Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Mol Neurosci ; 74(2): 55, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38776015

ABSTRACT

The dysregulation of lipid metabolism has been strongly associated with Alzheimer's disease (AD) and has intricate connections with various aspects of disease progression, such as amyloidogenesis, bioenergetic deficit, oxidative stress, neuroinflammation, and myelin degeneration. Here, a comprehensive bioinformatic assessment was conducted on lipid metabolism genes in the brains and peripheral blood of AD-derived transcriptome datasets, characterizing the correlation between differentially expressed genes (DEGs) of lipid metabolism and disease pathologies, as well as immune cell preferences. Through the application of weighted gene co-expression network analysis (WGCNA), modules eigengenes related to lipid metabolism were pinpointed, and the examination of their molecular functions within biological processes, molecular pathways, and their associations with pathological phenotypes and molecular networks has been characterized. Analysis of biological networks indicates notable discrepancies in the expression patterns of the DEGs between neuronal and immune cells, as well as variations in cell type enrichments within both brain tissue and peripheral blood. Additionally, drugs targeting the DEGs from central and peripheral and a diagnostic model for hub genes from the blood were retrieved and assessed, some of which were shown to be useful for therapeutic and diagnostic. These results revealed the distinctive pattern of transcriptionally abnormal lipid metabolism in central, peripheral, and immune cell activation, providing valuable insight into lipid metabolism for diagnosing and guiding more effective treatment for AD.


Subject(s)
Alzheimer Disease , Lipid Metabolism , Transcriptome , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Humans , Lipid Metabolism/genetics , Brain/metabolism , Gene Regulatory Networks
2.
Cell Commun Signal ; 22(1): 31, 2024 01 12.
Article in English | MEDLINE | ID: mdl-38216911

ABSTRACT

The intracellular deposition and intercellular transmission of α-synuclein (α-syn) are shared pathological characteristics among neurodegenerative disorders collectively known as α-synucleinopathies, including Parkinson's disease (PD). Although the precise triggers of α-synucleinopathies remain unclear, recent findings indicate that disruption of microglial homeostasis contributes to the pathogenesis of PD. Microglia play a crucial role in maintaining optimal neuronal function by ensuring a homeostatic environment, but this function is disrupted during the progression of α-syn pathology. The involvement of microglia in the accumulation, uptake, and clearance of aggregated proteins is critical for managing disease spread and progression caused by α-syn pathology. This review summarizes current knowledge on the interrelationships between microglia and α-synucleinopathies, focusing on the remarkable ability of microglia to recognize and internalize extracellular α-syn through diverse pathways. Microglia process α-syn intracellularly and intercellularly to facilitate the α-syn neuronal aggregation and cell-to-cell propagation. The conformational state of α-synuclein distinctly influences microglial inflammation, which can affect peripheral immune cells such as macrophages and lymphocytes and may regulate the pathogenesis of α-synucleinopathies. We also discuss ongoing research efforts to identify potential therapeutic approaches targeting both α-syn accumulation and inflammation in PD. Video Abstract.


Subject(s)
Parkinson Disease , Synucleinopathies , Humans , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism , alpha-Synuclein/therapeutic use , Synucleinopathies/metabolism , Synucleinopathies/pathology , Microglia/metabolism , Inflammation/metabolism , Homeostasis
3.
Transl Vis Sci Technol ; 10(6): 27, 2021 05 03.
Article in English | MEDLINE | ID: mdl-34015102

ABSTRACT

Purpose: Stem cell-based therapy has the potential to become one approach to regenerate the damaged trabecular meshwork (TM) in glaucoma. Co-culture of induced pluripotent stem cells (iPSCs) with human TM cells has been a successful approach to generate autologous TM resembling cells. However, the differentiated cells generated using this approach are still problematic for clinical usage. This study aimed to develop a clinically applicable strategy for generating TM-like cells from iPSCs. Methods: Highly expressed receptors during iPSC differentiation were identified by AutoSOME, Gene Ontology, and reverse transcription polymerase chain reaction (RT-PCR) analysis. The recombinant cytokines that bind to these receptors were used to generate a new differentiation protocol. The resultant TM-like cells were characterized morphologically, immunohistochemically, and transcriptionally. Results: We first determined two stages of iPSC differentiation and identified highly expressed receptors associated with the differentiation at each stage. The expression of these receptors was further confirmed by RT-PCR analysis. Exposure to the recombinant cytokines that bind to these receptors, including transforming growth factor beta 1, nerve growth factor beta, erythropoietin, prostaglandin F2 alpha, and epidermal growth factor, can efficiently differentiate iPSCs into TM-like cells, which express TM biomarkers and can form dexamethasone-inducible CLANs. Conclusions: We successfully generated a xeno- and feeder-free differentiation protocol with recombinant cytokines to generate the TM progenitor and TM-like cells from human iPSCs. Translational Relevance: The new approach minimizes the risks from contamination and also improves the differentiation efficiency and consistency, which are particularly crucial for clinical use of stem cells in glaucoma treatment.


Subject(s)
Induced Pluripotent Stem Cells , Cell Differentiation , Cells, Cultured , Cytokines/genetics , Humans , Trabecular Meshwork
4.
Int Immunopharmacol ; 87: 106792, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32707494

ABSTRACT

Chronic brain neuritis is important for the pathogenesis and progression of Alzheimer's disease (AD). Overexpression of tumor necrosis factor-α (TNFα)-inducible protein 8-like 2 (TIPE2), a novel immunoregulatory protein, reverses cognitive dysfunction in APP/PS1 mice. However, the mRNA profile changes in TIPE2 overexpression APP/PS1 mice and the molecular mechanism of cognitive attenuation remain unknown. In this study, after the Y-maze testing the spatial learning of the APP/PS1 mice and the TIPE2 overexpression APP/PS1 mice, high-throughput sequencing was performed on hippocampus tissues for analysis of mRNA profiles. A total of 183 differentially expressed genes (DEGs) were detected, of which 36 were down-regulated and 147 were up-regulated. Then, the mRNA profiles of the APP/PS1 mice and the wild-type mice were analyzed. A total of 196 DEGs were detected, of which 105 were down-regulated and 91 were up-regulated in the APP/PS1 mice. A comprehensive comparison of the mouse mRNAs showed that 20 genes were differentially expressed in both groups, among which, 19 genes showed an altered expression in the APP/PS1 mice, and the expression was recovered in TIPE2 overexpression APP/PS1 mice. We selected seven genes from these 19 genes, including Ttr, Lepr, Angptl2, Otx2, Clic6, Clo4a3 and Wfdc, for high-throughput sequencing. The results showed that, compared to the wild-type mice, these 7 genes were significantly down-regulated in the hippocampus of APP/PS1 mice. The expressions of a selected list of DEGs between APP/PS1 mice and APP/PS1 + OE mice were validated by quantitative real-time RT-PCR (qRT-PCR), and the results were consistent with the sequencing analysis. Taken together, increased adeno-associated virus (AAV)-mediated TIPE2 overexpression in the hippocampus of APP/PS1 mice reversed cognitive dysfunction. Transcriptional sequencing and bioinformatics analysis indicate that the attenuation of cognitive deficits was attributed to the recovery of certain genes.


Subject(s)
Cognitive Dysfunction/genetics , Intracellular Signaling Peptides and Proteins/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Female , Hippocampus , Male , Mice, Transgenic , Presenilin-1/genetics , RNA, Messenger
5.
J Neuroimmune Pharmacol ; 14(3): 519-529, 2019 09.
Article in English | MEDLINE | ID: mdl-31286344

ABSTRACT

Neuroinflammation plays an early and prominent role in the pathology of Alzheimer's disease (AD). Tumor necrosis factor-α-induced protein 8-like 2 (TIPE2) has been identified as a negative regulator of innate and adaptive immunity. However, whether TIPE2 affects cognitive functions in AD-like mouse models remains unknown. In this study, we compared the gene and protein expressions of TIPE2 between the APP/PS1 mice and the age-matched wild type (WT) mice at different stages of development using western blot and RT-qPCR. The hippocampal expression of the TIPE2 mRNA and protein in APP/PS1 mice was higher than that of the WT mice starting from 6 months to 10 months. However, the difference of the TIPE2 expression between the APP/PS1 mice and the WT mice declined in a time-dependent manner. The spatial learning and memory deficit from the 8-month-old APP/PS1 mice was observed in the Y-maze test and fear conditioning task. Interestingly, overexpression of TIPE2 by intra-hippocampal injection of AAV-TIPE2 into APP/PS1 mice resulted in an improvement of learning and memory and reduced expression of inflammatory cytokines, such as TNF-α, IL-6 and IL-1ß, and increased expression of anti-inflammatory cytokines, such as IL-10 and Arg-1. Taken together, our findings show that the TIPE2 expression level was negatively correlated with the pathogenesis of Alzheimer's disease, and overexpression of TIPE2 attenuates cognitive deficits in APP/PS1 mice, suggesting TIPE2 is a potential target for pharmacological intervention and improvement of cognitive deficits. Graphical Abstract .


Subject(s)
Alzheimer Disease/immunology , Intracellular Signaling Peptides and Proteins/physiology , Memory Disorders/prevention & control , Adaptive Immunity , Alzheimer Disease/psychology , Alzheimer Disease/therapy , Amyloid beta-Protein Precursor/genetics , Animals , Cognition Disorders/etiology , Cognition Disorders/immunology , Cognition Disorders/prevention & control , Cytokines/biosynthesis , Cytokines/genetics , Dependovirus/genetics , Disease Models, Animal , Fear , Gene Expression Regulation, Developmental , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/therapeutic use , Hippocampus , Immunity, Innate , Injections , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Maze Learning , Memory Disorders/etiology , Memory Disorders/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/genetics , Recombinant Proteins/metabolism , Up-Regulation
6.
Mol Vis ; 25: 255-265, 2019.
Article in English | MEDLINE | ID: mdl-31205407

ABSTRACT

Purpose: The ocular trabecular meshwork (TM) responsible for aqueous humor (AH) drainage is crucial for regulating intraocular pressure (IOP) of the eye. An IOP elevation that causes distended TM is involved in the pathogenesis of glaucoma, suggesting intercellular connections important for the TM pathophysiology. The goal of this study was to examine whether gap junction proteins between endothelial cells in the TM are expressional and functional. Methods: The expression levels of the gap junction channels in normal human TM cells were determined with real-time PCR and western blot assays. Immunohistochemistry (IHC) staining was performed to examine the localization of gap junction proteins in normal human TM cells and tissues. IOP and the outflow of AH were measured after intercameral injection of gap junction blockers in C57/BL6 mice. Results: Gap junction proteins GJA1, GJA8, GJB6, and GJC1 were robustly expressed in human TM cells from three individuals. Among the four gap junction channels, GJA1 and GJA8 exhibited the most abundance in the TM. The IHC analysis further confirmed that these proteins were expressed on the membrane between adjacent cells. In the human TM tissues, GJA1, GJA8, GJB6, and GJC1 were also observed along the trabecular beams. Inhibition of gap junctions with intracameral injection of blockers resulted in a statistically significant increase in aqueous humor outflow resistance and IOP elevation in mice. Conclusions: The GJA1 and GJA8 gap junction proteins, in particular, are robustly expressed in human TM cells and tissues. Pharmacological inhibition of gap junction channels causes an increased resistance of AH outflow and an elevation of IOP in mice. The present findings suggest the functional role of gap junction channels for regulation of AH outflow in the TM, and activation of gap junctions might represent a therapeutic strategy for treatment of glaucoma.


Subject(s)
Anterior Chamber/metabolism , Aqueous Humor/metabolism , Gap Junctions/metabolism , Trabecular Meshwork/metabolism , Animals , Connexins/genetics , Connexins/metabolism , Gene Expression Regulation , Humans , Male , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trabecular Meshwork/cytology
7.
Acta Pharmacol Sin ; 40(6): 737-745, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30333556

ABSTRACT

The α7 nicotinic acetylcholine receptor (α7 nAChR) is a ligand-gated Ca2+-permeable homopentameric ion channel implicated in cognition and neuropsychiatric disorders. Pharmacological enhancement of α7 nAChR function has been suggested for improvement of cognitive deficits. In the present study, we characterized a thiazolyl heterocyclic derivative, 6-(2-chloro-6-methylphenyl)-2-((3-fluoro-4-methylphenyl)amino)thiazolo[4,5-d]pyrimidin-7(6H)-one (JWX-A0108), as a novel type I α7 nAChR positive allosteric modulator (PAM), and evaluated its ability to reverse auditory gating and spatial working memory deficits in mice. In Xenopus oocytes expressing human nAChR channels, application of JWX-A0108 selectively enhanced α7 nAChR-mediated inward current in the presence of the agonist ACh (EC50 value = 4.35 ± 0.12 µM). In hippocampal slices, co-application of ACh and JWX-A0108 (10 µM for each) markedly increased both the frequency and amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs) recorded in pyramidal neurons, but JWX-A0108 did not affect GABA-induced current in oocytes expressing human GABAA receptor α1ß3γ2 and α5ß3γ2 subtypes. In mice with MK-801-induced deficits in auditory gating, administration of JWX-A0108 (1, 3, and 10 mg/kg, i.p.) dose-dependently attenuates MK-801-induced auditory gating deficits in five prepulse intensities (72, 76, 80, 84, and 88 dB). Furthermore, administration of JWX-A0108 (0.03, 0.1, or 0.3 mg/kg, i.p.) significantly reversed MK-801-induced impaired spatial working memory in mice. Our results demonstrate that JWX-A0108 is a novel type I PAM of α7 nAChR, which may be beneficial for improvement of cognitive deficits commonly found in neuropsychiatric disorders such as schizophrenia and Alzheimer's disease.


Subject(s)
Nootropic Agents/therapeutic use , Prepulse Inhibition/drug effects , Sensory Gating/drug effects , Thiazoles/therapeutic use , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Dizocilpine Maleate , Hippocampus/drug effects , Hippocampus/physiopathology , Humans , Interneurons/drug effects , Locomotion/drug effects , Male , Maze Learning/drug effects , Memory Disorders/drug therapy , Mice, Inbred C57BL , Nootropic Agents/pharmacokinetics , Nootropic Agents/pharmacology , Rats, Sprague-Dawley , Schizophrenia/chemically induced , Schizophrenia/drug therapy , Synaptic Transmission/drug effects , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...