-H.L.)
| |
Collapse
|
14
|
HMGB1 A box protects neurons by potently inhibiting both microglia and T cell-mediated inflammation in a mouse Parkinson's disease model. Clin Sci (Lond) 2020; 134:2075-2090. [PMID: 32706028 DOI: 10.1042/cs20200553] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 11/17/2022]
Abstract
In the subacute Parkinson's disease (PD) mice model of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), injection of HMGB1 competitive inhibitor protein HMGB1 A box and the ethyl pyruvate (EP) that inhibit the release of HMGB1 from cells restored the number of dopaminergic neurons and TH+ fibers in the SN and striatum. Our data show that A box up-regulated CD200-CD200R signal of microglia inhibited the activation of microglia mediated by HMGB1, and the production of TNF-α, IL-1β and IL-6 in vivo and in vitro mixed culture system. Microglia overexpressing CD200R produced less inflammatory chemokines and reduced the loss of TH+ neurons. In addition, HMGB1 A box decreased the level of CCL5 and significantly inhibited the infiltration of almost all T cells including Th17 and the proportion of Th17 in CD4+ T cells. In vitro MPP+ induced model and HMGB1-stimulated mesencephalic cell system activated microglia induced the differentiation of naïve T cells to Th17, and A box significantly inhibited this process. To sum up, our results show that HMGB1 A box targeting HMGB1, which effectively reduces the activation of microglia in MPTP PD model by restoring CD200-CD200R signal inhibit microglia mediated neuroinflammation and the differentiation of T cells to Th17.
Collapse
|
15
|
Iadecola C, Buckwalter MS, Anrather J. Immune responses to stroke: mechanisms, modulation, and therapeutic potential. J Clin Invest 2020; 130:2777-2788. [PMID: 32391806 PMCID: PMC7260029 DOI: 10.1172/jci135530] [Citation(s) in RCA: 426] [Impact Index Per Article: 85.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Stroke is the second leading cause of death worldwide and a leading cause of disability. Most strokes are caused by occlusion of a major cerebral artery, and substantial advances have been made in elucidating how ischemia damages the brain. In particular, increasing evidence points to a double-edged role of the immune system in stroke pathophysiology. In the acute phase, innate immune cells invade brain and meninges and contribute to ischemic damage, but may also be protective. At the same time, danger signals released into the circulation by damaged brain cells lead to activation of systemic immunity, followed by profound immunodepression that promotes life-threatening infections. In the chronic phase, antigen presentation initiates an adaptive immune response targeted to the brain, which may underlie neuropsychiatric sequelae, a considerable cause of poststroke morbidity. Here, we briefly review these pathogenic processes and assess the potential therapeutic value of targeting immunity in human stroke.
Collapse
Affiliation(s)
- Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Marion S. Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University Medical Center, Stanford, California, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
16
|
Chamera K, Trojan E, Szuster-Głuszczak M, Basta-Kaim A. The Potential Role of Dysfunctions in Neuron-Microglia Communication in the Pathogenesis of Brain Disorders. Curr Neuropharmacol 2020; 18:408-430. [PMID: 31729301 PMCID: PMC7457436 DOI: 10.2174/1570159x17666191113101629] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/15/2019] [Accepted: 11/10/2019] [Indexed: 12/18/2022] Open
Abstract
The bidirectional communication between neurons and microglia is fundamental for the proper functioning of the central nervous system (CNS). Chemokines and clusters of differentiation (CD) along with their receptors represent ligand-receptor signalling that is uniquely important for neuron - microglia communication. Among these molecules, CX3CL1 (fractalkine) and CD200 (OX-2 membrane glycoprotein) come to the fore because of their cell-type-specific localization. They are principally expressed by neurons when their receptors, CX3CR1 and CD200R, respectively, are predominantly present on the microglia, resulting in the specific axis which maintains the CNS homeostasis. Disruptions to this balance are suggested as contributors or even the basis for many neurological diseases. In this review, we discuss the roles of CX3CL1, CD200 and their receptors in both physiological and pathological processes within the CNS. We want to underline the critical involvement of these molecules in controlling neuron - microglia communication, noting that dysfunctions in their interactions constitute a key factor in severe neurological diseases, such as schizophrenia, depression and neurodegeneration-based conditions.
Collapse
Affiliation(s)
- Katarzyna Chamera
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St. 31-343Kraków, Poland
| | - Ewa Trojan
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St. 31-343Kraków, Poland
| | - Magdalena Szuster-Głuszczak
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St. 31-343Kraków, Poland
| | - Agnieszka Basta-Kaim
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St. 31-343Kraków, Poland
| |
Collapse
|
17
|
Ai QD, Chen C, Chu S, Zhang Z, Luo Y, Guan F, Lin M, Liu D, Wang S, Chen N. IMM-H004 therapy for permanent focal ischemic cerebral injury via CKLF1/CCR4-mediated NLRP3 inflammasome activation. Transl Res 2019; 212:36-53. [PMID: 31176667 DOI: 10.1016/j.trsl.2019.05.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/10/2019] [Accepted: 05/24/2019] [Indexed: 01/07/2023]
Abstract
Chemokine-like factor 1 (CKLF1) is a potential target for ischemic stroke therapy. The NOD-like receptor protein 3 (NLRP3) inflammasome has been postulated to mediate inflammatory responses during ischemic/reperfusion (I/R) injury. The compound IMM-H004 is a novel coumarin derivative that can improve cerebral I/R injury. This study aims to investigate the effects of IMM-H004 on ischemia stroke injury and further elucidate the molecular mechanisms. The standard pMCAO model of focal ischemia was used in this paper. Drugs were administered at 6 hours after ischemia, and behavioral assessment, euthanasia, and outcome measures were evaluated at 9 hours after ischemia. The effects of IMM-H004 on ischemic stroke injury were determined using 2,3,5-triphenyltetrazolium chloride (TTC) staining, behavioral tests, enzyme-linked immunosorbent assay (ELISA), and Nissl staining. Immunohistologic staining, immunofluorescence staining, quantitative RT-PCR (qPCR), western blotting, and coimmunoprecipitation (CO-IP) assays were used to elucidate the underlying mechanisms. IMM-H004 treatment provided significant protection against ischemia stroke through a CKLF1-dependent anti-inflammatory pathway in rats. IMM-H004 downregulated the amount of CKLF1 binding with C-C chemokine receptor type 4, further suppressing the activation of NLRP3 inflammasome and the following inflammatory response, ultimately protecting the ischemic brain. This preclinical study established the efficacy of IMM-H004 as a potential therapeutic medicine for permanent cerebral ischemia. These results support further efforts to develop IMM-H004 for human clinical trials in acute cerebral ischemia, particularly for patients who are not suitable for reperfusion therapy.
Collapse
Affiliation(s)
- Q D Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces and Hunan University of Chinese Medicine First-Class Disciple Construction Project of Chinese Materia Medica, Changsha, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Feifei Guan
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Peking Union Medicine College and Chinese Academy of Medical Sciences, Beijing, China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces and Hunan University of Chinese Medicine First-Class Disciple Construction Project of Chinese Materia Medica, Changsha, China
| | - Dandan Liu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shasha Wang
- School of Basic Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces and Hunan University of Chinese Medicine First-Class Disciple Construction Project of Chinese Materia Medica, Changsha, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
18
|
Sauter IP, Madrid KG, de Assis JB, Sá-Nunes A, Torrecilhas AC, Staquicini DI, Pasqualini R, Arap W, Cortez M. TLR9/MyD88/TRIF signaling activates host immune inhibitory CD200 in Leishmania infection. JCI Insight 2019; 4:126207. [PMID: 31092731 DOI: 10.1172/jci.insight.126207] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 04/11/2019] [Indexed: 02/06/2023] Open
Abstract
Virulent protozoans named Leishmania in tropical and subtropical areas produce devastating diseases by exploiting host immune responses. Amastigotes of Leishmania amazonensis stimulate macrophages to express CD200, an immunomodulatory ligand, which binds to its cognate receptor (CD200R) and inhibits the inducible nitric oxide synthase and nitric oxide (iNOS/NO) signaling pathways, thereby promoting intracellular survival. However, the mechanisms underlying CD200 induction in macrophages remain largely unknown. Here, we show that phagocytosis-mediated internalization of L. amazonensis amastigotes following activation of endosomal TLR9/MyD88/TRIF signaling is critical for inducing CD200 in infected macrophages. We also demonstrate that Leishmania microvesicles containing DNA fragments activate TLR9-dependent CD200 expression, which inhibits the iNOS/NO pathway and modulates the course of L. amazonensis infection in vivo. These findings demonstrate that Leishmania exploits TLR-signaling pathways not only to inhibit macrophage microbicidal function, but also to evade host systemic immune responses, which has many implications in the severity of the disease.
Collapse
Affiliation(s)
| | | | - Josiane B de Assis
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Anderson Sá-Nunes
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana C Torrecilhas
- Department of Pharmaceutical Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Daniela I Staquicini
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey and Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | | |
Collapse
|
19
|
Xie YZ, Zhang XJ, Zhang C, Yang Y, He JN, Chen YX. Protective effects of leonurine against ischemic stroke in mice by activating nuclear factor erythroid 2-related factor 2 pathway. CNS Neurosci Ther 2019; 25:1006-1017. [PMID: 31087454 PMCID: PMC6698971 DOI: 10.1111/cns.13146] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022] Open
Abstract
Aims Leonurine has been shown to trigger antioxidant responses during ischemic stroke, and nuclear factor erythroid 2‐related factor 2 (Nrf‐2) imparts protective effects against oxidative injury. The present study has determined that leonurine prevents ischemic injury of brain tissues via Nrf‐2 pathway activation. Methods Male ICR mice and Nrf‐2−/− mice were subjected to permanent middle cerebral artery occlusion (pMCAO) and received leonurine treatment at 2 hours after pMCAO by intraperitoneal injection. Neurological deficit scores as well as infarct volume were assessed to determine the neuroprotective role of leonurine. Nrf‐2 was investigated using Western blotting and real‐time polymerase chain reaction (RT‐PCR) analysis to elucidate the neuroprotective mechanism of leonurine. Commercial kits were employed to determine reactive oxygen species (ROS), superoxide (SOD), catalase (CAT), glutathione peroxidase (GSH‐Px), malonaldehyde (MDA), and glutathione (GSH). Vascular endothelial growth factor (VEGF) was evaluated by Western blotting and RT‐PCR analysis, and VEGF was localized using immunofluorescence. Results The application of leonurine on ICR mice resulted in an improvement in neurological deficit scores and a reduction in infarct volume. Leonurine upregulated nuclear Nrf‐2 protein and increased total Nrf‐2 protein expression and mRNA levels. Leonurine regulated SOD, MDA, CAT, GSH, and GSH‐Px, and it significantly inhibited ROS production in ICR mice. Leonurine improved VEGF expression and increased VEGF expression in neurons, astrocytes, and endothelial cells. However, leonurine had no obvious beneficial effects on Nrf‐2−/− mice. Conclusions Leonurine exerted neuroprotective effects, promoted antioxidant responses, and upregulated VEGF expression by activating the Nrf‐2 pathway.
Collapse
Affiliation(s)
- Yan-Zhao Xie
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China.,The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiang-Jian Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Cong Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Yang Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Jun-Na He
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Yan-Xia Chen
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China.,Department of Endocrinology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
20
|
Ritzel RM, Al Mamun A, Crapser J, Verma R, Patel AR, Knight BE, Harris N, Mancini N, Roy-O'Reilly M, Ganesh BP, Liu F, McCullough LD. CD200-CD200R1 inhibitory signaling prevents spontaneous bacterial infection and promotes resolution of neuroinflammation and recovery after stroke. J Neuroinflammation 2019; 16:40. [PMID: 30777093 PMCID: PMC6378746 DOI: 10.1186/s12974-019-1426-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/31/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Ischemic stroke results in a robust inflammatory response within the central nervous system. As the immune-inhibitory CD200-CD200 receptor 1 (CD200R1) signaling axis is a known regulator of immune homeostasis, we hypothesized that it may play a role in post-stroke immune suppression after stroke. METHODS In this study, we investigated the role of CD200R1-mediated signaling in stroke using CD200 receptor 1-deficient mice. Mice were subjected to a 60-min middle cerebral artery occlusion and evaluated at days 3 and 7, representing the respective peak and early resolution stages of neuroinflammation in this model of ischemic stroke. Infarct size and behavioral deficits were assessed at both time points. Central and peripheral cellular immune responses were measured using flow cytometry. Bacterial colonization was determined in lung tissue homogenates both after acute stroke and in an LPS model of systemic inflammation. RESULTS In wild-type (WT) animals, CD200R1 was expressed on infiltrating monocytes and lymphocytes after stroke but was absent on microglia. Early after ischemia (72 h), CD200R1-knockout (KO) mice had significantly poorer survival rates and an enhanced susceptibility to spontaneous bacterial colonization of the respiratory tract compared to wild-type (WT) controls, despite no difference in infarct or neurological deficits. While the CNS inflammation was resolved by day 7 post-stroke in WT mice, brain-resident microglia and monocyte activation persisted in CD200R1-KO mice, accompanied by a delayed, augmented lymphocyte response. At this time point, CD200R1-KO mice displayed greater weight loss, more severe neurological deficits, and impaired motor function compared to WT. Systemically, CD200R1-KO mice exhibited signs of persistent infection including lymphopenia, T cell activation and memory conversion, and narrowing of the TCR repertoire. These findings were confirmed in a second model of acute neuroinflammation induced by systemic endotoxin challenge. CONCLUSION This study defines an essential role of CD200-CD200R1 signaling in stroke. Loss of CD200R1 led to high mortality, increased rates of post-stroke infection, and enhanced entry of peripheral leukocytes into the brain after ischemia, with no increase in infarct size. This suggests that the loss of CD200 receptor leads to enhanced peripheral inflammation that is triggered by brain injury.
Collapse
Affiliation(s)
- Rodney M Ritzel
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Abdullah Al Mamun
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin Street, Houston, TX, 77370, USA
| | - Joshua Crapser
- Neuroscience Department, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Rajkumar Verma
- Neuroscience Department, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Anita R Patel
- Neuroscience Department, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Brittany E Knight
- Neuroscience Department, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Nia Harris
- Neuroscience Department, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Nickolas Mancini
- Neuroscience Department, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Meaghan Roy-O'Reilly
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin Street, Houston, TX, 77370, USA
| | - Bhanu Priya Ganesh
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin Street, Houston, TX, 77370, USA
| | - Fudong Liu
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin Street, Houston, TX, 77370, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin Street, Houston, TX, 77370, USA.
| |
Collapse
|
21
|
Szepesi Z, Manouchehrian O, Bachiller S, Deierborg T. Bidirectional Microglia-Neuron Communication in Health and Disease. Front Cell Neurosci 2018; 12:323. [PMID: 30319362 PMCID: PMC6170615 DOI: 10.3389/fncel.2018.00323] [Citation(s) in RCA: 306] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022] Open
Abstract
Microglia are ramified cells that exhibit highly motile processes, which continuously survey the brain parenchyma and react to any insult to the CNS homeostasis. Although microglia have long been recognized as a crucial player in generating and maintaining inflammatory responses in the CNS, now it has become clear, that their function are much more diverse, particularly in the healthy brain. The innate immune response and phagocytosis represent only a little segment of microglia functional repertoire that also includes maintenance of biochemical homeostasis, neuronal circuit maturation during development and experience-dependent remodeling of neuronal circuits in the adult brain. Being equipped by numerous receptors and cell surface molecules microglia can perform bidirectional interactions with other cell types in the CNS. There is accumulating evidence showing that neurons inform microglia about their status and thus are capable of controlling microglial activation and motility while microglia also modulate neuronal activities. This review addresses the topic: how microglia communicate with other cell types in the brain, including fractalkine signaling, secreted soluble factors and extracellular vesicles. We summarize the current state of knowledge of physiological role and function of microglia during brain development and in the mature brain and further highlight microglial contribution to brain pathologies such as Alzheimer’s and Parkinson’s disease, brain ischemia, traumatic brain injury, brain tumor as well as neuropsychiatric diseases (depression, bipolar disorder, and schizophrenia).
Collapse
Affiliation(s)
- Zsuzsanna Szepesi
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Oscar Manouchehrian
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sara Bachiller
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|