101
|
Wang D, Chen F, Han Z, Yin Z, Ge X, Lei P. Relationship Between Amyloid-β Deposition and Blood-Brain Barrier Dysfunction in Alzheimer's Disease. Front Cell Neurosci 2021; 15:695479. [PMID: 34349624 PMCID: PMC8326917 DOI: 10.3389/fncel.2021.695479] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
Amyloid-β (Aβ) is the predominant pathologic protein in Alzheimer's disease (AD). The production and deposition of Aβ are important factors affecting AD progression and prognosis. The deposition of neurotoxic Aβ contributes to damage of the blood-brain barrier. However, the BBB is also crucial in maintaining the normal metabolism of Aβ, and dysfunction of the BBB aggravates Aβ deposition. This review characterizes Aβ deposition and BBB damage in AD, summarizes their interactions, and details their respective mechanisms.
Collapse
Affiliation(s)
- Dong Wang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| | | | - Zhaoli Han
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| | - Zhenyu Yin
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| | - Xintong Ge
- Tianjin Neurological Institute, Tianjin, China
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| |
Collapse
|
102
|
Saitoh BY, Tanaka E, Yamamoto N, Kruining DV, Iinuma K, Nakamuta Y, Yamaguchi H, Yamasaki R, Matsumoto K, Kira JI. Early postnatal allergic airway inflammation induces dystrophic microglia leading to excitatory postsynaptic surplus and autism-like behavior. Brain Behav Immun 2021; 95:362-380. [PMID: 33862170 DOI: 10.1016/j.bbi.2021.04.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 04/01/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia play key roles in synaptic pruning, which primarily occurs from the postnatal period to adolescence. Synaptic pruning is essential for normal brain development and its impairment is implicated in neuropsychiatric developmental diseases such as autism spectrum disorders (ASD). Recent epidemiological surveys reported a strong link between ASD and atopic/allergic diseases. However, few studies have experimentally investigated the relationship between allergy and ASD-like manifestations, particularly in the early postnatal period, when allergic disorders occur frequently. Therefore, we aimed to characterize how allergic inflammation in the early postnatal period influences microglia and behavior using mouse models of short- and long-term airway allergy. Male mice were immunized by an intraperitoneal injection of aluminum hydroxide and ovalbumin (OVA) or phosphate-buffered saline (control) on postnatal days (P) 3, 7, and 11, followed by intranasal challenge with OVA or phosphate-buffered saline solution twice a week until P30 or P70. In the hippocampus, Iba-1-positive areas, the size of Iba-1-positive microglial cell bodies, and the ramification index of microglia by Sholl analysis were significantly smaller in the OVA group than in the control group on P30 and P70, although Iba-1-positive microglia numbers did not differ significantly between the two groups. In Iba-1-positive cells, postsynaptic density protein 95 (PSD95)-occupied areas and CD68-occupied areas were significantly decreased on P30 and P70, respectively, in the OVA group compared with the control group. Immunoblotting using hippocampal tissues demonstrated that amounts of PSD95, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor 2, and N-methyl-D-aspartate (NMDA) receptor 2B were significantly increased in the OVA group compared with the control group on P70, and a similar increasing trend for PSD95 was observed on P30. Neurogenesis was not significantly different between the two groups on P30 or P70 by doublecortin immunohistochemistry. The social preference index was significantly lower in the three chamber test and the number of buried marbles was significantly higher in the OVA group than in the control group on P70 but not on P30, whereas locomotion and anxiety were not different between the two groups. Compared with the control group, serum basal corticosterone levels were significantly elevated and hippocampal glucocorticoid receptor (GR) amounts and nuclear GR translocation in microglia, but not in neurons or astrocytes, were significantly decreased in the OVA group on P70 but not on P30. Gene set enrichment analysis of isolated microglia revealed that genes related to immune responses including Toll-like receptor signaling and chemokine signaling pathways, senescence, and glucocorticoid signaling were significantly upregulated in the OVA group compared with the control group on P30 and P70. These findings suggest that early postnatal allergic airway inflammation induces dystrophic microglia that exhibit defective synaptic pruning upon short- and long-term allergen exposure. Furthermore, long-term allergen exposure induced excitatory postsynaptic surplus and ASD-like behavior. Hypothalamo-pituitary-adrenal axis activation and the compensatory downregulation of microglial GR during long-term allergic airway inflammation may also facilitate these changes.
Collapse
Affiliation(s)
- Ban-Yu Saitoh
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eizo Tanaka
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Norio Yamamoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daan van Kruining
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, Netherlands
| | - Kyoko Iinuma
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuko Nakamuta
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichiro Matsumoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 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
|
103
|
Conte C. Possible Link between SARS-CoV-2 Infection and Parkinson's Disease: The Role of Toll-Like Receptor 4. Int J Mol Sci 2021; 22:7135. [PMID: 34281186 PMCID: PMC8269350 DOI: 10.3390/ijms22137135] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/19/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative motor disorder characterized by selective degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) of the midbrain, depletion of dopamine (DA), and impaired nigrostriatal pathway. The pathological hallmark of PD includes the aggregation and accumulation α-synuclein (α-SYN). Although the precise mechanisms underlying the pathogenesis of PD are still unknown, the activation of toll-like receptors (TLRs), mainly TLR4 and subsequent neuroinflammatory immune response, seem to play a significant role. Mounting evidence suggests that viral infection can concur with the precipitation of PD or parkinsonism. The recently identified coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of ongoing pandemic coronavirus disease 2019 (COVID-19), responsible for 160 million cases that led to the death of more than three million individuals worldwide. Studies have reported that many patients with COVID-19 display several neurological manifestations, including acute cerebrovascular diseases, conscious disturbance, and typical motor and non-motor symptoms accompanying PD. In this review, the neurotropic potential of SARS-CoV-2 and its possible involvement in the pathogenesis of PD are discussed. Specifically, the involvement of the TLR4 signaling pathway in mediating the virus entry, as well as the massive immune and inflammatory response in COVID-19 patients is explored. The binding of SARS-CoV-2 spike (S) protein to TLR4 and the possible interaction between SARS-CoV-2 and α-SYN as contributing factors to neuronal death are also considered.
Collapse
Affiliation(s)
- Carmela Conte
- Department of Pharmaceutical Sciences, University of Perugia, via Fabretti, 06123 Perugia, Italy
| |
Collapse
|
104
|
Baicalein, Baicalin, and Wogonin: Protective Effects against Ischemia-Induced Neurodegeneration in the Brain and Retina. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8377362. [PMID: 34306315 PMCID: PMC8263226 DOI: 10.1155/2021/8377362] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/08/2021] [Accepted: 06/19/2021] [Indexed: 12/17/2022]
Abstract
Ischemia is a common pathological condition present in many neurodegenerative diseases, including ischemic stroke, retinal vascular occlusion, diabetic retinopathy, and glaucoma, threatening the sight and lives of millions of people globally. Ischemia can trigger excessive oxidative stress, inflammation, and vascular dysfunction, leading to the disruption of tissue homeostasis and, ultimately, cell death. Current therapies are very limited and have a narrow time window for effective treatment. Thus, there is an urgent need to develop more effective therapeutic options for ischemia-induced neural injuries. With emerging reports on the pharmacological properties of natural flavonoids, these compounds present potent antioxidative, anti-inflammatory, and antiapoptotic agents for the treatment of ischemic insults. Three major active flavonoids, baicalein, baicalin, and wogonin, have been extracted from Scutellaria baicalensis Georgi (S. baicalensis); all of which are reported to have low cytotoxicity. They have been demonstrated to exert promising pharmacological capabilities in preventing cell and tissue damage. This review focuses on the therapeutic potentials of these flavonoids against ischemia-induced neurotoxicity and damage in the brain and retina. The bioactivity and bioavailability of baicalein, baicalin, and wogonin are also discussed. It is with hope that the therapeutic potential of these flavonoids can be utilized and developed as natural treatments for ischemia-induced injuries of the central nervous system (CNS).
Collapse
|
105
|
Song B, Lee SJ, Kim CH. Roles of Cytokines in the Temporal Changes of Microglial Membrane Currents and Neuronal Excitability and Synaptic Efficacy in ATP-Induced Cortical Injury Model. Int J Mol Sci 2021; 22:ijms22136853. [PMID: 34202215 PMCID: PMC8268462 DOI: 10.3390/ijms22136853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022] Open
Abstract
Cytokines are important neuroinflammatory modulators in neurodegenerative brain disorders including traumatic brain injury (TBI) and stroke. However, their temporal effects on the physiological properties of microglia and neurons during the recovery period have been unclear. Here, using an ATP-induced cortical injury model, we characterized selective effects of ATP injection compared to needle-control. In the damaged region, the fluorescent intensity of CX3CR1-GFP (+) cells, as well as the cell density, was increased and the maturation of newborn BrdU (+) cells continued until 28 day-post-injection (dpi) of ATP. The excitability and synaptic E/I balance of neurons and the inward and outward membrane currents of microglia were increased at 3 dpi, when expressions of tumor necrosis factor (TNF)-α/interleukin (IL)-1β and IL-10/IL-4 were also enhanced. These changes of both cells at 3 dpi were mostly decayed at 7 dpi and were suppressed by any of IL-10, IL-4, suramin (P2 receptor inhibitor) and 4-AP (K+ channel blocker). Acute ATP application alone induced only small effects from both naïve neurons and microglial cells in brain slice. However, TNF-α alone effectively increased the excitability of naïve neurons, which was blocked by suramin or 4-AP. TNF-α and IL-1β increased and decreased membrane currents of naïve microglia, respectively. Our results suggest that ATP and TNF-α dominantly induce the physiological activities of 3 dpi neurons and microglia, and IL-10 effectively suppresses such changes of both activated cells in K+ channel- and P2 receptor-dependent manner, while IL-4 suppresses neurons preferentially.
Collapse
Affiliation(s)
- Bokyung Song
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea;
- Neuroscience Program, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
| | - Sung-Joong Lee
- Program in Neuroscience, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea;
| | - Chong-Hyun Kim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea;
- Neuroscience Program, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
- Correspondence:
| |
Collapse
|
106
|
Chaudhary R, Morris RJ, Steinson E. The multifactorial roles of microglia and macrophages in the maintenance and progression of glioblastoma. J Neuroimmunol 2021; 357:577633. [PMID: 34153803 DOI: 10.1016/j.jneuroim.2021.577633] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/26/2021] [Accepted: 06/11/2021] [Indexed: 01/18/2023]
Abstract
The functional characteristics of glial cells, in particular microglia, have attained considerable importance in several diseases, including glioblastoma, the most hostile and malignant type of intracranial tumor. Microglia performs a highly significant role in the brain's inflammatory response mechanism. They exhibit anti-tumor properties via phagocytosis and the activation of a number of different cytotoxic substances. Some tumor-derived factors, however, transform these microglial cells into immunosuppressive and tumor-supportive, facilitating survival and progression of tumorigenic cells. Glioma-associated microglia and/or macrophages (GAMs) accounts for a large proportion of glioma infiltrating cells. Once within the tumor, GAMs exhibit a distinct phenotype of initiation that subsequently supports the growth and development of tumorigenic cells, angiogenesis and stimulates the infiltration of healthy brain regions. Interventions that suppress or prohibit the induction of GAMs at the tumor site or attenuate their immunological activities accommodating anti-tumor actions are likely to exert positive impact on glioblastoma treatment. In the present paper, we aim to summarize the most recent knowledge of microglia and its physiology, as well as include a very brief description of different molecular factors involved in microglia and glioblastoma interplay. We further address some of the major signaling pathways that regulate the baseline motility of glioblastoma progression. Finally, we discussed a number of therapeutic approaches regarding glioblastoma treatment.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, India.
| | - Rhianna J Morris
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Emma Steinson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
107
|
Redmon SN, Yarishkin O, Lakk M, Jo A, Mustafic E, Tvrdik P, Križaj D. TRPV4 channels mediate the mechanoresponse in retinal microglia. Glia 2021; 69:1563-1582. [PMID: 33624376 PMCID: PMC8989051 DOI: 10.1002/glia.23979] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022]
Abstract
The physiological and neurological correlates of plummeting brain osmolality during edema, traumatic CNS injury, and severe ischemia are compounded by neuroinflammation. Using multiple approaches, we investigated how retinal microglia respond to challenges mediated by increases in strain, osmotic gradients, and agonists of the stretch-activated cation channel TRPV4. Dissociated and intact microglia were TRPV4-immunoreactive and responded to the selective agonist GSK1016790A and substrate stretch with altered motility and elevations in intracellular calcium ([Ca2+ ]i ). Agonist- and hypotonicity-induced swelling was associated with a nonselective outwardly rectifying cation current, increased [Ca2+ ]i , and retraction of higher-order processes. The antagonist HC067047 reduced the extent of hypotonicity-induced microglial swelling and inhibited the suppressive effects of GSK1016790A and hypotonicity on microglial branching. Microglial TRPV4 signaling required intermediary activation of phospholipase A2 (PLA2), cytochrome P450, and epoxyeicosatrienoic acid production (EETs). The expression pattern of vanilloid thermoTrp genes in retinal microglia was markedly different from retinal neurons, astrocytes, and cortical microglia. These results suggest that TRPV4 represents a primary retinal microglial sensor of osmochallenges under physiological and pathological conditions. Its activation, associated with PLA2, modulates calcium signaling and cell architecture. TRPV4 inhibition might be a useful strategy to suppress microglial overactivation in the swollen and edematous CNS.
Collapse
Affiliation(s)
- Sarah N. Redmon
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, Salt Lake City, UT 84132
| | - Oleg Yarishkin
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, Salt Lake City, UT 84132
| | - Monika Lakk
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, Salt Lake City, UT 84132
| | - Andrew Jo
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, Salt Lake City, UT 84132
| | - Edin Mustafic
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, Salt Lake City, UT 84132
| | - Peter Tvrdik
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville VA 22908
| | - David Križaj
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, Salt Lake City, UT 84132
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT 84132
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84132
- Department of Neurobiology & Anatomy, University of Utah, Salt Lake City, UT 84132
| |
Collapse
|
108
|
The Microbiota-Gut-Brain Axis and Alzheimer Disease. From Dysbiosis to Neurodegeneration: Focus on the Central Nervous System Glial Cells. J Clin Med 2021; 10:jcm10112358. [PMID: 34072107 PMCID: PMC8199461 DOI: 10.3390/jcm10112358] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
The microbiota-gut system can be thought of as a single unit that interacts with the brain via the "two-way" microbiota-gut-brain axis. Through this axis, a constant interplay mediated by the several products originating from the microbiota guarantees the physiological development and shaping of the gut and the brain. In the present review will be described the modalities through which the microbiota and gut control each other, and the main microbiota products conditioning both local and brain homeostasis. Much evidence has accumulated over the past decade in favor of a significant association between dysbiosis, neuroinflammation and neurodegeneration. Presently, the pathogenetic mechanisms triggered by molecules produced by the altered microbiota, also responsible for the onset and evolution of Alzheimer disease, will be described. Our attention will be focused on the role of astrocytes and microglia. Numerous studies have progressively demonstrated how these glial cells are important to ensure an adequate environment for neuronal activity in healthy conditions. Furthermore, it is becoming evident how both cell types can mediate the onset of neuroinflammation and lead to neurodegeneration when subjected to pathological stimuli. Based on this information, the role of the major microbiota products in shifting the activation profiles of astrocytes and microglia from a healthy to a diseased state will be discussed, focusing on Alzheimer disease pathogenesis.
Collapse
|
109
|
Antimicrobial responses of peripheral and central nervous system glia against Staphylococcus aureus. Sci Rep 2021; 11:10722. [PMID: 34021227 PMCID: PMC8140078 DOI: 10.1038/s41598-021-90252-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 05/04/2021] [Indexed: 11/23/2022] Open
Abstract
Staphylococcus aureus infections of the central nervous system are serious and can be fatal. S. aureus is commonly present in the nasal cavity, and after injury to the nasal epithelium it can rapidly invade the brain via the olfactory nerve. The trigeminal nerve constitutes another potential route of brain infection. The glia of these nerves, olfactory ensheathing cells (OECs) and trigeminal nerve Schwann cells (TgSCs), as well as astrocytes populating the glia limitans layer, can phagocytose bacteria. Whilst some glial responses to S. aureus have been studied, the specific responses of different glial types are unknown. Here, we compared how primary mouse OECs, TgSCs, astrocytes and microglia responded to S. aureus. All glial types internalized the bacteria within phagolysosomes, and S. aureus-conjugated BioParticles could be tracked with subtle but significant differences in time-course of phagocytosis between glial types. Live bacteria could be isolated from all glia after 24 h in culture, and microglia, OECs and TgSCs exhibited better protection against intracellular S. aureus survival than astrocytes. All glial types responded to the bacteria by cytokine secretion. Overall, OECs secreted the lowest level of cytokines, suggesting that these cells, despite showing strong capacity for phagocytosis, have immunomodulatory functions that can be relevant for neural repair.
Collapse
|
110
|
Mouse primary microglia respond differently to LPS and poly(I:C) in vitro. Sci Rep 2021; 11:10447. [PMID: 34001933 PMCID: PMC8129154 DOI: 10.1038/s41598-021-89777-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 04/30/2021] [Indexed: 12/16/2022] Open
Abstract
Microglia, CNS resident innate immune cells, respond strongly to activation of TLR3 and TLR4, which recognize viral dsRNA poly(I:C) and bacterial endotoxin LPS, respectively. However, few studies have thoroughly and parallelly compared functional phenotypes and downstream mechanisms between LPS- and poly(I:C)-exposed primary microglia. Here, we investigated the responses of mouse primary microglia upon LPS and poly(I:C) stimulation by detecting various phenotypes ranging from morphology, proliferation, secretion, chemotaxis, to phagocytosis. Furthermore, we explored their sequential gene expression and the downstream signal cascades. Interestingly, we found that the microglial activation pattern induced by LPS was distinguished from that induced by poly(I:C). Regarding microglial morphology, LPS caused an ameboid-like shape while poly(I:C) induced a bushy shape. Microglial proliferation was also facilitated by LPS but not by poly(I:C). In addition, LPS and poly(I:C) modulated microglial chemotaxis and phagocytosis differently. Furthermore, genome-wide analysis provided gene-level support to these functional differences, which may be associated with NF-κb and type I interferon pathways. Last, LPS- and poly(I:C)-activated microglia mediated neurotoxicity in a co-culture system. This study extends our understanding of TLR roles in microglia and provides insights into selecting proper inflammatory microglial models, which may facilitate identification of new targets for therapeutic application.
Collapse
|
111
|
Liu J, Liu L, Wang X, Jiang R, Bai Q, Wang G. Microglia: A Double-Edged Sword in Intracerebral Hemorrhage From Basic Mechanisms to Clinical Research. Front Immunol 2021; 12:675660. [PMID: 34025674 PMCID: PMC8135095 DOI: 10.3389/fimmu.2021.675660] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS). It is well established that microglia are activated and polarized to acquire different inflammatory phenotypes, either pro-inflammatory or anti-inflammatory phenotypes, which act as a critical component in the neuroinflammation following intracerebral hemorrhage (ICH). Microglia produce pro-inflammatory mediators at the early stages after ICH onset, anti-inflammatory microglia with neuroprotective effects appear to be suppressed. Previous research found that driving microglia towards an anti-inflammatory phenotype could restrict inflammation and engulf cellular debris. The principal objective of this review is to analyze the phenotypes and dynamic profiles of microglia as well as their shift in functional response following ICH. The results may further the understanding of the body's self-regulatory functions involving microglia following ICH. On this basis, suggestions for future clinical development and research are provided.
Collapse
Affiliation(s)
- Jiachen Liu
- Xiangya Medical College of Central South University, Changsha, China
| | - Lirong Liu
- Department of Neurology, Shanxi Medical University, Taiyuan, China
| | - Xiaoyu Wang
- Xiangya Medical College of Central South University, Changsha, China
| | - Rundong Jiang
- Xiangya Medical College of Central South University, Changsha, China
| | - Qinqin Bai
- Department of Neurology, Shanxi Medical University, Taiyuan, China
| | - Gaiqing Wang
- Department of Neurology, Sanya Central Hospital (Hainan Third People's Hospital), Sanya, China
| |
Collapse
|
112
|
Radiation Triggers a Dynamic Sequence of Transient Microglial Alterations in Juvenile Brain. Cell Rep 2021; 31:107699. [PMID: 32492415 DOI: 10.1016/j.celrep.2020.107699] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 10/08/2019] [Accepted: 05/06/2020] [Indexed: 11/21/2022] Open
Abstract
Cranial irradiation (IR), an effective tool to treat malignant brain tumors, triggers a chronic pro-inflammatory microglial response, at least in the adult brain. Using single-cell and bulk RNA sequencing, combined with histology, we show that the microglial response in the juvenile mouse hippocampus is rapid but returns toward normal within 1 week. The response is characterized by a series of temporally distinct homeostasis-, sensome-, and inflammation-related molecular signatures. We find that a single microglial cell simultaneously upregulates transcripts associated with pro- and anti-inflammatory microglial phenotypes. Finally, we show that juvenile and adult irradiated microglia are already transcriptionally distinct in the early phase after IR. Our results indicate that microglia are involved in the initial stages but may not be responsible for driving long-term inflammation in the juvenile brain.
Collapse
|
113
|
Maurya SK, Bhattacharya N, Mishra S, Bhattacharya A, Banerjee P, Senapati S, Mishra R. Microglia Specific Drug Targeting Using Natural Products for the Regulation of Redox Imbalance in Neurodegeneration. Front Pharmacol 2021; 12:654489. [PMID: 33927630 PMCID: PMC8076853 DOI: 10.3389/fphar.2021.654489] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Microglia, a type of innate immune cell of the brain, regulates neurogenesis, immunological surveillance, redox imbalance, cognitive and behavioral changes under normal and pathological conditions like Alzheimer's, Parkinson's, Multiple sclerosis and traumatic brain injury. Microglia produces a wide variety of cytokines to maintain homeostasis. It also participates in synaptic pruning and regulation of neurons overproduction by phagocytosis of neural precursor cells. The phenotypes of microglia are regulated by the local microenvironment of neurons and astrocytes via interaction with both soluble and membrane-bound mediators. In case of neuron degeneration as observed in acute or chronic neurodegenerative diseases, microglia gets released from the inhibitory effect of neurons and astrocytes, showing activated phenotype either of its dual function. Microglia shows neuroprotective effect by secreting growths factors to heal neurons and clears cell debris through phagocytosis in case of a moderate stimulus. But the same microglia starts releasing pro-inflammatory cytokines like TNF-α, IFN-γ, reactive oxygen species (ROS), and nitric oxide (NO), increasing neuroinflammation and redox imbalance in the brain under chronic signals. Therefore, pharmacological targeting of microglia would be a promising strategy in the regulation of neuroinflammation, redox imbalance and oxidative stress in neurodegenerative diseases. Some studies present potentials of natural products like curcumin, resveratrol, cannabidiol, ginsenosides, flavonoids and sulforaphane to suppress activation of microglia. These natural products have also been proposed as effective therapeutics to regulate the progression of neurodegenerative diseases. The present review article intends to explain the molecular mechanisms and functions of microglia and molecular dynamics of microglia specific genes and proteins like Iba1 and Tmem119 in neurodegeneration. The possible interventions by curcumin, resveratrol, cannabidiol, ginsenosides, flavonoids and sulforaphane on microglia specific protein Iba1 suggest possibility of natural products mediated regulation of microglia phenotypes and its functions to control redox imbalance and neuroinflammation in management of Alzheimer's, Parkinson's and Multiple Sclerosis for microglia-mediated therapeutics.
Collapse
Affiliation(s)
| | - Neetu Bhattacharya
- Department of Zoology, Dyal Singh College, University of Delhi, Delhi, India
| | - Suman Mishra
- Department of Molecular Medicine and Biotechnology, SGPGI, Lucknow, India
| | - Amit Bhattacharya
- Department of Zoology, Ramjas College, University of Delhi, Delhi, India
| | - Pratibha Banerjee
- Immunogenomics Laboratory, Department of Human Genetics & Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Sabyasachi Senapati
- Immunogenomics Laboratory, Department of Human Genetics & Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Rajnikant Mishra
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
114
|
Adefegha SA, Assmann CE, Schetinger MRC, de Andrade CM, Emanuelli T. Moringa oleifera modulates cholinergic and purinergic enzymes activity in BV-2 microglial cells. Metab Brain Dis 2021; 36:627-638. [PMID: 33394288 DOI: 10.1007/s11011-020-00659-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/17/2020] [Indexed: 01/15/2023]
Abstract
Microglia are immune cells that are resident in central nervous system. Activation of microglial cells are detrimental to the survival of neurons. Thus, prevention of microglia activation and/or protection against microglia activation could be potential therapeutic strategy towards the management of inflammation-mediated neurodegenerative diseases. Moringa oleifera is widely consumed as food and used in folklore medicine for treating several diseases. This study was convened to investigate the effect of aqueous extract of Moringa oleifera on cell viability, cholinergic and purinergic enzymes in BV-2 microglial cultured cell. Aqueous extract of Moringa oleifera was prepared, lyophilized and reconstituted in 0.5% dimethylsulphoxide (DMSO). Cells were treated with Moringa oleifera extracts (0.1-100 μg/mL) and assessed for cell viability and nitric oxide production. Furthermore, the effect of Moringa oleifera on enzymes of cholinergic (acetylcholinesterase) and purinergic (nucleoside triphosphate diphosphohydrolase; NTPDase, 5' nucleotidase and adenosine deaminase; ADA) systems in BV-2 microglial cells were determined. Incubation of BV-2 microglia cell with M. oleifera extract maintained cell viability, modulated cholinergic and purinergic enzymes activity. The phenolic compounds found in M. oleifera extracts, include chlorogenic acid, rutin; quercetin pentoside, kaempferol derivative and quercetin derivative. Thus, this study suggest that the potential therapeutic effect of the phenolic compounds found in M. oleifera may have been responsible for the maintenance of cell viability in BV-2 microglia cells and modulation of cholinergic as well as purinergic enzymes activity.
Collapse
Affiliation(s)
- Stephen Adeniyi Adefegha
- Functional Foods, Nutraceuticals and Phytomedicine Laboratory, Department of Biochemistry, Federal University of Technology, Akure, Nigeria.
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Center of Natural and Exacts Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil.
- Graduate Program in Veterinary Medicine, Department of Small Animals, Center for Rural Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil.
- Department of Food Science and Technology, Center for Rural Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil.
| | - Charles Elias Assmann
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Center of Natural and Exacts Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Maria Rosa Chitolina Schetinger
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Center of Natural and Exacts Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Cinthia Melazzo de Andrade
- Graduate Program in Veterinary Medicine, Department of Small Animals, Center for Rural Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Tatiana Emanuelli
- Department of Food Science and Technology, Center for Rural Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| |
Collapse
|
115
|
Lindsay A, Hickman D, Srinivasan M. A nuclear factor-kappa B inhibiting peptide suppresses innate immune receptors and gliosis in a transgenic mouse model of Alzheimer's disease. Biomed Pharmacother 2021; 138:111405. [PMID: 33756153 DOI: 10.1016/j.biopha.2021.111405] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 12/17/2022] Open
Abstract
A disproportionate increase in activated nuclear factor-kappa B (NF-κB) has been shown to drive the Aβ deposition, neuroinflammation and neurodegeneration in Alzheimer's disease (AD). Hence, selective targeting of activated p65 represents an attractive therapeutic approach for AD. Glucocorticoid induced leucine zipper (GILZ) is a NF-κB interactant that binds and sequesters the activated p65 in the cytoplasm. The p65 binding domain of GILZ adopts a polyproline type II helical conformation, a motif that acts as an adaptable glove in the interface with the binding partner and constitutes an excellent template for drug design. Previously, peptide analogs of the p65 binding domain of GILZ, referred to as GA have been shown to suppress pathology in the lipopolysaccharide induced model of neuroinflammation. In this study, we investigated the CNS delivery of labeled GA administered intraperitoneally in adult mice for a period of upto 24 h. Further, we evaluated the suppressive potential of GA in 5xFAD mice, an aggressive model with five genetic mutations closely associated with human AD. Groups of 5xFAD mice administered GA or control peptide intraperitoneally on alternate days for six weeks were evaluated for Aβ deposition, microglia, inflammation and innate immune responses by immunohistochemistry and real time polymerase reaction. GA was observed in proximity with NeuN positive neurons suggesting that the compound crossed the blood brain barrier to reach the brain parenchyma. Further, GA treatment decreased Aβ load, reduced Iba1 + microglia and glial fibrillary acidic protein (GFAP)+ astrocytes, inhibited inflammatory cytokines and suppressed toll like receptor (TLR-2, TLR-4) expressions in 5xFAD mice.
Collapse
Affiliation(s)
- Alison Lindsay
- Department of Oral Pathology, Medicine and Radiology, Indiana University School of Dentistry, United States
| | - Deborah Hickman
- Laboratory of Animal Care and Research, Indiana University School of Medicine, Indiana University-Purdue University Indianapolis, United States
| | - Mythily Srinivasan
- Department of Oral Pathology, Medicine and Radiology, Indiana University School of Dentistry, United States; Provaidya LLC, Indianapolis, IN, United States.
| |
Collapse
|
116
|
Lyu J, Xie D, Bhatia TN, Leak RK, Hu X, Jiang X. Microglial/Macrophage polarization and function in brain injury and repair after stroke. CNS Neurosci Ther 2021; 27:515-527. [PMID: 33650313 PMCID: PMC8025652 DOI: 10.1111/cns.13620] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Stroke is a leading cause of disability and mortality, with limited treatment options. After stroke injury, microglia and CNS‐resident macrophages are rapidly activated and regulate neuropathological processes to steer the course of functional recovery. To accelerate this recovery, microglia can engulf dying cells and clear irreparably‐damaged tissues, thereby creating a microenvironment that is more suitable for the formation of new neural circuitry. In addition, monocyte‐derived macrophages cross the compromised blood‐brain barrier to infiltrate the injured brain. The specific functions of myeloid lineage cells in brain injury and repair are diverse and dependent on phenotypic polarization statuses. However, it remains to be determined to what degree the CNS‐invading macrophages occupy different functional niches from CNS‐resident microglia. In this review, we describe the physiological characteristics and functions of microglia in the developing and adult brain. We also review (a) the activation and phenotypic polarization of microglia and macrophages after stroke, (b) molecular mechanisms that control polarization status, and (c) the contribution of microglia to brain pathology versus repair. Finally, we summarize current breakthroughs in therapeutic strategies that calibrate microglia/macrophage responses after stroke. The present review summarizes recent advances in microglial research in relation to stroke with emphases on microglial/macrophage phenotypic polarization and function in brain injury and repair. It also reviews the physiological characteristics and functions of microglia in the developing and adult brain, and describes current breakthroughs in therapeutic strategies that calibrate microglia/macrophage responses after stroke.
![]()
Collapse
Affiliation(s)
- Junxuan Lyu
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Di Xie
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tarun N Bhatia
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Rehana K Leak
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Xiaoming Hu
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh, Pittsburgh, PA, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Xiaoyan Jiang
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh, Pittsburgh, PA, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| |
Collapse
|
117
|
Cyrino LAR, Delwing-de Lima D, Ullmann OM, Maia TP. Concepts of Neuroinflammation and Their Relationship With Impaired Mitochondrial Functions in Bipolar Disorder. Front Behav Neurosci 2021; 15:609487. [PMID: 33732117 PMCID: PMC7959852 DOI: 10.3389/fnbeh.2021.609487] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/18/2021] [Indexed: 12/24/2022] Open
Abstract
Bipolar disorder (BD) is a chronic psychiatric disease, characterized by frequent behavioral episodes of depression and mania, and neurologically by dysregulated neurotransmission, neuroplasticity, growth factor signaling, and metabolism, as well as oxidative stress, and neuronal apoptosis, contributing to chronic neuroinflammation. These abnormalities result from complex interactions between multiple susceptibility genes and environmental factors such as stress. The neurocellular abnormalities of BD can result in gross morphological changes, such as reduced prefrontal and hippocampal volume, and circuit reorganization resulting in cognitive and emotional deficits. The term "neuroprogression" is used to denote the progressive changes from early to late stages, as BD severity and loss of treatment response correlate with the number of past episodes. In addition to circuit and cellular abnormalities, BD is associated with dysfunctional mitochondria, leading to severe metabolic disruption in high energy-demanding neurons and glia. Indeed, mitochondrial dysfunction involving electron transport chain (ETC) disruption is considered the primary cause of chronic oxidative stress in BD. The ensuing damage to membrane lipids, proteins, and DNA further perpetuates oxidative stress and neuroinflammation, creating a perpetuating pathogenic cycle. A deeper understanding of BD pathophysiology and identification of associated biomarkers of neuroinflammation are needed to facilitate early diagnosis and treatment of this debilitating disorder.
Collapse
Affiliation(s)
- Luiz Arthur Rangel Cyrino
- Programa de Pós-Graduação em Saúde e Meio Ambiente, Laboratório de Práticas Farmacêuticas of Department of Pharmacy, University of Joinville Region—UNIVILLE, Joinville, Brazil
- Department of Psychology, University of Joinville—UNIVILLE, Joinville, Brazil
- Department of Pharmacy, University of Joinville—UNIVILLE, Joinville, Brazil
| | - Daniela Delwing-de Lima
- Programa de Pós-Graduação em Saúde e Meio Ambiente, Laboratório de Práticas Farmacêuticas of Department of Pharmacy, University of Joinville Region—UNIVILLE, Joinville, Brazil
- Department of Pharmacy, University of Joinville—UNIVILLE, Joinville, Brazil
- Department of Medicine, University of Joinville—UNIVILLE, Joinville, Brazil
| | | | | |
Collapse
|
118
|
The Neurovascular Unit Dysfunction in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22042022. [PMID: 33670754 PMCID: PMC7922832 DOI: 10.3390/ijms22042022] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/06/2021] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disease worldwide. Histopathologically, AD presents with two hallmarks: neurofibrillary tangles (NFTs), and aggregates of amyloid β peptide (Aβ) both in the brain parenchyma as neuritic plaques, and around blood vessels as cerebral amyloid angiopathy (CAA). According to the vascular hypothesis of AD, vascular risk factors can result in dysregulation of the neurovascular unit (NVU) and hypoxia. Hypoxia may reduce Aβ clearance from the brain and increase its production, leading to both parenchymal and vascular accumulation of Aβ. An increase in Aβ amplifies neuronal dysfunction, NFT formation, and accelerates neurodegeneration, resulting in dementia. In recent decades, therapeutic approaches have attempted to decrease the levels of abnormal Aβ or tau levels in the AD brain. However, several of these approaches have either been associated with an inappropriate immune response triggering inflammation, or have failed to improve cognition. Here, we review the pathogenesis and potential therapeutic targets associated with dysfunction of the NVU in AD.
Collapse
|
119
|
da Silva Chagas L, Sandre PC, de Velasco PC, Marcondes H, Ribeiro E Ribeiro NCA, Barreto AL, Alves Mauro LB, Ferreira JH, Serfaty CA. Neuroinflammation and Brain Development: Possible Risk Factors in COVID-19-Infected Children. Neuroimmunomodulation 2021; 28:22-28. [PMID: 33530091 PMCID: PMC7900470 DOI: 10.1159/000512815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/01/2020] [Indexed: 11/19/2022] Open
Abstract
COVID-19, a disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) betacoronavirus, affects children in a different way than it does in adults, with milder symptoms. However, several cases of neurological symptoms with neuroinflammatory syndromes, such as the multisystem inflammatory syndrome (MIS-C), following mild cases, have been reported. As with other viral infections, such as rubella, influenza, and cytomegalovirus, SARS-CoV-2 induces a surge of proinflammatory cytokines that affect microglial function, which can be harmful to brain development. Along with the viral induction of neuroinflammation, other noninfectious conditions may interact to produce additional inflammation, such as the nutritional imbalance of fatty acids and polyunsaturated fatty acids and alcohol consumption during pregnancy. Additionally, transient thyrotoxicosis induced by SARS-CoV-2 with secondary autoimmune hypothyroidism has been reported, which could go undetected during pregnancy. Together, those factors may pose additional risk factors for SARS-CoV-2 infection impacting mechanisms of neural development such as synaptic pruning and neural circuitry formation. The present review discusses those conditions in the perspective of the understanding of risk factors that should be considered and the possible emergence of neurodevelopmental disorders in COVID-19-infected children.
Collapse
Affiliation(s)
- Luana da Silva Chagas
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Poliana Capucho Sandre
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Patricia Coelho de Velasco
- Department of Applied Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Henrique Marcondes
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | | | - Aline Loureiro Barreto
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Luiza Beatriz Alves Mauro
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Julia Huber Ferreira
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Claudio A Serfaty
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil,
- National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil,
| |
Collapse
|
120
|
Farré-Alins V, Narros-Fernández P, Palomino-Antolín A, Decouty-Pérez C, Lopez-Rodriguez AB, Parada E, Muñoz-Montero A, Gómez-Rangel V, López-Muñoz F, Ramos E, González-Rodríguez Á, Gandía L, Romero A, Egea J. Melatonin Reduces NLRP3 Inflammasome Activation by Increasing α7 nAChR-Mediated Autophagic Flux. Antioxidants (Basel) 2020; 9:antiox9121299. [PMID: 33353046 PMCID: PMC7767051 DOI: 10.3390/antiox9121299] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 12/15/2020] [Indexed: 12/16/2022] Open
Abstract
Microglia controls the immune system response in the brain. Specifically, the activation and dysregulation of the NLRP3 inflammasome is responsible for the initiation of the inflammatory process through IL-1β and IL-18 release. In this work, we have focused on studying the effect of melatonin on the regulation of the NLRP3 inflammasome through α7 nicotinic receptor (nAChR) and its relationship with autophagy. For this purpose, we have used pharmacological and genetic approaches in lipopolysaccharide (LPS)-induced inflammation models in both in vitro and in vivo models. In the BV2 cell line, LPS inhibited autophagy, which increased NLRP3 protein levels. However, melatonin promoted an increase in the autophagic flux. Treatment of glial cultures from wild-type (WT) mice with LPS followed by extracellular adenosine triphosphate (ATP) produced the release of IL-1β, which was reversed by melatonin pretreatment. In cultures from α7 nAChR knock-out (KO) mice, melatonin did not reduce IL-1β release. Furthermore, melatonin decreased the expression of inflammasome components and reactive oxygen species (ROS) induced by LPS; co-incubation of melatonin with α-bungarotoxin (α-bgt) or luzindole abolished the anti-inflammatory and antioxidant effects. In vivo, melatonin reverted LPS-induced cognitive decline, reduced NLRP3 levels and promoted autophagic flux in the hippocampi of WT mice, whereas in α7 nAChR KO mice melatonin effect was not observed. These results suggest that melatonin may modulate the complex interplay between α7 nAChR and autophagy signaling.
Collapse
Affiliation(s)
- Víctor Farré-Alins
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Paloma Narros-Fernández
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Alejandra Palomino-Antolín
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Céline Decouty-Pérez
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Ana Belen Lopez-Rodriguez
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Esther Parada
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Alicia Muñoz-Montero
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Vanessa Gómez-Rangel
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Francisco López-Muñoz
- Faculty of Health Sciences, University Camilo José Cela, Villanueva de la Cañada, 28692 Madrid, Spain;
- Neuropsychopharmacology Unit, Hospital 12 de Octubre Research Institute (i+12), 28041 Madrid, Spain
- Portucalense Institute of Neuropsychology and Cognitive and Behavioural Neurosciences (INPP), Portucalense University, 4200-072 Porto, Portugal
- Thematic Network for Cooperative Health Research (RETICS), Addictive Disorders Network, Health Institute Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - Eva Ramos
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (E.R.); (A.R.)
| | - Águeda González-Rodríguez
- Research Unit, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD, ISCIII), 28029 Madrid, Spain
| | - Luis Gandía
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (E.R.); (A.R.)
| | - Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
- Correspondence: ; Tel.: +34-915574402
| |
Collapse
|
121
|
Glasnović A, O'Mara N, Kovačić N, Grčević D, Gajović S. RANK/RANKL/OPG Signaling in the Brain: A Systematic Review of the Literature. Front Neurol 2020; 11:590480. [PMID: 33329338 PMCID: PMC7710989 DOI: 10.3389/fneur.2020.590480] [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: 08/01/2020] [Accepted: 10/22/2020] [Indexed: 12/16/2022] Open
Abstract
Together with its dominant immunological and bone remodeling involvement, RRO axis, comprising of receptor activator of nuclear factor-κB (RANK), RANK ligand (RANKL), and osteoprotegerin (OPG) signaling, is as well-implicated in CNS functioning and corresponding pathologies. The CNS aspects of RANKL/RANK/OPG (RRO) axis were systematically reviewed. With search 10 databases, and 7 additional resources from first article publication to July 2019, resulted in total 2,222 hits, from which 33 relevant articles were selected. The elements of RRO axis in CNS include cells involved in neuroinflammation, predominantly in microglia, but as well in resident macrophages and inflammatory cells migrating across the blood-brain barrier. The expression in neurons and oligodendrocytes is mainly confined to processes of differentiation and cell death. RRO axis tunes the neuroinflammatory response, depending on the molecular, cellular and pathological context. RANK/RANKL signaling is neuroprotective in TLR-mediated inflammation, while OPG seems detrimental in stroke, but beneficial in multiple sclerosis. The levels of RRO axis elements can serve as biomarkers in the blood and cerebrospinal fluid. They act as neuroprotectant after brain damage even being implicated in body weight- and thermo-regulation. As derivatives of RRO axis already exist as therapeutic agents in bone remodeling, it would be intriquing to see if these or new RRO-based pharmaceuticals would appear effective in CNS therapies.
Collapse
Affiliation(s)
- Anton Glasnović
- Department of Histology and Embryology, Zagreb University School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, Zagreb University School of Medicine, Zagreb, Croatia
| | - Niall O'Mara
- Department of Medicine, Cork University Hospital, Cork, Ireland
| | - Nataša Kovačić
- Croatian Institute for Brain Research, Zagreb University School of Medicine, Zagreb, Croatia.,Department of Anatomy, Zagreb University School of Medicine, Zagreb, Croatia
| | - Danka Grčević
- Croatian Institute for Brain Research, Zagreb University School of Medicine, Zagreb, Croatia.,Department of Physiology and Immunology, Zagreb University School of Medicine, Zagreb, Croatia
| | - Srećko Gajović
- Department of Histology and Embryology, Zagreb University School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, Zagreb University School of Medicine, Zagreb, Croatia
| |
Collapse
|
122
|
Borsom EM, Lee K, Cope EK. Do the Bugs in Your Gut Eat Your Memories? Relationship between Gut Microbiota and Alzheimer's Disease. Brain Sci 2020; 10:E814. [PMID: 33153085 PMCID: PMC7693835 DOI: 10.3390/brainsci10110814] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 02/06/2023] Open
Abstract
The human microbiota is composed of trillions of microbial cells inhabiting the oral cavity, skin, gastrointestinal (GI) tract, airways, and reproductive organs. The gut microbiota is composed of dynamic communities of microorganisms that communicate bidirectionally with the brain via cytokines, neurotransmitters, hormones, and secondary metabolites, known as the gut microbiota-brain axis. The gut microbiota-brain axis is suspected to be involved in the development of neurological diseases, including Alzheimer's disease (AD), Parkinson's disease, and Autism Spectrum Disorder. AD is an irreversible, neurodegenerative disease of the central nervous system (CNS), characterized by amyloid-β plaques, neurofibrillary tangles, and neuroinflammation. Microglia and astrocytes, the resident immune cells of the CNS, play an integral role in AD development, as neuroinflammation is a driving factor of disease severity. The gut microbiota-brain axis is a novel target for Alzheimer's disease therapeutics to modulate critical neuroimmune and metabolic pathways. Potential therapeutics include probiotics, prebiotics, fecal microbiota transplantation, and dietary intervention. This review summarizes our current understanding of the role of the gut microbiota-brain axis and neuroinflammation in the onset and development of Alzheimer's disease, limitations of current research, and potential for gut microbiota-brain axis targeted therapies.
Collapse
|
123
|
Ramirez JM, Karlen-Amarante M, Wang JDJ, Bush NE, Carroll MS, Weese-Mayer DE, Huff A. The Pathophysiology of Rett Syndrome With a Focus on Breathing Dysfunctions. Physiology (Bethesda) 2020; 35:375-390. [PMID: 33052774 PMCID: PMC7864239 DOI: 10.1152/physiol.00008.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/21/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
Rett syndrome (RTT), an X-chromosome-linked neurological disorder, is characterized by serious pathophysiology, including breathing and feeding dysfunctions, and alteration of cardiorespiratory coupling, a consequence of multiple interrelated disturbances in the genetic and homeostatic regulation of central and peripheral neuronal networks, redox state, and control of inflammation. Characteristic breath-holds, obstructive sleep apnea, and aerophagia result in intermittent hypoxia, which, combined with mitochondrial dysfunction, causes oxidative stress-an important driver of the clinical presentation of RTT.
Collapse
Affiliation(s)
- Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
- Departments of Neurological Surgery and Pediatrics, University of Washington School of Medicine, Seattle, Washington
| | - Marlusa Karlen-Amarante
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
- Department of Physiology and Pathology, School of Dentistry of Araraquara, São Paulo State University (UNESP), Araraquara, Brazil
| | - Jia-Der Ju Wang
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
| | - Nicholas E Bush
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
| | - Michael S Carroll
- Data Analytics and Reporting, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Debra E Weese-Mayer
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Autonomic Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Alyssa Huff
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
124
|
Morales-Ropero JM, Arroyo-Urea S, Neubrand VE, Martín-Oliva D, Marín-Teva JL, Cuadros MA, Vangheluwe P, Navascués J, Mata AM, Sepúlveda MR. The endoplasmic reticulum Ca 2+ -ATPase SERCA2b is upregulated in activated microglia and its inhibition causes opposite effects on migration and phagocytosis. Glia 2020; 69:842-857. [PMID: 33105046 DOI: 10.1002/glia.23931] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/18/2022]
Abstract
Activation of microglia is an early immune response to damage in the brain. Although a key role for Ca2+ as trigger of microglial activation has been considered, little is known about the molecular scenario for regulating Ca2+ homeostasis in these cells. Taking into account the importance of the endoplasmic reticulum as a cellular Ca2+ store, the sarco(endo)plasmic reticulum Ca2+ -ATPase (SERCA2b) is an interesting target to modulate intracellular Ca2+ dynamics. We found upregulation of SERCA2b in activated microglia of human brain with Alzheimer's disease and we further studied the participation of SERCA2b in microglial functions by using the BV2 murine microglial cell line and primary microglia isolated from mouse brain. To trigger microglia activation, we used the bacterial lipopolysaccharide (LPS), which is known to induce an increase of cytosolic Ca2+ . Our results showed an upregulated expression of SERCA2b in LPS-induced activated microglia likely associated to an attempt to restore the increased cytosolic Ca2+ concentration. We analyzed SERCA2b contribution in microglial migration by using the specific SERCA inhibitor thapsigargin in scratch assays. Microglial migration was strongly stimulated with thapsigargin, even more than with LPS-induction, but delayed in time. However, phagocytic capacity of microglia was blocked in the presence of the SERCA inhibitor, indicating the importance of a tight control of cytosolic Ca2+ in these processes. All together, these results provide for the first time compelling evidence for SERCA2b as a major player regulating microglial functions, affecting migration and phagocytosis in an opposite manner.
Collapse
Affiliation(s)
- Juan M Morales-Ropero
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Sandra Arroyo-Urea
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Veronika E Neubrand
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - David Martín-Oliva
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - José L Marín-Teva
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Miguel A Cuadros
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Julio Navascués
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Ana M Mata
- Department of Biochemistry and Molecular Biology and Genetics, Faculty of Sciences, University of Extremadura, Badajoz, Spain
| | - M Rosario Sepúlveda
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| |
Collapse
|
125
|
Frühauf M, Zeitschel U, Höfling C, Ullm F, Rabiger FV, Alber G, Pompe T, Müller U, Roßner S. Construction of a 3D brain extracellular matrix model to study the interaction between microglia and T cells in co-culture. Eur J Neurosci 2020; 53:4034-4050. [PMID: 32954591 DOI: 10.1111/ejn.14978] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 12/24/2022]
Abstract
Neurodegenerative disorders are characterised by the activation of brain-resident microglia cells and by the infiltration of peripheral T cells. However, their interplay in disease has not been clarified yet. It is difficult to investigate complex cellular dynamics in living animals, and simple two-dimensional (2D) cell culture models do not resemble the soft 3D structure of brain tissue. Therefore, we developed a biomimetic 3D in vitro culture system for co-cultivation of microglia and T cells. As the activation and/or migration of immune cells in the brain might be affected by components of the extracellular matrix, defined 3D fibrillar collagen I-based matrices were constructed and modified with hyaluronan and/or chondroitin sulphate, resembling aspects of brain extracellular matrix. Murine microglia and spleen-derived T cells were cultured alone or in co-culture on the constructed matrices. Microglia exhibited in vivo-like morphology and T cells showed enhanced survival when co-cultured with microglia or to a minor degree in the presence of glia-conditioned medium. The open and porous fibrillar structure of the matrix allowed for cell invasion and direct cell-cell interaction, with stronger invasion of T cells. Both cell types showed no dependence on the matrix modifications. Microglia could be activated on the matrices by lipopolysaccharide resulting in interleukin-6 and tumour necrosis factor-α secretion. The findings herein indicate that biomimetic 3D matrices allow for co-cultivation and activation of primary microglia and T cells and provide useful tools to study their interaction in vitro.
Collapse
Affiliation(s)
- Marie Frühauf
- Paul Flechsig Institute for Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany.,Institute of Immunology/Molecular Pathogenesis, Centre for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany.,Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, Leipzig, Germany
| | - Ulrike Zeitschel
- Paul Flechsig Institute for Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Corinna Höfling
- Paul Flechsig Institute for Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Franziska Ullm
- Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, Leipzig, Germany
| | - Friederike V Rabiger
- Institute of Immunology/Molecular Pathogenesis, Centre for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Gottfried Alber
- Institute of Immunology/Molecular Pathogenesis, Centre for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Tilo Pompe
- Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, Leipzig, Germany
| | - Uwe Müller
- Institute of Immunology/Molecular Pathogenesis, Centre for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| |
Collapse
|
126
|
Gilchrist SE, Goudarzi S, Hafizi S. Gas6 Inhibits Toll-Like Receptor-Mediated Inflammatory Pathways in Mouse Microglia via Axl and Mer. Front Cell Neurosci 2020; 14:576650. [PMID: 33192322 PMCID: PMC7584110 DOI: 10.3389/fncel.2020.576650] [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: 06/26/2020] [Accepted: 09/08/2020] [Indexed: 12/30/2022] Open
Abstract
Background: Microglia are well known key regulators of neuroinflammation which feature in multiple neurodegenerative disorders. These cells survey the CNS and, under inflammatory conditions, become "activated" through stimulation of toll-like receptors (TLRs), resulting in changes in morphology and production and release of cytokines. In the present study, we examined the roles of the related TAM receptors, Mer and Axl, and of their ligand, Gas6, in the regulation of microglial pro-inflammatory TNF-α production and microglial morphology. Methods: Primary cultures of murine microglia of wild-type (WT), Mer-/- and Axl-/- backgrounds were stimulated by the TLR4 agonist, lipopolysaccharide (LPS) with or without pre-treatment with Gas6. Gene expression of TNF-α, Mer, and Axl was examined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and enzyme-linked immunosorbent assay (ELISA) was used to measure TNF-α release from microglia. Immunofluorescence staining of β-actin and the microglial marker Iba1 was performed to reveal microglial morphological changes, with cellular characteristics (area, perimeter, Feret's diameter, minimum Feret, roundness, and aspect ratio) being quantified using ImageJ software. Results: Under basal conditions, TNF-α gene expression was significantly lower in Axl-/- microglia compared to WT cells. However, all microglial cultures robustly responded to LPS stimulation with the upregulation of TNF-α expression to similar degrees. Furthermore, Mer receptor expression was less responsive to LPS stimulation when in Axl knockout cells. The presence of Gas6 consistently inhibited the LPS-induced upregulation of TNF-α in WT, Mer-/- and Axl-/- microglia. Moreover, Gas6 also inhibited LPS-induced changes in the microglial area, perimeter length, and cell roundness in wild-type cells. Conclusion: Gas6 can negatively regulate the microglial pro-inflammatory response to LPS as well as via stimulation of other TLRs, acting through either of the TAM receptors, Axl and Mer. This finding indicates an interaction between TLR and TAM receptor signaling pathways and reveals an anti-inflammatory role for the TAM ligand, Gas6, which could have therapeutic potential.
Collapse
Affiliation(s)
- Shannon E Gilchrist
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Salman Goudarzi
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Sassan Hafizi
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
127
|
Pinto B, Morelli G, Rastogi M, Savardi A, Fumagalli A, Petretto A, Bartolucci M, Varea E, Catelani T, Contestabile A, Perlini LE, Cancedda L. Rescuing Over-activated Microglia Restores Cognitive Performance in Juvenile Animals of the Dp(16) Mouse Model of Down Syndrome. Neuron 2020; 108:887-904.e12. [PMID: 33027640 PMCID: PMC7736620 DOI: 10.1016/j.neuron.2020.09.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/16/2020] [Accepted: 09/04/2020] [Indexed: 01/01/2023]
Abstract
Microglia are brain-resident immune cells and regulate mechanisms essential for cognitive functions. Down syndrome (DS), the most frequent cause of genetic intellectual disability, is caused by a supernumerary chromosome 21, containing also genes related to the immune system. In the hippocampus of the Dp(16) mouse model of DS and DS individuals, we found activated microglia, as assessed by their morphology; activation markers; and, for DS mice, electrophysiological profile. Accordingly, we found increased pro-inflammatory cytokine levels and altered interferon signaling in Dp(16) hippocampi. DS mice also showed decreased spine density and activity of hippocampal neurons and hippocampus-dependent cognitive behavioral deficits. Depletion of defective microglia or treatment with a commonly used anti-inflammatory drug rescued the neuronal spine and activity impairments and cognitive deficits in juvenile Dp(16) mice. Our results suggest an involvement of microglia in Dp(16)-mouse cognitive deficits and identify a new potential therapeutic approach for cognitive disabilities in DS individuals. DS mice display microglia alterations and cognitive impairment Depletion of microglia rescues cognitive impairment in DS mice Acetaminophen treatment rescues microglia and cognitive impairments in DS mice Brain samples of DS people recapitulate microglia alterations observed in DS mice
Collapse
Affiliation(s)
- Bruno Pinto
- BIO@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy; Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Giovanni Morelli
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Mohit Rastogi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Annalisa Savardi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Amos Fumagalli
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Emilio Varea
- Cellular Biology Department, University of Valencia, Valencia, Spain
| | - Tiziano Catelani
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Andrea Contestabile
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Laura E Perlini
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy; Dulbecco Telethon Institute, Rome, Italy.
| |
Collapse
|
128
|
Positron Emission Tomography in the Inflamed Cerebellum: Addressing Novel Targets among G Protein-Coupled Receptors and Immune Receptors. Pharmaceutics 2020; 12:pharmaceutics12100925. [PMID: 32998351 PMCID: PMC7601272 DOI: 10.3390/pharmaceutics12100925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 01/12/2023] Open
Abstract
Inflammatory processes preceding clinical manifestation of brain diseases are moving increasingly into the focus of positron emission tomographic (PET) investigations. A key role in inflammation and as a target of PET imaging efforts is attributed to microglia. Cerebellar microglia, with a predominant ameboid and activated subtype, is of special interest also regarding improved and changing knowledge on functional involvement of the cerebellum in mental activities in addition to its regulatory role in motor function. The present contribution considers small molecule ligands as potential PET tools for the visualization of several receptors recognized to be overexpressed in microglia and which can potentially serve as indicators of inflammatory processes in the cerebellum. The sphingosine 1 phosphate receptor 1 (S1P1), neuropeptide Y receptor 2 (NPY2) and purinoceptor Y12 (P2Y12) cannabinoid receptors and the chemokine receptor CX3CR1 as G-protein-coupled receptors and the ionotropic purinoceptor P2X7 provide structures with rather classical binding behavior, while the immune receptor for advanced glycation end products (RAGE) and the triggering receptor expressed on myeloid cells 2 (TREM2) might depend for instance on further accessory proteins. Improvement in differentiation between microglial functional subtypes in comparison to the presently used 18 kDa translocator protein ligands as well as of the knowledge on the role of polymorphisms are special challenges in such developments.
Collapse
|
129
|
Chidambaram H, Das R, Chinnathambi S. Interaction of Tau with the chemokine receptor, CX3CR1 and its effect on microglial activation, migration and proliferation. Cell Biosci 2020; 10:109. [PMID: 32944223 PMCID: PMC7493323 DOI: 10.1186/s13578-020-00474-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that leads to progressive loss of memory and dementia. The pathological hallmarks of AD include extracellular accumulation of amyloid-β peptides forming senile plaques and intracellular accumulation of Tau oligomers and filamentous species. Tau is a microtubule-binding protein that stabilizes tubulin to form microtubules under physiological condition. In AD/ pathological condition, Tau detaches from microtubules and aggregates to form oligomers of different sizes and filamentous species such as paired helical filaments. Microglia are the resident brain macrophages that are involved in the phagocytosis of microbes, cellular debris, misfolded and aggregated proteins. Chemokine receptor, CX3CR1 is mostly expressed on microglia and is involved in maintaining the microglia in a quiescent state by binding to its ligand, fractalkine (CX3CL1), which is expressed in neurons as both soluble or membrane-bound state. Hence, under physiological conditions, the CX3CR1/CX3CL1 axis plays a significant role in maintaining the central nervous system (CNS) homeostasis. Further, CX3CR1/CX3CL1 signalling is involved in the synthesis of anti-inflammatory cytokines and also has a significant role in cytoskeletal rearrangement, migration, apoptosis and proliferation. In AD brain, the expression level of fractalkine is reduced, and hence Tau competes to interact with its receptor, CX3CR1. In microglia, phagocytosis and internalization of extracellular Tau species occurs in the presence of a chemokine receptor, CX3CR1 which binds directly to Tau and promotes its internalization. In this review, the pathophysiological roles of CX3CR1/fractalkine signalling in microglia and neurons at different stages of Alzheimer's disease and the possible role of CX3CR1/Tau signalling has been widely discussed.
Collapse
Affiliation(s)
- Hariharakrishnan Chidambaram
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India
- Academy of Scientific and Innovative Research (AcSIR), 411008 Pune, India
| | - Rashmi Das
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India
- Academy of Scientific and Innovative Research (AcSIR), 411008 Pune, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India
- Academy of Scientific and Innovative Research (AcSIR), 411008 Pune, India
| |
Collapse
|
130
|
Avallone R, Lucchi C, Puja G, Codeluppi A, Filaferro M, Vitale G, Rustichelli C, Biagini G. BV-2 Microglial Cells Respond to Rotenone Toxic Insult by Modifying Pregnenolone, 5α-Dihydroprogesterone and Pregnanolone Levels. Cells 2020; 9:E2091. [PMID: 32933155 PMCID: PMC7563827 DOI: 10.3390/cells9092091] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 01/05/2023] Open
Abstract
Neuroinflammation, whose distinctive sign is the activation of microglia, is supposed to play a key role in the development and progression of neurodegenerative diseases. The aim of this investigation was to determine levels of neurosteroids produced by resting and injured BV-2 microglial cells. BV-2 cells were exposed to increasing concentrations of rotenone to progressively reduce their viability by increasing reactive oxygen species (ROS) production. BV-2 cell viability was significantly reduced 24, 48 and 72 h after rotenone (50-1000 nM) exposure. Concomitantly, rotenone (50-100 nM) determined a dose-independent augmentation of ROS production. Then, BV-2 cells were exposed to a single, threshold dose of rotenone (75 nM) to evaluate the overtime release of neurosteroids. In particular, pregnenolone, pregnenolone sulfate, progesterone, 5α-dihydroprogesterone (5α-DHP), allopregnanolone, and pregnanolone, were quantified in the culture medium by liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis. BV-2 cells synthesized all the investigated neurosteroids and, after exposure to rotenone, 5αDHP and pregnanolone production was remarkably increased. In conclusion, we found that BV-2 cells not only synthesize several neurosteroids, but further increase this production following oxidative damage. Pregnanolone and 5α-DHP may play a role in modifying the progression of neuroinflammation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Rossella Avallone
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Chiara Lucchi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.L.); (M.F.); (G.B.)
| | - Giulia Puja
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Alessandro Codeluppi
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Monica Filaferro
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.L.); (M.F.); (G.B.)
| | - Giovanni Vitale
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Cecilia Rustichelli
- Department of Life Sciences, Modena and Reggio Emilia University, 41125 Modena, Italy; (G.P.); (A.C.); (G.V.); (C.R.)
| | - Giuseppe Biagini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (C.L.); (M.F.); (G.B.)
| |
Collapse
|
131
|
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are, respectively, the most prevalent and fastest growing neurodegenerative diseases worldwide. The former is primarily characterized by memory loss and the latter by the motor symptoms of tremor and bradykinesia. Both AD and PD are progressive diseases that share several key underlying mitochondrial, inflammatory, and other metabolic pathologies. This review will detail how these pathologies intersect with ketone body metabolism and signaling, and how ketone bodies, particularly d-β-hydroxybutyrate (βHB), may serve as a potential adjunctive nutritional therapy for two of the world's most devastating conditions.
Collapse
|
132
|
Ambrose N, Rodriguez M, Waters KA, Machaalani R. Microglia in the human infant brain and factors that affect expression. Brain Behav Immun Health 2020; 7:100117. [PMID: 34589874 PMCID: PMC8474518 DOI: 10.1016/j.bbih.2020.100117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/21/2022] Open
Abstract
The present study reports on the microglial populations present in 34 regions of the human infant brain (1-11 months), and whether developmental parameters or extrinsic factors such as cigarette smoke exposure, prone sleeping and an upper respiratory tract infection (URTI) influence their expression. Further, we compare microglia populations amongst three sudden unexpected death in infancy (SUDI) sub-groups: explained SUDI (eSUDI, n = 7), sudden infant death syndrome (SIDS) I (n = 8) and SIDS II (n = 13). Ionised calcium binding adaptor molecule-1 (Iba1) was used to determine the morphology and area covered by microglia in a given brain region. Activation was explored using cluster-of-differentiation factor 68 (CD68) and human leukocyte antigen-DP,DQ,DR (HLA). We found regional heterogeneity in the area covered and activation status of microglia across the infant brain. The hippocampus, basal ganglia, white matter and dentate nucleus of the cerebellum showed larger areas of Iba1, while the brainstem had the smallest. Microglia in regions of the basal ganglia and cortex demonstrated positive correlations with infant developmental parameters, while in nuclei of the rostral medulla, negative correlations between microglia parameters were seen. URTI and cigarette smoke exposure were associated with a reduced microglial area in regions of the hippocampus and cortex (parietal and occipital), respectively. In the context of SIDS, a reduced microglial area was seen in SIDS II and fewer SIDS I infants demonstrated activated phenotypes in the hippocampus. Overall, we identify the distribution of microglia in the infant brain to be heterogenous, and influenced by intrinsic and extrinsic factors, and that the SIDS I group is a useful control group for future research into other infant CNS pathologies.
Collapse
Affiliation(s)
- Natalie Ambrose
- Discipline of Medicine, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
| | - Michael Rodriguez
- Department of Pathology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
| | - Karen A. Waters
- Discipline of Medicine, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
- Discipline of Child and Adolescent Health, Children’s Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
| | - Rita Machaalani
- Discipline of Medicine, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
- Discipline of Child and Adolescent Health, Children’s Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
| |
Collapse
|
133
|
Baker TL, Sun M, Semple BD, Tyebji S, Tonkin CJ, Mychasiuk R, Shultz SR. Catastrophic consequences: can the feline parasite Toxoplasma gondii prompt the purrfect neuroinflammatory storm following traumatic brain injury? J Neuroinflammation 2020; 17:222. [PMID: 32711529 PMCID: PMC7382044 DOI: 10.1186/s12974-020-01885-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/02/2020] [Indexed: 12/02/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality worldwide; however, treatment development is hindered by the heterogenous nature of TBI presentation and pathophysiology. In particular, the degree of neuroinflammation after TBI varies between individuals and may be modified by other factors such as infection. Toxoplasma gondii, a parasite that infects approximately one-third of the world’s population, has a tropism for brain tissue and can persist as a life-long infection. Importantly, there is notable overlap in the pathophysiology between TBI and T. gondii infection, including neuroinflammation. This paper will review current understandings of the clinical problems, pathophysiological mechanisms, and functional outcomes of TBI and T. gondii, before considering the potential synergy between the two conditions. In particular, the discussion will focus on neuroinflammatory processes such as microglial activation, inflammatory cytokines, and peripheral immune cell recruitment that occur during T. gondii infection and after TBI. We will present the notion that these overlapping pathologies in TBI individuals with a chronic T. gondii infection have the strong potential to exacerbate neuroinflammation and related brain damage, leading to amplified functional deficits. The impact of chronic T. gondii infection on TBI should therefore be investigated in both preclinical and clinical studies as the possible interplay could influence treatment strategies.
Collapse
Affiliation(s)
- Tamara L Baker
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Shiraz Tyebji
- Division of Infectious Diseases and Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Christopher J Tonkin
- Division of Infectious Diseases and Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, 6th Floor, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia. .,Department of Medicine, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
134
|
Ohgomori T, Jinno S. Modulation of neuropathology and cognitive deficits by lipopolysaccharide preconditioning in a mouse pilocarpine model of status epilepticus. Neuropharmacology 2020; 176:108227. [PMID: 32634527 DOI: 10.1016/j.neuropharm.2020.108227] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/15/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Recent studies indicate that microglia may play a critical role in epileptogenesis during the early post-status epilepticus (SE) period. In this study, we aimed to elucidate the effects of preconditioning of microglia with lipopolysaccharide (LPS) on neuropathology and cognitive deficits in a mouse pilocarpine model of SE. Mice were treated with an intraperitoneal injection of LPS 24 h before SE induction. The open field test at 13 days after SE showed that LPS preconditioning suppressed SE-induced hyperactivity. The Y-maze test at 14 days after SE showed that LPS preconditioning ameliorated SE-induced working memory impairment. The extent of neuronal damage was decreased by LPS preconditioning in the hippocampus of mice euthanized at 15 days after SE. Gene profile analysis of hippocampal microglia at 15 days after SE showed that the expression level of interleukin-1β was increased by SE induction but decreased by LPS preconditioning. By contrast, SE induction increased the expression levels of phagocytosis-related genes, and LPS preconditioning further enhanced their expression. Interestingly, LPS preconditioning increased the numerical density of hippocampal microglia expressing the 5D4 keratan sulfate epitope, a population of cells known to be involved in phagocytosis. The voxel density of glutamatergic synapses was increased by SE induction but decreased by LPS preconditioning, while GABAergic synapses were not affected by these procedures. Our findings indicate that LPS preconditioning may in part alleviate SE-related abnormal synaptogenesis and cognitive deficits, and also suggest that modulation of microglial activation during the early post-SE period may be a novel strategy for epilepsy treatment.
Collapse
Affiliation(s)
- Tomohiro Ohgomori
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan; Department of Rehabilitation, Faculty of Rehabilitation, Osaka Kawasaki Rehabilitation University, Kaizuka, 597-0104, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
135
|
Chidambaram H, Chinnathambi S. G-Protein Coupled Receptors and Tau-different Roles in Alzheimer’s Disease. Neuroscience 2020; 438:198-214. [DOI: 10.1016/j.neuroscience.2020.04.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 04/10/2020] [Accepted: 04/15/2020] [Indexed: 01/14/2023]
|
136
|
TLR-2 neutralization potentiates microglial M1 to M2 switching by the combinatorial treatment of ciprofloxacin and dexamethasone during S. aureus infection. J Neuroimmunol 2020; 344:577262. [DOI: 10.1016/j.jneuroim.2020.577262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022]
|
137
|
Rodríguez-Cruz A, Romo-Mancillas A, Mendiola-Precoma J, Escobar-Cabrera JE, García-Alcocer G, Berumen LC. "Effect of valerenic acid on neuroinflammation in a MPTP-induced mouse model of Parkinson's disease". IBRO Rep 2020; 8:28-35. [PMID: 31909290 PMCID: PMC6938966 DOI: 10.1016/j.ibror.2019.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022] Open
Abstract
Parkinson´s disease is the most important neuromotor pathology due to the prominent loss of dopaminergic neurons in the substantia nigra pars compacta. There is an inherent deficiency of dopamine in Parkinson´s disease, which is aggravated when neuroinflammatory processes are present. Several biomolecules are interesting candidates for the regulation of inflammation and possible neuroprotection, such as valerenic acid, one of the main components of Valeriana officinalis. A 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP)-induced mouse model of Parkinson's disease was developed to evaluate the motor effects of valerenic acid. The evaluation was carried out with four tests (an invert screen test for muscle strength, cross beam test, open field mobility test and lifting on hind legs test). Subsequently, the neuroinflammatory process was evaluated through ELISA of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α and IFN-γ). The decreases in the inflammatory and neurodegenerative processes were evaluated by Western blot and immunohistochemistry analyses of the tissues, which included an evaluation of the tyrosine hydroxylase and GFAP proteins. Finally, the predicted mechanism of action of valerenic acid was supported by molecular docking calculations with the 5-HT5A receptor. The results indicate that the use of valerenic acid as a co-treatment decreases the neuroinflammation in Parkinson's disease induced by MPTP and provides evidence of a decrease in the evaluated pro-inflammatory cytokines and in the amount of GFAP in the mesencephalic area. Valerenic acid prevents neuroinflammation in a Parkinson's disease mouse model, which might reflect the neuroprotection of dopaminergic neurons with the recovery of motor ability.
Collapse
Affiliation(s)
- Alfredo Rodríguez-Cruz
- Laboratorio de Investigación Genética, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
- Posgrado en Ciencias Químico-Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Antonio Romo-Mancillas
- Posgrado en Ciencias Químico-Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
- Laboratorio de Diseño Asistido por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Jesus Mendiola-Precoma
- Laboratorio de Investigación Genética, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
- Posgrado en Ciencias Químico-Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Jesica Esther Escobar-Cabrera
- Laboratorio de Investigación Genética, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
- Posgrado en Ciencias Químico-Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Guadalupe García-Alcocer
- Laboratorio de Investigación Genética, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
- Posgrado en Ciencias Químico-Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Laura Cristina Berumen
- Laboratorio de Investigación Genética, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
- Posgrado en Ciencias Químico-Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
| |
Collapse
|
138
|
Cui W, Sun C, Ma Y, Wang S, Wang X, Zhang Y. Inhibition of TLR4 Induces M2 Microglial Polarization and Provides Neuroprotection via the NLRP3 Inflammasome in Alzheimer's Disease. Front Neurosci 2020; 14:444. [PMID: 32508567 PMCID: PMC7251077 DOI: 10.3389/fnins.2020.00444] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/09/2020] [Indexed: 11/18/2022] Open
Abstract
Accumulating evidence has indicated that activation of microglia and neuroinflammation reaction play a prominent role in Alzheimer’s disease (AD). Inhibition of toll-like receptor 4 (TLR4) has been shown to be associated with immune responses and brain damage, but its effects on AD remain unclear. This study mainly aimed to investigate the protective effect of TAK-242 (TLR4-specific inhibitor) on microglial polarization and neuroprotection in an AD mouse model and the underlying mechanisms. We found that APP/PS1 transgenic AD mice exhibited a dramatic increase in TLR4 levels concomitant with a significantly higher expression of inflammatory microglia compared to C57BL/6 wild-type mice. Furthermore, inhibition of TLR4 by TAK-242 administration significantly improved neurological function, decreased the level of Bax, and caused a significant reduction in the levels of M1-markers (iNOS and TNFα), while the expressions of M2-phenotype markers (Trem-2 and Arg-1) were increased both in vivo and in vitro. Furthermore, TAK-242 treatment enhanced BV2 microglial phagocytosis. Moreover, Aβ25–35 caused the upregulation of inflammatory cytokine production, MyD88, NF-kappaB-p65, and NLRP3, which could be ameliorated by NLRP3-siRNA or TAK-242. These findings indicated that TLR4 inhibition provided neuroprotection and promoted a microglial switch from the inflammatory M1 phenotype to the protective M2 phenotype in AD. The mechanism involved may be related to modulation of the MyD88/NF-kappaB/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Weigang Cui
- Department of Human Anatomy, Xinxiang Medical University, Xinxiang, China.,Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Chunli Sun
- Department of Human Anatomy, Xinxiang Medical University, Xinxiang, China.,Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, China
| | - Yuqi Ma
- Department of Human Anatomy, Xinxiang Medical University, Xinxiang, China
| | - Songtao Wang
- Department of Human Anatomy, Xinxiang Medical University, Xinxiang, China.,Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, China
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Yinghua Zhang
- Department of Human Anatomy, Xinxiang Medical University, Xinxiang, China.,Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
139
|
Fontana IC, Zimmer AR, Rocha AS, Gosmann G, Souza DO, Lourenco MV, Ferreira ST, Zimmer ER. Amyloid-β oligomers in cellular models of Alzheimer's disease. J Neurochem 2020; 155:348-369. [PMID: 32320074 DOI: 10.1111/jnc.15030] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/21/2020] [Accepted: 04/17/2020] [Indexed: 12/22/2022]
Abstract
Amyloid-β (Aβ) dysmetabolism is tightly associated with pathological processes in Alzheimer's disease (AD). Currently, it is thought that, in addition to Aβ fibrils that give rise to plaque formation, Aβ aggregates into non-fibrillar soluble oligomers (AβOs). Soluble AβOs have been extensively studied for their synaptotoxic and neurotoxic properties. In this review, we discuss physicochemical properties of AβOs and their impact on different brain cell types in AD. Additionally, we summarize three decades of studies with AβOs, providing a compelling bulk of evidence regarding cell-specific mechanisms of toxicity. Cellular models may lead us to a deeper understanding of the detrimental effects of AβOs in neurons and glial cells, putatively shedding light on the development of innovative therapies for AD.
Collapse
Affiliation(s)
- Igor C Fontana
- Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Graduate Program in Biological Sciences: Biochemistry, UFRGS, Porto Alegre, Brazil
| | - Aline R Zimmer
- Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Andreia S Rocha
- Graduate Program in Biological Sciences: Biochemistry, UFRGS, Porto Alegre, Brazil
| | - Grace Gosmann
- Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, UFRGS, Porto Alegre, Brazil.,Department of Biochemistry, UFRGS, Porto Alegre, Brazil
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, UFRGS, Porto Alegre, Brazil.,Department of Pharmacology, UFRGS, Porto Alegre, Brazil.,Graduate Program in Biological Sciences: Pharmacology and Therapeutics,, UFRGS, Porto Alegre, Brazil
| |
Collapse
|
140
|
Matrisciano F, Pinna G. PPAR and functional foods: Rationale for natural neurosteroid-based interventions for postpartum depression. Neurobiol Stress 2020; 12:100222. [PMID: 32426424 PMCID: PMC7226878 DOI: 10.1016/j.ynstr.2020.100222] [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: 01/24/2020] [Revised: 03/23/2020] [Accepted: 04/08/2020] [Indexed: 02/07/2023] Open
Abstract
Allopregnanolone, a GABAergic neurosteroid and progesterone derivative, was recently approved by the Food and Drug Administration for the treatment of postpartum depression (PPD). Several mechanisms appear to be involved in the pathogenesis of PPD, including neuroendocrine dysfunction, neuroinflammation, neurotransmitter alterations, genetic and epigenetic modifications. Recent evidence highlights the higher risk for incidence of PPD in mothers exposed to unhealthy diets that negatively impact the microbiome composition and increase inflammation, all effects that are strongly correlated with mood disorders. Conversely, healthy diets have consistently been reported to decrease the risk of peripartum depression and to protect the body and brain against low-grade systemic chronic inflammation. Several bioactive micronutrients found in the so-called functional foods have been shown to play a relevant role in preventing neuroinflammation and depression, such as vitamins, minerals, omega-3 fatty acids and flavonoids. An intriguing molecular substrate linking functional foods with improvement of mood disorders may be represented by the peroxisome-proliferator activated receptor (PPAR) pathway, which can regulate allopregnanolone biosynthesis and brain-derived neurotropic factor (BDNF) and thereby may reduce inflammation and elevate mood. Herein, we discuss the potential connection between functional foods and PPAR and their role in preventing neuroinflammation and symptoms of PPD through neurosteroid regulation. We suggest that healthy diets by targeting the PPAR-neurosteroid axis and thereby decreasing inflammation may offer a suitable functional strategy to prevent and safely alleviate mood symptoms during the perinatal period.
Collapse
Affiliation(s)
- Francesco Matrisciano
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago (UIC), Chicago, IL, USA
| | - Graziano Pinna
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago (UIC), Chicago, IL, USA
| |
Collapse
|
141
|
Yuan Y, Wu C, Ling EA. Heterogeneity of Microglia Phenotypes: Developmental, Functional and Some Therapeutic Considerations. Curr Pharm Des 2020; 25:2375-2393. [PMID: 31584369 DOI: 10.2174/1381612825666190722114248] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Microglia play a pivotal role in maintaining homeostasis in complex brain environment. They first exist as amoeboid microglial cells (AMCs) in the developing brain, but with brain maturation, they transform into ramified microglial cells (RMCs). In pathological conditions, microglia are activated and have been classified into M1 and M2 phenotypes. The roles of AMCs, RMCs and M1/M2 microglia phenotypes especially in pathological conditions have been the focus of many recent studies. METHODS Here, we review the early development of the AMCs and RMCs and discuss their specific functions with reference to their anatomic locations, immunochemical coding etc. M1 and M2 microglia phenotypes in different neuropathological conditions are also reviewed. RESULTS Activated microglia are engaged in phagocytosis, production of proinflammatory mediators, trophic factors and synaptogenesis etc. Prolonged microglia activation, however, can cause damage to neurons and oligodendrocytes. The M1 and M2 phenotypes featured prominently in pathological conditions are discussed in depth. Experimental evidence suggests that microglia phenotype is being modulated by multiple factors including external and internal stimuli, local demands, epigenetic regulation, and herbal compounds. CONCLUSION Prevailing views converge that M2 polarization is neuroprotective. Thus, proper therapeutic designs including the use of anti-inflammatory drugs, herbal agents may be beneficial in suppression of microglial activation, especially M1 phenotype, for amelioration of neuroinflammation in different neuropathological conditions. Finally, recent development of radioligands targeting 18 kDa translocator protein (TSPO) in activated microglia may hold great promises clinically for early detection of brain lesion with the positron emission tomography.
Collapse
Affiliation(s)
- Yun Yuan
- Department of Anatomy and Histology/Embryology, Kunming Medical University, 1168 West Chunrong Road, Kunming, China
| | - Chunyun Wu
- Department of Anatomy and Histology/Embryology, Kunming Medical University, 1168 West Chunrong Road, Kunming, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, 4 Medical Drive, MD10, National University of Singapore, 117594, Singapore
| |
Collapse
|
142
|
O'Grady BJ, Lippmann ES. Recent Advancements in Engineering Strategies for Manipulating Neural Stem Cell Behavior. ACTA ACUST UNITED AC 2020; 1:41-47. [PMID: 33748772 DOI: 10.1007/s43152-020-00003-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Purpose of Review Stem cells are exquisitely sensitive to biophysical and biochemical cues within the native microenvironment. This review focuses on emerging strategies to manipulate neural cell behavior using these influences in three-dimensional (3D) culture systems. Recent Findings Traditional systems for neural cell differentiation typically produce heterogeneous populations with limited diversity rather than the complex, organized tissue structures observed in vivo. Advancements in developing engineering tools to direct neural cell fates can enable new applications in basic research, disease modeling, and regenerative medicine. Summary This review article highlights engineering strategies that facilitate controlled presentation of biophysical and biochemical cues to guide differentiation and impart desired phenotypes on neural cell populations. Specific highlighted examples include engineered biomaterials and microfluidic platforms for spatiotemporal control over the presentation of morphogen gradients.
Collapse
Affiliation(s)
- Brian J O'Grady
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
143
|
Desale SE, Chinnathambi S. Role of dietary fatty acids in microglial polarization in Alzheimer's disease. J Neuroinflammation 2020; 17:93. [PMID: 32209097 PMCID: PMC7093977 DOI: 10.1186/s12974-020-01742-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Microglial polarization is an utmost important phenomenon in Alzheimer’s disease that influences the brain environment. Polarization depends upon the types of responses that cells undergo, and it is characterized by receptors present on the cell surface and the secreted cytokines to the most. The expression of receptors on the surface is majorly influenced by internal and external factors such as dietary lipids. Types of fatty acids consumed through diet influence the brain environment and glial cell phenotype and types of receptors on microglia. Reports suggest that dietary habits influence microglial polarization and the switching of microglial phenotype is very important in neurodegenerative diseases. Omega-3 fatty acids have more influence on the brain, and they are found to regulate the inflammatory stage of microglia by fine-tuning the number of receptors expressed on microglia cells. In Alzheimer’s disease, one of the pathological proteins involved is Tau protein, and microtubule-associated protein upon abnormal phosphorylation detaches from the microtubule and forms insoluble aggregates. Aggregated proteins have a tendency to propagate within the neurons and also become one of the causes of neuroinflammation. We hypothesize that tuning microglia towards anti-inflammatory phenotype would reduce the propagation of Tau in Alzheimer’s disease.
Collapse
Affiliation(s)
- Smita Eknath Desale
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, 411008, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India. .,Academy of Scientific and Innovative Research (AcSIR), Pune, 411008, India.
| |
Collapse
|
144
|
Kuter KZ, Cenci MA, Carta AR. The role of glia in Parkinson's disease: Emerging concepts and therapeutic applications. PROGRESS IN BRAIN RESEARCH 2020; 252:131-168. [PMID: 32247363 DOI: 10.1016/bs.pbr.2020.02.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Originally believed to primarily affect neurons, Parkinson's disease (PD) has recently been recognized to also affect the functions and integrity of microglia and astroglia, two cell categories of fundamental importance to brain tissue homeostasis, defense, and repair. Both a loss of glial supportive-defensive functions and a toxic gain of glial functions are implicated in the neurodegenerative process. Moreover, the chronic treatment with L-DOPA may cause maladaptive glial plasticity favoring a development of therapy complications. This chapter focuses on the pathophysiology of PD from a glial point of view, presenting this rapidly growing field from the first discoveries made to the most recent developments. We report and compare histopathological and molecular findings from experimental models of PD and human studies. We moreover discuss the important role played by astrocytes in compensatory adaptations taking place during presymptomatic disease stages. We finally describe examples of potential therapeutic applications stemming from an increased understanding of the important roles of glia in PD.
Collapse
Affiliation(s)
- Katarzyna Z Kuter
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Cagliari, Italy.
| |
Collapse
|
145
|
Liu J, Tang M, Harkin K, Du X, Luo C, Chen M, Xu H. Single-cell RNA sequencing study of retinal immune regulators identified CD47 and CD59a expression in photoreceptors-Implications in subretinal immune regulation. J Neurosci Res 2020; 98:1498-1513. [PMID: 32166783 DOI: 10.1002/jnr.24618] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/21/2020] [Accepted: 02/29/2020] [Indexed: 12/29/2022]
Abstract
The neuroretina is protected by its own defense system, that is microglia and the complement system. Under normal physiological conditions, microglial activation is tightly regulated by the neurons although the underlying mechanism remains elusive. Using published single-cell RNA sequencing data sets, we found that immune regulatory molecules including CD200, CD47, CX3CL1, TGFβ, and complement inhibitor CD59a are expressed by various retinal neurons. Importantly, we found that photoreceptors express higher levels of CD47 and CD59a, which was further confirmed in cultured 661W cells, WERI-Rb1 cells, and microdissected photoreceptors from human eyes. The expression of CD59a mRNA in 661W cells was upregulated by TNFα and hypoxia, whereas LPS, hypoxia, and IL-4 upregulated CD47 mRNA expression in 661W cells. Immunofluorescence staining detected strong CD59a immunoreactivity in the outer nuclear layer, inner/outer segments, and discrete staining in ganglion cell layer (GCL), inner plexiform layer (IPL), and outer plexiform layer. The expression of CD59a in photoreceptors was increased in the detached retina, but decreased in retinas from experimental autoimmune uveoretinitis (EAU) mice. In EAU retina, CD59a was highly expressed by active immune cells. CD47 was detected in GCL, IPL, and inner nuclear layer and some photoreceptors. The expression of CD47 in photoreceptors was also increased in the detached retina but decreased in EAU retina. In a coculture system, 661W enhanced arginase-1 and reduced IL-6 mRNA expression in BV2 microglial cells. Our results suggest that photoreceptors express immune regulatory molecules and may have the potential to regulate immune activation in the outer retina/subretinal space under pathophysiological conditions.
Collapse
Affiliation(s)
- Jian Liu
- Aier Eye Institute, Aier School of Ophthalmology, Central South University, Changsha, P.R. China
| | - Miao Tang
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Kevin Harkin
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Xuan Du
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Chang Luo
- Aier Eye Institute, Aier School of Ophthalmology, Central South University, Changsha, P.R. China
| | - Mei Chen
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Heping Xu
- Aier Eye Institute, Aier School of Ophthalmology, Central South University, Changsha, P.R. China.,The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| |
Collapse
|
146
|
Buruiană A, Florian ȘI, Florian AI, Timiș TL, Mihu CM, Miclăuș M, Oșan S, Hrapșa I, Cataniciu RC, Farcaș M, Șușman S. The Roles of miRNA in Glioblastoma Tumor Cell Communication: Diplomatic and Aggressive Negotiations. Int J Mol Sci 2020; 21:ijms21061950. [PMID: 32178454 PMCID: PMC7139390 DOI: 10.3390/ijms21061950] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM) consists of a heterogeneous collection of competing cellular clones which communicate with each other and with the tumor microenvironment (TME). MicroRNAs (miRNAs) present various exchange mechanisms: free miRNA, extracellular vesicles (EVs), or gap junctions (GJs). GBM cells transfer miR-4519 and miR-5096 to astrocytes through GJs. Oligodendrocytes located in the invasion front present high levels of miR-219-5p, miR-219-2-3p, and miR-338-3p, all related to their differentiation. There is a reciprocal exchange between GBM cells and endothelial cells (ECs) as miR-5096 promotes angiogenesis after being transferred into ECs, whereas miR-145-5p acts as a tumor suppressor. In glioma stem cells (GSCs), miR-1587 and miR-3620-5p increase the proliferation and miR-1587 inhibits the hormone receptor co-repressor-1 (NCOR1) after EVs transfers. GBM-derived EVs carry miR-21 and miR-451 that are up-taken by microglia and monocytes/macrophages, promoting their proliferation. Macrophages release EVs enriched in miR-21 that are transferred to glioma cells. This bidirectional miR-21 exchange increases STAT3 activity in GBM cells and macrophages, promoting invasion, proliferation, angiogenesis, and resistance to treatment. miR-1238 is upregulated in resistant GBM clones and their EVs, conferring resistance to adjacent cells via the CAV1/EGFR signaling pathway. Decrypting these mechanisms could lead to a better patient stratification and the development of novel target therapies.
Collapse
Affiliation(s)
- Andrei Buruiană
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (A.B.); (S.O.); (I.H.); (R.C.C.); (M.F.)
| | - Ștefan Ioan Florian
- Department of Neurosurgery, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (Ș.I.F.); (A.I.F.)
- Department of Neurosurgery, Emergency County Hospital, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Alexandru Ioan Florian
- Department of Neurosurgery, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (Ș.I.F.); (A.I.F.)
- Department of Neurosurgery, Emergency County Hospital, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Teodora-Larisa Timiș
- Department of Physiology, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania;
| | - Carmen Mihaela Mihu
- Department of Morphological Sciences-Histology, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania;
| | - Maria Miclăuș
- Department of Medical Genetics, Emergency Hospital for Children, 68 Moților Street, 400370 Cluj-Napoca, Romania;
| | - Sergiu Oșan
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (A.B.); (S.O.); (I.H.); (R.C.C.); (M.F.)
| | - Iona Hrapșa
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (A.B.); (S.O.); (I.H.); (R.C.C.); (M.F.)
| | - Radu Constantin Cataniciu
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (A.B.); (S.O.); (I.H.); (R.C.C.); (M.F.)
| | - Marius Farcaș
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (A.B.); (S.O.); (I.H.); (R.C.C.); (M.F.)
- Department of Genetics, IMOGEN Research Center, Louis Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Sergiu Șușman
- Department of Morphological Sciences-Histology, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania;
- Department of Pathology, IMOGEN Research Center, Louis Pasteur Street, 400349 Cluj-Napoca, Romania
- Correspondence:
| |
Collapse
|
147
|
Rawlinson C, Jenkins S, Thei L, Dallas ML, Chen R. Post-Ischaemic Immunological Response in the Brain: Targeting Microglia in Ischaemic Stroke Therapy. Brain Sci 2020; 10:brainsci10030159. [PMID: 32168831 PMCID: PMC7139954 DOI: 10.3390/brainsci10030159] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/27/2020] [Accepted: 03/07/2020] [Indexed: 12/21/2022] Open
Abstract
Microglia, the major endogenous immune cells of the central nervous system, mediate critical degenerative and regenerative responses in ischaemic stroke. Microglia become "activated", proliferating, and undergoing changes in morphology, gene and protein expression over days and weeks post-ischaemia, with deleterious and beneficial effects. Pro-inflammatory microglia (commonly referred to as M1) exacerbate secondary neuronal injury through the release of reactive oxygen species, cytokines and proteases. In contrast, microglia may facilitate neuronal recovery via tissue and vascular remodelling, through the secretion of anti-inflammatory cytokines and growth factors (a profile often termed M2). This M1/M2 nomenclature does not fully account for the microglial heterogeneity in the ischaemic brain, with some simultaneous expression of both M1 and M2 markers at the single-cell level. Understanding and regulating microglial activation status, reducing detrimental and promoting repair behaviours, present the potential for therapeutic intervention, and open a longer window of opportunity than offered by acute neuroprotective strategies. Pharmacological modulation of microglial activation status to promote anti-inflammatory gene expression can increase neurogenesis and improve functional recovery post-stroke, based on promising preclinical data. Cell-based therapies, using preconditioned microglia, are of interest as a method of therapeutic modulation of the post-ischaemic inflammatory response. Currently, there are no clinically-approved pharmacological options targeting post-ischaemic inflammation. A major developmental challenge for clinical translation will be the selective suppression of the deleterious effects of microglial activity after stroke whilst retaining (or enhancing) the neurovascular repair and remodelling responses of microglia.
Collapse
Affiliation(s)
- Charlotte Rawlinson
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK;
| | - Stuart Jenkins
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK;
| | - Laura Thei
- School of Pharmacy, University of Reading, Reading RG6 6UB, UK; (L.T.); (M.L.D.)
| | - Mark L. Dallas
- School of Pharmacy, University of Reading, Reading RG6 6UB, UK; (L.T.); (M.L.D.)
| | - Ruoli Chen
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK;
- Correspondence: ; Tel.: +44-1782-733849; Fax: 44-1782-733326
| |
Collapse
|
148
|
De Luca SN, Miller AA, Sominsky L, Spencer SJ. Microglial regulation of satiety and cognition. J Neuroendocrinol 2020; 32:e12838. [PMID: 32097992 DOI: 10.1111/jne.12838] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/15/2020] [Accepted: 01/28/2020] [Indexed: 12/11/2022]
Abstract
Microglia have been known for decades as key immune cells that shape the central nervous system (CNS) during development and respond to brain pathogens and injury in adult life. Recent findings now suggest that these cells also play a highly complex role in several other functions of the CNS. In this review, we provide a brief overview of the established microglial functions in development and disease. We also discuss emerging research suggesting that microglia are important for both cognitive function and the regulation of food intake. With respect to cognitive function, current data suggest microglia are not indispensable for neurogenesis, synaptogenesis or cognition in the healthy young adult, although they crucially modulate and support these functions. In doing so, they are likely important in supporting the balance between apoptosis and survival of newborn neurones and in orchestrating appropriate synaptic remodelling in response to a learning stimulus. We also explore the possibility of a role for microglia in feeding and satiety. Microglia have been implicated in both appetite suppression with sickness and obesity and in promoting feeding under some conditions and we discuss these findings here, highlighting the contribution of these cells to healthy brain function.
Collapse
Affiliation(s)
- Simone N De Luca
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Alyson A Miller
- Institute of Cardiovascular & Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Luba Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
149
|
Coelho‐Santos V, Shih AY. Postnatal development of cerebrovascular structure and the neurogliovascular unit. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2020; 9:e363. [PMID: 31576670 PMCID: PMC7027551 DOI: 10.1002/wdev.363] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 12/22/2022]
Abstract
The unceasing metabolic demands of brain function are supported by an intricate three-dimensional network of arterioles, capillaries, and venules, designed to effectively distribute blood to all neurons and to provide shelter from harmful molecules in the blood. The development and maturation of this microvasculature involves a complex interplay between endothelial cells with nearly all other brain cell types (pericytes, astrocytes, microglia, and neurons), orchestrated throughout embryogenesis and the first few weeks after birth in mice. Both the expansion and regression of vascular networks occur during the postnatal period of cerebrovascular remodeling. Pial vascular networks on the brain surface are dense at birth and are then selectively pruned during the postnatal period, with the most dramatic changes occurring in the pial venular network. This is contrasted to an expansion of subsurface capillary networks through the induction of angiogenesis. Concurrent with changes in vascular structure, the integration and cross talk of neurovascular cells lead to establishment of blood-brain barrier integrity and neurovascular coupling to ensure precise control of macromolecular passage and metabolic supply. While we still possess a limited understanding of the rules that control cerebrovascular development, we can begin to assemble a view of how this complex process evolves, as well as identify gaps in knowledge for the next steps of research. This article is categorized under: Nervous System Development > Vertebrates: Regional Development Vertebrate Organogenesis > Musculoskeletal and Vascular Nervous System Development > Vertebrates: General Principles.
Collapse
Affiliation(s)
- Vanessa Coelho‐Santos
- Center for Developmental Biology and Regenerative MedicineSeattle Children's Research InstituteSeattleWashington
- Department of PediatricsUniversity of WashingtonSeattleWashington
| | - Andy Y. Shih
- Center for Developmental Biology and Regenerative MedicineSeattle Children's Research InstituteSeattleWashington
- Department of PediatricsUniversity of WashingtonSeattleWashington
| |
Collapse
|
150
|
De Luca SN, Soch A, Sominsky L, Nguyen TX, Bosakhar A, Spencer SJ. Glial remodeling enhances short-term memory performance in Wistar rats. J Neuroinflammation 2020; 17:52. [PMID: 32028971 PMCID: PMC7006153 DOI: 10.1186/s12974-020-1729-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/28/2020] [Indexed: 12/16/2022] Open
Abstract
Background Microglia play a key role in neuronal circuit and synaptic maturation in the developing brain. In the healthy adult, however, their role is less clear: microglial hyperactivation in adults can be detrimental to memory due to excessive synaptic pruning, yet learning and memory can also be impaired in the absence of these cells. In this study, we therefore aimed to determine how microglia contribute to short-term memory in healthy adults. Methods To this end, we developed a Cx3cr1-Dtr transgenic Wistar rat with a diphtheria toxin receptor (Dtr) gene inserted into the fractalkine receptor (Cx3cr1) promoter, expressed on microglia and monocytes. This model allows acute microglial and monocyte ablation upon application of diphtheria toxin, enabling us to directly assess microglia’s role in memory. Results Here, we show that short-term memory in the novel object and place recognition tasks is entirely unaffected by acute microglial ablation. However, when microglia repopulate the brain after depletion, learning and memory performance in these tasks is improved. This transitory memory enhancement is associated with an ameboid morphology in the newly repopulated microglial cells and increased astrocyte density that are linked with a higher density of mature hippocampal synaptic spines and differences in pre- and post-synaptic markers. Conclusions These data indicate that glia play a complex role in the healthy adult animal in supporting appropriate learning and memory and that subtle changes to the function of these cells may strategically enhance memory.
Collapse
Affiliation(s)
- Simone N De Luca
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, 3083, Australia
| | - Alita Soch
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, 3083, Australia
| | - Luba Sominsky
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, 3083, Australia
| | - Thai-Xinh Nguyen
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, 3083, Australia
| | - Abdulhameed Bosakhar
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, 3083, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, 3083, Australia. .,ARC Centre of Excellence for Nanoscale Biophotonics, RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|