101
|
Fatoba O, Itokazu T, Yamashita T. Microglia as therapeutic target in central nervous system disorders. J Pharmacol Sci 2020; 144:102-118. [PMID: 32921391 DOI: 10.1016/j.jphs.2020.07.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/19/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic microglial activation is associated with the pathogenesis of several CNS disorders. Microglia show phenotypic diversity and functional complexity in diseased CNS. Thus, understanding the pathology-specific heterogeneity of microglial behavior is crucial for the future development of microglia-modulating therapy for variety of CNS disorders. This review summarizes up-to-date knowledge on how microglia contribute to CNS homeostasis during development and throughout adulthood. We discuss the heterogeneity of microglial phenotypes in the context of CNS disorders with an emphasis on neurodegenerative diseases, demyelinating diseases, CNS trauma, and epilepsy. We conclude this review with a discussion about the disease-specific heterogeneity of microglial function and how it could be exploited for therapeutic intervention.
Collapse
Affiliation(s)
- Oluwaseun Fatoba
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Takahide Itokazu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
102
|
Michael J, Unger MS, Poupardin R, Schernthaner P, Mrowetz H, Attems J, Aigner L. Microglia depletion diminishes key elements of the leukotriene pathway in the brain of Alzheimer's Disease mice. Acta Neuropathol Commun 2020; 8:129. [PMID: 32771067 PMCID: PMC7414992 DOI: 10.1186/s40478-020-00989-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/24/2022] Open
Abstract
Leukotrienes (LTs) contribute to the neuropathology of chronic neurodegenerative disorders including Alzheimer's Disease (AD), where they mediate neuroinflammation and neuronal cell-death. In consequence, blocking the action of Leukotrienes (LTs) ameliorates pathologies and improves cognitive function in animal models of neurodegeneration. Surprisingly, the source of Leukotrienes (LTs) in the brain is largely unknown. Here, we identified the Leukotriene (LT) synthesis rate-limiting enzyme 5-Lipoxygenase (5-Lox) primarily in neurons and to a lesser extent in a subpopulation of microglia in human Alzheimer´s Disease (AD) hippocampus brain sections and in brains of APP Swedish PS1 dE9 (APP-PS1) mice, a transgenic model for Alzheimer´s Disease (AD) pathology. The 5-Lipoxygenase (5-Lox) activating protein (FLAP), which anchors 5-Lipoxygenase (5-Lox) to the membrane and mediates the contact to the substrate arachidonic acid, was confined exclusively to microglia with the entire microglia population expressing 5-Lipoxygenase activating protein (FLAP). To define the contribution of microglia in the Leukotriene (LT) biosynthesis pathway, we ablated microglia using the colony stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 in wildtype (WT) and APP-PS1 mice. Microglia ablation not only diminished the expression of FLAP and of the Leukotriene (LT) receptor Cysteinylleukotriene receptor 1 (CysLTR1), as expected based on their microglia cell type-specific expression, but also drastically reduced 5-Lipoxygenase (5-Lox) mRNA expression in the brain and its protein expression in neurons, in particular in wildtype (WT) mice. In conclusion i) microglia are key in Leukotriene (LT) biosynthesis, and ii) they regulate neuronal 5-Lipoxygenase (5-Lox) expression implying a yet unknown signaling mechanism between neurons and microglia.
Collapse
Affiliation(s)
- J Michael
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Strubergasse 21, 5020, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - M S Unger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Strubergasse 21, 5020, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - R Poupardin
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
- Experimental and Clinical Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - P Schernthaner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Strubergasse 21, 5020, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - H Mrowetz
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Strubergasse 21, 5020, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - J Attems
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - L Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Strubergasse 21, 5020, Salzburg, Austria.
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| |
Collapse
|
103
|
Burfeind KG, Zhu X, Norgard MA, Levasseur PR, Huisman C, Michaelis KA, Olson B, Marks DL. Microglia in the hypothalamus respond to tumor-derived factors and are protective against cachexia during pancreatic cancer. Glia 2020; 68:1479-1494. [PMID: 32039522 PMCID: PMC7205589 DOI: 10.1002/glia.23796] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/27/2020] [Accepted: 01/30/2020] [Indexed: 12/14/2022]
Abstract
Microglia in the mediobasal hypothalamus (MBH) respond to inflammatory stimuli and metabolic perturbations to mediate body composition. This concept is well studied in the context of high fat diet induced obesity (HFDO), yet has not been investigated in the context of cachexia, a devastating metabolic syndrome characterized by anorexia, fatigue, and muscle catabolism. We show that microglia accumulate specifically in the MBH early in pancreatic ductal adenocarcinoma (PDAC)-associated cachexia and assume an activated morphology. Furthermore, we observe astrogliosis in the MBH and hippocampus concurrent with cachexia initiation. We next show that circulating immune cells resembling macrophages infiltrate the MBH. PDAC-derived factors induced microglia to express a transcriptional profile in vitro that was distinct from that induced by lipopolysaccharide (LPS). Microglia depletion through CSF1-R antagonism resulted in accelerated cachexia onset and increased anorexia, fatigue, and muscle catabolism during PDAC. This corresponded with increased hypothalamic-pituitary-adrenal (HPA) axis activation. CSF1-R antagonism had little effect on inflammatory response in the circulation, liver, or tumor. These findings demonstrate that microglia are protective against PDAC cachexia and provide mechanistic insight into this function.
Collapse
Affiliation(s)
- Kevin G. Burfeind
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandOregon
- Medical Scientist Training Program, Oregon Health & Science UniversityPortlandOregon
| | - Xinxia Zhu
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandOregon
| | - Mason A. Norgard
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandOregon
| | - Peter R. Levasseur
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandOregon
| | - Christian Huisman
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandOregon
| | - Katherine A. Michaelis
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandOregon
- Medical Scientist Training Program, Oregon Health & Science UniversityPortlandOregon
| | - Brennan Olson
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandOregon
- Medical Scientist Training Program, Oregon Health & Science UniversityPortlandOregon
| | - Daniel L. Marks
- Papé Family Pediatric Research InstituteOregon Health & Science UniversityPortlandOregon
- Knight Cancer InstituteOregon Health & Science UniversityPortlandOregon
- Brenden‐Colson Center for Pancreatic CareOregon Health and & Science UniversityPortlandOregon
| |
Collapse
|
104
|
Hu X, Li S, Doycheva DM, Huang L, Lenahan C, Liu R, Huang J, Xie S, Tang J, Zuo G, Zhang JH. Rh-CSF1 attenuates neuroinflammation via the CSF1R/PLCG2/PKCε pathway in a rat model of neonatal HIE. J Neuroinflammation 2020; 17:182. [PMID: 32522286 PMCID: PMC7285566 DOI: 10.1186/s12974-020-01862-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/29/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) is a life-threatening cerebrovascular disease. Neuroinflammation plays an important role in the pathogenesis of HIE, in which microglia are key cellular mediators in the regulation of neuroinflammatory processes. Colony-stimulating factor 1 (CSF1), a specific endogenous ligand of CSF1 receptor (CSF1R), is crucial in microglial growth, differentiation, and proliferation. Recent studies showed that the activation of CSF1R with CSF1 exerted anti-inflammatory effects in a variety of nervous system diseases. This study aimed to investigate the anti-inflammatory effects of recombinant human CSF1 (rh-CSF1) and the underlying mechanisms in a rat model of HIE. METHODS A total of 202 10-day old Sprague Dawley rat pups were used. HI was induced by the right common carotid artery ligation with subsequent exposure of 2.5-h hypoxia. At 1 h and 24 h after HI induction, exogenous rh-CSF1 was administered intranasally. To explore the underlying mechanism, CSF1R inhibitor, BLZ945, and phospholipase C-gamma 2 (PLCG2) inhibitor, U73122, were injected intraperitoneally at 1 h before HI induction, respectively. Brain infarct area, brain water content, neurobehavioral tests, western blot, and immunofluorescence staining were performed. RESULTS The expressions of endogenous CSF1, CSF1R, PLCG2, protein kinase C epsilon type (PKCε), and cAMP response element-binding protein (CREB) were gradually increased after HIE. Rh-CSF1 significantly improved the neurological deficits at 48 h and 4 weeks after HI, which was accompanied by a reduction in the brain infarct area, brain edema, brain atrophy, and neuroinflammation. Moreover, activation of CSF1R by rh-CSF1 significantly increased the expressions of p-PLCG2, p-PKCε, and p-CREB, but inhibited the activation of neutrophil infiltration, and downregulated the expressions of IL-1β and TNF-α. Inhibition of CSF1R and PLCG2 abolished these neuroprotective effects of rh-CSF1 after HI. CONCLUSIONS Our findings demonstrated that the activation of CSF1R by rh-CSF1 attenuated neuroinflammation and improved neurological deficits after HI. The anti-inflammatory effects of rh-CSF1 partially acted through activating the CSF1R/PLCG2/PKCε/CREB signaling pathway after HI. These results suggest that rh-CSF1 may serve as a potential therapeutic approach to ameliorate injury in HIE patients.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, 550002, China.,Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Shirong Li
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, 550002, China.,Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Desislava Met Doycheva
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.,Bvrrell College of Osteopathic Medicine, Las Cruces, NM, 88003, USA
| | - Rui Liu
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, 550002, China.,Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Juan Huang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.,Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
| | - Shucai Xie
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.,Department of Hepatobiliary Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, Hainan, China
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Gang Zuo
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA. .,Department of Neurosurgery, Taicang Hospital Affiliated to Soochow University, Taicang, Suzhou, 215400, Jiangsu, China.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA. .,Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA. .,Department of Anesthesiology, Loma Linda University, Loma Linda, CA, 92350, USA.
| |
Collapse
|
105
|
Martin-Estebane M, Gomez-Nicola D. Targeting Microglial Population Dynamics in Alzheimer's Disease: Are We Ready for a Potential Impact on Immune Function? Front Cell Neurosci 2020; 14:149. [PMID: 32581720 PMCID: PMC7289918 DOI: 10.3389/fncel.2020.00149] [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: 03/29/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia, affecting two-thirds of people with dementia in the world. To date, no disease-modifying treatments are available to stop or delay the progression of AD. This chronic neurodegenerative disease is dominated by a strong innate immune response, whereby microglia plays a central role as the main resident macrophage of the brain. Recent genome-wide association studies (GWASs) have identified single-nucleotide polymorphisms (SNPs) located in microglial genes and associated with a delayed onset of AD, highlighting the important role of these cells on the onset and/or progression of the disease. These findings have increased the interest in targeting microglia-associated neuroinflammation as a potentially disease-modifying therapeutic approach for AD. In this review we provide an overview on the contribution of microglia to the pathophysiology of AD, focusing on the main regulatory pathways controlling microglial population dynamics during the neuroinflammatory response, such as the colony-stimulating factor 1 receptor (CSF1R), its ligands (the colony-stimulating factor 1 and interleukin 34) and the transcription factor PU.1. We also discuss the current therapeutic strategies targeting proliferation to modulate microglia-associated neuroinflammation and their potential impact on peripheral immune cell populations in the short and long-term. Understanding the effects of immunomodulatory approaches on microglia and other immune cell types might be critical for developing specific, effective, and safe therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Maria Martin-Estebane
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
106
|
Microglia depletion fails to abrogate inflammation-induced sickness in mice and rats. J Neuroinflammation 2020; 17:172. [PMID: 32475344 PMCID: PMC7262755 DOI: 10.1186/s12974-020-01832-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Background Production of inflammatory mediators by reactive microglial cells in the brain is generally considered the primary mechanism underlying the development of symptoms of sickness in response to systemic inflammation. Methods Depletion of microglia was achieved in C57BL/6 mice by chronic oral administration of PLX5622, a specific antagonist of colony stimulating factor-1 receptor, and in rats by a knock-in model in which the diphtheria toxin receptor was expressed under the control of the endogenous fractalkine receptor (CX3CR1) promoter sequence. After successful microglia depletion, mice and rats were injected with a sickness-inducing dose of lipopolysaccharide according to a 2 (depletion vs. control) × 2 (LPS vs. saline) factorial design. Sickness was measured by body weight loss and decreased locomotor activity in rats and mice, and reduced voluntary wheel running in mice. Results Chronic administration of PLX5622 in mice and administration of diphtheria toxin to knock-in rats depleted microglia and peripheral tissue macrophages. However, it did not abrogate the inducible expression of proinflammatory cytokines in the brain in response to LPS and even exacerbated it for some of the cytokines. In accordance with these neuroimmune effects, LPS-induced sickness was not abrogated, rather it was exacerbated when measured by running wheel activity in mice. Conclusions These findings reveal that the sickness-inducing effects of acute inflammation can develop independently of microglia activation.
Collapse
|
107
|
Chronic mild hypoxia accelerates recovery from preexisting EAE by enhancing vascular integrity and apoptosis of infiltrated monocytes. Proc Natl Acad Sci U S A 2020; 117:11126-11135. [PMID: 32371484 DOI: 10.1073/pnas.1920935117] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
While several studies have shown that hypoxic preconditioning suppresses development of the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (MS), no one has yet examined the important clinically relevant question of whether mild hypoxia can impact the progression of preexisting disease. Using a relapsing-remitting model of EAE, here we demonstrate that when applied to preexisting disease, chronic mild hypoxia (CMH, 10% O2) markedly accelerates clinical recovery, leading to long-term stable reductions in clinical score. At the histological level, CMH led to significant reductions in vascular disruption, leukocyte accumulation, and demyelination. Spinal cord blood vessels of CMH-treated mice showed reduced expression of the endothelial activation molecule VCAM-1 but increased expression of the endothelial tight junction proteins ZO-1 and occludin, key mechanisms underlying vascular integrity. Interestingly, while equal numbers of inflammatory leukocytes were present in the spinal cord at peak disease (day 14 postimmunization; i.e., 3 d after CMH started), apoptotic removal of infiltrated leukocytes during the remission phase was markedly accelerated in CMH-treated mice, as determined by increased numbers of monocytes positive for TUNEL and cleaved caspase-3. The enhanced monocyte apoptosis in CMH-treated mice was paralleled by increased numbers of HIF-1α+ monocytes, suggesting that CMH enhances monocyte removal by amplifying the hypoxic stress manifest within monocytes in acute inflammatory lesions. These data demonstrate that mild hypoxia promotes recovery from preexisting inflammatory demyelinating disease and suggest that this protection is primarily the result of enhanced vascular integrity and accelerated apoptosis of infiltrated monocytes.
Collapse
|
108
|
Scheiblich H, Trombly M, Ramirez A, Heneka MT. Neuroimmune Connections in Aging and Neurodegenerative Diseases. Trends Immunol 2020; 41:300-312. [DOI: 10.1016/j.it.2020.02.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 11/26/2022]
|
109
|
Guerrero BL, Sicotte NL. Microglia in Multiple Sclerosis: Friend or Foe? Front Immunol 2020; 11:374. [PMID: 32265902 PMCID: PMC7098953 DOI: 10.3389/fimmu.2020.00374] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/17/2020] [Indexed: 12/14/2022] Open
Abstract
Microglia originate from myeloid progenitors in the embryonic yolk sac and play an integral role in central nervous system (CNS) development, immune surveillance and repair. The role of microglia in multiple sclerosis (MS) has been complex and controversial, with evidence suggesting that these cells play key roles in both active inflammation and remyelination. Here we will review the most recent histological classification of MS lesions as well as the evidence supporting both inflammatory and reparative functions of these cells. We will also review how microglia may yield new biomarkers for MS activity and serve as a potential target for therapy.
Collapse
Affiliation(s)
- Brooke L Guerrero
- Multiple Sclerosis and Neuroimmunology Program, Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Nancy L Sicotte
- Multiple Sclerosis and Neuroimmunology Program, Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
110
|
Microglial Depletion with CSF1R Inhibitor During Chronic Phase of Experimental Traumatic Brain Injury Reduces Neurodegeneration and Neurological Deficits. J Neurosci 2020; 40:2960-2974. [PMID: 32094203 DOI: 10.1523/jneurosci.2402-19.2020] [Citation(s) in RCA: 213] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/03/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic neuroinflammation with sustained microglial activation occurs following severe traumatic brain injury (TBI) and is believed to contribute to subsequent neurodegeneration and neurological deficits. Microglia, the primary innate immune cells in brain, are dependent on colony stimulating factor 1 receptor (CSF1R) signaling for their survival. In this preclinical study, we examined the effects of delayed depletion of chronically activated microglia on functional recovery and neurodegeneration up to 3 months postinjury. A CSF1R inhibitor, Plexxikon (PLX) 5622, was administered to adult male C57BL/6J mice at 1 month after controlled cortical impact to remove chronically activated microglia, and the inhibitor was withdrawn 1-week later to allow for microglial repopulation. Following TBI, the repopulated microglia displayed a ramified morphology similar to that of Sham uninjured mice, whereas microglia in vehicle-treated TBI mice showed the typical chronic posttraumatic hypertrophic morphology. PLX5622 treatment limited TBI-associated neuropathological changes at 3 months postinjury; these included a smaller cortical lesion, reduced hippocampal neuron cell death, and decreased NOX2- and NLRP3 inflammasome-associated neuroinflammation. Furthermore, delayed depletion of chronically activated microglia after TBI led to widespread changes in the cortical transcriptome and altered gene pathways involved in neuroinflammation, oxidative stress, and neuroplasticity. Using a variety of complementary neurobehavioral tests, PLX5622-treated TBI mice also had improved long-term motor and cognitive function recovery through 3 months postinjury. Together, these studies demonstrate that chronic phase removal of neurotoxic microglia after TBI using CSF1R inhibitors markedly reduce chronic neuroinflammation and associated neurodegeneration, as well as related motor and cognitive deficits.SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) is a debilitating neurological disorder that can seriously impact the patient's quality of life. Microglial-mediated neuroinflammation is induced after severe TBI and contributes to neurological deficits and on-going neurodegenerative processes. Here, we investigated the effect of breaking the neurotoxic neuroinflammatory loop at 1-month after controlled cortical impact in mice by pharmacological removal of chronically activated microglia using a colony stimulating factor 1 receptor (CSF1R) inhibitor, Plexxikon 5622. Overall, we show that short-term elimination of microglia during the chronic phase of TBI followed by repopulation results in long-term improvements in neurological function, suppression of neuroinflammatory and oxidative stress pathways, and a reduction in persistent neurodegenerative processes. These studies are clinically relevant and support new concepts that the therapeutic window for TBI may be far longer than traditionally believed if chronic and evolving microglial-mediated neuroinflammation can be inhibited or regulated in a precise manner.
Collapse
|
111
|
Hupp S, Iliev AI. CSF-1 receptor inhibition as a highly effective tool for depletion of microglia in mixed glial cultures. J Neurosci Methods 2020; 332:108537. [PMID: 31790710 DOI: 10.1016/j.jneumeth.2019.108537] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/26/2019] [Accepted: 11/28/2019] [Indexed: 11/15/2022]
Abstract
BACKGROUND A breakthrough in the microglia and macrophages field was the identification of the macrophage colony stimulating factor-1 (CSF-1) as a pro-survival factor. Its pharmacological inhibition in animals depletes rapidly all microglia and macrophages. Microglial depletion in mixed glial cultures has always represented a challenge and none of the existing approaches delivers satisfactory results. NEW METHOD We applied a CSF-1R inhibitor (PLX5622) in primary mouse glial cultures, analyzing microglial dose-responses, starting at different time-points and incubating for various periods of time. RESULTS We used two treatment modalities with 10 μM PLX5622 to deplete microglia: i) immediately after brain homogenization and ii) at day in vitro 12. The application of the inhibitor immediately after cell preparation depleted microglia to 8% at 1 week, to 2% at 4 weeks and to 0.5% at 6 weeks (half-time 3.5 days). When mixed glial cultures were treated starting at day in vitro 12, microglia depletion was slower (half-time 6 days) and not complete, indicating a decreased sensitivity to CSF-1. The remaining astrocytes preserved their proliferation ability, their migration in a scratch wound assay, and their pro-inflammatory (IL-6) response towards lipopolysaccharide. COMPARISON TO EXISTING METHODS The proposed approach for microglial depletion in mixed glial cultures is more effective than other existing methods and is non-toxic to non-microglial cells. CONCLUSIONS CSF-1R inhibitors are effective tools for depleting microglia in mixed glial cultures. Longer maturation of the cultures leads to a diminished sensitivity of microglia towards CSF-1. Thus, the treatment should start as early as possible after glial culture preparation.
Collapse
Affiliation(s)
- Sabrina Hupp
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012, Bern, Switzerland.
| | - Asparouh I Iliev
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012, Bern, Switzerland.
| |
Collapse
|
112
|
Abstract
Microglia are resident macrophages of the CNS that are involved in its development, homeostasis and response to infection and damage. Microglial activation is a common feature of neurological disorders, and although in some instances this activation can be damaging, protective and regenerative functions of microglia have been revealed. The most prominent example of the regenerative functions is a role for microglia in supporting regeneration of myelin after injury, a process that is critical for axonal health and relevant to numerous disorders in which loss of myelin integrity is a prevalent feature, such as multiple sclerosis, Alzheimer disease and motor neuron disease. Although drugs that are intended to promote remyelination are entering clinical trials, the mechanisms by which remyelination is controlled and how microglia are involved are not completely understood. In this Review, we discuss work that has identified novel regulators of microglial activation - including molecular drivers, population heterogeneity and turnover - that might influence their pro-remyelination capacity. We also discuss therapeutic targeting of microglia as a potential approach to promoting remyelination.
Collapse
|
113
|
Brioschi S, Zhou Y, Colonna M. Brain Parenchymal and Extraparenchymal Macrophages in Development, Homeostasis, and Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:294-305. [PMID: 31907272 PMCID: PMC7034672 DOI: 10.4049/jimmunol.1900821] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/27/2019] [Indexed: 12/23/2022]
Abstract
Microglia are parenchymal macrophages of the CNS; as professional phagocytes they are important for maintenance of the brain's physiology. These cells are generated through primitive hematopoiesis in the yolk sac and migrate into the brain rudiment after establishment of embryonic circulation. Thereafter, microglia develop in a stepwise fashion, reaching complete maturity after birth. In the CNS, microglia self-renew without input from blood monocytes. Recent RNA-sequencing studies have defined a molecular signature for microglia under homeostasis. However, during disease, microglia undergo remarkable phenotypic changes, which reflect the acquisition of specialized functions tailored to the pathological context. In addition to microglia, the brain-border regions host populations of extraparenchymal macrophages with disparate origins and phenotypes that have recently been delineated. In this review we outline recent findings that provide a deeper understanding of both parenchymal microglia and extraparenchymal brain macrophages in homeostasis and during disease.
Collapse
Affiliation(s)
- Simone Brioschi
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| |
Collapse
|
114
|
Csf1 Deficiency Dysregulates Glial Responses to Demyelination and Disturbs CNS White Matter Remyelination. Cells 2019; 9:cells9010099. [PMID: 31906095 PMCID: PMC7017166 DOI: 10.3390/cells9010099] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/23/2019] [Accepted: 12/28/2019] [Indexed: 12/30/2022] Open
Abstract
Remyelination, a highly efficient central nervous system (CNS) regenerative process, is performed by oligodendrocyte progenitor cells (OPCs), which are recruited to the demyelination sites and differentiate into mature oligodendrocytes to form a new myelin sheath. Microglia, the specialized CNS-resident phagocytes, were shown to support remyelination through secretion of factors stimulating OPC recruitment and differentiation, and their pharmacological depletion impaired remyelination. Macrophage colony-stimulating factor (Csf1) has been implicated in the control of recruitment and polarization of microglia/macrophages in injury-induced CNS inflammation. However, it remains unclear how Csf1 regulates a glial inflammatory response to demyelination as well as axonal survival and new myelin formation. Here, we have investigated the effects of the inherent Csf1 deficiency in a murine model of remyelination. We showed that remyelination was severely impaired in Csf1-/- mutant mice despite the fact that reduction in monocyte/microglia accumulation affects neither the number of OPCs recruited to the demyelinating lesion nor their differentiation. We identified a specific inflammatory gene expression signature and found aberrant astrocyte activation in Csf1-/- mice. We conclude that Csf1-dependent microglia activity is essential for supporting the equilibrium between microglia and astrocyte pro-inflammatory vs. regenerative activation, demyelinated axons integration and, ultimately, reconstruction of damaged white matter.
Collapse
|
115
|
Healy LM, Yaqubi M, Ludwin S, Antel JP. Species differences in immune-mediated CNS tissue injury and repair: A (neuro)inflammatory topic. Glia 2019; 68:811-829. [PMID: 31724770 DOI: 10.1002/glia.23746] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/04/2019] [Accepted: 10/11/2019] [Indexed: 12/13/2022]
Abstract
Cells of the adaptive and innate immune systems in the brain parenchyma and in the meningeal spaces contribute to physiologic functions and disease states in the central nervous system (CNS). Animal studies have demonstrated the involvement of immune constituents, along with major histocompatibility complex (MHC) molecules, in neural development and rare genetic disorders (e.g., colony stimulating factor 1 receptor [CSF1R] deficiency). Genome wide association studies suggest a comparable role of the immune system in humans. Although the CNS can be the target of primary autoimmune disorders, no current experimental model captures all of the features of the most common human disorder placed in this category, multiple sclerosis (MS). Such features include spontaneous onset, environmental contributions, and a recurrent/progressive disease course in a genetically predisposed host. Numerous therapeutic interventions related to antigen and cytokine specific therapies have demonstrated effectiveness in experimental autoimmune encephalomyelitis (EAE), the animal model used to define principles underlying immune-mediated mechanisms in MS. Despite the similarities in the two diseases, most treatments used to ameliorate EAE have failed to translate to the human disease. As directly demonstrated in animal models and implicated by correlative studies in humans, adaptive and innate immune constituents within the systemic compartment and resident in the CNS contribute to the disease course of neurodegenerative and neurobehavioral disorders. The expanding knowledge of the molecular properties of glial cells provides increasing insights into species related variables. These variables affect glial bidirectional interactions with the immune system as well as their own production of "immune molecules" that mediate tissue injury and repair.
Collapse
Affiliation(s)
- Luke M Healy
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Moein Yaqubi
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Samuel Ludwin
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
116
|
Lassmann H. Pathology of inflammatory diseases of the nervous system: Human disease versus animal models. Glia 2019; 68:830-844. [PMID: 31605512 PMCID: PMC7065008 DOI: 10.1002/glia.23726] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022]
Abstract
Numerous recent studies have been performed to elucidate the function of microglia, macrophages, and astrocytes in inflammatory diseases of the central nervous system. Regarding myeloid cells a core pattern of activation has been identified, starting with the activation of resident homeostatic microglia followed by recruitment of blood borne myeloid cells. An initial state of proinflammatory activation is at later stages followed by a shift toward an‐anti‐inflammatory and repair promoting phenotype. Although this core pattern is similar between experimental models and inflammatory conditions in the human brain, there are important differences. Even in the normal human brain a preactivated microglia phenotype is evident, and there are disease specific and lesion stage specific differences in the contribution between resident and recruited myeloid cells and their lesion state specific activation profiles. Reasons for these findings reside in species related differences and in differential exposure to different environmental cues. Most importantly, however, experimental rodent studies on brain inflammation are mainly focused on autoimmune encephalomyelitis, while there is a very broad spectrum of human inflammatory diseases of the central nervous system, triggered and propagated by a variety of different immune mechanisms.
Collapse
Affiliation(s)
- Hans Lassmann
- Institut fur Hirnforschung, Medical University of Vienna, Wien, Austria
| |
Collapse
|
117
|
West PK, Viengkhou B, Campbell IL, Hofer MJ. Microglia responses to interleukin-6 and type I interferons in neuroinflammatory disease. Glia 2019; 67:1821-1841. [PMID: 31033014 DOI: 10.1002/glia.23634] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 04/07/2019] [Accepted: 04/10/2019] [Indexed: 01/03/2025]
Abstract
Microglia are the resident macrophages of the central nervous system (CNS). They are a heterogenous, exquisitely responsive, and highly plastic cell population, which enables them to perform diverse roles. They sense and respond to the local production of many different signals, including an assorted range of cytokines. Microglia respond strongly to interleukin-6 (IL-6) and members of the type I interferon (IFN-I) family, IFN-alpha (IFN-α), and IFN-beta (IFN-β). Although these cytokines are essential in maintaining homeostasis and for activating and regulating immune responses, their chronic production has been linked to the development of distinct human neurological diseases, termed "cerebral cytokinopathies." IL-6 and IFN-α have been identified as key mediators in the pathogenesis of neuroinflammatory disorders including neuromyelitis optica and Aicardi-Goutières syndrome, respectively, whereas IFN-β has an emerging role as a causal factor in age-associated cognitive decline. One of the key features that unites these diseases is the presence of highly reactive microglia. The current understanding is that microglia contribute to the development of cerebral cytokinopathies and represent an important therapeutic target. However, it remains to be resolved whether microglia have beneficial or detrimental effects. Here we review and discuss what is currently known about the microglial response to IL-6 and IFN-I, based on both animal models and clinical studies. Foundational knowledge regarding the microglial response to IL-6 and IFN-I is now being used to devise therapeutic strategies to ameliorate neuroinflammation and promote repair: either through targeting microglia, or by targeting the reduction of CNS levels or downstream biological pathways of IL-6 or IFN-I.
Collapse
Affiliation(s)
- Phillip K West
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Barney Viengkhou
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Iain L Campbell
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Markus J Hofer
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
118
|
Updating Neuroimmune Targets in Central Nervous System Dysfunction. Trends Pharmacol Sci 2019; 40:482-494. [DOI: 10.1016/j.tips.2019.04.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/15/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022]
|
119
|
Depletion of microglia ameliorates white matter injury and cognitive impairment in a mouse chronic cerebral hypoperfusion model. Biochem Biophys Res Commun 2019; 514:1040-1044. [DOI: 10.1016/j.bbrc.2019.05.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/06/2019] [Indexed: 02/07/2023]
|
120
|
Liu Y, Given KS, Dickson EL, Owens GP, Macklin WB, Bennett JL. Concentration-dependent effects of CSF1R inhibitors on oligodendrocyte progenitor cells ex vivo and in vivo. Exp Neurol 2019; 318:32-41. [PMID: 31029597 DOI: 10.1016/j.expneurol.2019.04.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 04/21/2019] [Accepted: 04/22/2019] [Indexed: 01/18/2023]
Abstract
Microglia are the principal resident immune cells in the central nervous system (CNS) and play important roles in CNS development, maintenance and repair. The survival and development of microglia depends on colony-stimulating factor 1 receptor (CSF1R), a member of the platelet-derived growth factor receptor (PDGFR) family of tyrosine kinases. Recently pharmacological CSF1R inhibition has been used to investigate the effects of microglial depletion in numerous animal models of CNS disease. However, the effects of CSF1R inhibitors on other cell types in the CNS remains incompletely characterized. In this report, we compared the effect of two commonly used CSF1R inhibitors, PLX5622 and PLX3397, on microglia and oligodendrocyte progenitor cell (OPC) numbers. In ex vivo cerebellar slices and adult mouse brain, both PLX compounds caused robust microglia loss; the kinetics of microglial depletion was more rapid with PLX5622. While high-doses of PLX5622 and PLX3397 reduced OPC number in primary cultures in vitro and ex vivo, low-doses of PLX5622 did not affect the number of OPCs or mature oligodendroglia in culture or in vivo. In adult mice, treatment with PLX5622 had no effect on OPC numbers for 7 days; however, a mild reduction was observed after 21 days in some CNS regions. In contrast, PLX3397 caused significant OPC loss after 7 days of treatment, despite only modest microglia depletion. Neither PLX compound had a remarkable effect on mature oligodendrocytes or myelin protein expression following long-term oral administration. Our results show that CSF1R inhibition with PLX5622 can selectively deplete microglia ex vivo and in vivo without affecting OPC number, demonstrating that microglia are not essential for OPC viability in ex vivo slice cultures or adult CNS tissues.
Collapse
Affiliation(s)
- Yiting Liu
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Katherine S Given
- Department of Cell & Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Erin L Dickson
- Department of Cell & Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Gregory P Owens
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Wendy B Macklin
- Department of Cell & Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA; Program in Neuroscience, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jeffrey L Bennett
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA; Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA; Program in Neuroscience, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
121
|
Kyle J, Wu M, Gourzi S, Tsirka SE. Proliferation and Differentiation in the Adult Subventricular Zone Are Not Affected by CSF1R Inhibition. Front Cell Neurosci 2019; 13:97. [PMID: 31001085 PMCID: PMC6454047 DOI: 10.3389/fncel.2019.00097] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/27/2019] [Indexed: 11/24/2022] Open
Abstract
Microglia are reported to have significant roles in regulating normal mammalian adult neurogenesis. There are two neurogenic niches in the adult mammal brain: the subgranular zone (SGZ) in the hippocampus, and the subventricular zone (SVZ), which makes up the lining of the lateral ventricles. While the microglia interactions on adult neurogenesis in the hippocampus have been characterized, the SVZ niche is not as well investigated. The SVZ niche is unique in that the newborn neurons migrate a much longer distance through multiple brain structures compared to newborn neurons in the hippocampus, making it more difficult to fully characterize how microglia influence this process. To examine the SVZ niche and migration pathway, we used the colony stimulating factor 1 receptor (CSF1R) antagonist PLX5622, which promotes brain wide microglia ablation. Microglia ablation resulted in no changes in the numbers of neural stem cells (NSCs), transient amplifying cells, and neuroblasts. Microglia ablation in the olfactory bulb (OB) was decreased compared to the SVZ. CSF1R inhibition had no effect on the ability of microglia to proliferate. Thus, our data suggest that microglia are not required for normal functioning SVZ adult neurogenesis.
Collapse
Affiliation(s)
- Jackson Kyle
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| | - Michelle Wu
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| | - Stefania Gourzi
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| | - Stella E Tsirka
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
122
|
Han J, Zhu K, Zhang X, Harris RA. Enforced microglial depletion and repopulation as a promising strategy for the treatment of neurological disorders. Glia 2019; 67:217-231. [PMID: 30378163 PMCID: PMC6635749 DOI: 10.1002/glia.23529] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/17/2018] [Accepted: 08/22/2018] [Indexed: 01/18/2023]
Abstract
Microglia are prominent immune cells in the central nervous system (CNS) and are critical players in both neurological development and homeostasis, and in neurological diseases when dysfunctional. Our previous understanding of the phenotypes and functions of microglia has been greatly extended by a dearth of recent investigations. Distinct genetically defined subsets of microglia are now recognized to perform their own independent functions in specific conditions. The molecular profiling of single microglial cells indicates extensively heterogeneous reactions in different neurological disorders, resulting in multiple potentials for crosstalk with other kinds of CNS cells such as astrocytes and neurons. In settings of neurological diseases it could thus be prudent to establish effective cell-based therapies by targeting entire microglial networks. Notably, activated microglial depletion through genetic targeting or pharmacological therapies within a suitable time window can stimulate replenishment of the CNS niche with new microglia. Additionally, enforced repopulation through provision of replacement cells also represents a potential means of exchanging dysfunctional with functional microglia. In each setting the newly repopulated microglia might have the potential to resolve ongoing neuroinflammation. In this review, we aim to summarize the most recent knowledge of microglia and to highlight microglial depletion and subsequent repopulation as a promising cell replacement therapy. Although glial cell replacement therapy is still in its infancy and future translational studies are still required, the approach is scientifically sound and provides new optimism for managing the neurotoxicity and neuroinflammation induced by activated microglia.
Collapse
Affiliation(s)
- Jinming Han
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Keying Zhu
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Xing‐Mei Zhang
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Robert A. Harris
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| |
Collapse
|
123
|
Wimmer I, Scharler C, Zrzavy T, Kadowaki T, Mödlagl V, Rojc K, Tröscher AR, Kitic M, Ueda S, Bradl M, Lassmann H. Microglia pre-activation and neurodegeneration precipitate neuroinflammation without exacerbating tissue injury in experimental autoimmune encephalomyelitis. Acta Neuropathol Commun 2019; 7:14. [PMID: 30704526 PMCID: PMC6357376 DOI: 10.1186/s40478-019-0667-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 01/26/2019] [Indexed: 12/26/2022] Open
Abstract
Human inflammatory or neurodegenerative diseases, such as progressive multiple sclerosis (MS), occur on a background of age-related microglia activation and iron accumulation as well as pre-existing neurodegeneration. Most experimental models for CNS diseases, however, are induced in rodents, which are naturally characterized by a homeostatic microglia phenotype, low cellular iron load and absence of neurodegeneration. Here, we show that naïve LEWzizi rats – Lewis rats with a zitter rat background – show a spontaneous phenotype partly mimicking the changes seen in human aging and particularly in the normal-appearing white and grey matter of patients with progressive MS. Using this model system, we further aimed to investigate (i) whether the acute monophasic MS model experimental autoimmune encephalomyelitis (EAE) transforms into chronic progressive disease and (ii) whether EAE-induced neuroinflammation and tissue damage aggravate on the LEWzizi background. We found that the pre-existing LEWzizi-specific pathology precipitated EAE-related neuroinflammation into forebrain areas, which are devoid of EAE lesions in normal Lewis rats. However, EAE-related tissue damage was neither modified by the LEWzizi-specific pathology nor did EAE-induced neuroinflammation modify the LEWzizi-related pathological process. Our data indicate that the interaction between pre-activated microglia and CD4+ autoreactive T cells during the induction and propagation of tissue damage in the CNS is limited.
Collapse
|
124
|
Funk KE, Klein RS. CSF1R antagonism limits local restimulation of antiviral CD8 + T cells during viral encephalitis. J Neuroinflammation 2019; 16:22. [PMID: 30704498 PMCID: PMC6354430 DOI: 10.1186/s12974-019-1397-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/02/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Microglia are resident macrophages of the central nervous system (CNS) locally maintained through colony-stimulating factor 1 receptor (CSF1R) signaling. Microglial depletion via CSF1R inactivation improves cognition in mouse models of neuroinflammation, but limits virologic control in the CNS of mouse models of neurotropic infections by unknown mechanisms. We hypothesize that CSF1R plays a critical role in myeloid cell responses that restrict viral replication and locally restimulate recruited antiviral T cells within the CNS. METHODS The impact of CSF1R signaling during West Nile virus infection was assessed in vivo using a mouse model of neurotropic infection. Pharmacological inactivation of CSF1R was achieved using PLX5622 prior to infection with virulent or attenuated strains of West Nile virus (WNV), an emerging neuropathogen. The subsequent effect of CSF1R antagonism on virologic control was assessed by measuring mortality and viral titers in the CNS and peripheral organs. Immune responses were assessed by flow cytometric-based phenotypic analyses of both peripheral and CNS immune cells. RESULTS Mice treated with CSF1R antagonist prior to infection exhibited higher susceptibility to lethal WNV infection and lack of virologic control in both the CNS and periphery. CSFR1 antagonism reduced B7 co-stimulatory signals on peripheral and CNS antigen-presenting cells (APCs) by depleting CNS cellular sources, which limited local reactivation of CNS-infiltrating virus-specific T cells and reduced viral clearance. CONCLUSIONS Our results demonstrate the impact of CSF1R antagonism on APC activation in the CNS and periphery and the importance of microglia in orchestrating the CNS immune response following neurotropic viral infection. These data will be an important consideration when assessing the benefit of CSF1R antagonism, which has been investigated as a therapeutic for neurodegenerative conditions, in which neuroinflammation is a contributing factor.
Collapse
Affiliation(s)
- Kristen E. Funk
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, MO 63110 USA
| | - Robyn S. Klein
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110 USA
- Department of Neurosciences, Washington University School of Medicine, Saint Louis, MO 63110 USA
| |
Collapse
|
125
|
Wlodarczyk A, Benmamar-Badel A, Cédile O, Jensen KN, Kramer I, Elsborg NB, Owens T. CSF1R Stimulation Promotes Increased Neuroprotection by CD11c+ Microglia in EAE. Front Cell Neurosci 2019; 12:523. [PMID: 30687013 PMCID: PMC6335250 DOI: 10.3389/fncel.2018.00523] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/14/2018] [Indexed: 12/27/2022] Open
Abstract
Microglia are resident immune cells of the central nervous system. Their development and maintenance depend on stimulation of Colony Stimulating Factor-1 receptor (CSF1R). Microglia play an important role in neurodevelopment and a population of microglia that expresses the complement receptor CD11c is critical for primary myelination. This population is virtually absent in the healthy adult brain but increases dramatically upon neuroinflammatory conditions, and these microglia are suggested to play a protective role in central nervous system (CNS) diseases. To date, the molecular trigger for their expansion is unknown. Here we showed that stimulation of CSF1R by either of its ligands, CSF1 and interleukin (IL)-34, can induce expansion of CD11c+ microglia. In addition, such stimulation resulted in amelioration of EAE symptoms and decreased demyelination. Treatment with CSF1R ligands also induced expression of the chemokine CCL2, and we showed that experimental overexpression of CCL2 in the brain led to a dramatic increase of CD11c+ microglia, independent of CCR2. Moreover, this led to elevated CSF1 expression, suggesting a positive feedback loop between CSF1R and CCL2. These data provide new insights to microglia biology and open new perspectives for modulating microglial activity in neuroinflammatory diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| | - Anouk Benmamar-Badel
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| | - Oriane Cédile
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Kirstine Nolling Jensen
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Isabella Kramer
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Nick Boe Elsborg
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| |
Collapse
|