Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
2.
Mol Neurodegener ; 18(1): 31, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37143090

ABSTRACT

BACKGROUND: Abnormal accumulation of amyloid beta peptide (Aß) in the brain induces a cascade of pathological changes in Alzheimer's disease (AD), and inhibiting BACE1, which is required for Aß generation, is therefore being explored for the treatment of AD by reducing Aß accumulation. As Bace1 knockout mice exhibit increased number of reactive astrocytes and AD brains have reactive astrocytes that surround amyloid plaques, we investigated the role of BACE1 in astrocytes and determined whether BACE1 regulates astrocytic functions. METHODS: We conducted unbiased single cell RNA-seq (scRNA-seq) using purified astrocytes from Bace1 KO mice and wild type control littermates. Similar scRNA-seq was also conducted using AD mice with conditional deletion of Bace1 in the adult stage (5xFAD;Bace1fl/fl;UBC-creER compared to 5xFAD;Bace1fl/fl controls). We compared the transcriptomes of astrocyte and reactive astrocyte clusters and identified several differentially expressed genes, which were further validated using Bace1 KO astrocyte cultures. Mice with astrocyte-specific Bace1 knockout in 5xFAD background were used to compare amyloid deposition. Mechanistic studies using cultured astrocytes were used to identify BACE1 substrates for changes in gene expression and signaling activity. RESULTS: Among altered genes, Clusterin (Clu) and Cxcl14 were significantly upregulated and validated by measuring protein levels. Moreover, BACE1 deficiency enhanced both astrocytic Aß uptake and degradation, and this effect was significantly attenuated by siRNA knockdown of Clu. Mechanistic study suggests that BACE1 deficiency abolishes cleavage of astrocytic insulin receptors (IR), and this may enhance expression of Clu and Cxcl14. Acutely isolated astrocytes from astrocyte-specific knockout of Bace1 mice (Bace1 fl/fl;Gfap-cre) show similar increases in CLU and IR. Furthermore, astrocyte-specific knockout of Bace1 in a 5xFAD background resulted in a significant attenuation in cortical Aß plaque load through enhanced clearance. CONCLUSION: Together, our study suggests that BACE1 in astrocytes regulates expression of Clu and Cxcl14, likely via the control of insulin receptor pathway, and inhibition of astrocytic BACE1 is a potential alternative strategy for enhancing Aß clearance.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Animals , Mice , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Astrocytes/metabolism , Clusterin/metabolism , Mice, Knockout , Mice, Transgenic
3.
J Biol Chem ; 298(11): 102532, 2022 11.
Article in English | MEDLINE | ID: mdl-36162508

ABSTRACT

CX3CL1, also known as fractalkine, is best known for its signaling activity through interactions with its cognate receptor CX3CR1. However, its intrinsic function that is independent of interaction with CX3CR1 remains to be fully understood. We demonstrate that the intracellular domain of CX3CL1 (CX3CL1-ICD), generated upon sequential cleavages by α-/ß-secretase and γ-secretase, initiates a back signaling activity, which mediates direct signal transmission to gene expression in the nucleus. To study this, we fused a synthetic peptide derived from CX3CL1-ICD, named Tet34, with a 13-amino acid tetanus sequence at the N terminus to facilitate translocation into neuronal cells. We show that treatment of mouse neuroblastoma Neuro-2A cells with Tet34, but not its scrambled control (Tet34s), induced cell proliferation, as manifested by changes in protein levels of transcription factors and progrowth molecules cyclin D1, PCNA, Sox5, and Cdk2. Further biochemical assays reveal elevation of phosphorylated insulin receptor ß subunit, insulin-like growth factor-1 receptor ß subunit, and insulin receptor substrates as well as activation of proliferation-linked kinase AKT. In addition, transgenic mice overexpressing membrane-anchored C-terminal CX3CL1 also exhibited activation of insulin/insulin-like growth factor-1 receptor signaling. Remarkably, we found that this Tet34 peptide, but not Tet34s, protected against endoplasmic reticulum stress and cellular apoptosis when Neuro-2A cells were challenged with toxic oligomers of ß-amyloid peptide or hydrogen peroxide. Taken together, our results suggest that CX3CL1-ICD may have translational potential for neuroprotection in Alzheimer's disease and for disorders resulting from insulin resistance.


Subject(s)
Chemokine CX3CL1 , Neuroprotection , Receptor, Insulin , Receptors, Somatomedin , Animals , Mice , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , CX3C Chemokine Receptor 1 , Mice, Transgenic , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism
4.
Exp Neurol ; 336: 113530, 2021 02.
Article in English | MEDLINE | ID: mdl-33221396

ABSTRACT

Microglia are one of the first responders to ischemic injury. Aged microglia acquire a senescent phenotype and produce more inflammatory cytokines after stroke. Depletion of microglia in young mice worsens post-ischemic damage by increasing inflammation. However, young mice do not have dysfunctional microglia. Hence, we hypothesized that depletion of microglia in older mice will contribute to improved early recovery after ischemic stroke injury. Aged (18-19 month) mice were fed with either control chow diet (CD) or PLX5622 chow diet (PLXD) for 21 days. On day 22, a 60-min middle cerebral artery occlusion (MCAo) surgery or sham surgery was performed. Twenty-four and 72 h after stroke immunohistochemistry and flow cytometry were performed. AFS98, a monoclonal antibody against CSF1R was used to specifically deplete brain macrophages by injection into the right hemisphere. Two days after AFS98 injections, mice underwent one-hour MCAo. Twenty-four hours later mice were euthanized and flow cytometry was performed. An increase in infarct volume (p < 0.05) was seen in the PLXD versus CD after stroke in aged mice at 24 and 72 h. An increase (p < 0.05) in infiltrating monocytes was observed after microglial depletion in aged stroke mice suggesting a differential monocyte response. An increase in astrocyte numbers was evident in the PLXD sham mice compared to CD sham, reflecting the off-target effects of PLX5622 treatment. In conclusion, PLX5622 and AFS98 treatment depleted microglia in aged animals but resulted in increased neuroinflammation after ischemic stroke.


Subject(s)
Brain/pathology , Ischemic Stroke/pathology , Microglia/pathology , Stroke/pathology , Aging , Animals , Astrocytes/pathology , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Infarction, Middle Cerebral Artery/pathology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Monocytes/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
5.
Gut Microbes ; 12(1): 1-14, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32897773

ABSTRACT

Aging is associated with cognitive decline and decreased concentrations of short-chain fatty acids (SCFAs) in the gut. SCFAs are significant in that they are protective to the gut and other organs. We tested the hypothesis that the aged gut microbiome alone is sufficient to decrease SCFAs in the host and produce cognitive decline. Fecal transplant gavages (FTGs) from aged (18-20 months) or young (2-3 months) male C57BL/6 mice into germ-free male C57BL/6 mice (N = 11 per group) were initiated at ~3 months of age. Fecal samples were collected and behavioral testing was performed over the study period. Bacterial community structures and relative abundances were measured in fecal samples by sequencing the bacterial 16S ribosomal RNA gene. Mice with aged and young microbiomes showed clear differences in bacterial ß diversity at 30, 60, and 90 d (P = .001 for each) after FTGs. The fecal SCFAs, acetate, propionate, and butyrate (microbiome effect, P < .01 for each) were decreased in mice with an aged microbiome. Mice with an aged microbiome demonstrated depressive-like behavior, impaired short-term memory, and impaired spatial memory over the 3 months following the initial FTG as assessed by the tail suspension (P = .008), the novel object recognition (P < .001), and the Barnes Maze (P = .030) tests, respectively. We conclude that an aged microbiome alone is sufficient to decrease SCFAs in the host and to produce cognitive decline.


Subject(s)
Aging , Cognition , Cognitive Dysfunction/therapy , Fatty Acids, Volatile/metabolism , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/physiology , Animals , Bacteria/classification , Bacteria/growth & development , Brain/immunology , Cognitive Dysfunction/etiology , Cytokines/blood , Depression , Fatty Acids, Volatile/analysis , Feces/chemistry , Feces/microbiology , Germ-Free Life , Leukocytes/immunology , Male , Memory , Mice , Mice, Inbred C57BL , T-Lymphocyte Subsets/immunology
6.
Brain Behav Immun ; 90: 235-247, 2020 11.
Article in English | MEDLINE | ID: mdl-32861719

ABSTRACT

Aging is associated with dysfunction of the gut microbiota-immune-brain axis, a major regulatory axis in both brain health and in central nervous system (CNS) diseases. Antigen presenting cells (APCs) play a major role in sensing changes in the gut microbiota and regulation of innate and adaptive immune responses. APCs have also been implicated in various chronic inflammatory conditions, including age-related neurodegenerative diseases. The increase in chronic low-level inflammation seen with aging has also been linked to behavioral decline. Despite their acknowledged importance along the gut microbiota-immune-brain axis, there is limited evidence on how APCs change with aging. In this study, we examined age-related changes in myeloid APCs in the gut, spleen, and brain as well as changes in the gut microbiota and behavioral phenotype in mice ranging in age from 2 months up to 32 months of both sexes. Our data show that the number of peripherally-sourced myeloid APCs significantly increases with advanced aging in the brain. In addition, our data showed that age-related changes in APCs are subset-specific in the gut and sexually dimorphic in the spleen. Our work highlights the importance of studying myeloid APCs in an age-, tissue-, and sex-specific manner.


Subject(s)
Central Nervous System Diseases , Gastrointestinal Microbiome , Aging , Animals , Antigen-Presenting Cells , Brain , Female , Male , Mice
7.
Aging (Albany NY) ; 12(9): 8049-8066, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32365331

ABSTRACT

Growth differentiation factor (GDF) 11 levels decline with aging. The age-related loss of GDF 11 has been implicated in the pathogenesis of a variety of age-related diseases. GDF11 supplementation reversed cardiac hypertrophy, bone loss, and pulmonary dysfunction in old mice, suggesting that GDF11 has a rejuvenating effect. Less is known about the potential of GDF11 to improve recovery after an acute injury, such as stroke, in aged mice. GDF11/8 levels were assessed in young and aged male mice and in postmortem human brain samples. Aged mice were subjected to a transient middle cerebral artery occlusion (MCAo). Five days after MCAo, mice received and bromodeoxyuridine / 5-Bromo-2'-deoxyuridine (BrdU) and either recombinant GDF11 or vehicle for five days and were assessed for recovery for one month following stroke. MRI was used to determine cerebrospinal fluid (CSF) volume, corpus callosum (CC) area, and brain atrophy at 30 days post-stroke. Immunohistochemistry was used to assess gliosis, neurogenesis, angiogenesis and synaptic density. Lower GDF11/8 levels were found with age in both mice and humans (p<0.05). GDF11 supplementation reduced mortality and improved sensorimotor deficits after stroke. Treatment also reduced brain atrophy and gliosis, increased angiogenesis, improved white matter integrity, and reduced inflammation after stroke. GDF11 may have a role in brain repair after ischemic injury.


Subject(s)
Aging , Bone Morphogenetic Proteins/pharmacokinetics , Brain/metabolism , Growth Differentiation Factors/pharmacokinetics , Ischemic Stroke/drug therapy , Recovery of Function/drug effects , Animals , Blotting, Western , Brain/drug effects , Dietary Supplements , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Ischemic Stroke/diagnosis , Ischemic Stroke/mortality , Male , Mice
8.
J Neuroinflammation ; 15(1): 148, 2018 May 17.
Article in English | MEDLINE | ID: mdl-29776451

ABSTRACT

BACKGROUND: Activation of transforming growth factor-ß-activated kinase 1 (TAK1) occurs after stroke and leads to an exacerbation of brain injury. TAK1 is involved in innate and adaptive immune responses, but it has divergent inflammatory effects that are dependent on the cell type in which it is activated. There is a robust infiltration of myeloid cells after stroke; however, the contribution of myeloid TAK1 to cerebral ischemia is currently unknown. We hypothesized that myeloid-specific deletion of TAK1 would protect against ischemic brain injury. METHODS: Myeloid TAK1ΔM and wild-type (WT) mice were subjected to middle cerebral artery occlusion (MCAo). Brain-infiltrating and splenic immune cells were evaluated at 3 days after stroke. Assessment of infarct size and behavioral deficits were performed on days 3 and 7 post-stroke. RESULTS: Infarcts were significantly smaller in TAK1ΔM mice (p < 0.01), and behavioral deficits were less severe despite equivalent reduction in cerebral blood flow. Flow cytometry demonstrated an increase in the frequency of splenic monocytes and neutrophils (p < 0.05) and a decrease in splenic CD3+ T (p < 0.01) and CD19+ B (p = 0.06) cells in TAK1ΔM mice compared to WT at baseline. Three days after stroke, a significant increase in the number of brain-infiltrating immune cell was observed in both TAK1ΔM (p < 0.05) and WT (p < 0.001) mice compared to their respective shams. However, there was a significant decrease in the infiltrating CD45hi immune cell counts (p < 0.05), with a pronounced reduction in infiltrating monocytes (p < 0.001) in TAK1ΔM after stroke compared to WT stroke mice. Additionally, a significant reduction in CD49d+ monocytes was seen in the brains of TAK1ΔM stroke mice compared to wild-type mice. Importantly, TAK1ΔM MCAo mice had smaller infarcts and improved behavioral outcomes at day 7 post-stroke. CONCLUSION: Our results showed that deletion of myeloid TAK1 resulted in smaller infarcts and improved functional outcomes at the peak of inflammation (day 3) and a reduction in brain-infiltrating immune cells that were primarily monocytes. Myeloid TAK1 deletion was also protective at 7 days post MCAo, reflecting a detrimental role of myeloid TAK1 in the progression of ischemic injury.


Subject(s)
Infarction, Middle Cerebral Artery/pathology , MAP Kinase Kinase Kinases/deficiency , Monocytes/pathology , Myeloid Cells/physiology , Neutrophils/physiology , Recovery of Function/genetics , Animals , Antigens, CD/metabolism , Cerebrovascular Circulation/genetics , Disease Models, Animal , Flow Cytometry , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/physiopathology , MAP Kinase Kinase Kinases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/metabolism , Muramidase/genetics , Muramidase/metabolism , Neutrophil Infiltration/genetics
9.
Brain Behav Immun ; 66: 302-312, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28751018

ABSTRACT

INTRODUCTION: Acute ischemic injury leads to severe neuronal loss. One of the key mechanisms responsible for this effect is inflammation, which is characterized by the activation of myeloid cells, including resident microglia and infiltrating monocytes/macrophages. P2X4 receptors (P2X4Rs) present on these immune cells modulate the inflammatory response. For example, excessive release of adenosine triphosphate during acute ischemic stroke triggers stimulation of P2X4Rs, leading to myeloid cell activation and proliferation and further exacerbating post-ischemic inflammation. In contrast, during recovery P2X4Rs activation on microglia leads to the release of brain-derived neurotrophic factor (BDNF), which alleviate depression, maintain synaptic plasticity and hasten post-stroke behavioral recovery. Therefore, we hypothesized that deletion of the P2X4R specifically from myeloid cells would have differential effects on acute versus chronic recovery following stroke. METHODS: We subjected global or myeloid-specific (MS) P2X4R knock-out (KO) mice and wild-type littermates of both sexes to right middle cerebral artery occlusion (60min). We performed histological, behavioral (sensorimotor and depressive), and biochemical (quantitative PCR and flow cytometry) analyses to determine the acute (three days after occlusion) and chronic (30days after occlusion) effects of receptor deletion. RESULTS: Global P2X4R deletion led to reduced infarct size in both sexes. In MS P2X4R KO mice, only females showed reduced infarct size, an effect that did not change with ovariectomy. MS P2X4R KO mice of both sexes showed swift recovery from sensorimotor deficits during acute recovery but exhibited a more pronounced post-stroke depressive behavior phenotype that was independent of infarct size. Quantitative PCR analysis of whole cell lysate as well as flow-sorted myeloid cells from the perilesional cortex showed increased cellular interleukin 1 beta (IL-1ß), interleukin 6 (IL-6), and tumor necrosis factor alpha (TNF-α) mRNA levels but reduced plasma levels of these cytokines in MS P2X4R KO mice after stroke. The expression levels of BDNF and other depression-associated genes were reduced in MS P2X4R KO mice after stroke. CONCLUSIONS: P2X4R deletion protects against stroke acutely but predisposes to depression-like behavior chronically after stroke. Thus, a time-sensitive approach should be considered when targeting P2X4Rs after stroke.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain/metabolism , Depression/complications , Receptors, Purinergic P2X4/physiology , Stroke/metabolism , Stroke/pathology , Animals , Behavior, Animal , Brain/pathology , Brain Ischemia/complications , Cytokines/metabolism , Depression/genetics , Female , Inflammation Mediators/metabolism , Male , Mice , Mice, Knockout , Microglia/pathology , Phenotype , RNA, Messenger/metabolism , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X4/metabolism , Recovery of Function , Stroke/complications
SELECTION OF CITATIONS
SEARCH DETAIL