Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Int J Radiat Oncol Biol Phys ; 119(3): 912-923, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38142839

ABSTRACT

PURPOSE: Cranial irradiation induces healthy tissue damage that can lead to neurocognitive complications, negatively affecting patient quality of life. One damage indicator associated with cognitive impairment is loss of neuronal spine density. We previously demonstrated that irradiation-mediated spine loss is microglial complement receptor 3 (CR3) and sex dependent. We hypothesized that these changes are associated with late-delayed cognitive deficits and amenable to pharmacologic intervention. METHODS AND MATERIALS: Our model of cranial irradiation (acute, 10 Gy gamma) used male and female CR3-wild type and CR3-deficient Thy-1 YFP mice of C57BL/6 background. Forty-five days after irradiation and behavioral testing, we quantified spine density and markers of microglial reactivity in the hippocampal dentate gyrus. In a separate experiment, male Thy-1 YFP C57BL/6 mice were treated with leukadherin-1, a modulator of CR3 function. RESULTS: We found that male mice demonstrate irradiation-mediated spine loss and cognitive deficits but that female and CR3 knockout mice do not. These changes were associated with greater reactivity of microglia in male mice. Pharmacologic manipulation of CR3 with LA1 prevented spine loss and cognitive deficits in irradiated male mice. CONCLUSIONS: This work improves our understanding of irradiation-mediated mechanisms and sex dependent responses and may help identify novel therapeutics to reduce irradiation-induced cognitive decline and improve patient quality of life.


Subject(s)
Cognitive Dysfunction , Cranial Irradiation , Dendritic Spines , Mice, Inbred C57BL , Microglia , Animals , Male , Female , Mice , Dendritic Spines/drug effects , Dendritic Spines/radiation effects , Cranial Irradiation/adverse effects , Microglia/drug effects , Microglia/radiation effects , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Macrophage-1 Antigen/metabolism , Mice, Knockout , Dentate Gyrus/drug effects , Dentate Gyrus/radiation effects , Sex Factors , Organic Chemicals
3.
Pharmaceutics ; 15(7)2023 Jun 24.
Article in English | MEDLINE | ID: mdl-37513996

ABSTRACT

Alzheimer's disease (AD) is a chronic neurodegenerative disorder that involves strong inflammatory components. Aberrant and prolonged inflammation in the CNS is thought to contribute to the development of the pathology. The use of single cytokine approaches to curb or leverage inflammatory mechanisms for disease modifying benefit has often resulted in conflicting data. Furthermore, these treatments were usually delivered locally into the CNS parenchyma, complicating translational efforts. To overcome these hurdles, we tested the use of glatiramer acetate (GA) in reducing amyloid beta (Aß) plaque pathology in the 5xFAD model of AD. GA immunizations were begun at the ages of 2.5 months, 5.5 months, and 8.5 months, and GA was delivered weekly for 8 weeks. While previous data describe potential benefits of GA immunization in decreasing Aß levels in murine models of AD, we found modest decreases in Aß levels if given during the development of pathology but, surprisingly, found increased Aß levels if GA was administered at later stages. The impact of GA treatment was only significant for female mice. Furthermore, we observed no changes between microglial uptake of plaque, CD11c immunopositivity of microglia, or levels of TMEM119 and P2Ry12 on microglia. Overall, these data warrant exercising caution when aiming to repurpose GA for AD.

4.
J Neuroinflammation ; 19(1): 38, 2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35130912

ABSTRACT

BACKGROUND: Alzheimer's disease is the leading cause of dementia worldwide. TAM receptor tyrosine kinases (Tyro3, Axl, MerTK) are known for their role in engagement of phagocytosis and modulation of inflammation, and recent evidence suggests a complex relationship between Axl, Mer, and microglial phagocytosis of amyloid plaques in AD. Gas6, the primary CNS TAM ligand, reduces neuroinflammation and improves outcomes in murine models of CNS disease. Therefore, we hypothesized that AAV-mediated overexpression of Gas6 would alleviate plaque pathology, reduce neuroinflammation, and improve behavior in the APP/PS1 model of Alzheimer's disease. METHODS: Adeno-associated viral vectors were used to overexpress Gas6 in the APP/PS1 model of Alzheimer's disease. Nine-month-old male and female APP/PS1 and nontransgenic littermates received bilateral stereotactic hippocampal injections of AAV-Gas6 or AAV-control, which expresses a non-functional Gas6 protein. One month after injections, mice underwent a battery of behavioral tasks to assess cognitive function and brains were processed for immunohistochemical and transcriptional analyses. RESULTS: Gas6 overexpression reduced plaque burden in male APP/PS1 mice. However, contrary to our hypothesis, Gas6 increased pro-inflammatory microglial gene expression and worsened contextual fear conditioning compared to control-treated mice. Gas6 overexpression appeared to have no effect on phagocytic mechanisms in vitro or in vivo as measured by CD68 immunohistochemistry, microglial methoxy-04 uptake, and primary microglial uptake of fluorescent fibrillar amyloid beta. CONCLUSION: Our data describes a triad of worsened behavior, reduced plaque number, and an increase in proinflammatory signaling in a sex-specific manner. While Gas6 has historically induced anti-inflammatory signatures in the peripheral nervous system, our data suggest an alternative, proinflammatory role in the context of Alzheimer's disease pathology.


Subject(s)
Alzheimer Disease , Intercellular Signaling Peptides and Proteins , Plaque, Amyloid , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Female , Inflammation/complications , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/pathology , Presenilin-1/genetics
5.
Front Neurosci ; 15: 758677, 2021.
Article in English | MEDLINE | ID: mdl-34744620

ABSTRACT

Neuroinflammation driven by the accumulation of amyloid ß (Aß) can lead to neurofibrillary tangle formation in Alzheimer's Disease (AD). To test the hypothesis that an anti-inflammatory immunomodulatory agent might have beneficial effects on amyloid and tau pathology, as well as microglial phenotype, we evaluated glatiramer acetate (GA), a multiple sclerosis drug thought to bias type 2 helper T (Th2) cell responses and alternatively activate myeloid cells. We administered weekly subcutaneous injections of GA or PBS to 15-month-old 3xTg AD mice, which develop both amyloid and tau pathology, for a period of 8 weeks. We found that subcutaneous administration of GA improved behavioral performance in novel object recognition and decreased Aß plaque in the 3xTg AD mice. Changes in tau phosphorylation were mixed with specific changes in phosphoepitopes seen in immunohistochemistry but not observed in western blot. In addition, we found that there was a trend toward increased microglia complexity in 3xTg mice treated with GA, suggesting a shift toward homeostasis. These findings correlated with subtle changes in the microglial transcriptome, in which the most striking difference was the upregulation of Dcstamp. Lastly, we found no evidence of changes in proportions of major helper T cell (Th) subtypes in the periphery. Overall, our study provides further evidence for the benefits of immunomodulatory therapies that alter the adaptive immune system with the goal of modifying microglia responses for the treatment of Alzheimer's Disease.

6.
Front Neurosci ; 14: 441, 2020.
Article in English | MEDLINE | ID: mdl-32528242

ABSTRACT

Chronic neuroinflammation has long been hypothesized to be involved in Alzheimer's Disease (AD) progression. Previous research suggests that both anti-inflammatory and inflammatory microglia ameliorate amyloid pathology, but the latter worsen tau pathology. In this study, we sought to determine whether induction of arginase-1 positive microglia with the anti-inflammatory cytokine IL-4 modulates pathology in the 3xTg mouse model of AD. Our findings indicate that a single intracranial IL-4 injection positively modulated performance of 3xTg AD mice in a Novel Object Recognition task, and locally increased the levels of arginase-1 positive myeloid cells when assessed one-week post injection. Furthermore, immunohistochemical analysis revealed decreased tau phosphorylation in IL-4 injected animals; however, we were not able to detect significant changes in tau phosphorylation utilizing Western blot. Lastly, IL-4 injection did not appear to cause significant changes in amyloid ß load. In conclusion, acute intracranial IL-4 led to some positive benefits in the 3xTg mouse model of AD. Although more work remains, these results support therapeutic strategies aimed at modifying microglial activation states in neurodegenerative diseases.

7.
Brain Behav Immun Health ; 4: 100057, 2020 Apr.
Article in English | MEDLINE | ID: mdl-34589843

ABSTRACT

Microglia, the resident immune cells of the central nervous system (CNS), play multiple roles in maintaining CNS homeostasis and mediating tissue repair, including proliferating in response to brain injury and disease. Cranial irradiation (CI), used for the treatment of brain tumors, has a long-lasting anti-proliferative effect on a number of cell types in the brain, including oligodendrocyte progenitor and neural progenitor cells; however, the effect of CI on CNS-resident microglial proliferation is not well characterized. Using a sterile cortical needle stab injury model in mice, we found that the ability of CNS-resident microglia to proliferate in response to injury was impaired by prior CI, in a dose-dependent manner, and was nearly abolished by a 20 â€‹Gy dose. Similarly, in a metastatic tumor model, prior CI (20 â€‹Gy) reduced microglial proliferation in response to tumor growth. The effect of irradiation was long-lasting; 20 â€‹Gy CI 6 months prior to stab injury significantly impaired microglial proliferation. We also investigated how stab and/or irradiation impacted levels of P2Y12R, CD68, CSF1, IL-34 and CSF1R, factors involved in the brain's normal response to injury. P2Y12R, CD68, CSF1, and IL-34 expression were altered by stab similarly in irradiated mice and controls; however, CSF1R was differentially affected. qRT-PCR and flow cytometry analyses demonstrated that CI reduced overall Csf1r mRNA levels and microglial specific CSF1R protein expression, respectively. Interestingly, Csf1r mRNA levels increased after injury in unirradiated controls; however, Csf1r levels were persistently decreased in irradiated mice, and did not increase in response to stab. Together, our data demonstrate that CI leads to a significant and lasting impairment of microglial proliferation, possibly through a CSF1R-mediated mechanism.

8.
Life Sci Space Res (Amst) ; 27: 89-98, 2020 Nov.
Article in English | MEDLINE | ID: mdl-34756235

ABSTRACT

Space radiation is comprised of highly charged ions (HZE particles) and protons that are able to pass through matter and cause radiation-induced injury, including neuronal damage and degeneration, glial activation, and oxidative stress. Previous work demonstrated a worsening of Alzheimer's disease pathology in the APP/PS1 transgenic mouse model, however effects of space radiation on tau pathology have not been studied. To determine whether tau pathology is altered by HZE particle or proton irradiation, we exposed 3xTg mice, which acquire both amyloid plaque and tau pathology with age, to iron, silicon, or solar particle event (SPE) irradiation at 9 months of age and evaluated behavior and brain pathology at 16 months of age. We found no differences in performance in fear conditioning and novel object recognition tasks between groups of mice exposed to sham, iron (10 and 100 cGy), silicon (10 and 100 cGy), or solar particle event radiation (200 cGy), though female mice had higher freezing responses than males. 200 cGy SPE irradiated female mice had fewer plaques than sham-irradiated females but had no differences in tau pathology. Overall, females had worse amyloid and tau pathology at 16 months of age and demonstrated a reduced neuroinflammatory gene expression response to radiation. These findings uncover differences between mouse models following radiation injury and corroborate prior reports of sex differences within the 3xTg mouse model.


Subject(s)
Alzheimer Disease , Alzheimer Disease/genetics , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , Animals , Female , Male , Mice , Mice, Transgenic , Presenilin-1 , tau Proteins
9.
J Neuroinflammation ; 16(1): 261, 2019 Dec 10.
Article in English | MEDLINE | ID: mdl-31822279

ABSTRACT

BACKGROUND: Neuroinflammation is thought to contribute to the pathogenesis of Alzheimer's disease (AD), yet numerous studies have demonstrated a beneficial role for neuroinflammation in amyloid plaque clearance. We have previously shown that sustained expression of IL-1ß in the hippocampus of APP/PS1 mice decreases amyloid plaque burden independent of recruited CCR2+ myeloid cells, suggesting resident microglia as the main phagocytic effectors of IL-1ß-induced plaque clearance. To date, however, the mechanisms of IL-1ß-induced plaque clearance remain poorly understood. METHODS: To determine whether microglia are involved in IL-1ß-induced plaque clearance, APP/PS1 mice induced to express mature human IL-1ß in the hippocampus via adenoviral transduction were treated with the Aß fluorescent probe methoxy-X04 (MX04) and microglial internalization of fibrillar Aß (fAß) was analyzed by flow cytometry and immunohistochemistry. To assess microglial proliferation, APP/PS1 mice transduced with IL-1ß or control were injected intraperitoneally with BrdU and hippocampal tissue was analyzed by flow cytometry. RNAseq analysis was conducted on microglia FACS sorted from the hippocampus of control or IL-1ß-treated APP/PS1 mice. These microglia were also sorted based on MX04 labeling (MX04+ and MX04- microglia). RESULTS: Resident microglia (CD45loCD11b+) constituted > 70% of the MX04+ cells in both Phe- and IL-1ß-treated conditions, and < 15% of MX04+ cells were recruited myeloid cells (CD45hiCD11b+). However, IL-1ß treatment did not augment the percentage of MX04+ microglia nor the quantity of fAß internalized by individual microglia. Instead, IL-1ß increased the total number of MX04+ microglia in the hippocampus due to IL-1ß-induced proliferation. In addition, transcriptomic analyses revealed that IL-1ß treatment was associated with large-scale changes in the expression of genes related to immune responses, proliferation, and cytokine signaling. CONCLUSIONS: These studies show that IL-1ß overexpression early in amyloid pathogenesis induces a change in the microglial gene expression profile and an expansion of microglial cells that facilitates Aß plaque clearance.


Subject(s)
Cellular Reprogramming/physiology , Interleukin-1beta/biosynthesis , Microglia/metabolism , Plaque, Amyloid/metabolism , Transcription, Genetic/physiology , Transcriptome/physiology , Animals , Cell Proliferation/physiology , Female , Interleukin-1beta/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/genetics
10.
Sci Rep ; 9(1): 18899, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31827187

ABSTRACT

Cranial irradiation is the main therapeutic treatment for primary and metastatic malignancies in the brain. However, cranial radiation therapy produces long-term impairment in memory, information processing, and attention that contribute to a decline in quality of life. The hippocampal neural network is fundamental for proper storage and retrieval of episodic and spatial memories, suggesting that hippocampal signaling dysfunction could be responsible for the progressive memory deficits observed following irradiation. Previous rodent studies demonstrated that irradiation induces significant loss in dendritic spine number, alters spine morphology, and is associated with behavioral task deficits. Additionally, the literature suggests a common mechanism in which synaptic elimination via microglial-mediated phagocytosis is complement dependent and associated with cognitive impairment in aging as well as disease. We demonstrate sexual dimorphisms in irradiation-mediated alterations of microglia activation markers and dendritic spine density. Further, we find that the significant dendritic spine loss observed in male mice following irradiation is microglia complement receptor 3 (CR3)-dependent. By identifying sex-dependent cellular and molecular factors underlying irradiation-mediated spine loss, therapies can be developed to counteract irradiation-induced cognitive decline and improve patient quality of life.


Subject(s)
Cranial Irradiation , Dendritic Spines/radiation effects , Hippocampus/radiation effects , Microglia/radiation effects , Receptors, Complement/metabolism , Animals , Cell Shape/radiation effects , Dendritic Spines/metabolism , Dendritic Spines/pathology , Female , Hippocampus/pathology , Male , Mice , Mice, Knockout , Microglia/pathology , Receptors, Complement/genetics , Sex Factors
11.
J Neuroinflammation ; 16(1): 74, 2019 Apr 05.
Article in English | MEDLINE | ID: mdl-30953557

ABSTRACT

Neuroinflammation is considered one of the cardinal features of Alzheimer's disease (AD). Neuritic plaques composed of amyloid ß and neurofibrillary tangle-laden neurons are surrounded by reactive astrocytes and microglia. Exposure of microglia, the resident myeloid cell of the CNS, to amyloid ß causes these cells to acquire an inflammatory phenotype. While these reactive microglia are important to contain and phagocytose amyloid plaques, their activated phenotype impacts CNS homeostasis. In rodent models, increased neuroinflammation promoted by overexpression of proinflammatory cytokines can cause an increase in hyperphosphorylated tau and a decrease in hippocampal function. The peripheral immune system can also play a detrimental or beneficial role in CNS inflammation. Systemic inflammation can increase the risk of developing AD dementia, and chemokines released directly by microglia or indirectly by endothelial cells can attract monocytes and T lymphocytes to the CNS. These peripheral immune cells can aid in amyloid ß clearance or modulate microglia responses, depending on the cell type. As such, several groups have targeted the peripheral immune system to modulate chronic neuroinflammation. In this review, we focus on the interplay of immunomodulating factors and cell types that are being investigated as possible therapeutic targets for the treatment or prevention of AD.


Subject(s)
Alzheimer Disease , Cytokines/metabolism , Microglia/physiology , Myeloid Cells/physiology , Alzheimer Disease/complications , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Animals , Cell Communication/physiology , Encephalitis/etiology , Humans
12.
Glia ; 66(4): 846-861, 2018 04.
Article in English | MEDLINE | ID: mdl-29288597

ABSTRACT

Ionizing radiation (IR) is commonly used to treat central nervous system (CNS) cancers and metastases. While IR promotes remission, frequent side effects including impaired cognition and white matter loss occur following treatment. Fractionation is used to minimize these CNS late side effects, as it reduces IR effects in differentiated normal tissue, but not rapidly proliferating normal or tumor tissue. However, side effects occur even with the use of fractionated paradigms. Oligodendrocyte progenitor cells (OPCs) are a proliferative population within the CNS affected by radiation. We hypothesized that fractionated radiation would lead to OPC loss, which could contribute to the delayed white matter loss seen after radiation exposure. We found that fractionated IR induced a greater early loss of OPCs than an equivalent single dose exposure. Furthermore, OPC recovery was impaired following fractionated IR. Finally, reduced OPC differentiation and mature oligodendrocyte numbers occurred in single dose and fractionated IR paradigms. This work demonstrates that fractionation does not spare normal brain tissue and, importantly, highlights the sensitivity of OPCs to fractionated IR, suggesting that fractionated schedules may promote white matter dysfunction, a point that should be considered in radiotherapy.


Subject(s)
Dose Fractionation, Radiation , Oligodendrocyte Precursor Cells/radiation effects , Radiation Tolerance , Animals , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain/radiation effects , Bromodeoxyuridine , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cesium Radioisotopes , Dose-Response Relationship, Drug , Female , Glutathione S-Transferase pi/genetics , Glutathione S-Transferase pi/metabolism , Immunohistochemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice, Transgenic , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/pathology , Radiation Tolerance/drug effects , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Recovery of Function , Sex Characteristics , Tamoxifen/pharmacology
13.
Radiat Res ; 186(6): 614-623, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27905869

ABSTRACT

Understanding the dose-toxicity profile of radiation is critical when evaluating potential health risks associated with natural and man-made sources in our environment. The purpose of this study was to evaluate the effects of low-dose whole-body high-energy charged (HZE) iron (Fe) ions and low-energy gamma exposure on proliferation and differentiation of adult-born neurons within the dentate gyrus of the hippocampus, cells deemed to play a critical role in memory regulation. To determine the dose-response characteristics of the brain to whole-body Fe-ion vs. gamma-radiation exposure, C57BL/6J mice were irradiated with 1 GeV/n Fe ions or a static 137Cs source (0.662 MeV) at doses ranging from 0 to 300 cGy. The neurogenesis was analyzed at 48 h and one month postirradiation. These experiments revealed that whole-body exposure to either Fe ions or gamma radiation leads to: 1. An acute decrease in cell division within the dentate gyrus of the hippocampus, detected at doses as low as 30 and 100 cGy for Fe ions and gamma radiation, respectively; and 2. A reduction in newly differentiated neurons (DCX immunoreactivity) at one month postirradiation, with significant decreases detected at doses as low as 100 cGy for both Fe ions and gamma rays. The data presented here contribute to our understanding of brain responses to whole-body Fe ions and gamma rays and may help inform health-risk evaluations related to systemic exposure during a medical or radiologic/nuclear event or as a result of prolonged space travel.


Subject(s)
Gamma Rays/adverse effects , Iron/adverse effects , Neurogenesis/radiation effects , Whole-Body Irradiation/adverse effects , Animals , Dose-Response Relationship, Radiation , Doublecortin Protein , Male , Mice , Mice, Inbred C57BL , Time Factors
14.
Int J Radiat Oncol Biol Phys ; 96(2): 449-457, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27478169

ABSTRACT

PURPOSE: To determine the late effects of fractionated versus single-dose cranial radiation on murine white matter. METHODS AND MATERIALS: Mice were exposed to 0 Gy, 6 × 6 Gy, or 1 × 20 Gy cranial irradiation at 10 to 12 weeks of age. Endpoints were assessed through 18 months from exposure using immunohistochemistry, electron microscopy, and electrophysiology. RESULTS: Weight gain was temporarily reduced after irradiation; greater loss was seen after single versus fractionated doses. Oligodendrocyte progenitor cells were reduced early and late after both single and fractionated irradiation. Both protocols also increased myelin g-ratio, reduced the number of nodes of Ranvier, and promoted a shift in the proportion of small, unmyelinated versus large, myelinated axon fibers. CONCLUSIONS: Fractionation does not adequately spare normal white matter from late radiation side effects.


Subject(s)
Cell Lineage/radiation effects , Cranial Irradiation/adverse effects , Dose Fractionation, Radiation , Oligodendroglia/radiation effects , Weight Gain/radiation effects , White Matter/radiation effects , Animals , Cells, Cultured , Dose-Response Relationship, Radiation , Mice , Oligodendroglia/pathology , Organ Sparing Treatments/methods , Organs at Risk/radiation effects , Radiation Dosage , Radiation Protection/methods , White Matter/pathology
15.
J Neuroinflammation ; 13: 30, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26842770

ABSTRACT

BACKGROUND: Cranial radiotherapy is used to treat tumors of the central nervous system (CNS), as well as non-neoplastic conditions such as arterio-venous malformations; however, its use is limited by the tolerance of adjacent normal CNS tissue, which can lead to devastating long-term sequelae for patients. Despite decades of research, the underlying mechanisms by which radiation induces CNS tissue injury remain unclear. Neuroinflammation and immune cell infiltration are a recognized component of the CNS radiation response; however, the extent and mechanisms by which bone marrow-derived (BMD) immune cells participate in late radiation injury is unknown. Thus, we set out to better characterize the response and tested the hypothesis that C-C chemokine receptor type 2 (CCR2) signaling was required for myeloid cell recruitment following brain irradiation. METHODS: We used young adult C57BL/6 male bone marrow chimeric mice created with donor mice that constitutively express enhanced green fluorescent protein (eGFP). The head was shielded to avoid brain radiation exposure during chimera construction. Radiation dose and time response studies were conducted in wild-type chimeras, and additional experiments were performed with chimeras created using donor marrow from CCR2 deficient, eGFP-expressing mice. Infiltrating eGFP+ cells were identified and quantified using immunofluorescent microscopy. RESULTS: Brain irradiation resulted in a dose- and time-dependent infiltration of BMD immune cells (predominately myeloid) that began at 1 month and persisted until 6 months following ≥15 Gy brain irradiation. Infiltration was limited to areas that were directly exposed to radiation. CCR2 signaling loss resulted in decreased numbers of infiltrating cells at 6 months that appeared to be restricted to cells also expressing major histocompatibility complex class II molecules. CONCLUSIONS: The potential roles played by infiltrating immune cells are of current importance due to increasing interest in immunotherapeutic approaches for cancer treatment and a growing clinical interest in survivorship and quality of life issues. Our findings demonstrate that injury from brain radiation facilitates a dose- and time-dependent recruitment of BMD cells that persists for at least 6 months and, in the case of myeloid cells, is dependent on CCR2 signaling.


Subject(s)
Brain Injuries/etiology , Brain Injuries/pathology , Myeloid Cells/radiation effects , Radiation Injuries/complications , Receptors, CCR2/metabolism , Signal Transduction/radiation effects , Animals , Bone Marrow Transplantation , Calcium-Binding Proteins/metabolism , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Radiation , Gene Expression Regulation/radiation effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Myosin Heavy Chains/metabolism , Neutrophil Infiltration/radiation effects , Radiation Chimera/physiology , Receptors, CCR2/genetics , Time Factors
16.
Dev Neurobiol ; 76(5): 533-50, 2016 May.
Article in English | MEDLINE | ID: mdl-26243376

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated member of the basic-helix-loop-helix/PER-ARNT-SIM(PAS) transcription factor superfamily that also mediates the toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Increasing evidence suggests that AhR influences the development of many tissues, including the central nervous system. Our previous studies suggest that sustained AhR activation by TCDD and/or AhR deletion disrupts cerebellar granule neuron precursor (GNP) development. In the current study, to determine whether endogenous AhR controls GNP development in a cell-autonomous manner, we created a GNP-specific AhR deletion mouse, AhR(fx/fx) /Math1(CRE/+) (AhR CKO). Selective AhR deletion in GNPs produced abnormalities in proliferation and differentiation. Specifically, fewer GNPs were engaged in S-phase, as demonstrated by ∼25% reductions in thymidine (in vitro) and Bromodeoxyuridine (in vivo) incorporation. Furthermore, total granule neuron numbers in the internal granule layer at PND21 and PND60 were diminished in AhR conditional knockout (CKO) mice compared with controls. Conversely, differentiation was enhanced, including ∼40% increase in neurite outgrowth and 50% increase in GABARα6 receptor expression in deletion mutants. Our results suggest that AhR activity plays a role in regulating granule neuron number and differentiation, possibly by coordinating this GNP developmental transition. These studies provide novel insights for understanding the normal roles of AhR signaling during cerebellar granule cell neurogenesis and may have important implications for the effects of environmental factors in cerebellar dysgenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cerebellum/growth & development , Cerebellum/physiopathology , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Receptors, Aryl Hydrocarbon/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Count , Cell Proliferation , Cells, Cultured , Cerebellum/pathology , Mice, Inbred C57BL , Mice, Knockout , Neurites/pathology , Neurites/physiology , Neurons/pathology , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Receptors, GABA-A/metabolism
17.
J Neuroinflammation ; 12: 203, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26538310

ABSTRACT

BACKGROUND: Neuroinflammation has long been considered a driver of Alzheimer's disease progression. However, experiments developed to explore the interaction between neuroinflammation and Alzheimer's disease (AD) pathology showed a surprising reduction in amyloid beta (Aß) plaque deposition. We sought to understand this unexpected outcome by examining microglia phenotypes during chronic neuroinflammation. METHODS: Using an adeno-associated virus vector carrying hIL-1ß cDNA, inflammation was induced in one hippocampus of 8-month-old amyloid precursor protein (APP)/PS1 mice for 4 weeks, while the other hemisphere received control injections. Bone marrow chimeras and staining analysis were used to identify the origins and types of immune cells present during sustained inflammation. Arginase 1 (Arg1) and inducible nitric oxide synthase (iNOS) immunoreactivity were used as markers of alternatively activated and classically activated cells, respectively, and changes in cellular uptake of Aß by Arg1+ or iNOS+ microglia was demonstrated by confocal microscopy. To determine if an anti-inflammatory phenotype was present during neuroinflammation, RNA was extracted on flow-sorted microglia and rt-PCR was performed. Interleukin-4 injection was used to induce alternatively activated cells, whereas a minipump and intrahippocampal cannula was used to deliver an interleukin (IL)-4Rα antibody to block the induction of Arg1+ cells in the setting of sustained IL-1ß expression. RESULTS: We observed a robust upregulation of centrally derived Arg1+ microglia present only in the inflamed hemisphere. Furthermore, in the inflamed hemisphere, greater numbers of Arg1+ microglia contained Aß when compared to iNOS+ microglia. RNA isolated from flow-sorted microglia from the inflamed hemisphere demonstrated elevation of mRNA species consistent with alternative activation as well as neuroprotective genes such as BDNF and IGF1. To explore if Arg1+ microglia mediated plaque reduction, we induced Arg1+ microglia with IL-4 and observed significant plaque clearance. Moreover, when we reduced Arg1+ microglia induction in the context of neuroinflammation using an anti-IL-4Rα antibody delivered via intrahippocampal cannula, we observed a clear correlation between numbers of Arg1+ microglia and plaque reduction. CONCLUSIONS: Together, these findings suggest that Arg1+ microglia are involved in Aß plaque reduction during sustained, IL-1ß-dependent neuroinflammation, opening up possible new avenues for immunomodulatory therapy of AD.


Subject(s)
Arginase/genetics , Inflammation/pathology , Interleukin-1beta/metabolism , Microglia/enzymology , Plaque, Amyloid/pathology , Adenoviridae/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , DNA, Complementary/genetics , Hippocampus/metabolism , Interleukin-1/genetics , Interleukin-4 Receptor alpha Subunit/immunology , Macrophage Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/biosynthesis , Presenilin-1/genetics
18.
Neurotoxicology ; 50: 131-41, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26283213

ABSTRACT

Methamphetamine (MA) is a potent, highly addictive psychostimulant abused by millions of people worldwide. MA induces neurotoxicity, damaging striatal dopaminergic terminals, and neuroinflammation, with striatal glial activation leading to pro-inflammatory cytokine and reactive oxygen species production. It is unclear whether MA-induced neuroinflammation contributes to MA-induced neurotoxicity. In the current study, we examined the linkage between the time course and dose response of MA-induced neurotoxicity and neuroinflammation. Adult male mice underwent a binge dosing regimen of four injections given every 2h with doses of 2, 4, 6, or 8 mg/kg MA per injection, and were sacrificed after 1, 3, 7, or 14 days. Binge MA treatment dose-dependently caused hyperthermia and induced hypoactivity after one day, though activity returned to control levels within one week. Striatal dopamine (DA) was diminished one day after treatment with at least 4 mg/kg MA, while DA turnover rates peaked after seven days. Although striatal tyrosine hydroxylase and DA transporter levels were also decreased one day after treatment with at least 4 mg/kg MA, they trended toward recovery by day 14. All doses of MA activated striatal glia within one day. While astrocyte activation persisted, microglial activation was attenuated over the two weeks of the study. These findings help clarify the relationship between MA-induced neuroinflammation and neurotoxicity, particularly regarding their temporal and dose-specific dynamics.


Subject(s)
Calcium-Binding Proteins/metabolism , Central Nervous System Stimulants/toxicity , Encephalitis/chemically induced , Methamphetamine/toxicity , Microfilament Proteins/metabolism , Neurotoxicity Syndromes/etiology , Animals , Biogenic Monoamines/metabolism , Body Temperature/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/metabolism , Dose-Response Relationship, Drug , Electrochemical Techniques , Encephalitis/physiopathology , Glial Fibrillary Acidic Protein/metabolism , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neurotoxicity Syndromes/physiopathology , Time Factors , Tyrosine 3-Monooxygenase/metabolism
19.
Front Immunol ; 5: 594, 2014.
Article in English | MEDLINE | ID: mdl-25477883
20.
J Neuroinflammation ; 11: 98, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24889886

ABSTRACT

The concept of multiple macrophage activation states is not new. However, extending this idea to resident tissue macrophages, like microglia, has gained increased interest in recent years. Unfortunately, the research on peripheral macrophage polarization does not necessarily translate accurately to their central nervous system (CNS) counterparts. Even though pro- and anti-inflammatory cytokines can polarize microglia to distinct activation states, the specific functions of these states is still an area of intense debate. This review examines the multiple possible activation states microglia can be polarized to. This is followed by a detailed description of microglial polarization and the functional relevance of this process in both acute and chronic CNS disease models described in the literature. Particular attention is given to utilizing M2 microglial polarization as a potential therapeutic option in treating diseases.


Subject(s)
Encephalitis/pathology , Microglia/physiology , Animals , Central Nervous System/immunology , Central Nervous System/pathology , Humans , Macrophage Activation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...