1
|
Reagin KL, Lee R, Williams LA, Cocciolone L, Funk KE. Compromised CD8+ T cell immunity in the aged brain increases severity of neurotropic coronavirus infection and postinfectious cognitive impairment. Aging Cell 2025; 24:e14409. [PMID: 39550693 PMCID: PMC11896202 DOI: 10.1111/acel.14409] [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: 07/11/2024] [Revised: 10/04/2024] [Accepted: 10/23/2024] [Indexed: 11/18/2024] Open
Abstract
Advanced age increases the risk of severe disease from SARS-CoV-2 infection, as well as incidence of long COVID and SARS-CoV-2 reinfection. We hypothesized that perturbations in the aged antiviral CD8+ T cell response predisposes elderly individuals to severe coronavirus infection, re-infection, and postinfectious cognitive sequelae. Using MHV-A59 as a murine model of respiratory coronavirus, we found that aging increased CNS infection and lethality to MHV infection. This was coupled with increased CD8+ T cells within the aged CNS but reduced antigen specificity. Aged animals also displayed a decreased proportion of CD103+ resident memory cells (TRM), which correlated with increased severity of secondary viral challenge. Using a reciprocal adoptive transfer paradigm, data show that not only were fewer aged CD8+ T cells retained within the adult brain post-infection, but also that adult CD8+ cells expressed lower levels of TRM marker CD103 when in the aged microenvironment. Furthermore, aged animals demonstrated spatial learning impairment following MHV infection, which worsened in both aged and adult animals following secondary viral challenge. Spatial learning impairment was accompanied by increased TUNEL positivity in hippocampal neurons, suggestive of neuronal apoptosis. Additionally, primary cell coculture showed that activated CD8+ T cells induced TUNEL positivity in neurons, independent of antigen-specificity. Altogether, these results show that non-antigen specific CD8+ T cells are recruited to the aged brain and cause broad neuronal death without establishing a TRM phenotype that confers lasting protection against a secondary infection.
Collapse
Affiliation(s)
- Katie L. Reagin
- Department of Biological SciencesUniversity of North Carolina at CharlotteCharlotteNorth CarolinaUSA
| | - Rae‐Ling Lee
- Department of Biological SciencesUniversity of North Carolina at CharlotteCharlotteNorth CarolinaUSA
| | - Luke A. Williams
- Department of Biological SciencesUniversity of North Carolina at CharlotteCharlotteNorth CarolinaUSA
| | - Loren Cocciolone
- Department of Biological SciencesUniversity of North Carolina at CharlotteCharlotteNorth CarolinaUSA
| | - Kristen E. Funk
- Department of Biological SciencesUniversity of North Carolina at CharlotteCharlotteNorth CarolinaUSA
| |
Collapse
|
2
|
Tang Y, Wu X, Li J, Li Y, Xu X, Li G, Zhang P, Qin C, Wu LJ, Tang Z, Tian DS. The Emerging Role of Microglial Hv1 as a Target for Immunomodulation in Myelin Repair. Aging Dis 2024; 15:1176-1203. [PMID: 38029392 PMCID: PMC11081154 DOI: 10.14336/ad.2023.1107] [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: 08/17/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
In the central nervous system (CNS), the myelin sheath ensures efficient interconnection between neurons and contributes to the regulation of the proper function of neuronal networks. The maintenance of myelin and the well-organized subtle process of myelin plasticity requires cooperation among myelin-forming cells, glial cells, and neural networks. The process of cooperation is fragile, and the balance is highly susceptible to disruption by microenvironment influences. Reactive microglia play a critical and complicated role in the demyelination and remyelination process. Recent studies have shown that the voltage-gated proton channel Hv1 is selectively expressed in microglia in CNS, which regulates intracellular pH and is involved in the production of reactive oxygen species, underlying multifaceted roles in maintaining microglia function. This paper begins by examining the molecular mechanisms of demyelination and emphasizes the crucial role of the microenvironment in demyelination. It focuses specifically on the role of Hv1 in myelin repair and its therapeutic potential in CNS demyelinating diseases.
Collapse
Affiliation(s)
- Yingxin Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoxiao Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gaigai Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Weerasinghe-Mudiyanselage PDE, Kang S, Kim JS, Kim SH, Wang H, Shin T, Moon C. Changes in structural plasticity of hippocampal neurons in an animal model of multiple sclerosis. Zool Res 2024; 45:398-414. [PMID: 38485508 PMCID: PMC11017077 DOI: 10.24272/j.issn.2095-8137.2023.309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/25/2023] [Indexed: 03/19/2024] Open
Abstract
Structural plasticity is critical for the functional diversity of neurons in the brain. Experimental autoimmune encephalomyelitis (EAE) is the most commonly used model for multiple sclerosis (MS), successfully mimicking its key pathological features (inflammation, demyelination, axonal loss, and gliosis) and clinical symptoms (motor and non-motor dysfunctions). Recent studies have demonstrated the importance of synaptic plasticity in EAE pathogenesis. In the present study, we investigated the features of behavioral alteration and hippocampal structural plasticity in EAE-affected mice in the early phase (11 days post-immunization, DPI) and chronic phase (28 DPI). EAE-affected mice exhibited hippocampus-related behavioral dysfunction in the open field test during both early and chronic phases. Dendritic complexity was largely affected in the cornu ammonis 1 (CA1) and CA3 apical and dentate gyrus (DG) subregions of the hippocampus during the chronic phase, while this effect was only noted in the CA1 apical subregion in the early phase. Moreover, dendritic spine density was reduced in the hippocampal CA1 and CA3 apical/basal and DG subregions in the early phase of EAE, but only reduced in the DG subregion during the chronic phase. Furthermore, mRNA levels of proinflammatory cytokines ( Il1β, Tnfα, and Ifnγ) and glial cell markers ( Gfap and Cd68) were significantly increased, whereas the expression of activity-regulated cytoskeleton-associated protein (ARC) was reduced during the chronic phase. Similarly, exposure to the aforementioned cytokines in primary cultures of hippocampal neurons reduced dendritic complexity and ARC expression. Primary cultures of hippocampal neurons also showed significantly reduced extracellular signal-regulated kinase (ERK) phosphorylation upon treatment with proinflammatory cytokines. Collectively, these results suggest that autoimmune neuroinflammation alters structural plasticity in the hippocampus, possibly through the ERK-ARC pathway, indicating that this alteration may be associated with hippocampal dysfunctions in EAE.
Collapse
Affiliation(s)
- Poornima D E Weerasinghe-Mudiyanselage
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Sohi Kang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Sung-Ho Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hongbing Wang
- Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Republic of Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea. E-mail:
| |
Collapse
|
4
|
Weerasinghe-Mudiyanselage PDE, Kim JS, Shin T, Moon C. Understanding the spectrum of non-motor symptoms in multiple sclerosis: insights from animal models. Neural Regen Res 2024; 19:84-91. [PMID: 37488849 PMCID: PMC10479859 DOI: 10.4103/1673-5374.375307] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 07/26/2023] Open
Abstract
Multiple sclerosis is a chronic autoimmune disease of the central nervous system and is generally considered to be a non-traumatic, physically debilitating neurological disorder. In addition to experiencing motor disability, patients with multiple sclerosis also experience a variety of non-motor symptoms, including cognitive deficits, anxiety, depression, sensory impairments, and pain. However, the pathogenesis and treatment of such non-motor symptoms in multiple sclerosis are still under research. Preclinical studies for multiple sclerosis benefit from the use of disease-appropriate animal models, including experimental autoimmune encephalomyelitis. Prior to understanding the pathophysiology and developing treatments for non-motor symptoms, it is critical to characterize the animal model in terms of its ability to replicate certain non-motor features of multiple sclerosis. As such, no single animal model can mimic the entire spectrum of symptoms. This review focuses on the non-motor symptoms that have been investigated in animal models of multiple sclerosis as well as possible underlying mechanisms. Further, we highlighted gaps in the literature to explain the non-motor aspects of multiple sclerosis in experimental animal models, which will serve as the basis for future studies.
Collapse
Affiliation(s)
- Poornima D. E. Weerasinghe-Mudiyanselage
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR program, Chonnam National University, Gwangju, Republic of Korea
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR program, Chonnam National University, Gwangju, Republic of Korea
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR program, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
5
|
Vecchiarelli HA, Lopes LT, Paolicelli RC, Stevens B, Wake H, Tremblay MÈ. Synapse Regulation. ADVANCES IN NEUROBIOLOGY 2024; 37:179-208. [PMID: 39207693 DOI: 10.1007/978-3-031-55529-9_11] [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
Microglia are the resident immune cells of the brain. As such, they rapidly detect changes in normal brain homeostasis and accurately respond by fine-tuning in a tightly regulated manner their morphology, gene expression, and functional behavior. Depending on the nature of these changes, microglia can thicken and retract their processes, proliferate and migrate, release numerous signaling factors and compounds influencing neuronal physiology (e.g., cytokines and trophic factors), in addition to secreting proteases able to transform the extracellular matrix, and phagocytosing various types of cellular debris, etc. Because microglia also transform rapidly (on a time scale of minutes) during experimental procedures, studying these very special cells requires methods that are specifically non-invasive. The development of such methods has provided unprecedented insights into the roles of microglia during normal physiological conditions. In particular, transcranial two-photon in vivo imaging revealed that presumably "resting" microglia continuously survey the brain parenchyma with their highly motile processes, in addition to modulating their structural and functional interactions with neuronal circuits along the changes in neuronal activity and behavioral experience occurring throughout the lifespan. In this chapter, we will describe how surveillant microglia interact with synaptic elements and modulate the number, maturation, function, and plasticity of synapses in the healthy developing, mature, and aging brain, with consequences on neuronal activity, learning and memory, and the behavioral outcome.
Collapse
Affiliation(s)
| | | | - Rosa C Paolicelli
- Division of Psychiatry Research, University of Zurich, Schlieren, Switzerland
| | - Beth Stevens
- Department of Neurology, Harvard Medical School, Center for Life Science, Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA
| | - Hiroaki Wake
- Division of Brain Circuits, National Institute for Basic Biology, Myodaiji-cho, Okazaki, Japan
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
6
|
von Bernhardi R, Eugenín J. Aging Microglia and Their Impact in the Nervous System. ADVANCES IN NEUROBIOLOGY 2024; 37:379-395. [PMID: 39207703 DOI: 10.1007/978-3-031-55529-9_21] [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
Aging is the greatest risk factor for neurodegenerative diseases. Microglia are the resident immune cells in the central nervous system (CNS), playing key roles in its normal functioning, and as mediators for age-dependent changes of the CNS, condition at which they generate a hostile environment for neurons. Transforming Growth Factor β1 (TGFβ1) is a regulatory cytokine involved in immuneregulation and neuroprotection, affecting glial cell inflammatory activation, neuronal survival, and function. TGFβ1 signaling undergoes age-dependent changes affecting the regulation of microglial cells and can contribute to the pathophysiology of neurodegenerative diseases. This chapter focuses on assessing the role of age-related changes on the regulation of microglial cells and their impact on neuroinflammation and neuronal function, for understanding age-dependent changes of the nervous system.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Faculty of Odontology and Rehabilitation Sciences, Universidad San Sebastian, Santiago, Chile.
| | - Jaime Eugenín
- Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
7
|
Bellingacci L, Canonichesi J, Mancini A, Parnetti L, Di Filippo M. Cytokines, synaptic plasticity and network dynamics: a matter of balance. Neural Regen Res 2023; 18:2569-2572. [PMID: 37449591 DOI: 10.4103/1673-5374.371344] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
The modern view of the immune system as a sensitizing and modulating machinery of the central nervous system is now well recognized. However, the specific mechanisms underlying this fine crosstalk have yet to be fully disentangled. To control cognitive function and behavior, the two systems are engaged in a subtle interacting act. In this scenario, a dual action of pro-inflammatory cytokines in the modulation of brain network connections is emerging. Pro-inflammatory cytokines are indeed required to express physiological plasticity in the hippocampal network while being detrimental when over-expressed during uncontrolled inflammatory processes. In this dynamic equilibrium, synaptic functioning and the performance of neural networks are ensured by maintaining an appropriate balance between pro- and anti-inflammatory molecules in the central nervous system microenvironment.
Collapse
Affiliation(s)
- Laura Bellingacci
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Jacopo Canonichesi
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
8
|
Zilberter Y, Tabuena DR, Zilberter M. NOX-induced oxidative stress is a primary trigger of major neurodegenerative disorders. Prog Neurobiol 2023; 231:102539. [PMID: 37838279 PMCID: PMC11758986 DOI: 10.1016/j.pneurobio.2023.102539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023]
Abstract
Neurodegenerative diseases (NDDs) causing cognitive impairment and dementia are difficult to treat due to the lack of understanding of primary initiating factors. Meanwhile, major sporadic NDDs share many risk factors and exhibit similar pathologies in their early stages, indicating the existence of common initiation pathways. Glucose hypometabolism associated with oxidative stress is one such primary, early and shared pathology, and a likely major cause of detrimental disease-associated cascades; targeting this common pathology may therefore be an effective preventative strategy for most sporadic NDDs. However, its exact cause and trigger remain unclear. Recent research suggests that early oxidative stress caused by NADPH oxidase (NOX) activation is a shared initiating mechanism among major sporadic NDDs and could prove to be the long-sought ubiquitous NDD trigger. We focus on two major NDDs - Alzheimer's disease (AD) and Parkinson's disease (PD), as well as on acquired epilepsy which is an increasingly recognized comorbidity in NDDs. We also discuss available data suggesting the relevance of the proposed mechanisms to other NDDs. We delve into the commonalities among these NDDs in neuroinflammation and NOX involvement to identify potential therapeutic targets and gain a deeper understanding of the underlying causes of NDDs.
Collapse
Affiliation(s)
- Yuri Zilberter
- Aix-Marseille Université, INSERM UMR1106, Institut de Neurosciences des Systèmes, Marseille, France
| | - Dennis R Tabuena
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Misha Zilberter
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA.
| |
Collapse
|
9
|
Menculini G, Mancini A, Gaetani L, Bellingacci L, Tortorella A, Parnetti L, Di Filippo M. Psychiatric symptoms in multiple sclerosis: a biological perspective on synaptic and network dysfunction. J Neurol Neurosurg Psychiatry 2023; 94:389-395. [PMID: 36653171 DOI: 10.1136/jnnp-2022-329806] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 12/30/2022] [Indexed: 01/20/2023]
Abstract
Psychiatric symptoms frequently occur in multiple sclerosis (MS), presenting with a complex phenomenology that encompasses a large clinical spectrum from clear-cut psychiatric disorders up to isolated psychopathological manifestations. Despite their relevant impact on the overall disease burden, such clinical features are often misdiagnosed, receive suboptimal treatment and are not systematically evaluated in the quantification of disease activity. The development of psychiatric symptoms in MS underpins a complex pathogenesis involving both emotional reactions to a disabling disease and structural multifocal central nervous system damage. Here, we review MS psychopathological manifestations under a biological perspective, highlighting the pathogenic relevance of synaptic and neural network dysfunction. Evidence obtained from human and experimental disease models suggests that MS-related psychiatric phenomenology is part of a disconnection syndrome due to diffuse inflammatory and neurodegenerative brain damage.
Collapse
Affiliation(s)
- Giulia Menculini
- Section of Psychiatry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Laura Bellingacci
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alfonso Tortorella
- Section of Psychiatry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
10
|
Kann O, Almouhanna F, Chausse B. Interferon γ: a master cytokine in microglia-mediated neural network dysfunction and neurodegeneration. Trends Neurosci 2022; 45:913-927. [PMID: 36283867 DOI: 10.1016/j.tins.2022.10.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
Traditionally, lymphocytic interferon γ (IFN-γ) was considered to be a simple 'booster' of proinflammatory responses by microglia (brain-resident macrophages) during bacterial or viral infection. Recent slice culture (in situ) and in vivo studies suggest, however, that IFN-γ has a unique role in microglial activation. Priming by IFN-γ results in proliferation (microgliosis), enhanced synapse elimination, and moderate nitric oxide release sufficient to impair synaptic transmission, gamma rhythm activity, and cognitive functions. Moreover, IFN-γ is pivotal for driving Toll-like receptor (TLR)-activated microglia into neurotoxic phenotypes that induce energetic and oxidative stress, severe network dysfunction, and neuronal death. Pharmacological targeting of activated microglia could be beneficial during elevated IFN-γ levels, blood-brain barrier leakage, and parenchymal T lymphocyte infiltration associated with, for instance, encephalitis, multiple sclerosis, and Alzheimer's disease.
Collapse
Affiliation(s)
- Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany; Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, D-69120 Heidelberg, Germany.
| | - Fadi Almouhanna
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Bruno Chausse
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| |
Collapse
|
11
|
Voskuhl RR, MacKenzie-Graham A. Chronic experimental autoimmune encephalomyelitis is an excellent model to study neuroaxonal degeneration in multiple sclerosis. Front Mol Neurosci 2022; 15:1024058. [PMID: 36340686 PMCID: PMC9629273 DOI: 10.3389/fnmol.2022.1024058] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 09/30/2022] [Indexed: 08/19/2023] Open
Abstract
Animal models of multiple sclerosis (MS), specifically experimental autoimmune encephalomyelitis (EAE), have been used extensively to develop anti-inflammatory treatments. However, the similarity between MS and one particular EAE model does not end at inflammation. MS and chronic EAE induced in C57BL/6 mice using myelin oligodendrocyte glycoprotein (MOG) peptide 35-55 share many neuropathologies. Beyond both having white matter lesions in spinal cord, both also have widespread neuropathology in the cerebral cortex, hippocampus, thalamus, striatum, cerebellum, and retina/optic nerve. In this review, we compare neuropathologies in each of these structures in MS with chronic EAE in C57BL/6 mice, and find evidence that this EAE model is well suited to study neuroaxonal degeneration in MS.
Collapse
Affiliation(s)
- Rhonda R. Voskuhl
- UCLA MS Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | | |
Collapse
|
12
|
Yang K, He X, Wu Z, Yin Y, Pan H, Zhao X, Sun T. The emerging roles of piezo1 channels in animal models of multiple sclerosis. Front Immunol 2022; 13:976522. [PMID: 36177027 PMCID: PMC9513475 DOI: 10.3389/fimmu.2022.976522] [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: 06/23/2022] [Accepted: 08/24/2022] [Indexed: 11/23/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory, demyelinating, and neurodegenerative disease in the central nervous system (CNS). Its pathogenesis is quite complex: Accumulated evidence suggests that biochemical signals as well as mechanical stimuli play important roles in MS. In both patients and animal models of MS, brain viscoelasticity is reduced during disease progression. Piezo mechanosensitive channels are recently discovered, and their three-dimensional structure has been solved. Both the membrane dome mechanism and the membrane footprint hypothesis have been proposed to explain their mechanosensitivity. While membrane-mediated forces alone appear to be sufficient to induce Piezo gating, tethers attached to the membrane or to the channel itself also seem to play a role. Current research indicates that Piezo1 channels play a key role in multiple aspects of MS pathogenesis. Activation of Piezo1 channels in axon negatively regulates CNS myelination. in addition, the inhibition of Piezo1 in CD4+ T cells and/or T regulatory cells (Treg) attenuates experimental autoimmune encephalitis (EAE) symptoms. Although more work has to be done to clarify the roles of Piezo1 channels in MS, they might be a promising future drug target for MS treatment.
Collapse
Affiliation(s)
- Kai Yang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
- *Correspondence: Kai Yang, ; Taolei Sun,
| | - Xueai He
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Zhengqi Wu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Yimeng Yin
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Hanyu Pan
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Xinyue Zhao
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
- *Correspondence: Kai Yang, ; Taolei Sun,
| |
Collapse
|
13
|
Collongues N, Becker G, Jolivel V, Ayme-Dietrich E, de Seze J, Binamé F, Patte-Mensah C, Monassier L, Mensah-Nyagan AG. A Narrative Review on Axonal Neuroprotection in Multiple Sclerosis. Neurol Ther 2022; 11:981-1042. [PMID: 35610531 PMCID: PMC9338208 DOI: 10.1007/s40120-022-00363-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/03/2022] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) resulting in demyelination and neurodegeneration. The therapeutic strategy is now largely based on reducing inflammation with immunosuppressive drugs. Unfortunately, when disease progression is observed, no drug offers neuroprotection apart from its anti-inflammatory effect. In this review, we explore current knowledge on the assessment of neurodegeneration in MS and look at putative targets that might prove useful in protecting the axon from degeneration. Among them, Bruton's tyrosine kinase inhibitors, anti-apoptotic and antioxidant agents, sex hormones, statins, channel blockers, growth factors, and molecules preventing glutamate excitotoxicity have already been studied. Some of them have reached phase III clinical trials and carry a great message of hope for our patients with MS.
Collapse
Affiliation(s)
- Nicolas Collongues
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France. .,Center for Clinical Investigation, INSERM U1434, Strasbourg, France. .,Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France. .,University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.
| | - Guillaume Becker
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Valérie Jolivel
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Estelle Ayme-Dietrich
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Jérôme de Seze
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France.,Center for Clinical Investigation, INSERM U1434, Strasbourg, France.,Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Fabien Binamé
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Christine Patte-Mensah
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| | - Laurent Monassier
- University Department of Pharmacology, Addictology, Toxicology and Therapeutic, Strasbourg University, Strasbourg, France.,NeuroCardiovascular Pharmacology and Toxicology Laboratory, UR7296, University Hospital of Strasbourg, Strasbourg, France
| | - Ayikoé Guy Mensah-Nyagan
- Biopathology of Myelin, Neuroprotection and Therapeutic Strategy, INSERM U1119, Strasbourg, France
| |
Collapse
|
14
|
Scaroni F, Visconte C, Serpente M, Golia MT, Gabrielli M, Huiskamp M, Hulst HE, Carandini T, De Riz M, Pietroboni A, Rotondo E, Scarpini E, Galimberti D, Teunissen CE, van Dam M, de Jong BA, Fenoglio C, Verderio C. miR-150-5p and let-7b-5p in Blood Myeloid Extracellular Vesicles Track Cognitive Symptoms in Patients with Multiple Sclerosis. Cells 2022; 11:cells11091551. [PMID: 35563859 PMCID: PMC9104242 DOI: 10.3390/cells11091551] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 02/04/2023] Open
Abstract
Cognitive deficits strongly affect the quality of life of patients with multiple sclerosis (MS). However, no cognitive MS biomarkers are currently available. Extracellular vesicles (EVs) contain markers of parental cells and are able to pass from the brain into blood, representing a source of disease biomarkers. The aim of this study was to investigate whether small non-coding microRNAs (miRNAs) targeting synaptic genes and packaged in plasma EVs may reflect cognitive deficits in MS patients. Total EVs were precipitated by Exoquick from the plasma of twenty-six cognitively preserved (CP) and twenty-three cognitively impaired (CI) MS patients belonging to two independent cohorts. Myeloid EVs were extracted by affinity capture from total EVs using Isolectin B4 (IB4). Fourteen miRNAs targeting synaptic genes were selected and measured by RT-PCR in both total and myeloid EVs. Myeloid EVs from CI patients expressed higher levels of miR-150-5p and lower levels of let-7b-5p compared to CP patients. Stratification for progressive MS (PMS) and relapsing-remitting MS (RRMS) and correlation with clinical parameters suggested that these alterations might be attributable to cognitive deficits rather than disease progression. This study identifies miR-150-5p and let-7b-5p packaged in blood myeloid EVs as possible biomarkers for cognitive deficits in MS.
Collapse
Affiliation(s)
- Federica Scaroni
- Institute of Neuroscience, CNR, Via Follereau 3, 20854 Vedano al Lambro, Italy; (F.S.); (M.T.G.); (M.G.)
| | - Caterina Visconte
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via F. Sforza 35, 20122 Milan, Italy; (C.V.); (E.S.); (D.G.)
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
| | - Maria Serpente
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Maria Teresa Golia
- Institute of Neuroscience, CNR, Via Follereau 3, 20854 Vedano al Lambro, Italy; (F.S.); (M.T.G.); (M.G.)
| | - Martina Gabrielli
- Institute of Neuroscience, CNR, Via Follereau 3, 20854 Vedano al Lambro, Italy; (F.S.); (M.T.G.); (M.G.)
| | - Marijn Huiskamp
- MS Center Amsterdam, Amsterdam Neuroscience, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam UMC, De Boelelaan 1117, 1081 Amsterdam, The Netherlands; (M.H.); (M.v.D.)
| | - Hanneke E. Hulst
- Health-, Medical- and Neuropsychology Unit, Institute of Psychology, Leiden University, 2300 Leiden, The Netherlands;
| | - Tiziana Carandini
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Milena De Riz
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Anna Pietroboni
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Emanuela Rotondo
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Elio Scarpini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via F. Sforza 35, 20122 Milan, Italy; (C.V.); (E.S.); (D.G.)
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via F. Sforza 35, 20122 Milan, Italy; (C.V.); (E.S.); (D.G.)
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Charlotte E. Teunissen
- MS Center Amsterdam, Amsterdam Neuroscience, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 Amsterdam, The Netherlands; (C.E.T.); (B.A.d.J.)
| | - Maureen van Dam
- MS Center Amsterdam, Amsterdam Neuroscience, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam UMC, De Boelelaan 1117, 1081 Amsterdam, The Netherlands; (M.H.); (M.v.D.)
| | - Brigit A. de Jong
- MS Center Amsterdam, Amsterdam Neuroscience, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 Amsterdam, The Netherlands; (C.E.T.); (B.A.d.J.)
| | - Chiara Fenoglio
- MS Center Amsterdam, Amsterdam Neuroscience, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 Amsterdam, The Netherlands; (C.E.T.); (B.A.d.J.)
- Department of Neuropathology and Transplantation, University of Milan, Via F. Sforza 35, 20122 Milan, Italy
- Correspondence: (C.F.); (C.V.); Tel.: +39-0264488386 (C.V.)
| | - Claudia Verderio
- Institute of Neuroscience, CNR, Via Follereau 3, 20854 Vedano al Lambro, Italy; (F.S.); (M.T.G.); (M.G.)
- Correspondence: (C.F.); (C.V.); Tel.: +39-0264488386 (C.V.)
| |
Collapse
|
15
|
Démosthènes A, Sion B, Giraudet F, Moisset X, Daulhac L, Eschalier A, Bégou M. In-Depth Characterization of Somatic and Orofacial Sensitive Dysfunctions and Interfering-Symptoms in a Relapsing-Remitting Experimental Autoimmune Encephalomyelitis Mouse Model. Front Neurol 2022; 12:789432. [PMID: 35111128 PMCID: PMC8801881 DOI: 10.3389/fneur.2021.789432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/16/2021] [Indexed: 11/23/2022] Open
Abstract
Among the many symptoms (motor, sensory, and cognitive) associated with multiple sclerosis (MS), chronic pain is a common disabling condition. In particular, neuropathic pain symptoms are very prevalent and debilitating, even in early stages of the disease. Unfortunately, chronic pain still lacks efficient therapeutic agents. Progress is needed (i) clinically by better characterizing pain symptoms in MS and understanding the underlying mechanisms, and (ii) preclinically by developing a more closely dedicated model to identify new therapeutic targets and evaluate new drugs. In this setting, new variants of experimental autoimmune encephalomyelitis (EAE) are currently developed in mice to exhibit less severe motor impairments, thereby avoiding confounding factors in assessing pain behaviors over the disease course. Among these, the optimized relapsing-remitting EAE (QuilA-EAE) mouse model, induced using myelin oligodendrocyte glycoprotein peptide fragment (35–55), pertussis toxin, and quillaja bark saponin, seems very promising. Our study sought (i) to better define sensitive dysfunctions and (ii) to extend behavioral characterization to interfering symptoms often associated with pain during MS, such as mood disturbances, fatigue, and cognitive impairment, in this optimized QuilA-EAE model. We made an in-depth characterization of this optimized QuilA-EAE model, describing for the first time somatic thermal hyperalgesia associated with mechanical and cold allodynia. Evaluation of orofacial pain sensitivity showed no mechanical or thermal allodynia. Detailed evaluation of motor behaviors highlighted slight defects in fine motor coordination in the QuilA-EAE mice but without impact on pain evaluation. Finally, no anxiety-related or cognitive impairment was observed during the peak of sensitive symptoms. Pharmacologically, as previously described, we found that pregabalin, a treatment commonly used in neuropathic pain patients, induced an analgesic effect on mechanical allodynia. In addition, we showed an anti-hyperalgesic thermal effect on this model. Our results demonstrate that this QuilA-EAE model is clearly of interest for studying pain symptom development and so could be used to identify and evaluate new therapeutic targets. The presence of interfering symptoms still needs to be further characterized.
Collapse
Affiliation(s)
- Amélie Démosthènes
- Université Clermont Auvergne, Inserm, Neuro-Dol, Faculté de Pharmacie, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
| | - Benoît Sion
- Université Clermont Auvergne, Inserm, Neuro-Dol, Faculté de Pharmacie, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
| | - Fabrice Giraudet
- Université Clermont Auvergne, Inserm, Neuro-Dol, Faculté de Pharmacie, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
| | - Xavier Moisset
- Université Clermont Auvergne, CHU de Clermont-Ferrand, Inserm, Neuro-Dol, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
| | - Laurence Daulhac
- Université Clermont Auvergne, Inserm, Neuro-Dol, Faculté de Pharmacie, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
| | - Alain Eschalier
- Université Clermont Auvergne, Inserm, Neuro-Dol, Faculté de Pharmacie, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
| | - Mélina Bégou
- Université Clermont Auvergne, Inserm, Neuro-Dol, Faculté de Pharmacie, Faculté de Médecine, Institut Analgesia, BP38, Clermont-Ferrand, France
- *Correspondence: Mélina Bégou
| |
Collapse
|
16
|
Di Filippo M, Mancini A, Bellingacci L, Gaetani L, Mazzocchetti P, Zelante T, La Barbera L, De Luca A, Tantucci M, Tozzi A, Durante V, Sciaccaluga M, Megaro A, Chiasserini D, Salvadori N, Lisetti V, Portaccio E, Costa C, Sarchielli P, Amato MP, Parnetti L, Viscomi MT, Romani L, Calabresi P. Interleukin-17 affects synaptic plasticity and cognition in an experimental model of multiple sclerosis. Cell Rep 2021; 37:110094. [PMID: 34879272 DOI: 10.1016/j.celrep.2021.110094] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/06/2021] [Accepted: 11/12/2021] [Indexed: 12/20/2022] Open
Abstract
Cognitive impairment (CI) is a disabling concomitant of multiple sclerosis (MS) with a complex and controversial pathogenesis. The cytokine interleukin-17A (IL-17A) is involved in the immune pathogenesis of MS, but its possible effects on synaptic function and cognition are still largely unexplored. In this study, we show that the IL-17A receptor (IL-17RA) is highly expressed by hippocampal neurons in the CA1 area and that exposure to IL-17A dose-dependently disrupts hippocampal long-term potentiation (LTP) through the activation of its receptor and p38 mitogen-activated protein kinase (MAPK). During experimental autoimmune encephalomyelitis (EAE), IL-17A overexpression is paralleled by hippocampal LTP dysfunction. An in vivo behavioral analysis shows that visuo-spatial learning abilities are preserved when EAE is induced in mice lacking IL-17A. Overall, this study suggests a key role for the IL-17 axis in the neuro-immune cross-talk occurring in the hippocampal CA1 area and its potential involvement in synaptic dysfunction and MS-related CI.
Collapse
MESH Headings
- Animals
- Behavior, Animal
- CA1 Region, Hippocampal/metabolism
- CA1 Region, Hippocampal/pathology
- CA1 Region, Hippocampal/physiopathology
- Cognition
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Encephalomyelitis, Autoimmune, Experimental/psychology
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Long-Term Potentiation
- Male
- Mice, Biozzi
- Mice, Inbred C57BL
- Mice, Knockout
- Neuronal Plasticity
- Receptors, Interleukin-17/genetics
- Receptors, Interleukin-17/metabolism
- Signal Transduction
- Spatial Learning
- Synapses/metabolism
- Synapses/pathology
- p38 Mitogen-Activated Protein Kinases
- Mice
Collapse
Affiliation(s)
- Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| | - Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Laura Bellingacci
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Petra Mazzocchetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Teresa Zelante
- Section of Pathology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Livia La Barbera
- Unit of Molecular Neurosciences, Department of Medicine, University Campus-Biomedico, Rome, Italy
| | - Antonella De Luca
- Section of Pathology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Michela Tantucci
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alessandro Tozzi
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Valentina Durante
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Miriam Sciaccaluga
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alfredo Megaro
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Davide Chiasserini
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Nicola Salvadori
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Viviana Lisetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emilio Portaccio
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | - Cinzia Costa
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paola Sarchielli
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Maria Pia Amato
- Department of NEUROFARBA, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Maria Teresa Viscomi
- Section of Histology and Embryology, Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luigina Romani
- Section of Pathology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paolo Calabresi
- Neurology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy; Section of Neurology, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
17
|
Bourel J, Planche V, Dubourdieu N, Oliveira A, Séré A, Ducourneau EG, Tible M, Maitre M, Lesté-Lasserre T, Nadjar A, Desmedt A, Ciofi P, Oliet SH, Panatier A, Tourdias T. Complement C3 mediates early hippocampal neurodegeneration and memory impairment in experimental multiple sclerosis. Neurobiol Dis 2021; 160:105533. [PMID: 34673149 DOI: 10.1016/j.nbd.2021.105533] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/09/2021] [Accepted: 10/17/2021] [Indexed: 12/26/2022] Open
Abstract
Memory impairment is one of the disabling manifestations of multiple sclerosis (MS) possibly present from the early stages of the disease and for which there is no specific treatment. Hippocampal synaptic dysfunction and dendritic loss, associated with microglial activation, can underlie memory deficits, yet the molecular mechanisms driving such hippocampal neurodegeneration need to be elucidated. In early-stage experimental autoimmune encephalomyelitis (EAE) female mice, we assessed the expression level of molecules involved in microglia-neuron interactions within the dentate gyrus and found overexpression of genes of the complement pathway. Compared to sham immunized mice, the central element of the complement cascade, C3, showed the strongest and 10-fold upregulation, while there was no increase of downstream factors such as the terminal component C5. The combination of in situ hybridization with immunofluorescence showed that C3 transcripts were essentially produced by activated microglia. Pharmacological inhibition of C3 activity, by daily administration of rosmarinic acid, was sufficient to prevent early dendritic loss, microglia-mediated phagocytosis of synapses in the dentate gyrus, and memory impairment in EAE mice, while morphological markers of microglial activation were still observed. In line, when EAE was induced in C3 deficient mice (C3KO), dendrites and spines of the dentate gyrus as well as memory abilities were preserved. Altogether, these data highlight the central role of microglial C3 in early hippocampal neurodegeneration and memory impairment in EAE and, therefore, pave the way toward new neuroprotective strategies in MS to prevent cognitive deficit using complement inhibitors.
Collapse
Affiliation(s)
- Julien Bourel
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France
| | - Vincent Planche
- Univ. Bordeaux, CNRS, UMR 5293, Institut des Maladies Neurodégénératives, F-33000 Bordeaux, France
| | - Nadège Dubourdieu
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France
| | - Aymeric Oliveira
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France
| | - Alexandra Séré
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | | | - Marion Tible
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France
| | - Marlène Maitre
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France
| | | | - Agnes Nadjar
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France; Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - Aline Desmedt
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France
| | - Philippe Ciofi
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France
| | - Stéphane H Oliet
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France
| | - Aude Panatier
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France
| | - Thomas Tourdias
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300 Bordeaux, France; CHU de Bordeaux, Neuroimagerie diagnostique et thérapeutique, F-33000 Bordeaux, France.
| |
Collapse
|
18
|
Kumar SP, Babu PP. NADPH Oxidase: a Possible Therapeutic Target for Cognitive Impairment in Experimental Cerebral Malaria. Mol Neurobiol 2021; 59:800-820. [PMID: 34782951 DOI: 10.1007/s12035-021-02598-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/12/2021] [Indexed: 12/19/2022]
Abstract
Long-term cognitive impairment associated with seizure-induced hippocampal damage is the key feature of cerebral malaria (CM) pathogenesis. One-fourth of child survivors of CM suffer from long-lasting neurological deficits and behavioral anomalies. However, mechanisms on hippocampal dysfunction are unclear. In this study, we elucidated whether gp91phox isoform of nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) (a potent marker of oxidative stress) mediates hippocampal neuronal abnormalities and cognitive dysfunction in experimental CM (ECM). Mice symptomatic to CM were rescue treated with artemether monotherapy (ARM) and in combination with apocynin (ARM + APO) adjunctive based on scores of Rapid Murine Come behavior Scale (RMCBS). After a 30-day survivability period, we performed Barnes maze, T-maze, and novel object recognition cognitive tests to evaluate working and reference memory in all the experimental groups except CM. Sensorimotor tests were conducted in all the cohorts to assess motor coordination. We performed Golgi-Cox staining to illustrate cornu ammonis-1 (CA1) pyramidal neuronal morphology and study overall hippocampal neuronal density changes. Further, expression of NOX2, NeuN (neuronal marker) in hippocampal CA1 and dentate gyrus was determined using double immunofluorescence experiments in all the experimental groups. Mice administered with ARM monotherapy and APO adjunctive treatment exhibited similar survivability. The latter showed better locomotor and cognitive functions, reduced ROS levels, and hippocampal NOX2 immunoreactivity in ECM. Our results show a substantial increase in hippocampal NeuN immunoreactivity and dendritic arborization in ARM + APO cohorts compared to ARM-treated brain samples. Overall, our study suggests that overexpression of NOX2 could result in loss of hippocampal neuronal density and dendritic spines of CA1 neurons affecting the spatial working and reference memory during ECM. Notably, ARM + APO adjunctive therapy reversed the altered neuronal morphology and oxidative damage in hippocampal neurons restoring long-term cognitive functions after CM.
Collapse
Affiliation(s)
- Simhadri Praveen Kumar
- F-23/71, Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India
| | - Phanithi Prakash Babu
- F-23/71, Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India.
| |
Collapse
|
19
|
Boyd A, Byrne S, Middleton RJ, Banati RB, Liu GJ. Control of Neuroinflammation through Radiation-Induced Microglial Changes. Cells 2021; 10:2381. [PMID: 34572030 PMCID: PMC8468704 DOI: 10.3390/cells10092381] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 12/15/2022] Open
Abstract
Microglia, the innate immune cells of the central nervous system, play a pivotal role in the modulation of neuroinflammation. Neuroinflammation has been implicated in many diseases of the CNS, including Alzheimer's disease and Parkinson's disease. It is well documented that microglial activation, initiated by a variety of stressors, can trigger a potentially destructive neuroinflammatory response via the release of pro-inflammatory molecules, and reactive oxygen and nitrogen species. However, the potential anti-inflammatory and neuroprotective effects that microglia are also thought to exhibit have been under-investigated. The application of ionising radiation at different doses and dose schedules may reveal novel methods for the control of microglial response to stressors, potentially highlighting avenues for treatment of neuroinflammation associated CNS disorders, such as Alzheimer's disease and Parkinson's disease. There remains a need to characterise the response of microglia to radiation, particularly low dose ionising radiation.
Collapse
Affiliation(s)
- Alexandra Boyd
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia; (A.B.); (S.B.); (R.J.M.); (R.B.B.)
| | - Sarah Byrne
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia; (A.B.); (S.B.); (R.J.M.); (R.B.B.)
| | - Ryan J. Middleton
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia; (A.B.); (S.B.); (R.J.M.); (R.B.B.)
| | - Richard B. Banati
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia; (A.B.); (S.B.); (R.J.M.); (R.B.B.)
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, Sydney, NSW 2050, Australia
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia; (A.B.); (S.B.); (R.J.M.); (R.B.B.)
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, Sydney, NSW 2050, Australia
| |
Collapse
|
20
|
Bellingacci L, Mancini A, Gaetani L, Tozzi A, Parnetti L, Di Filippo M. Synaptic Dysfunction in Multiple Sclerosis: A Red Thread from Inflammation to Network Disconnection. Int J Mol Sci 2021; 22:ijms22189753. [PMID: 34575917 PMCID: PMC8469646 DOI: 10.3390/ijms22189753] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 12/24/2022] Open
Abstract
Multiple sclerosis (MS) has been clinically considered a chronic inflammatory disease of the white matter; however, in the last decade growing evidence supported an important role of gray matter pathology as a major contributor of MS-related disability and the involvement of synaptic structures assumed a key role in the pathophysiology of the disease. Synaptic contacts are considered central units in the information flow, involved in synaptic transmission and plasticity, critical processes for the shaping and functioning of brain networks. During the course of MS, the immune system and its diffusible mediators interact with synaptic structures leading to changes in their structure and function, influencing brain network dynamics. The purpose of this review is to provide an overview of the existing literature on synaptic involvement during experimental and human MS, in order to understand the mechanisms by which synaptic failure eventually leads to brain networks alterations and contributes to disabling MS symptoms and disease progression.
Collapse
Affiliation(s)
- Laura Bellingacci
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (L.B.); (A.M.); (L.G.); (L.P.)
| | - Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (L.B.); (A.M.); (L.G.); (L.P.)
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (L.B.); (A.M.); (L.G.); (L.P.)
| | - Alessandro Tozzi
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy;
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (L.B.); (A.M.); (L.G.); (L.P.)
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (L.B.); (A.M.); (L.G.); (L.P.)
- Correspondence: ; Tel.: +39-075-578-3830
| |
Collapse
|
21
|
d'Isa R, Comi G, Leocani L. The 4-Hole-Board Test for Assessment of Long-Term Spatial Memory in Mice. Curr Protoc 2021; 1:e228. [PMID: 34432376 DOI: 10.1002/cpz1.228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The hole-board test has been used in rodents since the early 60s to measure exploratory behavior, locomotor activity and cognitive function. The test is based on rodents' natural curiosity and attraction for novelty. Basically, the hole-board consists of a small square arena with an extractable platform as floor, which has a set of equally spaced circular holes on its surface. In this article, we describe the protocol of a 4-hole-board test allowing the assessment of long-term spatial memory in mice without the employment of water or food restriction, painful stimuli (as electrical shocks) or any aversive condition (as forced swimming or exposure to intense light). Four holes are present on the floor of the square arena (one for each of its four quadrants). Mice released in the arena spontaneously approach the holes and explore them by briefly inserting the snout inside, a behavior defined as nose-poking (or head-dipping). If, after 24 hr, rodents are re-exposed to the hole-board, the novelty of the holes decreases. Animals with an intact long-term memory will show a reduction of the frequency of nose-poking into the holes. The total number of nose-pokes on day 1 is an index of exploration, while the percentage of decrease in nose-poking on day 2 represents an index of long-term spatial memory. Number of quadrant crossings is scored as a control measure for locomotor activity, which with the present protocol should remain stable across the days of testing. Indeed, the 4-hole-board test represents a stress-free and animal-friendly option to evaluate long-term spatial memory. In the present paper, we provide detailed description of the hole-board apparatus and step-by-step protocol for assessment of spatial memory in mice. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Validation of the 4-hole-board Basic Protocol 2: Evaluation of long-term spatial memory through the 4-hole-board test.
Collapse
Affiliation(s)
- Raffaele d'Isa
- Experimental Neurophysiology Unit, Institute of Experimental Neurology (INSPE), San Raffaele Scientific Institute, IRCCS-San Raffaele Hospital, Milan, Italy
| | - Giancarlo Comi
- Vita-Salute San Raffaele University, Milan, Italy.,Casa di Cura del Policlinico, Milan, Italy
| | - Letizia Leocani
- Experimental Neurophysiology Unit, Institute of Experimental Neurology (INSPE), San Raffaele Scientific Institute, IRCCS-San Raffaele Hospital, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
22
|
Kalafatakis I, Karagogeos D. Oligodendrocytes and Microglia: Key Players in Myelin Development, Damage and Repair. Biomolecules 2021; 11:1058. [PMID: 34356682 PMCID: PMC8301746 DOI: 10.3390/biom11071058] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes, the myelin-making cells of the CNS, regulate the complex process of myelination under physiological and pathological conditions, significantly aided by other glial cell types such as microglia, the brain-resident, macrophage-like innate immune cells. In this review, we summarize how oligodendrocytes orchestrate myelination, and especially myelin repair after damage, and present novel aspects of oligodendroglial functions. We emphasize the contribution of microglia in the generation and regeneration of myelin by discussing their beneficial and detrimental roles, especially in remyelination, underlining the cellular and molecular components involved. Finally, we present recent findings towards human stem cell-derived preclinical models for the study of microglia in human pathologies and on the role of microbiome on glial cell functions.
Collapse
Affiliation(s)
- Ilias Kalafatakis
- Laboratory of Neuroscience, Department of Basic Science, University of Crete Medical School, 70013 Heraklion, Greece;
- IMBB FORTH, Nikolaou Plastira 100, Vassilika Vouton, 70013 Heraklion, Greece
| | - Domna Karagogeos
- Laboratory of Neuroscience, Department of Basic Science, University of Crete Medical School, 70013 Heraklion, Greece;
- IMBB FORTH, Nikolaou Plastira 100, Vassilika Vouton, 70013 Heraklion, Greece
| |
Collapse
|
23
|
Une H, Yamasaki R, Nagata S, Yamaguchi H, Nakamuta Y, Indiasari UC, Cui Y, Shinoda K, Masaki K, Götz M, Kira JI. Brain gray matter astroglia-specific connexin 43 ablation attenuates spinal cord inflammatory demyelination. J Neuroinflammation 2021; 18:126. [PMID: 34090477 PMCID: PMC8180177 DOI: 10.1186/s12974-021-02176-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/19/2021] [Indexed: 11/10/2022] Open
Abstract
Background Brain astroglia are activated preceding the onset of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). We characterized the effects of brain astroglia on spinal cord inflammation, focusing on astroglial connexin (Cx)43, because we recently reported that Cx43 has a critical role in regulating neuroinflammation. Methods Because glutamate aspartate transporter (GLAST)+ astroglia are enriched in the brain gray matter, we generated Cx43fl/fl;GLAST-CreERT2/+ mice that were brain gray matter astroglia-specific Cx43 conditional knockouts (Cx43 icKO). EAE was induced by immunization with myelin oligodendroglia glycoprotein (MOG) 35–55 peptide 10 days after tamoxifen injection. Cx43fl/fl mice were used as controls. Results Acute and chronic EAE signs were significantly milder in Cx43 icKO mice than in controls whereas splenocyte MOG-specific responses were unaltered. Histologically, Cx43 icKO mice showed significantly less demyelination and fewer CD45+ infiltrating immunocytes, including F4/80+ macrophages, and Iba1+ microglia in the spinal cord than controls. Microarray analysis of the whole cerebellum revealed marked upregulation of anti-inflammatory A2-specific astroglia gene sets in the pre-immunized phase and decreased proinflammatory A1-specific and pan-reactive astroglial gene expression in the onset phase in Cx43 icKO mice compared with controls. Astroglia expressing C3, a representative A1 marker, were significantly decreased in the cerebrum, cerebellum, and spinal cord of Cx43 icKO mice compared with controls in the peak phase. Isolated Cx43 icKO spinal microglia showed more anti-inflammatory and less proinflammatory gene expression than control microglia in the pre-immunized phase. In particular, microglial expression of Ccl2, Ccl5, Ccl7, and Ccl8 in the pre-immunized phase and of Cxcl9 at the peak phase was lower in Cx43 icKO than in controls. Spinal microglia circularity was significantly lower in Cx43 icKO than in controls in the peak phase. Significantly lower interleukin (IL)-6, interferon-γ, and IL-10 levels were present in cerebrospinal fluid from Cx43 icKO mice in the onset phase compared with controls. Conclusions The ablation of Cx43 in brain gray matter astroglia attenuates EAE by promoting astroglia toward an anti-inflammatory phenotype and suppressing proinflammatory activation of spinal microglia partly through depressed cerebrospinal fluid proinflammatory cytokine/chemokine levels. Brain astroglial Cx43 might be a novel therapeutic target for MS. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02176-1.
Collapse
Affiliation(s)
- Hayato Une
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Satoshi Nagata
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuko Nakamuta
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ulfa Camelia Indiasari
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yiwen Cui
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koji Shinoda
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan. .,Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Ookawa, Fukuoka, 831-8501, Japan. .,Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, International University of Health and Welfare, 2-6-11 Yakuin, Chuo-ku, Fukuoka, 810-0022, Japan.
| |
Collapse
|
24
|
Behl T, Kaur G, Sehgal A, Zengin G, Singh S, Ahmadi A, Bungau S. Flavonoids, the Family of Plant-derived Antioxidants making inroads into Novel Therapeutic Design against IR-induced Oxidative Stress in Parkinson's Disease. Curr Neuropharmacol 2021; 20:324-343. [PMID: 34030619 PMCID: PMC9413797 DOI: 10.2174/1570159x19666210524152817] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/17/2021] [Accepted: 05/05/2021] [Indexed: 11/22/2022] Open
Abstract
Background: Ionizing radiation from telluric sources is unceasingly an unprotected pitfall to humans. Thus, the foremost contributors to human exposure are global and medical radiations. Various evidences assembled during preceding years reveal the pertinent role of ionizing radiation-induced oxidative stress in the progression of neurodegenerative insults, such as Parkinson’s disease, which have been contributing to increased proliferation and generation of reactive oxygen species. Objective: This review delineates the role of ionizing radiation-induced oxidative stress in Parkinson’s disease and proposes novel therapeutic interventions of flavonoid family, offering effective management and slowing down the progression of Parkinson’s disease. Methods: Published papers were searched in MEDLINE, PubMed, etc., published to date for in-depth database collection. Results: The oxidative damage may harm the non-targeted cells. It can also modulate the functions of the central nervous system, such as protein misfolding, mitochondria dysfunction, increased levels of oxidized lipids, and dopaminergic cell death, which accelerate the progression of Parkinson’s disease at the molecular, cellular, or tissue levels. In Parkinson’s disease, reactive oxygen species exacerbate the production of nitric oxides and superoxides by activated microglia, rendering death of dopaminergic neuronal cell through different mechanisms. Conclusion: Rising interest has extensively engrossed in the clinical trial designs based on the plant-derived family of antioxidants. They are known to exert multifarious impact on neuroprotection via directly suppressing ionizing radiation-induced oxidative stress and reactive oxygen species production or indirectly increasing the dopamine levels and activating the glial cells.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Gagandeep Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, Konya, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Centre, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari. Iran
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea. Romania
| |
Collapse
|
25
|
Revi N, Rengan AK. Impact of dietary polyphenols on neuroinflammation-associated disorders. Neurol Sci 2021; 42:3101-3119. [PMID: 33988799 DOI: 10.1007/s10072-021-05303-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022]
Abstract
Neurodegenerative disorders like Alzheimer's, Parkinson's, and associated dementia typically originate with altered protein folding and aggregation of their β structures in the neurons. This self-aggregation leads to glial activation in the brain, causing neuroinflammation and leads to neuronal death. According to statistics provided by WHO, there are around 50 million people with dementia worldwide and every year, 10 million more cases are projected to increase. Also, around 5-8 percentage of people who are aged above 60 globally has dementia or associated disorders. Over 82 million in 2030 and 152 in 2050 are expected to have dementia. Most of these patients fall into low-middle-income countries which makes it even more essential to find an affordable and effective treatment method. Polyphenols of different origin are studied for their potential role as anti-neuro-inflammatory molecules. This review would summarize recent advances in three widely researched dietary polyphenols projected as potential therapeutic agents for disorders like Alzheimer's, Parkinson's, etc. They are Resveratrol, Catechins, and Tannins. The review would discuss the recent advances and challenges in using these polyphenols using specific examples as potential therapeutic agents against neuroinflammation associated disorders. An abstract of neuroinflammation-associated events and the effects by selected polyphenols.
Collapse
Affiliation(s)
- Neeraja Revi
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, India.
| |
Collapse
|
26
|
Insights into the Pathophysiology of Psychiatric Symptoms in Central Nervous System Disorders: Implications for Early and Differential Diagnosis. Int J Mol Sci 2021; 22:ijms22094440. [PMID: 33922780 PMCID: PMC8123079 DOI: 10.3390/ijms22094440] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Different psychopathological manifestations, such as affective, psychotic, obsessive-compulsive symptoms, and impulse control disturbances, may occur in most central nervous system (CNS) disorders including neurodegenerative and neuroinflammatory diseases. Psychiatric symptoms often represent the clinical onset of such disorders, thus potentially leading to misdiagnosis, delay in treatment, and a worse outcome. In this review, psychiatric symptoms observed along the course of several neurological diseases, namely Alzheimer’s disease, fronto-temporal dementia, Parkinson’s disease, Huntington’s disease, and multiple sclerosis, are discussed, as well as the involved brain circuits and molecular/synaptic alterations. Special attention has been paid to the emerging role of fluid biomarkers in early detection of these neurodegenerative diseases. The frequent occurrence of psychiatric symptoms in neurological diseases, even as the first clinical manifestations, should prompt neurologists and psychiatrists to share a common clinico-biological background and a coordinated diagnostic approach.
Collapse
|
27
|
Hu CF, Wu SP, Lin GJ, Shieh CC, Hsu CS, Chen JW, Chen SH, Hong JS, Chen SJ. Microglial Nox2 Plays a Key Role in the Pathogenesis of Experimental Autoimmune Encephalomyelitis. Front Immunol 2021; 12:638381. [PMID: 33868265 PMCID: PMC8050344 DOI: 10.3389/fimmu.2021.638381] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
While oxidative stress has been linked to multiple sclerosis (MS), the role of superoxide-producing phagocyte NADPH oxidase (Nox2) in central nervous system (CNS) pathogenesis remains unclear. This study investigates the impact of Nox2 gene ablation on pro- and anti-inflammatory cytokine and chemokine production in a mouse experimental autoimmune encephalomyelitis (EAE) model. Nox2 deficiency attenuates EAE-induced neural damage and reduces disease severity, pathogenic immune cells infiltration, demyelination, and oxidative stress in the CNS. The number of autoreactive T cells, myeloid cells, and activated microglia, as well as the production of cytokines and chemokines, including GM-CSF, IFNγ, TNFα, IL-6, IL-10, IL-17A, CCL2, CCL5, and CXCL10, were much lower in the Nox2-/- CNS tissues but remained unaltered in the peripheral lymphoid organs. RNA-seq profiling of microglial transcriptome identified a panel of Nox2 dependent proinflammatory genes: Pf4, Tnfrsf9, Tnfsf12, Tnfsf13, Ccl7, Cxcl3, and Cxcl9. Furthermore, gene ontology and pathway enrichment analyses revealed that microglial Nox2 plays a regulatory role in multiple pathways known to be important for MS/EAE pathogenesis, including STAT3, glutathione, leukotriene biosynthesis, IL-8, HMGB1, NRF2, systemic lupus erythematosus in B cells, and T cell exhaustion signaling. Taken together, our results provide new insights into the critical functions performed by microglial Nox2 during the EAE pathogenesis, suggesting that Nox2 inhibition may represent an important therapeutic target for MS.
Collapse
Affiliation(s)
- Chih-Fen Hu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Gu-Jiun Lin
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Chang Shieh
- Institute of Clinical Medicine, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Chih-Sin Hsu
- Genomics Center for Clinical and Biotechnological Applications of Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jing-Wun Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Heng Chen
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Shyi-Jou Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
28
|
Gaetani L, Salvadori N, Chipi E, Gentili L, Borrelli A, Parnetti L, Di Filippo M. Cognitive impairment in multiple sclerosis: lessons from cerebrospinal fluid biomarkers. Neural Regen Res 2021; 16:36-42. [PMID: 32788445 PMCID: PMC7818856 DOI: 10.4103/1673-5374.286949] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cognitive impairment is a common clinical manifestation of multiple sclerosis, but its pathophysiology is not completely understood. White and grey matter injury together with synaptic dysfunction do play a role. The measurement of biomarkers in the cerebrospinal fluid and the study of their association with cognitive impairment may provide interesting in vivo evidence of the biological mechanisms underlying multiple sclerosis-related cognitive impairment. So far, only a few studies on this topic have been published, giving interesting results that deserve further investigation. Cerebrospinal fluid biomarkers of different pathophysiological mechanisms seem to reflect different neuropsychological patterns of cognitive deficits in multiple sclerosis. The aim of this review is to discuss the studies that have correlated cerebrospinal fluid markers of immune, glial and neuronal pathology with cognitive impairment in multiple sclerosis. Although preliminary, these findings suggest that cerebrospinal fluid biomarkers show some correlation with cognitive performance in multiple sclerosis, thus providing interesting insights into the mechanisms underlying the involvement of specific cognitive domains.
Collapse
Affiliation(s)
- Lorenzo Gaetani
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Nicola Salvadori
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Elena Chipi
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Lucia Gentili
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Angela Borrelli
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | | |
Collapse
|
29
|
Marino J, Maubert ME, Mele AR, Spector C, Wigdahl B, Nonnemacher MR. Functional impact of HIV-1 Tat on cells of the CNS and its role in HAND. Cell Mol Life Sci 2020; 77:5079-5099. [PMID: 32577796 PMCID: PMC7674201 DOI: 10.1007/s00018-020-03561-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/08/2020] [Accepted: 05/25/2020] [Indexed: 02/07/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) transactivator of transcription (Tat) is a potent mediator involved in the development of HIV-1-associated neurocognitive disorders (HAND). Tat is expressed even in the presence of antiretroviral therapy (ART) and is able to enter the central nervous system (CNS) through a variety of ways, where Tat can interact with microglia, astrocytes, brain microvascular endothelial cells, and neurons. The presence of low concentrations of extracellular Tat alone has been shown to lead to dysregulated gene expression, chronic cell activation, inflammation, neurotoxicity, and structural damage in the brain. The reported effects of Tat are dependent in part on the specific HIV-1 subtype and amino acid length of Tat used. HIV-1 subtype B Tat is the most common subtype in North American and therefore, most studies have been focused on subtype B Tat; however, studies have shown many genetic, biologic, and pathologic differences between HIV subtype B and subtype C Tat. This review will focus primarily on subtype B Tat where the full-length protein is 101 amino acids, but will also consider variants of Tat, such as Tat 72 and Tat 86, that have been reported to exhibit a number of distinctive activities with respect to mediating CNS damage and neurotoxicity.
Collapse
Affiliation(s)
- Jamie Marino
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Monique E Maubert
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Anthony R Mele
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Cassandra Spector
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N. 15th St, Philadelphia, PA, 19102, USA.
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
30
|
Stojić-Vukanić Z, Hadžibegović S, Nicole O, Nacka-Aleksić M, Leštarević S, Leposavić G. CD8+ T Cell-Mediated Mechanisms Contribute to the Progression of Neurocognitive Impairment in Both Multiple Sclerosis and Alzheimer's Disease? Front Immunol 2020; 11:566225. [PMID: 33329528 PMCID: PMC7710704 DOI: 10.3389/fimmu.2020.566225] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022] Open
Abstract
Neurocognitive impairment (NCI) is one of the most relevant clinical manifestations of multiple sclerosis (MS). The profile of NCI and the structural and functional changes in the brain structures relevant for cognition in MS share some similarities to those in Alzheimer's disease (AD), the most common cause of neurocognitive disorders. Additionally, despite clear etiopathological differences between MS and AD, an accumulation of effector/memory CD8+ T cells and CD8+ tissue-resident memory T (Trm) cells in cognitively relevant brain structures of MS/AD patients, and higher frequency of effector/memory CD8+ T cells re-expressing CD45RA (TEMRA) with high capacity to secrete cytotoxic molecules and proinflammatory cytokines in their blood, were found. Thus, an active pathogenetic role of CD8+ T cells in the progression of MS and AD may be assumed. In this mini-review, findings supporting the putative role of CD8+ T cells in the pathogenesis of MS and AD are displayed, and putative mechanisms underlying their pathogenetic action are discussed. A special effort was made to identify the gaps in the current knowledge about the role of CD8+ T cells in the development of NCI to "catalyze" translational research leading to new feasible therapeutic interventions.
Collapse
Affiliation(s)
- Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Senka Hadžibegović
- Institut des Maladies Neurodégénératives, CNRS, UMR5293, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR5293, Bordeaux, France
| | - Olivier Nicole
- Institut des Maladies Neurodégénératives, CNRS, UMR5293, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR5293, Bordeaux, France
| | - Mirjana Nacka-Aleksić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Sanja Leštarević
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| |
Collapse
|
31
|
Aging-Exacerbated Acute Axon and Myelin Injury Is Associated with Microglia-Derived Reactive Oxygen Species and Is Alleviated by the Generic Medication Indapamide. J Neurosci 2020; 40:8587-8600. [PMID: 33060175 DOI: 10.1523/jneurosci.1098-20.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/26/2020] [Accepted: 09/24/2020] [Indexed: 11/21/2022] Open
Abstract
Age is a critical risk factor for many neurologic conditions, including progressive multiple sclerosis. Yet the mechanisms underlying the relationship are unknown. Using lysolecithin-induced demyelinating injury to the mouse spinal cord, we characterized the acute lesion and investigated the mechanisms of increased myelin and axon damage with age. We report exacerbated myelin and axon loss in middle-aged (8-10 months of age) compared with young (6 weeks of age) female C57BL/6 mice by 1-3 d of lesion evolution in the white matter. Transcriptomic analysis linked elevated injury to increased expression of Cybb, the gene encoding the catalytic subunit of NADPH oxidase gp91phox. Immunohistochemistry in male and female Cx3cr1 CreER/+ :Rosa26 tdTom/+ mice for gp91phox revealed that the upregulation in middle-aged animals occurred primarily in microglia and not infiltrated monocyte-derived macrophages. Activated NADPH oxidase generates reactive oxygen species and elevated oxidative damage was corroborated by higher malondialdehyde immunoreactivity in lesions from middle-aged compared with young mice. From a previously conducted screen for generic drugs with antioxidant properties, we selected the antihypertensive CNS-penetrant medication indapamide for investigation. We report that indapamide reduced superoxide derived from microglia cultures and that treatment of middle-aged mice with indapamide was associated with a decrease in age-exacerbated lipid peroxidation, demyelination and axon loss. In summary, age-exacerbated acute injury following lysolecithin administration is mediated in part by microglia NADPH oxidase activation, and this is alleviated by the CNS-penetrant antioxidant, indapamide.SIGNIFICANCE STATEMENT Age is associated with an increased risk for the development of several neurologic conditions including progressive multiple sclerosis, which is represented by substantial microglia activation. We demonstrate that in the lysolecithin demyelination model in young and middle-aged mice, the latter group developed greater acute axonal and myelin loss attributed to elevated oxidative stress through NADPH oxidase in lineage-traced microglia. We thus used a CNS-penetrant generic medication used in hypertension, indapamide, as we found it to have antioxidant properties in a previous drug screen. Following lysolecithin demyelination in middle-aged mice, indapamide treatment was associated with decreased oxidative stress and axon/myelin loss. We propose indapamide as a potential adjunctive therapy in aging-associated neurodegenerative conditions such as Alzheimer's disease and progressive multiple sclerosis.
Collapse
|
32
|
Eugenol-Encapsulated Nanocarriers for Microglial Polarisation: a Promising Therapeutic Application for Neuroprotection. BIONANOSCIENCE 2020. [DOI: 10.1007/s12668-020-00789-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
33
|
Kwilasz AJ, Todd LS, Duran-Malle JC, Schrama AEW, Mitten EH, Larson TA, Clements MA, Harris KM, Litwiler ST, Wang X, Van Dam AM, Maier SF, Rice KC, Watkins LR, Barrientos RM. Experimental autoimmune encephalopathy (EAE)-induced hippocampal neuroinflammation and memory deficits are prevented with the non-opioid TLR2/TLR4 antagonist (+)-naltrexone. Behav Brain Res 2020; 396:112896. [PMID: 32905811 DOI: 10.1016/j.bbr.2020.112896] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/12/2020] [Accepted: 08/30/2020] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis (MS) is associated with burdensome memory impairments and preclinical literature suggests that these impairments are linked to neuroinflammation. Previously, we have shown that toll-like receptor 4 (TLR4) antagonists, such as (+)-naltrexone [(+)-NTX], block neuropathic pain and associated spinal inflammation in rats. Here we extend these findings to first demonstrate that (+)-NTX blocks TLR2 in addition to TLR4. Additionally, we examined in two rat strains whether (+)-NTX could attenuate learning and memory disturbances and associated neuroinflammation using a low-dose experimental autoimmune encephalomyelitis (EAE) model of MS. EAE is the most commonly used experimental model for the human inflammatory demyelinating disease, MS. This low-dose model avoided motor impairments that would confound learning and memory measurements. Fourteen days later, daily subcutaneous (+)-NTX or saline injections began and continued throughout the study. Contextual and auditory-fear conditioning were conducted at day 21 to assess hippocampal and amygdalar function. With this low-dose model, EAE impaired long-term, but not short-term, contextual fear memory; both long-term and short-term auditory-cued fear memory were spared. This was associated with increased mRNA for hippocampal interleukin-1β (IL-1β), TLR2, TLR4, NLRP3, and IL-17 and elevated expression of the microglial marker Iba1 in CA1 and DG regions of the hippocampus, confirming the neuroinflammation observed in higher-dose EAE models. Importantly, (+)-NTX completely prevented the EAE-induced memory impairments and robustly attenuated the associated proinflammatory effects. These findings suggest that (+)-NTX may exert therapeutic effects on memory function by dampening the neuroinflammatory response in the hippocampus through blockade of TLR2/TLR4. This study suggests that TLR2 and TLR4 antagonists may be effective at treating MS-related memory deficits.
Collapse
Affiliation(s)
- Andrew J Kwilasz
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Laurel S Todd
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Julissa C Duran-Malle
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Anouk E W Schrama
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Eric H Mitten
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Tracey A Larson
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Madison A Clements
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Kevin M Harris
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Scott T Litwiler
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of the Sciences, Changchun, Jilin 130022, China
| | - Anne-Marie Van Dam
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Kenner C Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Linda R Watkins
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; The Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Ruth M Barrientos
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; The Center for Neuroscience, University of Colorado, Boulder, CO, USA; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
34
|
Plastini MJ, Desu HL, Brambilla R. Dynamic Responses of Microglia in Animal Models of Multiple Sclerosis. Front Cell Neurosci 2020; 14:269. [PMID: 32973458 PMCID: PMC7468479 DOI: 10.3389/fncel.2020.00269] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/31/2020] [Indexed: 12/20/2022] Open
Abstract
Microglia play an essential role in maintaining central nervous system (CNS) homeostasis, as well as responding to injury and disease. Most neurological disorders feature microglial activation, a process whereby microglia undergo profound morphological and transcriptional changes aimed at containing CNS damage and promoting repair, but often resulting in overt inflammation that sustains and propagates the neurodegenerative process. This is especially evident in multiple sclerosis (MS), were microglial activation and microglia-driven neuroinflammation are considered key events in the onset, progression, and resolution of the disease. Our understanding of microglial functions in MS has widened exponentially in the last decade by way of new tools and markers to discriminate microglia from other myeloid populations. Consequently, the complex functional and phenotypical diversity of microglia can now be appreciated. This, in combination with a variety of animal models that mimic specific features and processes of MS, has contributed to filling the gap of knowledge in the cascade of events underlying MS pathophysiology. The purpose of this review is to present the most up to date knowledge of the dynamic responses of microglia in the commonly used animal models of MS, specifically the immune-mediated experimental autoimmune encephalomyelitis (EAE) model, and the chemically-induced cuprizone and lysolecithin models. Elucidating the spectrum of microglial functions in these models, from detrimental to protective, is essential to identify emerging targets for therapy and guide drug discovery efforts.
Collapse
Affiliation(s)
- Melanie J Plastini
- The Miami Project To Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States.,The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Haritha L Desu
- The Miami Project To Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States.,The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Roberta Brambilla
- The Miami Project To Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States.,The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, United States.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE-Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
35
|
Inhibition of the NLRP3-inflammasome prevents cognitive deficits in experimental autoimmune encephalomyelitis mice via the alteration of astrocyte phenotype. Cell Death Dis 2020; 11:377. [PMID: 32415059 PMCID: PMC7229224 DOI: 10.1038/s41419-020-2565-2] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 01/15/2023]
Abstract
Multiple sclerosis (MS) is a chronic disease that is characterized by demyelination and axonal damage in the central nervous system. Cognitive deficits are recognized as one of the features of MS, and these deficits affect the patients’ quality of life. Increasing evidence from experimental autoimmune encephalomyelitis (EAE), the animal model of MS, has suggested that EAE mice exhibit hippocampal impairment and cognitive deficits. However, the underlying mechanisms are still unclear. The NLRP3 inflammasome is a key contributor to neuroinflammation and is involved in the development of MS and EAE. Activation of the NLRP3 inflammasome in microglia is fundamental for subsequent inflammatory events. Activated microglia can convert astrocytes to the neurotoxic A1 phenotype in a variety of neurological diseases. However, it remains unknown whether the NLRP3 inflammasome contributes to cognitive deficits and astrocyte phenotype alteration in EAE. In this study, we demonstrated that severe memory deficits occurred in the late phase of EAE, and cognitive deficits were ameliorated by treatment with MCC950, an inhibitor of the NLRP3 inflammasome. In addition, MCC950 alleviated hippocampal pathology and synapse loss. Astrocytes from EAE mice were converted to the neurotoxic A1 phenotype, and this conversion was prevented by MCC950 treatment. IL-18, which is the downstream of NLRP3 inflammasome, was sufficient to induce the conversion of astrocytes to the A1 phenotype through the NF-κB pathway. IL-18 induced A1 type reactive astrocytes impaired hippocampal neurons through the release of complement component 3 (C3). Altogether, our present data suggest that the NLRP3 inflammasome plays an important role in cognitive deficits in EAE, possibly via the alteration of astrocyte phenotypes. Our study provides a novel therapeutic strategy for hippocampal impairment in EAE and MS.
Collapse
|
36
|
Inflammasome and Cognitive Symptoms in Human Diseases: Biological Evidence from Experimental Research. Int J Mol Sci 2020; 21:ijms21031103. [PMID: 32046097 PMCID: PMC7036918 DOI: 10.3390/ijms21031103] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
Cognitive symptoms are prevalent in the elderly and are associated with an elevated risk of developing dementia. Disease-driven changes can cause cognitive disabilities in memory, attention, and language. The inflammasome is an innate immune intracellular complex that has a critical role in the host defense system, in that it senses infectious pathogen-associated and endogenous danger-associated molecular patterns. An unbalanced or dysregulated inflammasome is associated with infectious, inflammatory, and neurodegenerative diseases. Due to its importance in such pathological conditions, the inflammasome is an emerging drug target for human diseases. A growing number of studies have revealed links between cognitive symptoms and the inflammasome. Several studies have shown that reducing the inflammasome component mitigates cognitive symptoms in diseased states. Therefore, understanding the inflammasome regulatory mechanisms may be required for the prevention and treatment of cognitive symptoms. The purpose of this review is to discuss the current understanding of the inflammasome and its relationships with cognitive symptoms in various human diseases.
Collapse
|
37
|
Nox2 dependent redox-regulation of microglial response to amyloid-β stimulation and microgliosis in aging. Sci Rep 2020; 10:1582. [PMID: 32005915 PMCID: PMC6994719 DOI: 10.1038/s41598-020-58422-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 12/12/2019] [Indexed: 12/19/2022] Open
Abstract
Microglia express constitutively a Nox2 enzyme that is involved in neuroinflammation by the generation of reactive oxygen species (ROS). Amyloid β (Aβ) plays a crucial role in Alzheimer's disease. However, the mechanism of Aβ-induced microglial dysfunction and redox-regulation of microgliosis in aging remains unclear. In this study, we examined Nox2-derived ROS in mediating microglial response to Aβ peptide 1-42 (Aβ42) stimulation in vitro, in aging-associated microgliosis in vivo and in post-mortem human samples. Compared to controls, Aβ42 markedly induced BV2 cell ROS production, Nox2 expression, p47phox and ERK1/2 phosphorylation, cell proliferation and IL-1β secretion. All these changes could be inhibited to the control levels in the presence of Nox2 inhibitor or superoxide scavenger. Compared to young (3-4 months) controls, midbrain tissues from wild-type aging mice (20-22 months) had significantly higher levels of Nox2-derived ROS production, Aβ deposition, microgliosis and IL-1β production. However, these aging-related changes were reduced or absent in Nox2 knockout aging mice. Clinical significance of aging-associated Nox2 activation, microgliosis and IL-1β production was investigated using post-mortem midbrain tissues of humans at young (25-38 years) and old age (61-85 years). In conclusion, Nox2-dependent redox-signalling is crucial in microglial response to Aβ42 stimulation and in aging-associated microgliosis and brain inflammation.
Collapse
|
38
|
Morris G, Berk M, Maes M, Carvalho AF, Puri BK. Socioeconomic Deprivation, Adverse Childhood Experiences and Medical Disorders in Adulthood: Mechanisms and Associations. Mol Neurobiol 2019; 56:5866-5890. [PMID: 30685844 PMCID: PMC6614134 DOI: 10.1007/s12035-019-1498-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 01/15/2019] [Indexed: 12/30/2022]
Abstract
Severe socioeconomic deprivation (SED) and adverse childhood experiences (ACE) are significantly associated with the development in adulthood of (i) enhanced inflammatory status and/or hypothalamic-pituitary-adrenal (HPA) axis dysfunction and (ii) neurological, neuroprogressive, inflammatory and autoimmune diseases. The mechanisms by which these associations take place are detailed. The two sets of consequences are themselves strongly associated, with the first set likely contributing to the second. Mechanisms enabling bidirectional communication between the immune system and the brain are described, including complex signalling pathways facilitated by factors at the level of immune cells. Also detailed are mechanisms underpinning the association between SED, ACE and the genesis of peripheral inflammation, including epigenetic changes to immune system-related gene expression. The duration and magnitude of inflammatory responses can be influenced by genetic factors, including single nucleotide polymorphisms, and by epigenetic factors, whereby pro-inflammatory cytokines, reactive oxygen species, reactive nitrogen species and nuclear factor-κB affect gene DNA methylation and histone acetylation and also induce several microRNAs including miR-155, miR-181b-1 and miR-146a. Adult HPA axis activity is regulated by (i) genetic factors, such as glucocorticoid receptor polymorphisms; (ii) epigenetic factors affecting glucocorticoid receptor function or expression, including the methylation status of alternative promoter regions of NR3C1 and the methylation of FKBP5 and HSD11β2; (iii) chronic inflammation and chronic nitrosative and oxidative stress. Finally, it is shown how severe psychological stress adversely affects mitochondrial structure and functioning and is associated with changes in brain mitochondrial DNA copy number and transcription; mitochondria can act as couriers of childhood stress into adulthood.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, P.O. Box 291, Geelong, Victoria, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Level 1 North, Main Block, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, 35 Poplar Rd, Parkville, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - André F Carvalho
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Centre for Addiction & Mental Health (CAMH), Toronto, ON, Canada
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
39
|
High dose of dexamethasone protects against EAE-induced motor deficits but impairs learning/memory in C57BL/6 mice. Sci Rep 2019; 9:6673. [PMID: 31040362 PMCID: PMC6491620 DOI: 10.1038/s41598-019-43217-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 03/17/2019] [Indexed: 12/24/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune and neuroinflammatory disease characterized by demyelination of the Central Nervous System. Immune cells activation and release of pro-inflammatory cytokines play a crucial role in the disease modulation, decisively contributing to the neurodegeneration observed in MS and the experimental autoimmune encephalomyelitis (EAE), the widely used MS animal model. Synthetic glucocorticoids, commonly used to treat the MS attacks, have controversial effects on neuroinflammation and cognition. We sought to verify the influence of dexamethasone (DEX) on the EAE progression and on EAE-induced cognitive deficits. In myelin oligodendrocyte glycoprotein peptide (MOG35-55)-induced EAE female mice, treated once with DEX (50 mg/kg) or not, on the day of immunization, DEX decreased EAE-induced motor clinical scores, infiltrating cells in the spinal cord and delayed serum corticosterone peak. At the asymptomatic phase (8-day post-immunization), DEX did not protected from the EAE-induced memory consolidation deficits, which were accompanied by increased glucocorticoid receptor (GR) activity and decreased EGR-1 expression in the hippocampus. Blunting hippocampal GR genomic activation with DnGR vectors prevented DEX effects on EAE-induced memory impairment. These data suggest that, although DEX improves clinical signs, it decreases cognitive and memory capacity by diminishing neuronal activity and potentiating some aspects of neuroinflammation in EAE.
Collapse
|
40
|
Neuregulin-1 Fosters Supportive Interactions between Microglia and Neural Stem/Progenitor Cells. Stem Cells Int 2019; 2019:8397158. [PMID: 31089334 PMCID: PMC6476022 DOI: 10.1155/2019/8397158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/31/2018] [Accepted: 02/13/2019] [Indexed: 01/23/2023] Open
Abstract
Microglia play diverse roles in homeostasis and pathology of the central nervous system (CNS). Their response to injury or insult is critical for initiating neuroinflammation and tissue damage as well as resolution of inflammation and wound healing. Changes to the microenvironment of microglia appear to be a key determinant of their phenotype and their role in the endogenous repair process in the injured or diseased CNS. Our recent findings have identified a positive role for neuregulin-1 (Nrg-1) in regulating immune response in spinal cord injury and focal demyelinating lesions. We show that increasing the tissue availability of Nrg-1 after injury can promote endogenous repair by modulating neuroinflammation. In the present study, we sought to elucidate the specific role of Nrg-1 in regulating microglial activity and more importantly their influence on the behavior of neural stem/progenitor cells (NPCs). Using injury-relevant in vitro systems, we demonstrate that Nrg-1 attenuates the expression of proinflammatory mediators in activated microglia. Moreover, we provide novel evidence that availability of Nrg-1 can restore the otherwise suppressed phagocytic ability of proinflammatory microglia. Interestingly, the presence of Nrg-1 in the microenvironment of proinflammatory microglia mitigates their inhibitory effects on NPC proliferation. Nrg-1 treated proinflammatory microglia also augment mobilization of NPCs, while they had no influence on their suppressive effects on NPC differentiation. Mechanistically, we show that Nrg-1 enhances the interactions of proinflammatory microglia and NPCs, at least in part, through reduction of TNF-α expression in microglia. These findings provide new insights into the endogenous regulation of microglia-NPC interactions and identify new potential targets for optimizing this important crosstalk during the regenerative process after CNS injury and neuroinflammatory conditions.
Collapse
|
41
|
Mancini A, Gaetani L, Gentili L, Di Filippo M. Finding a way to preserve mitochondria: new pathogenic pathways in experimental multiple sclerosis. Neural Regen Res 2019; 14:77-78. [PMID: 30531078 PMCID: PMC6262986 DOI: 10.4103/1673-5374.243707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- Andrea Mancini
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - Lorenzo Gaetani
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - Lucia Gentili
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - Massimiliano Di Filippo
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| |
Collapse
|
42
|
Enhanced GABAergic Tonic Inhibition Reduces Intrinsic Excitability of Hippocampal CA1 Pyramidal Cells in Experimental Autoimmune Encephalomyelitis. Neuroscience 2018; 395:89-100. [PMID: 30447391 DOI: 10.1016/j.neuroscience.2018.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 11/03/2018] [Accepted: 11/05/2018] [Indexed: 11/24/2022]
Abstract
Cognitive impairment (CI), a debilitating and pervasive feature of multiple sclerosis (MS), is correlated with hippocampal atrophy. Findings from postmortem MS hippocampi indicate that expression of genes involved in both excitatory and inhibitory neurotransmission are altered in MS, and although deficits in excitatory neurotransmission have been reported in the MS model experimental autoimmune encephalomyelitis (EAE), the functional consequence of altered inhibitory neurotransmission remains poorly understood. In this study, we used electrophysiological and biochemical techniques to examine inhibitory neurotransmission in the CA1 region of the hippocampus in EAE. We find that tonic, GABAergic inhibition is enhanced in CA1 pyramidal cells from EAE mice. Although plasma membrane expression of the GABA transporter GAT-3 was decreased in the EAE hippocampus, an increased surface expression of α5 subunit-containing GABAA receptors appears to be primarily responsible for the increase in tonic inhibition during EAE. Enhanced tonic inhibition during EAE was associated with decreased CA1 pyramidal cell excitability and inhibition of α5 subunit-containing GABAA receptors with the negative allosteric modulator L-655,708 enhanced pyramidal cell excitability in EAE mice. Together, our results suggest that altered GABAergic neurotransmission may underlie deficits in hippocampus-dependent cognitive function in EAE and MS.
Collapse
|
43
|
Bellizzi MJ, Hammond JW, Li H, Gantz Marker MA, Marker DF, Freeman RS, Gelbard HA. The Mixed-Lineage Kinase Inhibitor URMC-099 Protects Hippocampal Synapses in Experimental Autoimmune Encephalomyelitis. eNeuro 2018; 5:ENEURO.0245-18.2018. [PMID: 30627663 PMCID: PMC6325567 DOI: 10.1523/eneuro.0245-18.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/31/2018] [Accepted: 11/07/2018] [Indexed: 12/04/2022] Open
Abstract
Treatments to stop gray matter degeneration are needed to prevent progressive disability in multiple sclerosis (MS). We tested whether inhibiting mixed-lineage kinases (MLKs), which can drive inflammatory microglial activation and neuronal degeneration, could protect hippocampal synapses in C57BL/6 mice with experimental autoimmune encephalomyelitis (EAE), a disease model that recapitulates the excitatory synaptic injury that occurs widely within the gray matter in MS. URMC-099, a broad spectrum MLK inhibitor with additional activity against leucine-rich repeat kinase 2 (LRRK2) and other kinases, prevented loss of PSD95-positive postsynaptic structures, shifted activated microglia toward a less inflammatory phenotype, and reversed deficits in hippocampal-dependent contextual fear conditioning in EAE mice when administered after the onset of motor symptoms. A narrow spectrum inhibitor designed to be highly selective for MLK3 failed to protect synapses in EAE hippocampi, and could not rescue cultured neurons from trophic deprivation in an in vitro model of MLK-driven neuronal degeneration. These results suggest that URMC-099 may have potential as a neuroprotective treatment in MS and demonstrate that a broad spectrum of inhibition against a combination of MLK and other kinases is more effective in neuroinflammatory disease than selectively targeting a single kinase.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Calcium-Binding Proteins/metabolism
- Cells, Cultured
- Conditioning, Psychological/drug effects
- Cytokines/genetics
- Cytokines/metabolism
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Enzyme Inhibitors/therapeutic use
- Fear/drug effects
- Fear/psychology
- Female
- Hippocampus/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Microfilament Proteins/metabolism
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Neurons/drug effects
- Neuroprotective Agents/therapeutic use
- Peptide Fragments/toxicity
- Pyridines/therapeutic use
- Pyrroles/therapeutic use
- Superior Cervical Ganglion/cytology
- Synapses/drug effects
Collapse
Affiliation(s)
- Matthew J. Bellizzi
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY 14642
| | - Jennetta W. Hammond
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
| | - Herman Li
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
| | - Mary A. Gantz Marker
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642
| | - Daniel F. Marker
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
| | - Robert S. Freeman
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642
| | - Harris A. Gelbard
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY 14642
- Departments of Pediatrics and Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
44
|
Yang L, Jin P, Wang X, Zhou Q, Lin X, Xi S. Fluoride activates microglia, secretes inflammatory factors and influences synaptic neuron plasticity in the hippocampus of rats. Neurotoxicology 2018; 69:108-120. [PMID: 30273629 DOI: 10.1016/j.neuro.2018.09.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 09/16/2018] [Accepted: 09/24/2018] [Indexed: 11/18/2022]
Abstract
Epidemiological studies have reported that highly fluoridated drinking water may significantly decrease the Intelligence Quotient (IQ) of exposed children. It is thought that synaptic plasticity is the basis of learning and memory skills in developing children. However, the effect on synaptic plasticity by activated microglia induced via fluoride treatment is less clear. Our previous research showed that fluoride ions activated microglia which then released pro-inflammatory cytokines. In this study, hippocampal-dependent memory status was evaluated in rat models sub-chronically exposed to fluoride in their drinking water. Microglial activation in the hippocampus was examined using immunofluorescence staining and the expression of synaptophysin (SYP) and postsynaptic density protein 95 (PSD-95), Long-term potentiation (LTP) and the expression of Amino-3-hydroxy-5-methy-4-isoxazole propionate (AMPA) receptor subunit GluR2 as well as N-methyl-d-aspartate (NMDA) receptor subunit NMDAR2β of exposed rats. We found that fluoride exposure activated microglia and increased the expression of DAP12 and TREM2, as well as promoted pro-inflammatory cytokines secretion via ERK/MAPK and P38/MAPK signal pathways. Furthermore fluoride depressed LTP and decreased PSD-95 protein levels as well as expression of ionotropic glutamate receptors GluR2 and NMDAR2β. We concluded that the role of fluoride on synaptic plasticity may be associated with neuroinflammation induced by microglia.
Collapse
Affiliation(s)
- Li Yang
- Department of Environmental and Occupational Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China.
| | - Peiyu Jin
- Department of Environmental and Occupational Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China.
| | - Xiaoyan Wang
- Department of Environmental and Occupational Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China.
| | - Qing Zhou
- Department of Environmental and Occupational Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China.
| | - Xiaoli Lin
- Department of Environmental and Occupational Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China.
| | - Shuhua Xi
- Department of Environmental and Occupational Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China.
| |
Collapse
|
45
|
Tumor Necrosis Factor and Interleukin-1 β Modulate Synaptic Plasticity during Neuroinflammation. Neural Plast 2018; 2018:8430123. [PMID: 29861718 PMCID: PMC5976900 DOI: 10.1155/2018/8430123] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 03/28/2018] [Indexed: 11/25/2022] Open
Abstract
Cytokines are constitutively released in the healthy brain by resident myeloid cells to keep proper synaptic plasticity, either in the form of Hebbian synaptic plasticity or of homeostatic plasticity. However, when cytokines dramatically increase, establishing a status of neuroinflammation, the synaptic action of such molecules remarkably interferes with brain circuits of learning and cognition and contributes to excitotoxicity and neurodegeneration. Among others, interleukin-1β (IL-1β) and tumor necrosis factor (TNF) are the best studied proinflammatory cytokines in both physiological and pathological conditions and have been invariably associated with long-term potentiation (LTP) (Hebbian synaptic plasticity) and synaptic scaling (homeostatic plasticity), respectively. Multiple sclerosis (MS) is the prototypical neuroinflammatory disease, in which inflammation triggers excitotoxic mechanisms contributing to neurodegeneration. IL-β and TNF are increased in the brain of MS patients and contribute to induce the changes in synaptic plasticity occurring in MS patients and its animal model, the experimental autoimmune encephalomyelitis (EAE). This review will introduce and discuss current evidence of the role of IL-1β and TNF in the regulation of synaptic strength at both physiological and pathological levels, in particular speculating on their involvement in the synaptic plasticity changes observed in the EAE brain.
Collapse
|
46
|
Oswald MCW, Garnham N, Sweeney ST, Landgraf M. Regulation of neuronal development and function by ROS. FEBS Lett 2018; 592:679-691. [PMID: 29323696 PMCID: PMC5888200 DOI: 10.1002/1873-3468.12972] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 01/02/2018] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species (ROS) have long been studied as destructive agents in the context of nervous system ageing, disease and degeneration. Their roles as signalling molecules under normal physiological conditions is less well understood. Recent studies have provided ample evidence of ROS-regulating neuronal development and function, from the establishment of neuronal polarity to growth cone pathfinding; from the regulation of connectivity and synaptic transmission to the tuning of neuronal networks. Appreciation of the varied processes that are subject to regulation by ROS might help us understand how changes in ROS metabolism and buffering could progressively impact on neuronal networks with age and disease.
Collapse
Affiliation(s)
| | - Nathan Garnham
- Department of BiologyUniversity of YorkHeslington YorkUK
| | | | | |
Collapse
|
47
|
Mancini A, Tantucci M, Mazzocchetti P, de Iure A, Durante V, Macchioni L, Giampà C, Alvino A, Gaetani L, Costa C, Tozzi A, Calabresi P, Di Filippo M. Microglial activation and the nitric oxide/cGMP/PKG pathway underlie enhanced neuronal vulnerability to mitochondrial dysfunction in experimental multiple sclerosis. Neurobiol Dis 2018; 113:97-108. [PMID: 29325869 DOI: 10.1016/j.nbd.2018.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 12/07/2017] [Accepted: 01/04/2018] [Indexed: 12/24/2022] Open
Abstract
During multiple sclerosis (MS), a close link has been demonstrated to occur between inflammation and neuro-axonal degeneration, leading to the hypothesis that immune mechanisms may promote neurodegeneration, leading to irreversible disease progression. Energy deficits and inflammation-driven mitochondrial dysfunction seem to be involved in this process. In this work we investigated, by the use of striatal electrophysiological field-potential recordings, if the inflammatory process associated with experimental autoimmune encephalomyelitis (EAE) is able to influence neuronal vulnerability to the blockade of mitochondrial complex IV, a crucial component for mitochondrial activity responsible of about 90% of total cellular oxygen consumption. We showed that during the acute relapsing phase of EAE, neuronal susceptibility to mitochondrial complex IV inhibition is markedly enhanced. This detrimental effect was counteracted by the pharmacological inhibition of microglia, of nitric oxide (NO) synthesis and its intracellular pathway (involving soluble guanylyl cyclase, sGC, and protein kinase G, PKG). The obtained results suggest that mitochondrial complex IV exerts an important role in maintaining neuronal energetic homeostasis during EAE. The pathological processes associated with experimental MS, and in particular the activation of microglia and of the NO pathway, lead to an increased neuronal vulnerability to mitochondrial complex IV inhibition, representing promising pharmacological targets.
Collapse
Affiliation(s)
- Andrea Mancini
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Michela Tantucci
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Petra Mazzocchetti
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Antonio de Iure
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Valentina Durante
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Lara Macchioni
- Sezione di Fisiologia e Biochimica, Dipartimento di Medicina Sperimentale, Università degli Studi di Perugia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Carmela Giampà
- Università Cattolica del Sacro Cuore, Istituto di Anatomia Umana e Biologia Cellulare, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Alessandra Alvino
- Università Cattolica del Sacro Cuore, Istituto di Anatomia Umana e Biologia Cellulare, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Lorenzo Gaetani
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Cinzia Costa
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Alessandro Tozzi
- Sezione di Fisiologia e Biochimica, Dipartimento di Medicina Sperimentale, Università degli Studi di Perugia, S. Andrea delle Fratte, 06132 Perugia, Italy; IRCCS, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Paolo Calabresi
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy; IRCCS, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Massimiliano Di Filippo
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy.
| |
Collapse
|
48
|
Drago F, Lombardi M, Prada I, Gabrielli M, Joshi P, Cojoc D, Franck J, Fournier I, Vizioli J, Verderio C. ATP Modifies the Proteome of Extracellular Vesicles Released by Microglia and Influences Their Action on Astrocytes. Front Pharmacol 2017; 8:910. [PMID: 29321741 PMCID: PMC5733563 DOI: 10.3389/fphar.2017.00910] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 11/29/2017] [Indexed: 11/23/2022] Open
Abstract
Extracellular ATP is among molecules promoting microglia activation and inducing the release of extracellular vesicles (EVs), which are potent mediators of intercellular communication between microglia and the microenvironment. We previously showed that EVs produced under ATP stimulation (ATP-EVs) propagate a robust inflammatory reaction among astrocytes and microglia in vitro and in mice with subclinical neuroinflammation (Verderio et al., 2012). However, the proteome of EVs released upon ATP stimulation has not yet been elucidated. In this study we applied a label free proteomic approach to characterize the proteome of EVs released constitutively and during microglia activation with ATP. We show that ATP drives sorting in EVs of a set of proteins implicated in cell adhesion/extracellular matrix organization, autophagy-lysosomal pathway and cellular metabolism, that may influence the response of recipient astrocytes to EVs. These data provide new clues to molecular mechanisms involved in microglia response to ATP and in microglia signaling to the environment via EVs.
Collapse
Affiliation(s)
- Francesco Drago
- Univ. Lille, INSERM, U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse - PRISM, Lille, France.,Fondazione Istituto Oncologico del Mediterraneo, Viagrande, Italy
| | | | | | | | - Pooja Joshi
- Institute of Neuroscience (CNR), Milan, Italy
| | - Dan Cojoc
- Institute of Materials (CNR), Trieste, Italy
| | - Julien Franck
- Univ. Lille, INSERM, U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse - PRISM, Lille, France
| | - Isabelle Fournier
- Univ. Lille, INSERM, U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse - PRISM, Lille, France
| | - Jacopo Vizioli
- Univ. Lille, INSERM, U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse - PRISM, Lille, France
| | - Claudia Verderio
- IRCCS Humanitas, Rozzano, Italy.,Institute of Neuroscience (CNR), Milan, Italy
| |
Collapse
|
49
|
Stampanoni Bassi M, Gilio L, Buttari F, Maffei P, Marfia GA, Restivo DA, Centonze D, Iezzi E. Remodeling Functional Connectivity in Multiple Sclerosis: A Challenging Therapeutic Approach. Front Neurosci 2017; 11:710. [PMID: 29321723 PMCID: PMC5733539 DOI: 10.3389/fnins.2017.00710] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/04/2017] [Indexed: 11/13/2022] Open
Abstract
Neurons in the central nervous system are organized in functional units interconnected to form complex networks. Acute and chronic brain damage disrupts brain connectivity producing neurological signs and/or symptoms. In several neurological diseases, particularly in Multiple Sclerosis (MS), structural imaging studies cannot always demonstrate a clear association between lesion site and clinical disability, originating the "clinico-radiological paradox." The discrepancy between structural damage and disability can be explained by a complex network perspective. Both brain networks architecture and synaptic plasticity may play important roles in modulating brain networks efficiency after brain damage. In particular, long-term potentiation (LTP) may occur in surviving neurons to compensate network disconnection. In MS, inflammatory cytokines dramatically interfere with synaptic transmission and plasticity. Importantly, in addition to acute and chronic structural damage, inflammation could contribute to reduce brain networks efficiency in MS leading to worse clinical recovery after a relapse and worse disease progression. These evidence suggest that removing inflammation should represent the main therapeutic target in MS; moreover, as synaptic plasticity is particularly altered by inflammation, specific strategies aimed at promoting LTP mechanisms could be effective for enhancing clinical recovery. Modulation of plasticity with different non-invasive brain stimulation (NIBS) techniques has been used to promote recovery of MS symptoms. Better knowledge of features inducing brain disconnection in MS is crucial to design specific strategies to promote recovery and use NIBS with an increasingly tailored approach.
Collapse
Affiliation(s)
- Mario Stampanoni Bassi
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Luana Gilio
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Pierpaolo Maffei
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | - Girolama A Marfia
- Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | | | - Diego Centonze
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Ennio Iezzi
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| |
Collapse
|
50
|
Choi DH, Lee J. A Mini-Review of the NADPH oxidases in Vascular Dementia: Correlation with NOXs and Risk Factors for VaD. Int J Mol Sci 2017; 18:ijms18112500. [PMID: 29165383 PMCID: PMC5713465 DOI: 10.3390/ijms18112500] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress (OS) is one of the factors that cause dementia conditions such as Alzheimer’s disease and vascular dementia (VaD). In the pathogenesis of VaD, OS is associated with risk factors that include increased age, hypertension, and stroke. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are a molecular source of reactive oxygen species (ROS). According to recent studies, inhibition of NOX activity can reduce cognitive impairment in animal models of VaD. In this article, we review the evidence linking cognitive impairment with NOX-dependent OS, including the vascular NOX and non-vascular NOX systems, in VaD.
Collapse
Affiliation(s)
- Dong-Hee Choi
- Center for Neuroscience Research, Institute of Biomedical Science & Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 143701, Korea.
- Department of Medical Science, Konkuk University School of Medicine, 120 Neungdong-ro, Gwangjin-gu, Seoul 143701, Korea.
| | - Jongmin Lee
- Department of Medical Science, Konkuk University School of Medicine, 120 Neungdong-ro, Gwangjin-gu, Seoul 143701, Korea.
- Department of Rehabilitation Medicine, Konkuk University School of Medicine, 120 Neungdong-ro, Gwangjin-gu, Seoul 143701, Korea.
| |
Collapse
|