Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Mol Life Sci ; 80(9): 262, 2023 Aug 19.
Article in English | MEDLINE | ID: mdl-37597109

ABSTRACT

The constitutive photomorphogenesis 9 (COP9) signalosome (CSN) is a deNEDDylase controlling ubiquitination activity of cullin-RING-E3 ligases (CRLs) and thus the levels of key cellular proteins. While the CSN and its catalytic subunit CSN5 have been extensively studied in cancer, its role in inflammatory and neurological diseases is less understood. Following verification that CSN5 is expressed in mouse and human brain, here we studied the role of the CSN in neuroinflammation and ischemic neuronal damage employing models of relevant brain-resident cell types, an ex vivo organotypic brain slice culture model, and the CRL NEDDylation state-modifying drugs MLN4924 and CSN5i-3, which mimic and inhibit, respectively, CSN5 deNEDDylase activity. Untargeted mass spectrometry-based proteomics revealed that MLN4924 and CSN5i-3 substantially alter the microglial proteome, including inflammation-related proteins. Applying these drugs and mimicking microglial and endothelial inflammation as well as ischemic neuronal stress by TNF and oxygen-glucose-deprivation/reoxygenation (OGD/RO) treatment, respectively, we could link CSN5/CSN-mediated cullin deNEDDylation to reduction of microglial inflammation, attenuated cerebral endothelial inflammation, improved barrier integrity, as well as protection from ischemic stress-induced neuronal cell death. Specifically, MLN4924 reduced phagocytic activity, motility, and inflammatory cytokine expression of microglial cells, and this was linked to inhibition of inflammation-induced NF-κB and Akt signaling. Inversely, Csn5 knockdown and CSN5i-3 increased NF-κB signaling. Moreover, MLN4924 abrogated TNF-induced NF-κB signaling in cerebral microvascular endothelial cells (hCMECs) and rescued hCMEC monolayers from OGD/RO-triggered barrier leakage, while CSN5i-3 exacerbated permeability. In an ex vivo organotypic brain slice model of ischemia/reperfusion stress, MLN4924 protected from neuronal death, while CSN5i-3 impaired neuronal survival. Neuronal damage was attributable to microglial activation and inflammatory cytokines, as indicated by microglial shape tracking and TNF-blocking experiments. Our results indicate a protective role of the CSN in neuroinflammation via brain-resident cell types involved in ischemic brain disease and implicate CSN activity-mimicking deNEDDylating drugs as potential therapeutics.


Subject(s)
NF-kappa B , Neuroinflammatory Diseases , Humans , Animals , Mice , COP9 Signalosome Complex , Cullin Proteins , Endothelial Cells , Brain , Inflammation/drug therapy , Cytokines
2.
J Nucl Med ; 63(1): 117-124, 2022 01.
Article in English | MEDLINE | ID: mdl-34016733

ABSTRACT

ß-amyloid (Aß) PET is an important tool for quantification of amyloidosis in the brain of suspected Alzheimer disease (AD) patients and transgenic AD mouse models. Despite the excellent correlation of Aß PET with gold standard immunohistochemical assessments, the relative contributions of fibrillar and nonfibrillar Aß components to the in vivo Aß PET signal remain unclear. Thus, we obtained 2 murine cerebral amyloidosis models that present with distinct Aß plaque compositions and performed regression analysis between immunohistochemistry and Aß PET to determine the biochemical contributions to Aß PET signal in vivo. Methods: We investigated groups of AppNL-G-F and APPPS1 mice at 3, 6, and 12 mo of age by longitudinal 18F-florbetaben Aß PET and with immunohistochemical analysis of the fibrillar and total Aß burdens. We then applied group-level intermodality regression models using age- and genotype-matched sets of fibrillar and nonfibrillar Aß data (predictors) and Aß PET results (outcome) for both Aß mouse models. An independent group of double-hit APPPS1 mice with dysfunctional microglia due to knockout of triggering receptor expression on myeloid cells 2 (Trem2-/-) served for validation and evaluation of translational impact. Results: Neither fibrillar nor nonfibrillar Aß content alone sufficed to explain the Aß PET findings in either AD model. However, a regression model compiling fibrillar and nonfibrillar Aß together with the estimate of individual heterogeneity and age at scanning could explain a 93% of variance of the Aß PET signal (P < 0.001). Fibrillar Aß burden had a 16-fold higher contribution to the Aß PET signal than nonfibrillar Aß. However, given the relatively greater abundance of nonfibrillar Aß, we estimate that nonfibrillar Aß produced 79% ± 25% of the net in vivo Aß PET signal in AppNL-G-F mice and 25% ± 12% in APPPS1 mice. Corresponding results in separate groups of APPPS1/Trem2-/- and APPPS1/Trem2+/+ mice validated the calculated regression factors and revealed that the altered fibrillarity due to Trem2 knockout impacts the Aß PET signal. Conclusion: Taken together, the in vivo Aß PET signal derives from the composite of fibrillar and nonfibrillar Aß plaque components. Although fibrillar Aß has inherently higher PET tracer binding, the greater abundance of nonfibrillar Aß plaque in AD-model mice contributes importantly to the PET signal.


Subject(s)
Plaque, Amyloid
3.
Nat Neurosci ; 25(1): 20-25, 2022 01.
Article in English | MEDLINE | ID: mdl-34811521

ABSTRACT

Microglia appear activated in the vicinity of amyloid beta (Aß) plaques, but whether microglia contribute to Aß propagation into unaffected brain regions remains unknown. Using transplantation of wild-type (WT) neurons, we show that Aß enters WT grafts, and that this is accompanied by microglia infiltration. Manipulation of microglia function reduced Aß deposition within grafts. Furthermore, in vivo imaging identified microglia as carriers of Aß pathology in previously unaffected tissue. Our data thus argue for a hitherto unexplored mechanism of Aß propagation.


Subject(s)
Amyloid beta-Peptides , Microglia , Amyloid beta-Peptides/metabolism , Brain/metabolism , Humans , Microglia/metabolism , Neurons/metabolism , Plaque, Amyloid/pathology
4.
Biomed Pharmacother ; 144: 112239, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34601192

ABSTRACT

Inflammatory bowel diseases (IBD), represented by ulcerative colitis (UC) and Crohn's disease (CD), are characterized by chronic inflammation of the gastrointestinal tract, what leads to diarrhea, malnutrition, and weight loss. Depression of the growth hormone-insulin-like growth factor-1 axis (GH-IGF-1 axis) could be responsible of these symptoms. We demonstrate that long-term treatment (54 weeks) of adult CD patients with adalimumab (ADA) results in a decrease in serum IGF-1 without changes in serum IGF-1 binding protein (IGF1BP4). These results prompted us to conduct a preclinical study to test the efficiency of IGF-1 in the medication for experimental colitis. IGF-1 treatment of rats with DSS-induced colitis has a beneficial effect on the following circulating biochemical parameters: glucose, albumin, and total protein levels. In this experimental group we also observed healthy maintenance of colon size, body weight, and lean mass in comparison with the DSS-only group. Histological analysis revealed restoration of the mucosal barrier with the IGF-1 treatment, which was characterized by healthy quantities of mucin production, structural maintenance of adherers junctions (AJs), recuperation of E-cadherin and ß-catenin levels and decrease in infiltrating immune cells and in metalloproteinase-2 levels. The experimentally induced colitis caused activation of apoptosis markers, including cleaved caspase 3, caspase 8, and PARP and decreases cell-cycle checkpoint activators including phosphorylated Rb, cyclin E, and E2F1. The IGF-1 treatment inhibited cyclin E depletion and partially protects PARP levels. The beneficial effects of IGF-1 in experimental colitis could be explained by a re-sensitization of the IGF-1/IRS-1/AKT cascade to exogenous IGF-1. Given these results, we postulate that IGF-1 treatment of IBD patients could prove to be successful in reducing disease pathology.


Subject(s)
Body Weight/drug effects , Colitis/prevention & control , Colon/drug effects , Insulin-Like Growth Factor I/pharmacology , Intestinal Mucosa/drug effects , Adalimumab/therapeutic use , Adult , Animals , Biomarkers/blood , Colitis/metabolism , Colitis/pathology , Colitis, Ulcerative/blood , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/drug therapy , Colon/metabolism , Colon/pathology , Crohn Disease/blood , Crohn Disease/diagnosis , Crohn Disease/drug therapy , Disease Models, Animal , Female , Humans , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Longitudinal Studies , Male , Middle Aged , Prospective Studies , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar , Signal Transduction , Spain , Time Factors , Treatment Outcome , Tumor Necrosis Factor Inhibitors/therapeutic use
5.
Nat Neurosci ; 22(2): 191-204, 2019 02.
Article in English | MEDLINE | ID: mdl-30617257

ABSTRACT

Coding variants in the triggering receptor expressed on myeloid cells 2 (TREM2) are associated with late-onset Alzheimer's disease (AD). We demonstrate that amyloid plaque seeding is increased in the absence of functional Trem2. Increased seeding is accompanied by decreased microglial clustering around newly seeded plaques and reduced plaque-associated apolipoprotein E (ApoE). Reduced ApoE deposition in plaques is also observed in brains of AD patients carrying TREM2 coding variants. Proteomic analyses and microglia depletion experiments revealed microglia as one origin of plaque-associated ApoE. Longitudinal amyloid small animal positron emission tomography demonstrates accelerated amyloidogenesis in Trem2 loss-of-function mutants at early stages, which progressed at a lower rate with aging. These findings suggest that in the absence of functional Trem2, early amyloidogenesis is accelerated due to reduced phagocytic clearance of amyloid seeds despite reduced plaque-associated ApoE.


Subject(s)
Alzheimer Disease/genetics , Amyloid/metabolism , Apolipoproteins E/metabolism , Brain/pathology , Membrane Glycoproteins/genetics , Plaque, Amyloid/genetics , Receptors, Immunologic/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Genotype , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Phagocytosis/physiology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Receptors, Immunologic/metabolism
6.
Alzheimers Dement ; 15(3): 453-464, 2019 03.
Article in English | MEDLINE | ID: mdl-30442540

ABSTRACT

INTRODUCTION: Murine microglia expressing the Alzheimer's disease-linked TREM2R47H mutation display variable decrease in phagocytosis, while impaired phagocytosis is reported following loss of TREM2. However, no data exist on TREM2+/R47H human microglia. Therefore, we created human pluripotent stem cell (hPSC) monocytes and transdifferentiated microglia-like cells (tMGs) to examine the effect of the TREM2+/R47H mutation and loss of TREM2 on phagocytosis. METHODS: We generated isogenic TREM2+/R47H, TREM2+/-, and TREM2-/- hPSCs using CRISPR/Cas9. Following differentiation to monocytes and tMGs, we studied the uptake of Escherichia coli fragments and analyzed amyloid plaque clearance from cryosections of APP/PS1+/- mouse brains. RESULTS: We demonstrated that tMGs resemble cultured human microglia. TREM2+/- and TREM2-/- hPSC monocytes and tMGs phagocytosed significantly less E. coli fragments and cleared less amyloid plaques than wild-type hPSC progeny, with no difference for TREM2+/R47H progeny. DISCUSSION: In vitro phagocytosis of hPSC monocytes and tMGs was not affected by the TREM2+/R47H mutation but was significantly impaired in TREM2+/- and TREM2-/- progeny.


Subject(s)
Membrane Glycoproteins/deficiency , Microglia/metabolism , Monocytes/metabolism , Plaque, Amyloid/metabolism , Receptors, Immunologic/deficiency , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain , CRISPR-Cas Systems , Cells, Cultured , Escherichia coli , Membrane Glycoproteins/genetics , Mice, Transgenic , Phagocytosis , Pluripotent Stem Cells , Presenilin-1/genetics , Presenilin-1/metabolism , Receptors, Immunologic/genetics
7.
Sci Rep ; 8(1): 11335, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054579

ABSTRACT

Diabetes mellitus is a group of disorders characterized by prolonged high levels of circulating blood glucose. Type 1 diabetes is caused by decreased insulin production in the pancreas whereas type 2 diabetes may develop due to obesity and lack of exercise; it begins with insulin resistance whereby cells fail to respond properly to insulin and it may also progress to decreased insulin levels. The brain is an important target for insulin, and there is great interest in understanding how diabetes affects the brain. In addition to the direct effects of insulin on the brain, diabetes may also impact the brain through modulation of the inflammatory system. Here we investigate how perturbation of circulating insulin levels affects the expression of Hes3, a transcription factor expressed in neural stem and progenitor cells that is involved in tissue regeneration. Our data show that streptozotocin-induced ß-cell damage, high fat diet, as well as metformin, a common type 2 diabetes medication, regulate Hes3 levels in the brain. This work suggests that Hes3 is a valuable biomarker helping to monitor the state of endogenous neural stem and progenitor cells in the context of diabetes mellitus.


Subject(s)
Aging/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/metabolism , Diet, High-Fat , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Metformin/administration & dosage , Nerve Tissue Proteins/metabolism , Streptozocin/toxicity , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation/drug effects , Insulin-Secreting Cells/drug effects , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Phenotype , Repressor Proteins
8.
EMBO J ; 32(24): 3145-60, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24240175

ABSTRACT

Transcriptome analysis of somatic stem cells and their progeny is fundamental to identify new factors controlling proliferation versus differentiation during tissue formation. Here, we generated a combinatorial, fluorescent reporter mouse line to isolate proliferating neural stem cells, differentiating progenitors and newborn neurons that coexist as intermingled cell populations during brain development. Transcriptome sequencing revealed numerous novel long non-coding (lnc)RNAs and uncharacterized protein-coding transcripts identifying the signature of neurogenic commitment. Importantly, most lncRNAs overlapped neurogenic genes and shared with them a nearly identical expression pattern suggesting that lncRNAs control corticogenesis by tuning the expression of nearby cell fate determinants. We assessed the power of our approach by manipulating lncRNAs and protein-coding transcripts with no function in corticogenesis reported to date. This led to several evident phenotypes in neurogenic commitment and neuronal survival, indicating that our study provides a remarkably high number of uncharacterized transcripts with hitherto unsuspected roles in brain development. Finally, we focussed on one lncRNA, Miat, whose manipulation was found to trigger pleiotropic effects on brain development and aberrant splicing of Wnt7b. Hence, our study suggests that lncRNA-mediated alternative splicing of cell fate determinants controls stem-cell commitment during neurogenesis.


Subject(s)
Brain/embryology , Gene Expression Profiling/methods , Neural Stem Cells/physiology , RNA, Long Noncoding/genetics , Alternative Splicing , Animals , Brain/cytology , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Mice , Mice, Transgenic , Neurogenesis , Neurons , Phenotype , Proteins/genetics , Proto-Oncogene Proteins/genetics , Wnt Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...