1
|
Komiya H, Takeuchi H, Ogasawara A, Ogawa Y, Kubota S, Hashiguchi S, Takahashi K, Kunii M, Tanaka K, Tada M, Doi H, Tanaka F. Siponimod inhibits microglial inflammasome activation. Neurosci Res 2025; 213:138-145. [PMID: 39921000 DOI: 10.1016/j.neures.2025.02.002] [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: 09/15/2024] [Revised: 01/16/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Siponimod is the first oral drug approved for active secondary progressive multiple sclerosis. It acts as a functional antagonist of sphingosine-1-phosphate (S1P) receptor 1 (S1P1) through S1P1 internalization, and also serves an agonist of S1P5; however, the detailed mechanisms of its therapeutic effects on glial cells have yet to be elucidated. In this study, we investigated the anti-inflammatory mechanism of siponimod in microglia. Pretreatment with either siponimod or the S1P1 antagonist W146 significantly suppressed the production of interleukin-1β in activated microglia stimulated with lipopolysaccharide plus nigericin, an inflammasome activator. Furthermore, siponimod treatment reduced the protein levels of cleaved caspase-1 and inhibited the formation of aggregates of apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC specks) in microglia. Our data indicate that siponimod achieves its anti-inflammatory effects by inhibiting inflammasome activation in microglia via S1P1 antagonism. This process is inferred to play a crucial role in mitigating the secondary progression of multiple sclerosis, where microglial activation in the gray matter is considered a key pathological factor.
Collapse
Affiliation(s)
- Hiroyasu Komiya
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hideyuki Takeuchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Department of Neurology, Graduate School of Medicine, International University of Health and Welfare, Narita, Japan; Center for Intractable Neurological Diseases and Dementia, International University of Health and Welfare Atami Hospital, Atami, Japan.
| | - Akihiro Ogasawara
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuki Ogawa
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shun Kubota
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shunta Hashiguchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Keita Takahashi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Misako Kunii
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kenichi Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mikiko Tada
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroshi Doi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| |
Collapse
|
2
|
Kawahara K, Hasegawa T, Hasegawa N, Izumi T, Sato K, Sakamaki T, Ando M, Maeda T. Truncated GPNMB, a microglial transmembrane protein, serves as a scavenger receptor for oligomeric β-amyloid peptide 1-42 in primary type 1 microglia. J Neurochem 2024; 168:1317-1339. [PMID: 38361142 DOI: 10.1111/jnc.16078] [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: 10/24/2023] [Revised: 01/27/2024] [Accepted: 01/31/2024] [Indexed: 02/17/2024]
Abstract
Glycoprotein non-metastatic melanoma protein B (GPNMB) is up-regulated in one subtype of microglia (MG) surrounding senile plaque depositions of amyloid-beta (Aβ) peptides. However, whether the microglial GPNMB can recognize the fibrous Aβ peptides as ligands remains unknown. In this study, we report that the truncated form of GPNMB, the antigen for 9F5, serves as a scavenger receptor for oligomeric Aβ1-42 (o-Aβ1-42) in rat primary type 1 MG. 125I-labeled o-Aβ1-42 exhibited specific and saturable endosomal/lysosomal degradation in primary-cultured type 1 MG from GPNMB-expressing wild-type mice, whereas the degradation activity was markedly reduced in cells from Gpnmb-knockout mice. The Gpnmb-siRNA significantly inhibits the degradation of 125I-o-Aβ1-42 by murine microglial MG5 cells. Therefore, GPNMB contributes to mouse MG's o-Aβ1-42 clearance. In rat primary type 1 MG, the cell surface expression of truncated GPNMB was confirmed by a flow cytometric analysis using a previously established 9F5 antibody. 125I-labeled o-Aβ1-42 underwent endosomal/lysosomal degradation by rat primary type 1 MG in a dose-dependent fashion, while the 9F5 antibody inhibited the degradation. The binding of 125I-o-Aβ1-42 to the rat primary type 1 MG was inhibited by 42% by excess unlabeled o-Aβ1-42, and by 52% by the 9F5 antibody. Interestingly, the 125I-o-Aβ1-42 degradations by MG-like cells from human-induced pluripotent stem cells was inhibited by the 9F5 antibody, suggesting that truncated GPNMB also serve as a scavenger receptor for o-Aβ1-42 in human MG. Our study demonstrates that the truncated GPNMB (the antigen for 9F5) binds to oligomeric form of Aβ1-42 and functions as a scavenger receptor on MG, and 9F5 antibody can act as a blocking antibody for the truncated GPNMB.
Collapse
Affiliation(s)
- Kohichi Kawahara
- Department of Pharmacology, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, Japan
- Department of Bio-analytical Chemistry, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, Japan
| | - Takuya Hasegawa
- Department of Pharmacology, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, Japan
| | - Noa Hasegawa
- Department of Pharmacology, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, Japan
| | - Taisei Izumi
- Department of Pharmacology, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, Japan
| | - Koji Sato
- Laboratory of Health Chemistry, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, Japan
| | - Toshiyuki Sakamaki
- Laboratory of Health Chemistry, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, Japan
| | - Masayuki Ando
- Education Center for Pharmacy, Faculty of Pharmacy, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, Japan
| | - Takehiko Maeda
- Department of Pharmacology, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, Japan
| |
Collapse
|
3
|
Rezaie P, Hanisch UK. History of Microglia. ADVANCES IN NEUROBIOLOGY 2024; 37:15-37. [PMID: 39207684 DOI: 10.1007/978-3-031-55529-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The term 'microglia' was first introduced into the scientific literature a century ago. The various eras of microglial research have been defined not only by the number of reports subsequently generated but, more critically, also by the concepts that have shaped our present-day views and understanding of microglia. Key methods, technologies, and models, as well as seminal discoveries made possible through their deployment have enabled breakthroughs, and now pave the way for lines of investigation that could not have been anticipated even a decade ago. Advances in our understanding of the microglial origin, forms, and functions have relied fundamentally on parallel developments in immunology. As the 'neuro-immune' cells of the brain, microglia are now under the spotlight in various disciplines. This chapter surveys the gradual processes and precipitous events that helped form ideas concerning the developmental origin of microglia and their roles in health and disease. It first covers the dawning phase during which the early pioneers of microglial research discovered cellular entities and already assigned functions to them. Following a recess period, the 1960s brought about a renaissance of active interest, with the development of tools and models-and fundamental notions on microglial contributions to central nervous system (CNS) pathologies. These seminal efforts laid the foundation for the awakening of a sweeping research era beginning in the 1980s and spurred on by a blast of immunological discoveries. Finally, this chapter stresses the advancements in molecular, genetic, and imaging approaches to the study of microglia with the turn of the millennium, enabling insights into virtually all facets of microglial physiology. Moving forward, it is clear that the future holds substantial promise for further discoveries. The next epoch in the history of microglial research has just begun.
Collapse
Affiliation(s)
- Payam Rezaie
- School of Life, Health & Chemical Sciences, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, UK.
| | | |
Collapse
|
4
|
Ogasawara A, Takeuchi H, Komiya H, Ogawa Y, Nishimura K, Kubota S, Hashiguchi S, Takahashi K, Kunii M, Tanaka K, Tada M, Doi H, Tanaka F. Anti-inflammatory effects of siponimod on astrocytes. Neurosci Res 2022; 184:38-46. [DOI: 10.1016/j.neures.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/27/2022] [Accepted: 08/04/2022] [Indexed: 10/31/2022]
|
5
|
Soares NL, Vieira HLA. Microglia at the Centre of Brain Research: Accomplishments and Challenges for the Future. Neurochem Res 2021; 47:218-233. [PMID: 34586585 DOI: 10.1007/s11064-021-03456-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023]
Abstract
Microglia are the immune guardians of the central nervous system (CNS), with critical functions in development, maintenance of homeostatic tissue balance, injury and repair. For a long time considered a forgotten 'third element' with basic phagocytic functions, a recent surge in interest, accompanied by technological progress, has demonstrated that these distinct myeloid cells have a wide-ranging importance for brain function. This review reports microglial origins, development, and function in the healthy brain. Moreover, it also targets microglia dysfunction and how it contributes to the progression of several neurological disorders, focusing on particular molecular mechanisms and whether these may present themselves as opportunities for novel, microglia-targeted therapeutic approaches, an ever-enticing prospect. Finally, as it has been recently celebrated 100 years of microglia research, the review highlights key landmarks from the past century and looked into the future. Many challenging problems have arisen, thus it points out some of the most pressing questions and experimental challenges for the ensuing century.
Collapse
Affiliation(s)
- Nuno L Soares
- Chronic Diseases Research Center (CEDOC) - Faculdade de Ciências Médicas/NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria 130, 1169-056, Lisboa, Portugal.
| | - Helena L A Vieira
- Chronic Diseases Research Center (CEDOC) - Faculdade de Ciências Médicas/NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria 130, 1169-056, Lisboa, Portugal.,Department of Chemistry, UCIBIO, Applied Molecular Biosciences Unit, NOVA School of Science and Technology, Universidade Nova de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, Lisboa, Portugal
| |
Collapse
|
6
|
Al-Awadi AMI, AlJawder AI, Mousa A, Taha S, Bakhiet M. A role for the immune system-released activating agent (ISRAA) in the ontogenetic development of brain astrocytes. PLoS One 2021; 16:e0248455. [PMID: 33970944 PMCID: PMC8109834 DOI: 10.1371/journal.pone.0248455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/25/2021] [Indexed: 11/23/2022] Open
Abstract
The Immune System-Released Activating Agent (ISRAA) was discovered as a novel molecule that functions as a mediator between the nervous and immune systems in response to a nervous stimulus following an immune challenge. This research investigated the role of ISRAA) in promoting the ontogeny of the mouse brain astrocytes. Astrocyte cultures were prepared from two-month-old BALB/c mice. Recombinant ISRAA protein was used to stimulate astrocyte cultures. Immunohistochemistry and ELISA were utilized to measure ISRAA and IFN-γ levels, IFN-γR expression and STAT1 nuclear translocation. MTT-assay was used to evaluate cellular survival and proliferation. To assess astrocyte cell lysates and tyrosine-phosphorylated proteins, SDS-PAGE and western blot were used. ISRAA was highly expressed in mouse embryonic astrocytes, depending on cell age. Astrocytes aged seven days (E7) showed increased proliferation and diminished differentiation, while 21-day-old (E21) astrocytes depicted reversed effects. IFN-γ was involved in the ISRAA action as ISRAA induced IFN-γ in both age groups, but only E21 astrocytes expressed IFN-γR. ISRAA stimulation of E21 resulted in tyrosine phosphorylation of numerous cellular proteins and the nuclear translocation of STAT1, a signalling pathway utilized by IFN-γ. The results suggest that ISRAA is involved in mouse brain development through the cytokine network involving IFN-γ.
Collapse
Affiliation(s)
- Aminah M. I. Al-Awadi
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Abdulaziz Isa AlJawder
- Department of Physiology, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Alyaa Mousa
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Safa Taha
- Department of Molecular Medicine, College of Medicine and Medical Sciences, Princess Al Jawhara Center for Molecular Medicine, Genetics and Inherited Diseases, Arabian Gulf University, Manama, Bahrain
| | - Moiz Bakhiet
- Department of Molecular Medicine, College of Medicine and Medical Sciences, Princess Al Jawhara Center for Molecular Medicine, Genetics and Inherited Diseases, Arabian Gulf University, Manama, Bahrain
| |
Collapse
|
7
|
Komiya H, Takeuchi H, Ogawa Y, Suzuki K, Ogasawara A, Takahashi K, Azuma YT, Doi H, Tanaka F. Ablation of interleukin-19 improves motor function in a mouse model of amyotrophic lateral sclerosis. Mol Brain 2021; 14:74. [PMID: 33931083 PMCID: PMC8086093 DOI: 10.1186/s13041-021-00785-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 04/24/2021] [Indexed: 11/25/2022] Open
Abstract
Neuroinflammation by activated microglia and astrocytes plays a critical role in progression of amyotrophic lateral sclerosis (ALS). Interleukin-19 (IL-19) is a negative-feedback regulator that limits pro-inflammatory responses of microglia in an autocrine and paracrine manner, but it remains unclear how IL-19 contributes to ALS pathogenesis. We investigated the role of IL-19 in ALS using transgenic mice carrying human superoxide dismutase 1 with the G93A mutation (SOD1G93A Tg mice). We generated IL-19-deficient SOD1G93A Tg (IL-19-/-/SOD1G93A Tg) mice by crossing SOD1G93A Tg mice with IL-19-/- mice, and then evaluated disease progression, motor function, survival rate, and pathological and biochemical alternations in the resultant mice. In addition, we assessed the effect of IL-19 on glial cells using primary microglia and astrocyte cultures from the embryonic brains of SOD1G93A Tg mice and IL-19-/-/SOD1G93A Tg mice. Expression of IL-19 in primary microglia and lumbar spinal cord was higher in SOD1G93A Tg mice than in wild-type mice. Unexpectedly, IL-19-/-/SOD1G93A Tg mice exhibited significant improvement of motor function. Ablation of IL-19 in SOD1G93A Tg mice increased expression of both neurotoxic and neuroprotective factors, including tumor necrosis factor-α (TNF-α), IL-1β, glial cell line-derived neurotrophic factor (GDNF), and transforming growth factor β1, in lumbar spinal cord. Primary microglia and astrocytes from IL-19-/-/SOD1G93A Tg mice expressed higher levels of TNF-α, resulting in release of GDNF from astrocytes. Inhibition of IL-19 signaling may alleviate ALS symptoms.
Collapse
Affiliation(s)
- Hiroyasu Komiya
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Hideyuki Takeuchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| | - Yuki Ogawa
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Kosuke Suzuki
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Akihiro Ogasawara
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Keita Takahashi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Yasu-Taka Azuma
- Laboratory of Veterinary Pharmacology, Division of Veterinary Science, Osaka Prefecture University Graduate School of Life and Environmental Science, Izumisano, Osaka, 598-9531, Japan
| | - Hiroshi Doi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| |
Collapse
|
8
|
Vedam-Mai V. Harnessing the immune system for the treatment of Parkinson's disease. Brain Res 2021; 1758:147308. [PMID: 33524380 DOI: 10.1016/j.brainres.2021.147308] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/11/2020] [Accepted: 01/16/2021] [Indexed: 01/03/2023]
Abstract
Current treatment options for Parkinson's disease (PD) typically aim to replace dopamine, and hence only provide symptomatic relief. However, in the long run, this approach alone loses its efficacy as it is associated with debilitating side effects. Hence there is an unmet clinical need for addressing levodopa resistant symptoms, and an urgency to develop therapies that can halt or prevent the course of PD. The premise that α-syn can transmit from cell-to-cell in a prion like manner has opened up the possibility for the use of immunotherapy in PD. There is evidence for inflammation in PD as is evidenced by microglial activation, as well as the involvement of the peripheral immune system in PD, and peripheral inflammation can exacerbate dopaminergic degeneration as seen in animal models of the disease. However, mechanisms that link the immune system with PD are not clear, and the sequence of immune responses with respect to PD are still unknown. Nevertheless, our present knowledge offers avenues for the development of immune-based therapies for PD. In order to successfully employ such strategies, we must comprehend the state of the peripheral immune system during the course of PD. This review describes the developments in the field of both active and passive immunotherapies in the treatment of PD, and highlights the crucial need for future research for clarifying the role of inflammation and immunity in this debilitating disease.
Collapse
|
9
|
Generation of CSF1-Independent Ramified Microglia-Like Cells from Leptomeninges In Vitro. Cells 2020; 10:cells10010024. [PMID: 33375610 PMCID: PMC7824226 DOI: 10.3390/cells10010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 11/16/2022] Open
Abstract
Although del Río-Hortega originally reported that leptomeningeal cells are the source of ramified microglia in the developing brain, recent views do not seem to pay much attention to this notion. In this study, in vitro experiments were conducted to determine whether leptomeninges generate ramified microglia. The leptomeninges of neonatal rats containing Iba1+ macrophages were peeled off the brain surface. Leptomeningeal macrophages strongly expressed CD68 and CD163, but microglia in the brain parenchyma did not. Leptomeningeal macrophages expressed epidermal growth factor receptor (EGFR) as revealed by RT-PCR and immunohistochemical staining. Cells obtained from the peeled-off leptomeninges were cultured in a serum-free medium containing EGF, resulting in the formation of large cell aggregates in which many proliferating macrophages were present. In contrast, colony-stimulating factor 1 (CSF1) did not enhance the generation of Iba1+ cells from the leptomeningeal culture. The cell aggregates generated ramified Iba1+ cells in the presence of serum, which express CD68 and CD163 at much lower levels than primary microglia isolated from a mixed glial culture. Therefore, the leptomeningeal-derived cells resembled parenchymal microglia better than primary microglia. This study suggests that microglial progenitors expressing EGFR reside in the leptomeninges and that there is a population of microglia-like cells that grow independently of CSF1.
Collapse
|
10
|
Sierra A, Paolicelli RC, Kettenmann H. Cien Años de Microglía: Milestones in a Century of Microglial Research. Trends Neurosci 2019; 42:778-792. [PMID: 31635851 DOI: 10.1016/j.tins.2019.09.004] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022]
Abstract
The year 2019 marks the 100-year anniversary of the discovery of microglia by Pío del Río-Hortega. We will recount the state of neuroscience research at the beginning of the 20th century and the heated scientific dispute regarding microglial identity. We will then walk through some of the milestones of microglial research in the decades since then. In the last 20 years, the field has grown exponentially. Researchers have shown that microglia are unlike any other resident macrophages: they have a unique origin and distinguishing features. Microglia are extraordinarily motile cells and constantly survey their environment, interacting with neurons, astrocytes, oligodendrocytes, neural stem cells, and infiltrating immune cells. We finally highlight some open questions for future research regarding microglia's identity, population dynamics, and dual (beneficial and detrimental) role in pathology.
Collapse
Affiliation(s)
- Amanda Sierra
- Achucarro Basque Center for Neuroscience, Ikerbasque Foundation, University of the Basque Country UPV/EHU, Parque Científico UPV/EHU, Barrio Sarriena s/n, Leioa, Bizkaia, 48940, Spain.
| | - Rosa C Paolicelli
- Department of Physiology, University of Lausanne, Rue du Bugnon 7, 1005 Lausanne, Switzerland.
| | - Helmut Kettenmann
- Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Cellular Neurosciences, Robert Roessle Str 10, 13092 Berlin, Germany.
| |
Collapse
|
11
|
Men Y, Yelick J, Jin S, Tian Y, Chiang MSR, Higashimori H, Brown E, Jarvis R, Yang Y. Exosome reporter mice reveal the involvement of exosomes in mediating neuron to astroglia communication in the CNS. Nat Commun 2019; 10:4136. [PMID: 31515491 PMCID: PMC6742670 DOI: 10.1038/s41467-019-11534-w] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 07/10/2019] [Indexed: 12/20/2022] Open
Abstract
Astroglia play active and diverse roles in modulating neuronal/synaptic functions in the CNS. How these astroglial functions are regulated, especially by neuronal signals, remains largely unknown. Exosomes, a major type of extracellular vesicles (EVs) that originate from endosomal intraluminal vesicles (ILVs), have emerged as a new intercellular communication process. By generating cell-type-specific ILVs/exosome reporter (CD63-GFPf/f) mice and immuno-EM/confocal image analysis, we found that neuronal CD63-GFP+ ILVs are primarily localized in soma and dendrites, but not in axonal terminals in vitro and in vivo. Secreted neuronal exosomes contain a subset of microRNAs (miRs) that is distinct from the miR profile of neurons. These miRs, especially the neuron-specific miR-124-3p, are potentially internalized into astrocytes. MiR-124-3p further up-regulates the predominant glutamate transporter GLT1 by suppressing GLT1-inhibiting miRs. Our findings suggest a previously undescribed neuronal exosomal miR-mediated genetic regulation of astrocyte functions, potentially opening a new frontier in understanding CNS intercellular communication. Our current understanding of exosome signaling among CNS cells is mostly limited to culture models. In this study, authors generated a new cell-type specific exosome reporter mouse line which allows the first in vivo investigation of the localization of neuronal exosomes in the CNS, and also potentially highlights the role of exosomally transferred miR-124-3p in mediating astroglial glutamate uptake function
Collapse
Affiliation(s)
- Yuqin Men
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Avenue, Boston, MA, 02111, USA.,Tufts University, Sackler School of Biomedical Sciences, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Julia Yelick
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Shijie Jin
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Yang Tian
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Avenue, Boston, MA, 02111, USA.,Dongfang Hospital of University of Chinese Medicine, No.6, District 1, Fangxingyuan, Fangzhuang, Fengtai District, 100078, Beijing, People's Republic of China
| | - Ming Sum R Chiang
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Haruki Higashimori
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Eoin Brown
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Rachel Jarvis
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Yongjie Yang
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Avenue, Boston, MA, 02111, USA. .,Tufts University, Sackler School of Biomedical Sciences, 136 Harrison Avenue, Boston, MA, 02111, USA.
| |
Collapse
|
12
|
Silvin A, Ginhoux F. Microglia heterogeneity along a spatio-temporal axis: More questions than answers. Glia 2018; 66:2045-2057. [PMID: 30144321 DOI: 10.1002/glia.23458] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/05/2018] [Accepted: 05/04/2018] [Indexed: 12/29/2022]
Abstract
Microglia are resident macrophages of the central nervous system; they arise during early embryonic development and persist throughout adulthood. These unique cells provide developmental support, contribute to adult brain homeostasis and impart immune protection during infection. Dysregulated microglia are implicated in the pathophysiology of several neurological disorders, including Alzheimer disease, and as such, a better understanding of their regulation and function is required for rational therapeutic design. Recent studies have highlighted the various heterogeneous aspects of microglia, such as their wide differentiation spectrum from early embryogenesis to adulthood, their location in different brain regions and their responses to ageing, infection and inflammation. In this review, we discuss microglial heterogeneity in time and space and highlight the remaining questions arising from such heterogeneity.
Collapse
Affiliation(s)
- Aymeric Silvin
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, IMMUNOS Building #3-4, BIOPOLIS, 138648, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, IMMUNOS Building #3-4, BIOPOLIS, 138648, Singapore.,Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| |
Collapse
|
13
|
Weller M, Esser P, Heimann K, Wiedemann P. Mononuclear Phagocytes in Proliferative Vitreoretinopathy (PVR). A Specific Role of Microglial Cells in Non-Traumatic Disease? Eur J Ophthalmol 2018; 1:161-6. [PMID: 1821209 DOI: 10.1177/112067219100100401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mononuclear phagocytes have been a focus of attention in the cellular biology of proliferative vitreoretinopathy (PVR) for more than ten years. The pattern of phagocyte participation in periretinal traction membrane formation in PVR depends on the etiology, i.e. trauma, rhegmatogenous retinal detachment, previous therapy, i.e. multiple surgical interventions, and the clinical stage of the disease. We have recently identified microglial cells as a distinct cellular population, in membranes from patients with non-traumatic PVR. Current evidence of mononuclear phagocyte function in PVR suggests a role for resident phagocytes of the vitreous and retina in PVR subsequent to rhegmatogenous detachment, and a role for blood-derived monocytes in post-traumatic PVR. The cellular biology of PVR may be much more heterogeneous than previously assumed.
Collapse
Affiliation(s)
- M Weller
- Department of Vitreoretinal Surgery, University Eye Hospital, Cologne, Germany
| | | | | | | |
Collapse
|
14
|
Wu R, Li X, Xu P, Huang L, Cheng J, Huang X, Jiang J, Wu LJ, Tang Y. TREM2 protects against cerebral ischemia/reperfusion injury. Mol Brain 2017; 10:20. [PMID: 28592261 PMCID: PMC5461720 DOI: 10.1186/s13041-017-0296-9] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 04/20/2017] [Indexed: 01/01/2023] Open
Abstract
Although post-ischemic inflammation induced by the innate immune response is considered an essential step in the progression of cerebral ischemia injury, the role of triggering receptor expressed on myeloid cells 2 (TREM2) in the pathogenesis of ischemic stroke remains to be elucidated. Here, we found that the transcriptional and post-transcriptional levels of TREM2 were increased in cultured primary microglia after oxygen-glucose deprivation and reoxygenation and in the ischemic penumbra of the cerebral cortex after middle cerebral artery occlusion (MCAO) and reperfusion in mice. TREM2 was mainly expressed in microglia, but not in astrocytes, neurons, or oligodendrocytes in mice subjected to MCAO. Manipulating TREM2 expression levels in vitro and in vivo significantly regulated the production of pro- and anti-inflammatory mediators after ischemic stroke. TREM2 overexpression markedly suppressed the inflammatory response and neuronal apoptosis. By contrast, TREM2 gene silencing intensified the inflammatory response, increased neuronal apoptosis and infarct volume, and further exacerbated neurological dysfunction. Our study demonstrated that TREM2 protects against cerebral ischemia/reperfusion injury through the aspect of post-ischemic inflammatory response and neuronal apoptosis. Pharmacological targeting of TREM2 to suppress the inflammatory response may provide a new approach for developing therapeutic strategies in the treatment of ischemic stroke and other cerebrovascular diseases.
Collapse
Affiliation(s)
- Rong Wu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiangpen Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Pengfei Xu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Likui Huang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jinping Cheng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiaolong Huang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jingru Jiang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Long-Jun Wu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA.,Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, Guangdong Province, 510120, China. .,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510120, China. .,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.
| |
Collapse
|
15
|
Marcet P, Santos N, Borlongan CV. When friend turns foe: central and peripheral neuroinflammation in central nervous system injury. ACTA ACUST UNITED AC 2017; 4:82-92. [PMID: 29670933 PMCID: PMC5901724 DOI: 10.20517/2347-8659.2017.07] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Injury to the central nervous system (CNS) is common, and though it has been well studied, many aspects of traumatic brain injury (TBI) and stroke are poorly understood. TBI and stroke are two pathologic events that can cause severe, immediate impact to the neurostructure and function of the CNS, which has been recognized recently to be exacerbated by the body’s own immune response. Although the brain damage induced by the initial trauma is most likely unsalvageable, the secondary immunologic deterioration of neural tissue gives ample opportunity for therapeutic strategists seeking to mitigate TBI’s secondary detrimental effects. The purpose of this paper is to highlight the cell death mechanisms associated with CNS injury with special emphasis on inflammation. The authors discuss sources of inflammation, and introduce the role of the spleen in the systemic response to inflammation after CNS injury.
Collapse
Affiliation(s)
- Paul Marcet
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Nicole Santos
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
16
|
Shanshan Y, Beibei J, Li T, Minna G, Shipeng L, Li P, Yong Z. Phospholipase A2 of Peroxiredoxin 6 Plays a Critical Role in Cerebral Ischemia/Reperfusion Inflammatory Injury. Front Cell Neurosci 2017; 11:99. [PMID: 28424593 PMCID: PMC5380807 DOI: 10.3389/fncel.2017.00099] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
Microglia-mediated inflammation is an important step in the progression of cerebral ischemia/reperfusion injury and the associated production of receptors of immunomoudulation, including Toll-like receptors (TLRs). Peroxiredoxin 6 (Prdx6) has been demonstrated as the endogenous antioxidant protein for its peroxidase properties. However, the role of the independent phospholipase A2 (iPLA2) activity of Prdx6 in stroke has not been well studied. In this study, we evaluated whether blocking the calcium-iPLA2 activity of Prdx6 using siRNA and inhibitors (1-hexadecyl-3-(trifluoroethgl)-sn-glycerol-2 phosphomethanol, MJ33) would have a critical effect on inflammatory brain damage. We conducted oxygen-glucose deprivation (OGD)/recovery (R) in vitro and middle cerebral artery occlusion (MCAO) in vivo in a microglia/neuron co-culture system and in rats. In vitro, we found that Prdx6-iPLA2 activity was associated with the secretion of neurotoxic inflammatory mediators interleukin1β (IL-1β), interleukin-17 (IL-17) and interleukin-23 (IL-23) and elevated expression of Toll-like receptor 2/4 (TLR2/4), leading to the formation of nuclear factor-kappa B (NF-κB), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in microglial cells. In vivo, combined treatment with Prdx6-iPLA2 activity inhibitor MJ33 showed a greater diminution in neurologic deficits, cerebral infarction, brain water content and inflammatory molecules than Prdx6-siRNA treatment alone. Our findings provide new insight into Prdx6-iPLA2 function in the brain. Inhibition of Prdx6-iPLA2 activity by gene therapy and/or pharmacology may constitute a promising new therapeutic approach to the treatment of stroke.
Collapse
Affiliation(s)
- Yu Shanshan
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Jiang Beibei
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Tan Li
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Gao Minna
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Lei Shipeng
- Department of Respiratory Medicine, Jiangjin Center HospitalChongqing, China
| | - Peng Li
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Zhao Yong
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| |
Collapse
|
17
|
Shimizu T, Wisessmith W, Li J, Abe M, Sakimura K, Chetsawang B, Sahara Y, Tohyama K, Tanaka KF, Ikenaka K. The balance between cathepsin C and cystatin F controls remyelination in the brain ofPlp1-overexpressing mouse, a chronic demyelinating disease model. Glia 2017; 65:917-930. [DOI: 10.1002/glia.23134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 02/03/2017] [Accepted: 02/10/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Takahiro Shimizu
- Division of Neurobiology and Bioinformatics; National Institute for Physiological Sciences; Okazaki Japan
| | - Wilaiwan Wisessmith
- Division of Neurobiology and Bioinformatics; National Institute for Physiological Sciences; Okazaki Japan
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University; Salaya Nakhonpathom Thailand
| | - Jiayi Li
- Division of Neurobiology and Bioinformatics; National Institute for Physiological Sciences; Okazaki Japan
- Department of Physiological Sciences; Graduate University for Advanced Studies (SOKENDAI); Okazaki Japan
| | - Manabu Abe
- Brain Research Institute, Niigata University; Niigata Japan
| | - Kenji Sakimura
- Brain Research Institute, Niigata University; Niigata Japan
| | - Banthit Chetsawang
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University; Salaya Nakhonpathom Thailand
| | - Yoshinori Sahara
- Department of Physiology; Iwate Medical University School of Dentistry; Iwate Japan
| | - Koujiro Tohyama
- Department of Physiology; Iwate Medical University School of Dentistry; Iwate Japan
- Center for Electron Microscopy and Bio-Imaging Research, Iwate Medical University; Iwate Japan
| | - Kenji F. Tanaka
- Division of Neurobiology and Bioinformatics; National Institute for Physiological Sciences; Okazaki Japan
- Department of Neuropsychiatry; Keio University; Tokyo Japan
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics; National Institute for Physiological Sciences; Okazaki Japan
- Department of Physiological Sciences; Graduate University for Advanced Studies (SOKENDAI); Okazaki Japan
| |
Collapse
|
18
|
Alqinyah M, Maganti N, Ali MW, Yadav R, Gao M, Cacan E, Weng HR, Greer SF, Hooks SB. Regulator of G Protein Signaling 10 (Rgs10) Expression Is Transcriptionally Silenced in Activated Microglia by Histone Deacetylase Activity. Mol Pharmacol 2017; 91:197-207. [PMID: 28031332 PMCID: PMC5325084 DOI: 10.1124/mol.116.106963] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 12/12/2016] [Indexed: 02/06/2023] Open
Abstract
RGS10 has emerged as a key regulator of proinflammatory cytokine production in microglia, functioning as an important neuroprotective factor. Although RGS10 is normally expressed in microglia at high levels, expression is silenced in vitro following activation of TLR4 receptor. Given the ability of RGS10 to regulate inflammatory signaling, dynamic regulation of RGS10 levels in microglia may be an important mechanism to tune inflammatory responses. The goals of the current study were to confirm that RGS10 is suppressed in an in vivo inflammatory model of microglial activation and to determine the mechanism for activation-dependent silencing of Rgs10 expression in microglia. We demonstrate that endogenous RGS10 is present in spinal cord microglia, and RGS10 protein levels are suppressed in the spinal cord in a nerve injury-induced neuropathic pain mouse model. We show that the histone deacetylase (HDAC) enzyme inhibitor trichostatin A blocks the ability of lipopolysaccharide (LPS) to suppress Rgs10 transcription in BV-2 and primary microglia, demonstrating that HDAC enzymes are required for LPS silencing of Rgs10 Furthermore, we used chromatin immunoprecipitation to demonstrate that H3 histones at the Rgs10 proximal promoter are deacetylated in BV-2 microglia following LPS activation, and HDAC1 association at the Rgs10 promoter is enhanced following LPS stimulation. Finally, we have shown that sphingosine 1-phosphate, an endogenous microglial signaling mediator that inhibits HDAC activity, enhances basal Rgs10 expression in BV-2 microglia, suggesting that Rgs10 expression is dynamically regulated in microglia in response to multiple signals.
Collapse
Affiliation(s)
- Mohammed Alqinyah
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia (M.A., M.W.A., R.Y., M.G., H.-R.W., S.B.H.); and Department of Biology, Georgia State University, Atlanta, Georgia (N.M., E.C., S.F.G.)
| | - Nagini Maganti
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia (M.A., M.W.A., R.Y., M.G., H.-R.W., S.B.H.); and Department of Biology, Georgia State University, Atlanta, Georgia (N.M., E.C., S.F.G.)
| | - Mourad W Ali
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia (M.A., M.W.A., R.Y., M.G., H.-R.W., S.B.H.); and Department of Biology, Georgia State University, Atlanta, Georgia (N.M., E.C., S.F.G.)
| | - Ruchi Yadav
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia (M.A., M.W.A., R.Y., M.G., H.-R.W., S.B.H.); and Department of Biology, Georgia State University, Atlanta, Georgia (N.M., E.C., S.F.G.)
| | - Mei Gao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia (M.A., M.W.A., R.Y., M.G., H.-R.W., S.B.H.); and Department of Biology, Georgia State University, Atlanta, Georgia (N.M., E.C., S.F.G.)
| | - Ercan Cacan
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia (M.A., M.W.A., R.Y., M.G., H.-R.W., S.B.H.); and Department of Biology, Georgia State University, Atlanta, Georgia (N.M., E.C., S.F.G.)
| | - Han-Rong Weng
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia (M.A., M.W.A., R.Y., M.G., H.-R.W., S.B.H.); and Department of Biology, Georgia State University, Atlanta, Georgia (N.M., E.C., S.F.G.)
| | - Susanna F Greer
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia (M.A., M.W.A., R.Y., M.G., H.-R.W., S.B.H.); and Department of Biology, Georgia State University, Atlanta, Georgia (N.M., E.C., S.F.G.)
| | - Shelley B Hooks
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia (M.A., M.W.A., R.Y., M.G., H.-R.W., S.B.H.); and Department of Biology, Georgia State University, Atlanta, Georgia (N.M., E.C., S.F.G.)
| |
Collapse
|
19
|
Kawahara K, Hirata H, Ohbuchi K, Nishi K, Maeda A, Kuniyasu A, Yamada D, Maeda T, Tsuji A, Sawada M, Nakayama H. The novel monoclonal antibody 9F5 reveals expression of a fragment of GPNMB/osteoactivin processed by furin-like protease(s) in a subpopulation of microglia in neonatal rat brain. Glia 2016; 64:1938-61. [PMID: 27464357 PMCID: PMC5129557 DOI: 10.1002/glia.23034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 07/02/2016] [Accepted: 07/07/2016] [Indexed: 12/19/2022]
Abstract
To differentiate subtypes of microglia (MG), we developed a novel monoclonal antibody, 9F5, against one subtype (type 1) of rat primary MG. The 9F5 showed high selectivity for this cell type in Western blot and immunocytochemical analyses and no cross-reaction with rat peritoneal macrophages (Mφ). We identified the antigen molecule for 9F5: the 50- to 70-kDa fragments of rat glycoprotein nonmetastatic melanoma protein B (GPNMB)/osteoactivin, which started at Lys(170) . In addition, 9F5 immunoreactivity with GPNMB depended on the activity of furin-like protease(s). More important, rat type 1 MG expressed the GPNMB fragments, but type 2 MG and Mφ did not, although all these cells expressed mRNA and the full-length protein for GPNMB. These results suggest that 9F5 reactivity with MG depends greatly on cleavage of GPNMB and that type 1 MG, in contrast to type 2 MG and Mφ, may have furin-like protease(s) for GPNMB cleavage. In neonatal rat brain, amoeboid 9F5+ MG were observed in specific brain areas including forebrain subventricular zone, corpus callosum, and retina. Double-immunοstaining with 9F5 antibody and anti-Iba1 antibody, which reacts with MG throughout the CNS, revealed that 9F5+ MG were a portion of Iba1+ MG, suggesting that MG subtype(s) exist in vivo. We propose that 9F5 is a useful tool to discriminate between rat type 1 MG and other subtypes of MG/Mφ and to reveal the role of the GPNMB fragments during developing brain. GLIA 2016;64:1938-1961.
Collapse
Affiliation(s)
- Kohichi Kawahara
- Department of Molecular Cell Function, Faculty of Life Sciences, Kumamoto University, 5-1 Ohe-Honmachi, Kumamoto, 862-0973, Japan. .,Department of Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, 265-1 Higashijima, Niigata, 956-8603, Japan.
| | - Hiroshi Hirata
- Department of Molecular Cell Function, Faculty of Life Sciences, Kumamoto University, 5-1 Ohe-Honmachi, Kumamoto, 862-0973, Japan
| | - Kengo Ohbuchi
- Department of Molecular Cell Function, Faculty of Life Sciences, Kumamoto University, 5-1 Ohe-Honmachi, Kumamoto, 862-0973, Japan
| | - Kentaro Nishi
- Department of Molecular Cell Function, Faculty of Life Sciences, Kumamoto University, 5-1 Ohe-Honmachi, Kumamoto, 862-0973, Japan
| | - Akira Maeda
- Department of Molecular Cell Function, Faculty of Life Sciences, Kumamoto University, 5-1 Ohe-Honmachi, Kumamoto, 862-0973, Japan
| | - Akihiko Kuniyasu
- Department of Molecular Cell Pharmacology, Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto, 860-0082, Japan
| | - Daisuke Yamada
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, 265-1 Higashijima, Niigata, 956-8603, Japan
| | - Takehiko Maeda
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, 265-1 Higashijima, Niigata, 956-8603, Japan
| | - Akihiko Tsuji
- Department of Biological Science and Technology, the University of Tokushima Graduate School, 2-1 Minamijosanjima, Tokushima, 770-8506, Japan
| | - Makoto Sawada
- Department of Brain Functions, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan
| | - Hitoshi Nakayama
- Department of Molecular Cell Function, Faculty of Life Sciences, Kumamoto University, 5-1 Ohe-Honmachi, Kumamoto, 862-0973, Japan.
| |
Collapse
|
20
|
Xu P, Xu Y, Hu B, Wang J, Pan R, Murugan M, Wu LJ, Tang Y. Extracellular ATP enhances radiation-induced brain injury through microglial activation and paracrine signaling via P2X7 receptor. Brain Behav Immun 2015; 50:87-100. [PMID: 26122280 DOI: 10.1016/j.bbi.2015.06.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/24/2015] [Accepted: 06/24/2015] [Indexed: 12/14/2022] Open
Abstract
Activation of purinergic receptors by extracellular ATP (eATP) released from injured cells has been implicated in the pathogenesis of many neuronal disorders. The P2X7 receptor (P2X7R), an ion-selective purinergic receptor, is associated with microglial activation and paracrine signaling. However, whether ATP and P2X7R are involved in radiation-induced brain injury (RBI) remains to be determined. Here, we found that the eATP level was elevated in the cerebrospinal fluid (CSF) of RBI patients and was associated with the clinical severity of the disorder. In our experimental model, radiation treatment increased the level of eATP in the supernatant of primary cultures of neurons and glial cells and in the CSF of irradiated mice. In addition, ATP administration activated microglia, induced the release of the inflammatory mediators such as cyclooxygenase-2, tumor necrosis factor α and interleukin 6, and promoted neuronal apoptosis. Furthermore, blockade of ATP-P2X7R interaction using P2X7 antagonist Brilliant Blue G or P2X7 knockdown suppressed radiation-induced microglial activation and proliferation in the hippocampus, and restored the spatial memory of irradiated mice. Finally, we found that the PI3K/AKT and nuclear factor κB mediated pathways were downstream of ATP-P2X7R signaling in RBI. Taken together, our results unveiled the critical role of ATP-P2X7R in brain damage in RBI, suggesting that inhibition of ATP-P2X7R axis might be a potential strategy for the treatment of patients with RBI.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yongteng Xu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Bin Hu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jue Wang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Rui Pan
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Madhuvika Murugan
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, United States
| | - Long-Jun Wu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, United States
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
21
|
Ottum PA, Arellano G, Reyes LI, Iruretagoyena M, Naves R. Opposing Roles of Interferon-Gamma on Cells of the Central Nervous System in Autoimmune Neuroinflammation. Front Immunol 2015; 6:539. [PMID: 26579119 PMCID: PMC4626643 DOI: 10.3389/fimmu.2015.00539] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/08/2015] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is the principal cause of autoimmune neuroinflammation in humans, and its animal model, experimental autoimmune encephalomyelitis (EAE), is widely used to gain insight about their immunopathological mechanisms for and the development of novel therapies for MS. Most studies on the role of interferon (IFN)-γ in the pathogenesis and progression of EAE have focused on peripheral immune cells, while its action on central nervous system (CNS)-resident cells has been less explored. In addition to the well-known proinflammatory and damaging effects of IFN-γ in the CNS, evidence has also endowed this cytokine both a protective and regulatory role in autoimmune neuroinflammation. Recent investigations performed in this research field have exposed the complex role of IFN-γ in the CNS uncovering unexpected mechanisms of action that underlie these opposing activities on different CNS-resident cell types. The mechanisms behind these two-faced effects of IFN-γ depend on dose, disease phase, and cell development stage. Here, we will review and discuss the dual role of IFN-γ on CNS-resident cells in EAE highlighting its protective functions and the mechanisms proposed.
Collapse
Affiliation(s)
- Payton A Ottum
- Immunology Program, Biomedical Sciences Institute, School of Medicine, Universidad de Chile , Santiago , Chile
| | - Gabriel Arellano
- Immunology Program, Biomedical Sciences Institute, School of Medicine, Universidad de Chile , Santiago , Chile
| | - Lilian I Reyes
- Faculty of Science, Universidad San Sebastián , Santiago , Chile
| | - Mirentxu Iruretagoyena
- Department of Clinical Immunology and Rheumatology, School of Medicine, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Rodrigo Naves
- Immunology Program, Biomedical Sciences Institute, School of Medicine, Universidad de Chile , Santiago , Chile
| |
Collapse
|
22
|
Parajuli B, Horiuchi H, Mizuno T, Takeuchi H, Suzumura A. CCL11 enhances excitotoxic neuronal death by producing reactive oxygen species in microglia. Glia 2015; 63:2274-84. [PMID: 26184677 DOI: 10.1002/glia.22892] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 06/05/2015] [Accepted: 06/30/2015] [Indexed: 12/28/2022]
Abstract
The chemokine CCL11 (also known as eotaxin-1) is a potent eosinophil chemoattractant that mediates allergic diseases such as asthma, atopic dermatitis, and inflammatory bowel diseases. Previous studies demonstrated that concentrations of CCL11 are elevated in the sera and cerebrospinal fluids (CSF) of patients with neuroinflammatory disorders, including multiple sclerosis. Moreover, the levels of CCL11 in plasma and CSF increase with age, and CCL11 suppresses adult neurogenesis in the central nervous system (CNS), resulting in memory impairment. However, the precise source and function of CCL11 in the CNS are not fully understood. In this study, we found that activated astrocytes release CCL11, whereas microglia predominantly express the CCL11 receptor. CCL11 significantly promoted the migration of microglia, and induced microglial production of reactive oxygen species by upregulating nicotinamide adenine dinucleotide phosphate-oxidase 1 (NOX1), thereby promoting excitotoxic neuronal death. These effects were reversed by inhibition of NOX1. Our findings suggest that CCL11 released from activated astrocytes triggers oxidative stress via microglial NOX1 activation and potentiates glutamate-mediated neurotoxicity, which may be involved in the pathogenesis of various neurological disorders.
Collapse
Affiliation(s)
- Bijay Parajuli
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-Ku, Nagoya, Japan
| | - Hiroshi Horiuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-Ku, Nagoya, Japan
| | - Tetsuya Mizuno
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-Ku, Nagoya, Japan
| | - Hideyuki Takeuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-Ku, Nagoya, Japan
| | - Akio Suzumura
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-Ku, Nagoya, Japan
| |
Collapse
|
23
|
The protective effect of lactoferrin on ventral mesencephalon neurons against MPP + is not connected with its iron binding ability. Sci Rep 2015; 5:10729. [PMID: 26035688 PMCID: PMC4451802 DOI: 10.1038/srep10729] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 04/21/2015] [Indexed: 11/20/2022] Open
Abstract
Lactoferrin (Lf) can bind to lactoferrin receptor (LfR), leading to iron transport through the plasma membrane. Besides iron transportation, Lf also has antioxidant and anti-inflammatory properties. In the brain, Lf is only synthesized by activated microglia. LfR is present in blood vessels and nigral dopaminergic neurons. Both nigral iron accumulation and microglia activation is believed to be involved in Parkinson’s disease (PD), moreover, increased Lf and LfR in dopaminergic neurons were found in PD cases and MPTP-intoxicated mice. How iron influences microglia to release Lf? Does Lf tend to transport iron to dopaminergic neurons leading to cell death or to protect dopaminergic neuron from neurotoxin? In this study, we observed that iron increased Lf synthesis in activated microglia. In ventral mesencephalon neurons, both iron-free Lf (apo-Lf) and iron-saturated Lf (holo-Lf) exerted neuroprotective effects against MPP+ by mechanisms, believed to enhance the mitochondrial transmembrane potential, improve Cu/Zn-superoxide dismutase activity, increase Bcl-2 expression. Although apo-Lf but not holo-Lf chelated cellular iron, there was no difference between the two types of Lf in the neuroprotection. Our data indicate that iron overload increases the activated microglia releasing Lf. Lf plays protective role on ventral mesencephalon neurons against MPP+, which is iron-chelating independent.
Collapse
|
24
|
Ohkawa Y, Momota H, Kato A, Hashimoto N, Tsuda Y, Kotani N, Honke K, Suzumura A, Furukawa K, Ohmi Y, Natsume A, Wakabayashi T, Furukawa K. Ganglioside GD3 Enhances Invasiveness of Gliomas by Forming a Complex with Platelet-derived Growth Factor Receptor α and Yes Kinase. J Biol Chem 2015; 290:16043-58. [PMID: 25940087 DOI: 10.1074/jbc.m114.635755] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Indexed: 11/06/2022] Open
Abstract
There have been a few studies on the ganglioside expression in human glioma tissues. However, the role of these gangliosides such as GD3 and GD2 has not been well understood. In this study we employed a genetically engineered mouse model of glioma to clarify the functions of GD3 in gliomas. Forced expression of platelet-derived growth factor B in cultured astrocytes derived from p53-deficient mice resulted in the expression of GD3 and GD2. GD3-positive astrocytes exhibited increased cell growth and invasion activities along with elevated phosphorylation of Akt and Yes kinase. By enzyme-mediated activation of radical sources reaction and mass spectrometry, we identified PDGF receptor α (PDGFRα) as a GD3-associated molecule. GD3-positive astrocytes showed a significant amount of PDGFRα in glycolipid-enriched microdomains/rafts compared with GD3-negative cells. Src kinase family Yes was co-precipitated with PDGFRα, and its pivotal role in the increased cell invasion of GD3-positive astrocytes was demonstrated by silencing with anti-Yes siRNA. Direct association between PDGFRα and GD3 was also shown, suggesting that GD3 forms ternary complex with PDGFRα and Yes. The fact that GD3, PDGFRα, and activated Yes were colocalized in lamellipodia and the edge of tumors in cultured cells and glioma tissues, respectively, suggests that GD3 induced by platelet-derived growth factor B enhances PDGF signals in glycolipid-enriched microdomain/rafts, leading to the promotion of malignant phenotypes such as cell proliferation and invasion in gliomas.
Collapse
Affiliation(s)
- Yuki Ohkawa
- From the Department of Biochemistry II, the Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-0065, Japan, the Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, 1200 Matsumoto-cho, Kasugai 487-8501, Japan
| | - Hiroyuki Momota
- the Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-0065, Japan
| | - Akira Kato
- the Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-0065, Japan
| | | | | | - Norihiro Kotani
- the Department of Biochemistry, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan
| | - Koichi Honke
- the Department of Biochemistry, Kochi University Medical School, Kohasu, Okou-cho, Nankoku, Kochi 783-8505, Japan
| | - Akio Suzumura
- the Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furou-cho, Chikusa-ku, Nagoya 464-8601, Japan, and
| | - Keiko Furukawa
- From the Department of Biochemistry II, the Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, 1200 Matsumoto-cho, Kasugai 487-8501, Japan
| | | | - Atsushi Natsume
- the Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-0065, Japan
| | - Toshihiko Wakabayashi
- the Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-0065, Japan
| | - Koichi Furukawa
- From the Department of Biochemistry II, the Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, 1200 Matsumoto-cho, Kasugai 487-8501, Japan
| |
Collapse
|
25
|
Horiuchi H, Parajuli B, Kawanokuchi J, Jin S, Mizuno T, Takeuchi H, Suzumura A. Oligomeric amyloid β facilitates microglial excitotoxicity by upregulating tumor necrosis factor‐α and downregulating excitatory amino acid transporter 2 in astrocytes. CLINICAL AND EXPERIMENTAL NEUROIMMUNOLOGY 2015; 6:183-190. [DOI: 10.1111/cen3.12192] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
AbstractObjectivesSoluble oligomeric amyloid β (oAβ) directly causes synaptic dysfunction and neuronal death, which are involved in the pathogenesis of Alzheimer's disease. In contrast, several lines of evidence have shown that glia‐mediated excitotoxicity is also involved in the disease progression of Alzheimer's disease. However, it is still unclear how oAβ induces glia‐mediated excitotoxicity. Therefore, we tried to clarify the mechanism of oAβ‐induced glia‐mediated excitotoxicity using mouse primary cultures.MethodsGlial glutamate production was assessed using a colorimetric assay kit. Glial production of tumor necrosis factor‐α (TNF‐α) was evaluated using a specific enzyme‐linked immunosorbent assay kit. Microglial survival was examined using the MTS assay. mRNA and protein expression levels of excitatory amino acid transporter 2 were determined using quantitative polymerase chain reaction and western blotting, respectively.ResultsWe showed that oAβ‐stimulation induces glutamate release from astrocyte/microglia co‐cultures, but not from single culture of astrocytes or microglia. oAβ increased TNF‐α release from astrocytes, but not from microglia, and the astrocyte‐derived TNF‐α enhanced glutamate release from microglia. TNF‐α elongated microglial survival and triggered sustained microglial glutamate release in a positive feedback mechanism through TNF‐α receptor 1. Furthermore, treatment of oAβ decreased astrocytic expression of excitatory amino acid transporter 2, which plays a pivotal role in homeostasis of the extracellular glutamate level.ConclusionsThe present findings suggest that oAβ contributes to glia‐mediated excitotoxicity through the two‐hit mechanism by suppressing astrocytic glutamate uptake through excitatory amino acid transporter 2 and enhancing microglial glutamate release, and triggers chronic glial neuroinflammation through the TNF‐α/TNF‐α receptor 1 positive feedback loop in Alzheimer's disease.
Collapse
Affiliation(s)
- Hiroshi Horiuchi
- Department of Neuroimmunology Research Institute of Environmental Medicine Nagoya University Nagoya Japan
| | - Bijay Parajuli
- Department of Neuroimmunology Research Institute of Environmental Medicine Nagoya University Nagoya Japan
| | - Jun Kawanokuchi
- Department of Neuroimmunology Research Institute of Environmental Medicine Nagoya University Nagoya Japan
| | - Shijie Jin
- Department of Neuroimmunology Research Institute of Environmental Medicine Nagoya University Nagoya Japan
| | - Tetsuya Mizuno
- Department of Neuroimmunology Research Institute of Environmental Medicine Nagoya University Nagoya Japan
| | - Hideyuki Takeuchi
- Department of Neuroimmunology Research Institute of Environmental Medicine Nagoya University Nagoya Japan
| | - Akio Suzumura
- Department of Neuroimmunology Research Institute of Environmental Medicine Nagoya University Nagoya Japan
| |
Collapse
|
26
|
Dysregulation of glutamine transporter SNAT1 in Rett syndrome microglia: a mechanism for mitochondrial dysfunction and neurotoxicity. J Neurosci 2015; 35:2516-29. [PMID: 25673846 DOI: 10.1523/jneurosci.2778-14.2015] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Rett syndrome (RTT) is an autism spectrum disorder caused by loss-of-function mutations in the gene encoding MeCP2, an epigenetic modulator that binds the methyl CpG dinucleotide in target genes to regulate transcription. Previously, we and others reported a role of microglia in the pathophysiology of RTT. To understand the mechanism of microglia dysfunction in RTT, we identified a MeCP2 target gene, SLC38A1, which encodes a major glutamine transporter (SNAT1), and characterized its role in microglia. We found that MeCP2 acts as a microglia-specific transcriptional repressor of SNAT1. Because glutamine is mainly metabolized in the mitochondria, where it is used as an energy substrate and a precursor for glutamate production, we hypothesize that SNAT1 overexpression in MeCP2-deficient microglia would impair the glutamine homeostasis, resulting in mitochondrial dysfunction as well as microglial neurotoxicity because of glutamate overproduction. Supporting this hypothesis, we found that MeCP2 downregulation or SNAT1 overexpression in microglia resulted in (1) glutamine-dependent decrease in microglial viability, which was corroborated by reduced microglia counts in the brains of MECP2 knock-out mice; (2) proliferation of mitochondria and enhanced mitochondrial production of reactive oxygen species; (3) increased oxygen consumption but decreased ATP production (an energy-wasting state); and (4) overproduction of glutamate that caused NMDA receptor-dependent neurotoxicity. The abnormalities could be rectified by mitochondria-targeted expression of catalase and a mitochondria-targeted peptide antioxidant, Szeto-Schiller 31. Our results reveal a novel mechanism via which MeCP2 regulates bioenergetic pathways in microglia and suggest a therapeutic potential of mitochondria-targeted antioxidants for RTT.
Collapse
|
27
|
Horiuchi H, Parajuli B, Wang Y, Azuma YT, Mizuno T, Takeuchi H, Suzumura A. Interleukin-19 acts as a negative autocrine regulator of activated microglia. PLoS One 2015; 10:e0118640. [PMID: 25794104 PMCID: PMC4368203 DOI: 10.1371/journal.pone.0118640] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 01/12/2015] [Indexed: 11/19/2022] Open
Abstract
Activated microglia can exert either neurotoxic or neuroprotective effects, and they play pivotal roles in the pathogenesis and progression of various neurological diseases. In this study, we used cDNA microarrays to show that interleukin-19 (IL-19), an IL-10 family cytokine, is markedly upregulated in activated microglia. Furthermore, we found that microglia are the only cells in the nervous system that express the IL-19 receptor, a heterodimer of the IL-20Rα and IL-20Rβ subunits. IL-19 deficiency increased the production of such pro-inflammatory cytokines as IL-6 and tumor necrosis factor-α in activated microglia, and IL-19 treatment suppressed this effect. Moreover, in a mouse model of Alzheimer's disease, we observed upregulation of IL-19 in affected areas in association with disease progression. Our findings demonstrate that IL-19 is an anti-inflammatory cytokine, produced by activated microglia, that acts negatively on microglia in an autocrine manner. Thus, microglia may self-limit their inflammatory response by producing the negative regulator IL-19.
Collapse
Affiliation(s)
- Hiroshi Horiuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464–8601, Japan
| | - Bijay Parajuli
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464–8601, Japan
| | - Yue Wang
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464–8601, Japan
| | - Yasu-Taka Azuma
- Laboratory of Veterinary Pharmacology, Division of Veterinary Science, Osaka Prefecture University Graduate School of Life and Environmental Science, Izumisano, Osaka, 598–8531, Japan
| | - Tetsuya Mizuno
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464–8601, Japan
| | - Hideyuki Takeuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464–8601, Japan
- * E-mail:
| | - Akio Suzumura
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464–8601, Japan
| |
Collapse
|
28
|
Ishihara Y, Itoh K, Ishida A, Yamazaki T. Selective estrogen-receptor modulators suppress microglial activation and neuronal cell death via an estrogen receptor-dependent pathway. J Steroid Biochem Mol Biol 2015; 145:85-93. [PMID: 25305410 DOI: 10.1016/j.jsbmb.2014.10.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/25/2014] [Accepted: 10/05/2014] [Indexed: 01/02/2023]
Abstract
Growing evidence shows that steroid hormones, especially 17β-estradiol (E2), protect neuronal cells by attenuating excess activation of microglia. However, the use of E2 in the clinic is controversial because of its peripheral actions in reproductive organs and its potential to increase risk for endometrial cancer and breast cancer. Selective estrogen-receptor modulators (SERMs) bind to estrogen receptors (ERs), but their effects as ER agonists or antagonists are dependent on the target tissue. SERMs pose very little cancer risk as a result of their anti-estrogen action in reproductive organs, but their action in the brain is not well understood. In this study, we investigated the effects of SERMs tamoxifen (Tam) and raloxifene (Rlx) on microglial activation and subsequent neuronal injury. Tam and Rlx suppressed the increases in proinflammatory cytokines and chemokine expression that were induced by lipopolysaccharide (LPS) in rat primary microglia cultures. The microglial-conditioned media pretreated with Tam or Rlx significantly attenuated cellular injury in SH-SY5Y cells elicited by microglial-conditioned media treated with LPS alone. Rat primary microglia expressed ERα and ERβ primarily in the nucleus, and thus we examined the involvement of ERs in the suppressive action of Tam and Rlx on microglial activation using a pure ER antagonist, ICI182,780. Pretreatment with ICI182,780 abolished the suppressive effects of SERMs on microglial activation, as well as their protective action on SH-SY5Y cells. A luciferase assay using a vector with three estrogen response elements (EREs) revealed that Tam and Rlx activated ERE-mediated transcription in rat primary microglia. Taken together, these results suggest that Tam and Rlx suppress microglial activation and subsequent neuronal cell death via an ER-mediated transcription pathway. SERMs could represent a novel therapeutic strategy for disorders of the central nervous system based on their ability to suppress neuroinflammation.
Collapse
Affiliation(s)
- Yasuhiro Ishihara
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan.
| | - Kouichi Itoh
- Laboratory for Brain Science, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa 769-2193, Japan
| | - Atsuhiko Ishida
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
| | - Takeshi Yamazaki
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
| |
Collapse
|
29
|
Mutnal MB, Hu S, Schachtele SJ, Lokensgard JR. Infiltrating regulatory B cells control neuroinflammation following viral brain infection. THE JOURNAL OF IMMUNOLOGY 2014; 193:6070-80. [PMID: 25385825 DOI: 10.4049/jimmunol.1400654] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Previous studies have demonstrated the existence of a subset of B lymphocytes, regulatory B cells (Bregs), which modulate immune function. In this study, in vivo and in vitro experiments were undertaken to elucidate the role of these Bregs in controlling neuroinflammation following viral brain infection. We used multicolor flow cytometry to phenotype lymphocyte subpopulations infiltrating the brain, along with in vitro cocultures to assess their anti-inflammatory and immunoregulatory roles. This distinctive subset of CD19(+)CD1d(hi)CD5(+) B cells was found to infiltrate the brains of chronically infected animals, reaching highest levels at the latest time point tested (30 d postinfection). B cell-deficient Jh(-/-) mice were found to develop exacerbated neuroimmune responses as measured by enhanced accumulation and/or retention of CD8(+) T cells within the brain, as well as increased levels of microglial activation (MHC class II). Conversely, levels of Foxp3(+) regulatory T cells were found to be significantly lower in Jh(-/-) mice when compared with wild-type (Wt) animals. Further experiments showed that in vitro-generated IL-10-secreting Bregs (B10) were able to inhibit cytokine responses from microglia following stimulation with viral Ags. These in vitro-generated B10 cells were also found to promote proliferation of regulatory T cells in coculture studies. Finally, gain-of-function experiments demonstrated that reconstitution of Wt B cells into Jh(-/-) mice restored neuroimmune responses to levels exhibited by infected Wt mice. Taken together, these results demonstrate that Bregs modulate T lymphocyte as well as microglial cell responses within the infected brain and promote CD4(+)Foxp3(+) T cell proliferation in vitro.
Collapse
Affiliation(s)
- Manohar B Mutnal
- Neuroimmunology Laboratory, Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, MN 55455
| | - Shuxian Hu
- Neuroimmunology Laboratory, Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, MN 55455
| | - Scott J Schachtele
- Neuroimmunology Laboratory, Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, MN 55455
| | - James R Lokensgard
- Neuroimmunology Laboratory, Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
30
|
Jadhav VS, Krause KH, Singh SK. HIV-1 Tat C modulates NOX2 and NOX4 expressions through miR-17 in a human microglial cell line. J Neurochem 2014; 131:803-15. [PMID: 25146963 DOI: 10.1111/jnc.12933] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/12/2014] [Accepted: 08/14/2014] [Indexed: 12/22/2022]
Abstract
HIV-1 invades CNS in the early course of infection, which can lead to the cascade of neuroinflammation. NADPH oxidases (NOXs) are the major producers of reactive oxygen species (ROS), which play important roles during pathogenic insults. The molecular mechanism of ROS generation via microRNA-mediated pathway in human microglial cells in response to HIV-1 Tat protein has been demonstrated in this study. Over-expression and knockdown of microRNAs, luciferase reporter assay, and site-directed mutagenesis are main molecular techniques used in this study. A significant reduction in miR-17 levels and increased NOX2, NOX4 expression levels along with ROS production were observed in human microglial cells upon HIV-1 Tat C exposure. The validation of NOX2 and NOX4 as direct targets of miR-17 was done by luciferase reporter assay. The over-expression and knockdown of miR-17 in human microglial cells showed the direct role of miR-17 in regulation of NOX2, NOX4 expression and intracellular ROS generation. We demonstrated the regulatory role of cellular miR-17 in ROS generation through over-expression and knockdown of miR-17 in human microglial cells exposed to HIV-1 Tat C protein. Activated microglial cells mediated neuroinflammatory events are observed in HIV-associated neurological disorders. The reduction in miR-17 levels was observed in microglial cells exposed to HIV-1 Tat C protein. miR-17 regulated the expression of NOX2 and NOX4, which in turn regulated the reactive oxygen species (ROS) production in microglial cells. Increased ROS production led to the activation of microglial cells and increased cytokine production. This study thus demonstrated a novel miR-17-mediated regulatory pathway of ROS production in microglial cells. HMC3 = human microglia clone 3 cell lines.
Collapse
Affiliation(s)
- Vaishnavi Sunil Jadhav
- Laboratory of Neurovirology and Inflammation Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad, India
| | | | | |
Collapse
|
31
|
Takeuchi H, Suzumura A. Gap junctions and hemichannels composed of connexins: potential therapeutic targets for neurodegenerative diseases. Front Cell Neurosci 2014; 8:189. [PMID: 25228858 PMCID: PMC4151093 DOI: 10.3389/fncel.2014.00189] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/19/2014] [Indexed: 12/03/2022] Open
Abstract
Microglia are macrophage-like resident immune cells that contribute to the maintenance of homeostasis in the central nervous system (CNS). Abnormal activation of microglia can cause damage in the CNS, and accumulation of activated microglia is a characteristic pathological observation in neurologic conditions such as trauma, stroke, inflammation, epilepsy, and neurodegenerative diseases. Activated microglia secrete high levels of glutamate, which damages CNS cells and has been implicated as a major cause of neurodegeneration in these conditions. Glutamate-receptor blockers and microglia inhibitors (e.g., minocycline) have been examined as therapeutic candidates for several neurodegenerative diseases; however, these compounds exerted little therapeutic benefit because they either perturbed physiological glutamate signals or suppressed the actions of protective microglia. The ideal therapeutic approach would hamper the deleterious roles of activated microglia without diminishing their protective effects. We recently found that abnormally activated microglia secrete glutamate via gap-junction hemichannels on the cell surface. Moreover, administration of gap-junction inhibitors significantly suppressed excessive microglial glutamate release and improved disease symptoms in animal models of neurologic conditions such as stroke, multiple sclerosis, amyotrophic lateral sclerosis, and Alzheimer's disease. Recent evidence also suggests that neuronal and glial communication via gap junctions amplifies neuroinflammation and neurodegeneration. Elucidation of the precise pathologic roles of gap junctions and hemichannels may lead to a novel therapeutic strategies that can slow and halt the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Hideyuki Takeuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University Nagoya, Japan
| | - Akio Suzumura
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University Nagoya, Japan
| |
Collapse
|
32
|
Assessment of tumor cells in a mouse model of diffuse infiltrative glioma by Raman spectroscopy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:860241. [PMID: 25247190 PMCID: PMC4163456 DOI: 10.1155/2014/860241] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 06/30/2014] [Accepted: 07/04/2014] [Indexed: 12/13/2022]
Abstract
Glioma of infiltrative nature is challenging for surgeons to achieve tumor-specific and maximal resection. Raman spectroscopy provides structural information on the targeted materials as vibrational shifts. We utilized Raman spectroscopy to distinguish invasive tumors from normal tissues. Spectra obtained from replication-competent avian sarcoma-(RCAS-) based infiltrative glioma cells and glioma tissues (resembling low-grade human glioma) were compared with those obtained from normal mouse astrocytes and normal tissues. In cell analysis, the spectra at 950-1000, 1030, 1050-1100, 1120-1130, 1120-1200, 1200-1300, 1300-1350, and 1450 cm(-1) were significantly higher in infiltrative glioma cells than in normal astrocytes. In brain tissue analysis, the spectra at 1030, 1050-1100, and 1200-1300 cm(-1) were significantly higher in infiltrative glioma tissues than in normal brain tissues. These spectra reflect the structures of proteins, lipids, and DNA content. The sensitivity and specificity to predict glioma cells by distinguishing normal cells were 98.3% and 75.0%, respectively. Principal component analysis elucidated the significance of spectral difference between tumor tissues and normal tissues. It is possible to distinguish invasive tumors from normal tissues by using Raman spectroscopy.
Collapse
|
33
|
Rotunno MS, Auclair JR, Maniatis S, Shaffer SA, Agar J, Bosco DA. Identification of a misfolded region in superoxide dismutase 1 that is exposed in amyotrophic lateral sclerosis. J Biol Chem 2014; 289:28527-38. [PMID: 25164820 DOI: 10.1074/jbc.m114.581801] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mutations and aberrant post-translational modifications within Cu,Zn-superoxide dismutase (SOD1) cause this otherwise protective enzyme to misfold, leading to amyotrophic lateral sclerosis (ALS). The C4F6 antibody selectively binds misfolded SOD1 in spinal cord tissues from postmortem human ALS cases, as well as from an ALS-SOD1 mouse model, suggesting that the C4F6 epitope reports on a pathogenic conformation that is common to misfolded SOD1 variants. To date, the residues and structural elements that comprise this epitope have not been elucidated. Using a chemical cross-linking and mass spectrometry approach, we identified the C4F6 epitope within several ALS-linked SOD1 variants, as well as an oxidized form of WT SOD1, supporting the notion that a similar misfolded conformation is shared among pathological SOD1 proteins. Exposure of the C4F6 epitope was modulated by the SOD1 electrostatic (loop VII) and zinc binding (loop IV) loops and correlated with SOD1-induced toxicity in a primary microglia activation assay. Site-directed mutagenesis revealed Asp(92) and Asp(96) as key residues within the C4F6 epitope required for the SOD1-C4F6 binding interaction. We propose that stabilizing the functional loops within SOD1 and/or obscuring the C4F6 epitope are viable therapeutic strategies for treating SOD1-mediated ALS.
Collapse
Affiliation(s)
- Melissa S Rotunno
- From the Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Jared R Auclair
- the Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115
| | - Stephanie Maniatis
- the Proteomics and Mass Spectrometry Facility, University of Massachusetts Medical School, Shrewsbury, Massachusetts 01545, and
| | - Scott A Shaffer
- the Proteomics and Mass Spectrometry Facility, University of Massachusetts Medical School, Shrewsbury, Massachusetts 01545, and the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Jeffrey Agar
- the Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115
| | - Daryl A Bosco
- From the Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
34
|
Abstract
Glioblastoma (GBM) is the most malignant brain tumor where patients' survival is only 14.6 months, despite multimodal therapy with debulking surgery, concurrent chemotherapy and radiotherapy. There is an urgent, unmet need for novel, effective therapeutic strategies for this devastating disease. Although several immunotherapies are under development for the treatment of GBM patients, the use of natural killer (NK) cells is still marginal despite this being a promising approach to treat cancer. In regard of our knowledge on the role of NG2/CSPG4 in promoting GBM aggressiveness we investigated the potential of an innovative immunotherapeutic strategy combining mAb9.2.27 against NG2/CSPG4 and NK cells in preclinical animal models of GBM. Multiple immune escape mechanisms maintain the tumor microenvironment in an anti-inflammatory state to promote tumor growth, however, the distinct roles of resident microglia versus recruited macrophages is not elucidated. We hypothesized that exploiting the cytokine release capabilities of activated (NK) cells to reverse the anti-inflammatory axis combined with mAb9.2.27 targeting the NG2/CSPG4 may favor tumor destruction by editing pro-GBM immune responses. Combination treatment with NK+mAb9.2.27 diminished tumor growth that was associated with reduced tumor proliferation, increased cellular apoptosis and prolonged survival compared to vehicle and monotherapy controls. The therapeutic efficacy was mediated by recruitment of CCR2low macrophages into the tumor microenvironment, increased ED1 and MHC class II expression on microglia that might render them competent for GBM antigen presentation, as well as elevated IFN-γ and TNF-α levels in the cerebrospinal fluid compared to controls. Depletion of systemic macrophages by liposome-encapsulated clodronate decreased the CCR2low macrophages recruited to the brain and abolished the beneficial outcomes. Moreover, mAb9.2.27 reversed tumor-promoting effects of patient-derived tumor-associated macrophage/microglia(TAM) ex vivo.Taken together, these findings indicate thatNK+mAb9.2.27 treatment may be an amenable therapeutic strategy to treat NG2/CSPG4 expressing GBMs. We provide a novel conceptual approach of combination immunotherapy for glioblastoma. The results traverse beyond the elucidation of NG2/CSPG4 as a therapeutic target, but demonstrate a proof of concept that this antibody may hold potential for the treatment of GBM by activation of tumor infiltrated microglia/macrophages.
Collapse
|
35
|
Noda M, Takii K, Parajuli B, Kawanokuchi J, Sonobe Y, Takeuchi H, Mizuno T, Suzumura A. FGF-2 released from degenerating neurons exerts microglial-induced neuroprotection via FGFR3-ERK signaling pathway. J Neuroinflammation 2014; 11:76. [PMID: 24735639 PMCID: PMC4022102 DOI: 10.1186/1742-2094-11-76] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/26/2014] [Indexed: 12/02/2022] Open
Abstract
Background The accumulation of activated microglia is a hallmark of various neurodegenerative diseases. Microglia may have both protective and toxic effects on neurons through the production of various soluble factors, such as chemokines. Indeed, various chemokines mediate the rapid and accurate migration of microglia to lesions. In the zebra fish, another well-known cellular migrating factor is fibroblast growth factor-2 (FGF-2). Although FGF-2 does exist in the mammalian central nervous system (CNS), it is unclear whether FGF-2 influences microglial function. Methods The extent of FGF-2 release was determined by ELISA, and the expression of its receptors was examined by immunocytochemistry. The effect of several drug treatments on a neuron and microglia co-culture system was estimated by immunocytochemistry, and the neuronal survival rate was quantified. Microglial phagocytosis was evaluated by immunocytochemistry and quantification, and microglial migration was estimated by fluorescence-activated cell sorting (FACS). Molecular biological analyses, such as Western blotting and promoter assay, were performed to clarify the FGF-2 downstream signaling pathway in microglia. Results Fibroblast growth factor-2 is secreted by neurons when damaged by glutamate or oligomeric amyloid β 1-42. FGF-2 enhances microglial migration and phagocytosis of neuronal debris, and is neuroprotective against glutamate toxicity through FGFR3-extracellular signal-regulated kinase (ERK) signaling pathway, which is directly controlled by Wnt signaling in microglia. Conclusions FGF-2 secreted from degenerating neurons may act as a ‘help-me’ signal toward microglia by inducing migration and phagocytosis of unwanted debris.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tetsuya Mizuno
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan.
| | | |
Collapse
|
36
|
Sirtuin 1 attenuates oxidative stress via upregulation of superoxide dismutase 2 and catalase in astrocytes. J Neuroimmunol 2014; 269:38-43. [DOI: 10.1016/j.jneuroim.2014.02.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/30/2014] [Accepted: 02/05/2014] [Indexed: 12/11/2022]
|
37
|
Yao Y, Tsirka SE. Monocyte chemoattractant protein-1 and the blood-brain barrier. Cell Mol Life Sci 2014; 71:683-97. [PMID: 24051980 PMCID: PMC3946874 DOI: 10.1007/s00018-013-1459-1] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 07/20/2013] [Accepted: 08/19/2013] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB) is a dynamic structure that maintains the homeostasis of the brain and thus proper neurological functions. BBB compromise has been found in many pathological conditions, including neuroinflammation. Monocyte chemoattractant protein-1 (MCP1), a chemokine that is transiently and significantly up-regulated during inflammation, is able to disrupt the integrity of BBB and modulate the progression of various diseases, including excitotoxic injury and hemorrhage. In this review, we first introduce the biochemistry and biology of MCP1, and then summarize the effects of MCP1 on BBB integrity as well as individual BBB components.
Collapse
Affiliation(s)
- Yao Yao
- Program in Molecular and Cellular Pharmacology, Department of Pharmacological Sciences, BST8-192, Stony Brook University, Stony Brook, NY 11794-8651 USA
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065 USA
| | - Stella E. Tsirka
- Program in Molecular and Cellular Pharmacology, Department of Pharmacological Sciences, BST8-192, Stony Brook University, Stony Brook, NY 11794-8651 USA
| |
Collapse
|
38
|
Olsson T. Role of cytokines in multiple sclerosis and experimental autoimmune encephalomyelitis. Eur J Neurol 2013; 1:7-19. [PMID: 24283424 DOI: 10.1111/j.1468-1331.1994.tb00045.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- T Olsson
- Division of Neurology, Department of Clinical Neuroscience and Family Medicine, Karolinska Institute, Huddinge Hospital, S-141 86 Huddinge, Sweden
| |
Collapse
|
39
|
Affiliation(s)
- Hideyuki Takeuchi
- Department of Neuroimmunology; Research Institute of Environmental Medicine; Nagoya University; Nagoya Japan
| |
Collapse
|
40
|
Hernandez-Ontiveros DG, Tajiri N, Acosta S, Giunta B, Tan J, Borlongan CV. Microglia activation as a biomarker for traumatic brain injury. Front Neurol 2013; 4:30. [PMID: 23531681 PMCID: PMC3607801 DOI: 10.3389/fneur.2013.00030] [Citation(s) in RCA: 201] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 03/10/2013] [Indexed: 12/24/2022] Open
Abstract
Traumatic brain injury (TBI) has become the signature wound of wars in Afghanistan and Iraq. Injury may result from a mechanical force, a rapid acceleration-deceleration movement, or a blast wave. A cascade of secondary cell death events ensues after the initial injury. In particular, multiple inflammatory responses accompany TBI. A series of inflammatory cytokines and chemokines spreads to normal brain areas juxtaposed to the core impacted tissue. Among the repertoire of immune cells involved, microglia is a key player in propagating inflammation to tissues neighboring the core site of injury. Neuroprotective drug trials in TBI have failed, likely due to their sole focus on abrogating neuronal cell death and ignoring the microglia response despite these inflammatory cells’ detrimental effects on the brain. Another relevant point to consider is the veracity of results of animal experiments due to deficiencies in experimental design, such as incomplete or inadequate method description, data misinterpretation, and reporting may introduce bias and give false-positive results. Thus, scientific publications should follow strict guidelines that include randomization, blinding, sample-size estimation, and accurate handling of all data (Landis et al., 2012). A prolonged state of inflammation after brain injury may linger for years and predispose patients to develop other neurological disorders, such as Alzheimer’s disease. TBI patients display progressive and long-lasting impairments in their physical, cognitive, behavioral, and social performance. Here, we discuss inflammatory mechanisms that accompany TBI in an effort to increase our understanding of the dynamic pathological condition as the disease evolves over time and begin to translate these findings for defining new and existing inflammation-based biomarkers and treatments for TBI.
Collapse
Affiliation(s)
- Diana G Hernandez-Ontiveros
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | | | | | | | | | | |
Collapse
|
41
|
Kawanokuchi J, Takeuchi H, Sonobe Y, Mizuno T, Suzumura A. Interleukin-27 promotes inflammatory and neuroprotective responses in microglia. ACTA ACUST UNITED AC 2013. [DOI: 10.1111/cen3.12005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Jun Kawanokuchi
- Department of Neuroimmunology; Research Institute of Environmental Medicine; Nagoya University; Nagoya; Japan
| | - Hideyuki Takeuchi
- Department of Neuroimmunology; Research Institute of Environmental Medicine; Nagoya University; Nagoya; Japan
| | - Yoshifumi Sonobe
- Department of Neuroimmunology; Research Institute of Environmental Medicine; Nagoya University; Nagoya; Japan
| | - Tetsuya Mizuno
- Department of Neuroimmunology; Research Institute of Environmental Medicine; Nagoya University; Nagoya; Japan
| | - Akio Suzumura
- Department of Neuroimmunology; Research Institute of Environmental Medicine; Nagoya University; Nagoya; Japan
| |
Collapse
|
42
|
Noda H, Takeuchi H, Mizuno T, Suzumura A. Fingolimod phosphate promotes the neuroprotective effects of microglia. J Neuroimmunol 2013; 256:13-8. [PMID: 23290828 DOI: 10.1016/j.jneuroim.2012.12.005] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 11/19/2012] [Accepted: 12/11/2012] [Indexed: 12/25/2022]
Abstract
Fingolimod phosphate (FTY720) is a sphingosine 1-phosphate (S1P) receptor agonist that is being used as a new oral drug for multiple sclerosis. FTY720 prevents lymphocytes from moving out of the lymphoid organs and inhibits autoreactive lymphocytes from infiltrating the central nervous system. Whether FTY720 directly affects microglia-the innate immune cells of the central nervous system-is unclear. Here we show that FTY720 binds S1P1 receptors to downregulate activated microglial production of such pro-inflammatory cytokines as tumor necrosis factor-α, interleukin-1β, and interleukin-6. FTY720 also upregulates microglial production of brain-derived neurotrophic factor and glial cell-derived neurotrophic factor. These results suggested that FTY720 directly promotes the neuroprotective effects of microglia. Therefore, FTY720 may be a potent therapeutic agent for not only multiple sclerosis but also other neurologic diseases associated with microglial activation.
Collapse
Affiliation(s)
- Hiromi Noda
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | | | | | | |
Collapse
|
43
|
Horiuchi M, Wakayama K, Itoh A, Kawai K, Pleasure D, Ozato K, Itoh T. Interferon regulatory factor 8/interferon consensus sequence binding protein is a critical transcription factor for the physiological phenotype of microglia. J Neuroinflammation 2012; 9:227. [PMID: 23020843 PMCID: PMC3546867 DOI: 10.1186/1742-2094-9-227] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 08/13/2012] [Indexed: 01/15/2023] Open
Abstract
Background Recent fate-mapping studies establish that microglia, the resident mononuclear phagocytes of the CNS, are distinct in origin from the bone marrow-derived myeloid lineage. Interferon regulatory factor 8 (IRF8, also known as interferon consensus sequence binding protein) plays essential roles in development and function of the bone marrow-derived myeloid lineage. However, little is known about its roles in microglia. Methods The CNS tissues of IRF8-deficient mice were immunohistochemically analyzed. Pure microglia isolated from wild-type and IRF8-deficient mice were studied in vitro by proliferation, immunocytochemical and phagocytosis assays. Microglial response in vivo was compared between wild-type and IRF8-deficient mice in the cuprizon-induced demyelination model. Results Our analysis of IRF8-deficient mice revealed that, in contrast to compromised development of IRF8-deficient bone marrow myeloid lineage cells, development and colonization of microglia are not obviously affected by loss of IRF8. However, IRF8-deficient microglia demonstrate several defective phenotypes. In vivo, IRF8-deficient microglia have fewer elaborated processes with reduced expression of IBA1/AIF1 compared with wild-type microglia, suggesting a defective phenotype. IRF8-deficient microglia are significantly less proliferative in mixed glial cultures than wild-type microglia. Unlike IRF8-deficient bone marrow myeloid progenitors, exogenous macrophage colony stimulating factor (colony stimulating factor 1) (M-CSF (CSF1)) restores their proliferation in mixed glial cultures. In addition, IRF8-deficient microglia exhibit an exaggerated growth response to exogenous granulocyte-macrophage colony stimulating factor (colony stimulating factor 2) (GM-CSF (CSF2)) in the presence of other glial cells. IRF8-deficient microglia also demonstrate altered cytokine expressions in response to interferon-gamma and lipopolysaccharide in vitro. Moreover, the maximum phagocytic capacity of IRF8-deficient microglia is reduced, although their engulfment of zymosan particles is not overtly impaired. Defective scavenging activity of IRF8-deficient microglia was further confirmed in vivo in the cuprizone-induced demyelination model in mice. Conclusions This study is the first to demonstrate the essential contribution of IRF8-mediated transcription to a broad range of microglial phenotype. Microglia are distinct from the bone marrow myeloid lineage with respect to their dependence on IRF8-mediated transcription.
Collapse
Affiliation(s)
- Makoto Horiuchi
- Department of Neurology, University of California Davis, School of Medicine, 4860 Y Street, Sacramento, CA 95817, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
The neurotoxic effect of astrocytes activated with toll-like receptor ligands. J Neuroimmunol 2012; 254:10-8. [PMID: 22999806 DOI: 10.1016/j.jneuroim.2012.08.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/17/2012] [Accepted: 08/20/2012] [Indexed: 12/25/2022]
Abstract
Toll-like receptors (TLRs) are key molecules in the innate immune system in the central nervous system. Although astrocytes are believed to play physiological roles in regulating neuronal activity and synaptic transmission, activated astrocytes may also be toxic to neurons. Here, we show that the ligands for TLRs 2, 4, 5 and 6 induce neuronal cell death in neuron-astrocytes co-cultures through the production of reactive oxygen species (ROS). Inhibition of ROS production by NADPH oxidase inhibitor apocynin significantly suppresses neuronal cell death. ROS induced in astrocytes via TLRs may be involved in neuroinflammation and a therapeutic target for neurotoxicity by activated astrocytes.
Collapse
|
45
|
Ma D, Doi Y, Jin S, Li E, Sonobe Y, Takeuchi H, Mizuno T, Suzumura A. TGF-β induced by interleukin-34-stimulated microglia regulates microglial proliferation and attenuates oligomeric amyloid β neurotoxicity. Neurosci Lett 2012; 529:86-91. [PMID: 22985514 DOI: 10.1016/j.neulet.2012.08.071] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/16/2012] [Accepted: 08/26/2012] [Indexed: 12/22/2022]
Abstract
Microglia play critical roles in the pathogenesis of Alzheimer's disease (AD). We have previously shown that interleukin-34 (IL-34) enhances microglial proliferation and induces microglial neuroprotective properties against oligomeric amyloid β (oAβ) toxicity by producing insulin degrading enzyme, an Aβ degrading enzyme, and anti-oxidant enzyme heme oxygenase-1. In this study, we found that IL-34 dose-dependently induces TGF-β in microglia, and that TGF-β attenuates oAβ neurotoxicity in neuron microglial co-cultures. The TGF-β 1 receptor kinase inhibitor SD208 enhances microglial proliferation by IL-34 and suppresses the neuroprotective effect of IL-34-treated microglia. These findings suggest that TGF-β produced by IL-34-treated microglia is a negative regulator of microglial proliferation and enhances the neuroprotective property of microglia.
Collapse
Affiliation(s)
- Di Ma
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | | | | | | | | | | | | | | |
Collapse
|
46
|
The neuroprotective effects of milk fat globule-EGF factor 8 against oligomeric amyloid β toxicity. J Neuroinflammation 2012; 9:148. [PMID: 22742657 PMCID: PMC3418568 DOI: 10.1186/1742-2094-9-148] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 05/01/2012] [Indexed: 12/25/2022] Open
Abstract
Background Phosphatidylserine receptor is a key molecule that mediates the phagocytosis of apoptotic cells. Milk fat globule-EGF factor 8 (MFG-E8) is a phosphatidylserine receptor that is expressed on various macrophage lineage cells, including microglia in the central nervous system (CNS). Targeted clearance of degenerated neurons by microglia is essential to maintain healthy neural networks. We previously showed that the CX3C chemokine fractalkine is secreted from degenerated neurons and accelerates microglial clearance of neuronal debris via inducing the release of MFG-E8. However, the mechanisms by which microglia produce MFG-E8 and the precise functions of MFG-E8 are unknown. Methods The release of MFG-E8 from microglia treated with conditioned medium from neurons exposed to neurotoxic substances, glutamate or oligomeric amyloid β (oAβ) was measured by ELISA. The neuroprotective effects of MFG-E8 and MFG-E8 − induced microglial phagocytosis of oAβ were assessed by immunocytochemistry. The effects of MFG-E8 on the production of the anti-oxidative enzyme hemeoxygenase-1 (HO-1) were determined by ELISA and immunocytochemisty. Results MFG-E8 was induced in microglia treated with conditioned medium from neurons that had been exposed to neurotoxicants, glutamate or oAβ. MFG-E8 significantly attenuated oAβ-induced neuronal cell death in a primary neuron − microglia coculture system. Microglial phagocytosis of oAβ was accelerated by MFG-E8 treatment due to increased CD47 expression in the absence of neurotoxic molecule production, such as tumor necrosis factor-α, nitric oxide, and glutamate. MFG-E8 − treated microglia induced nuclear factor E(2) − related factor 2 (Nrf2) − mediated HO-1 production, which also contributed to neuroprotection. Conclusions These results suggest that microglia release MFG-E8 in response to signals from degenerated neurons and that MFG-E8 protects oAβ-induced neuronal cell death by promoting microglial phagocytic activity and activating the Nrf2-HO-1 pathway. Thus, MFG-E8 may have novel roles as a neuroprotectant in neurodegenerative conditions.
Collapse
|
47
|
Parajuli B, Sonobe Y, Kawanokuchi J, Doi Y, Noda M, Takeuchi H, Mizuno T, Suzumura A. Immunoglobulin G(1) immune complex upregulates interferon-γ-induced nitric oxide production via ERK1/2 activation in murine microglia. J Neuroimmunol 2012; 244:57-62. [PMID: 22306300 DOI: 10.1016/j.jneuroim.2012.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 12/28/2011] [Accepted: 01/09/2012] [Indexed: 01/05/2023]
Abstract
Intrathecal Immunoglobulin G (IgG) is elevated in some central nervous system (CNS) diseases and microglia upregulate Fcγ receptors in various neurological disorders. However, the interaction between IgG or IgG immune complexes and microglial Fcγ receptors is not fully understood. In this study, the effect of IgG(1) immune complexes on microglia was investigated. IgG(1) immune complexes increased nitric oxide production in murine microglia in the presence of interferon (IFN)-γ. These effects were dependent upon IgG(1) immune complex-induced activation of spleen tyrosine kinase with subsequent activation of extracellular signal regulated kinase1/2. Collectively, these results indicate that IgG(1) immune complexes can exert immunomodulatory effects in various central nervous system disorders.
Collapse
Affiliation(s)
- Bijay Parajuli
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Leitner DF, Connor JR. Functional roles of transferrin in the brain. Biochim Biophys Acta Gen Subj 2011; 1820:393-402. [PMID: 22138408 DOI: 10.1016/j.bbagen.2011.10.016] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 10/13/2011] [Accepted: 10/24/2011] [Indexed: 12/11/2022]
Abstract
BACKGROUND Transferrin is synthesized in the brain by choroid plexus and oligodendrocytes, but only that in the choroid plexus is secreted. Transferrin is a major iron delivery protein to the brain, but the amount transcytosed across the brain microvasculature is minimal. Transferrin is the major source of iron delivery to neurons. It may deliver iron to immature oligodendrocytes but this trophic effect declines over time while iron requirements for maintaining myelination continue. Finally, transferrin may play an important role in neurodegenerative diseases through its ability to mobilize iron. SCOPE OF REVIEW The role of transferrin in maintaining brain iron homeostasis and the mechanism by which it enters the brain and delivers iron will be discussed. Its relevance to neurological disorders will also be addressed. MAJOR CONCLUSIONS Transferrin is the major iron delivery protein for neurons and the microvasculature, but has a limited role for glial cells. The main source of transferrin in the brain is likely from the choroid plexus although the concentration of transferrin at any given time in the brain includes that synthesized in oligodendrocytes. Little is known about brain iron egress or the role of transferrin in this process. GENERAL SIGNIFICANCE Neuron survival requires iron, which is predominantly delivered by transferrin. The concentration of transferrin in the cerebrospinal fluid is reflective of brain iron availability and can function as a biomarker in disease. Accumulation of iron in the brain contributes to neurodegenerative processes, thus an understanding of the role that transferrin plays in regulating brain iron homeostasis is essential. This article is part of a Special Issue entitled Transferrins: Molecular mechanisms of iron transport and disorders.
Collapse
Affiliation(s)
- Dominique F Leitner
- Department of Neurosurgery, Penn State University, M.S. Hershey Medical Center, 500 University Dr., Hershey, PA 17033-0850, USA
| | | |
Collapse
|
49
|
Endong L, Shijie J, Sonobe Y, Di M, Hua L, Kawanokuchi J, Mizuno T, Suzumura A. The gap-junction inhibitor carbenoxolone suppresses the differentiation of Th17 cells through inhibition of IL-23 expression in antigen presenting cells. J Neuroimmunol 2011; 240-241:58-64. [PMID: 22036952 DOI: 10.1016/j.jneuroim.2011.09.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 09/27/2011] [Accepted: 09/27/2011] [Indexed: 01/08/2023]
Abstract
Carbenoxolone (CBX) is a widely used gap-junction inhibitor. We have previously shown that treatment with CBX significantly delayed the onset of experimental autoimmune encephalomyelitis (EAE). However, the mechanism by which CBX delays the onset of EAE remains to be elucidated. Here, we show that CBX specifically inhibits the production of IL-23 by dendritic cells (DCs) and microglia in vitro. CBX treatment significantly reduced the population of Th17 cells in EAE mice. Furthermore, CBX downregulated the expression of IL-23 p19 via increased production of protein phosphatase 2A (PP2A). Thus, CBX may be an effective therapeutic strategy against Th17-mediated autoimmune diseases, such as multiple sclerosis.
Collapse
Affiliation(s)
- Li Endong
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Mizuno T, Doi Y, Mizoguchi H, Jin S, Noda M, Sonobe Y, Takeuchi H, Suzumura A. Interleukin-34 selectively enhances the neuroprotective effects of microglia to attenuate oligomeric amyloid-β neurotoxicity. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2016-27. [PMID: 21872563 DOI: 10.1016/j.ajpath.2011.06.011] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 06/14/2011] [Accepted: 06/17/2011] [Indexed: 12/11/2022]
Abstract
Microglia, macrophage-like resident immune cells in the brain, possess both neurotoxic and neuroprotective properties and have a critical role in the development of Alzheimer's disease (AD). We examined the function of Interleukin-34 (IL-34), a newly discovered cytokine, on microglia because it reportedly induces proliferation of monocytes and macrophages. We observed that the neuronal cells primarily produce IL-34 and that microglia express its receptor, colony-stimulating factor 1 receptor. IL-34 promoted microglial proliferation and clearance of soluble oligomeric amyloid-β (oAβ), which mediates synaptic dysfunction and neuronal damage in AD. IL-34 increased the expression of insulin-degrading enzyme, aiding the clearance of oAβ, and induced the antioxidant enzyme heme oxygenase-1 in microglia to reduce oxidative stress, without producing neurotoxic molecules. Consequently, microglia treated with IL-34 attenuated oAβ neurotoxicity in primary neuron-microglia co-cultures. In vivo, intracerebroventricular administration of IL-34 ameliorated impairment of associative learning and reduced oAβ levels through up-regulation of insulin-degrading enzyme and heme oxygenase-1 in an APP/PS1 transgenic mouse model of AD. These findings support the idea that enhancement of the neuroprotective property of microglia by IL-34 may be an effective approach against oAβ neurotoxicity in AD.
Collapse
Affiliation(s)
- Tetsuya Mizuno
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.
| | | | | | | | | | | | | | | |
Collapse
|