Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Aging (Albany NY) ; 13(9): 12973-12995, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33962394

ABSTRACT

The ketogenic diet has been widely used in the treatment of various nervous system and metabolic-related diseases. Our previous research found that a ketogenic diet exerts a protective effect and promotes functional recovery after spinal cord injury. However, the mechanism of action is still unclear. In this study, different dietary feeding methods were used, and myelin expression and gene level changes were detected among different groups. We established 15 RNA-seq cDNA libraries from among 4 different groups. First, KEGG pathway enrichment of upregulated differentially expressed genes and gene set enrichment analysis of the ketogenic diet and normal diet groups indicated that a ketogenic diet significantly improved the steroid anabolic pathway in rats with spinal cord injury. Through cluster analysis, protein-protein interaction analysis and visualization of iPath metabolic pathways, it was determined that Sqle, Sc5d, Cyp51, Dhcr24, Msmo1, Hsd17b7, and Fdft1 expression changed significantly. Second, through weighted gene co-expression network analysis showed that rats fed a ketogenic diet showed a significant reduction in the expression of genes involved in immune-related pathways, including those associated with immunity and infectious diseases. A ketogenic diet may improve the immune microenvironment and myelin growth in rats with spinal cord injury through reprogramming of steroid metabolism.


Subject(s)
Diet, Ketogenic , Myelin Sheath/metabolism , Spinal Cord Injuries/diet therapy , Steroids/metabolism , Animals , Disease Models, Animal , Gene Regulatory Networks , Humans , Male , Metabolic Networks and Pathways/genetics , Metabolic Networks and Pathways/immunology , Myelin Sheath/immunology , Myelin Sheath/pathology , Protein Interaction Maps , RNA-Seq , Rats , Recovery of Function , Spinal Cord Injuries/immunology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
2.
Aging (Albany NY) ; 12(21): 22313-22334, 2020 11 07.
Article in English | MEDLINE | ID: mdl-33188159

ABSTRACT

α-Synuclein (α-Syn) is a small, soluble, disordered protein that is widely expressed in the nervous system. Although its physiological functions are not yet fully understood, it is mainly involved in synaptic vesicle transport, neurotransmitter synthesis and release, cell membrane homeostasis, lipid synthesis, mitochondrial and lysosomal activities, and heavy metal removal. The complex and inconsistent pathological manifestations of α-Syn are attributed to its structural instability, mutational complexity, misfolding, and diverse posttranslational modifications. These effects trigger mitochondrial dysfunction, oxidative stress, and neuroinflammatory responses, resulting in neuronal death and neurodegeneration. Several recent studies have discovered the pathogenic roles of α-Syn in traumatic and vascular central nervous system diseases, such as traumatic spinal cord injury, brain injury, and stroke, and in aggravating the processes of neurodegeneration. This review aims to highlight the structural and pathophysiological changes in α-Syn and its mechanism of action in traumatic and vascular diseases of the central nervous system.


Subject(s)
Central Nervous System Diseases/metabolism , Cerebrovascular Disorders/metabolism , Trauma, Nervous System/metabolism , alpha-Synuclein/metabolism , Animals , Central Nervous System Diseases/pathology , Cerebrovascular Disorders/pathology , Humans , Trauma, Nervous System/pathology
3.
BMC Genomics ; 20(1): 851, 2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31726970

ABSTRACT

BACKGROUND: Endogenous α-synuclein (α-Syn) is involved in many pathophysiological processes in the secondary injury stage after acute spinal cord injury (SCI), and the mechanism governing these functions has not been thoroughly elucidated to date. This research aims to characterize the effect of α-Syn knockdown on transcriptional levels after SCI and to determine the mechanisms underlying α-Syn activity based on RNA-seq. RESULT: The establishment of a rat model of lentiviral vector-mediated knockdown of α-Syn in Sprague-Dawley rats with T3 spinal cord contusion (LV_SCI group). The results of the RNA-seq analysis showed that there were 337 differentially expressed genes (DEGs) between the SCI group and the LV_SCI group, and 153 DEGs specific to LV_SCI between the (SCI vs LV_SCI) and (SCI vs CON) comparisons. The top 20 biological transition terms were identified by Gene ontology (GO) analysis. The Kyoto Gene and Genomic Encyclopedia (KEGG) analysis showed that the LV_SCI group significantly upregulated the cholinergic synaptic & nicotine addiction and the neuroactive ligand receptor interaction signaling pathway. Enriched chord analysis analyzes key genes. Further cluster analysis, gene and protein interaction network analysis and RT-qPCR results showed that Chrm2 and Chrnb2 together significantly in both pathways. The proliferation of muscarinic cholinergic receptor subtype 2 (Chrm2) and nicotinic cholinergic receptor subtype ß2 (Chrnb2), and the neurogenesis were elevated in the injury site of LV_SCI group by immunofluorescence. Further by subcellular localization, the LV_SCI group enhanced the expression of Chrnb2 at the cell membrane. CONCLUSION: Knockdown of α-Syn after SCI enhance motor function and promote neurogenesis probably through enhancing cholinergic signaling pathways and neuroreceptor interactions. This study not only further clarifies the understanding of the mechanism of knockdown of α-Syn on SCI but also helps to guide the treatment strategy for SCI.


Subject(s)
Gene Expression Profiling , Spinal Cord Injuries/genetics , Transcriptome , alpha-Synuclein/genetics , Animals , Animals, Genetically Modified , Biomarkers , Cholinergic Neurons/metabolism , Computational Biology/methods , Gene Expression Profiling/methods , Gene Expression Regulation , Gene Knockdown Techniques , Gene Ontology , Gene Regulatory Networks , Neurogenesis/genetics , RNA, Messenger , Rats , Signal Transduction , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
4.
Neural Plast ; 2019: 9610687, 2019.
Article in English | MEDLINE | ID: mdl-30911293

ABSTRACT

MicroRNA-219 (miR-219) regulates the proliferation and differentiation of oligodendrocyte precursor cells (OPCs) during central nervous system (CNS) development. OPCs only differentiate into oligodendrocytes (OLs) in the healthy CNS, but can generate astrocytes (As) after injury. We hypothesized that miR-219 may modulate OPC proliferation and differentiation in a cervical C5 contusion spinal cord injury (SCI) model. After injury, we observed a decrease in the miR-219 level and quantity of OLs and an increase in the number of OPCs and As. Silencing of miR-219 by its antagomir in vivo produced similar results, but of greater magnitude. Overexpression of miR-219 by its agomir in vivo increased the number of OLs and suppressed generation of OPCs and As. Luxol fast blue staining confirmed that SCI caused demyelination and that the extent of demyelination was attenuated by miR-219 overexpression, but aggravated by miR-219 reduction. Monocarboxylate transporter 1 (MCT-1) may be implicated in the regulation of OPC proliferation and differentiation mediated by miR-219 following contusion SCI. Collectively, our data suggest that miR-219 may mediate SCI-induced OPC proliferation and differentiation, and MCT-1 may participate in this process as a target of miR-219.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , MicroRNAs/pharmacology , Oligodendrocyte Precursor Cells/drug effects , Spinal Cord Injuries/pathology , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Male , Oligodendrocyte Precursor Cells/cytology , Oligodendrocyte Precursor Cells/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/metabolism
5.
J Neuroinflammation ; 16(1): 283, 2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31888724

ABSTRACT

BACKGROUND: The prognosis of spinal cord injury (SCI) is closely related to secondary injury, which is dominated by neuroinflammation. There is evidence that α-synuclein aggregates after SCI and that inhibition of α-synuclein aggregation can improve the survival of neurons after SCI, but the mechanism is still unclear. This study was designed to investigate the effects of α-synuclein on neuroinflammation after SCI and to determine the underlying mechanisms. METHOD: A T3 spinal cord contusion model was established in adult male Sprague-Dawley rats. An SNCA-shRNA-carrying lentivirus (LV-SNCA-shRNA) was injected into the injury site to block the expression of α-synuclein (forming the SCI+KD group), and the SCI and sham groups were injected with an empty vector. Basso-Beattie-Bresnahan (BBB) behavioural scores and footprint analysis were used to detect motor function. Inflammatory infiltration and myelin loss were measured in the spinal cord tissues of each group by haematoxylin-eosin (HE) and Luxol Fast Blue (LFB) staining, respectively. Immunohistochemistry, Western blot analysis, and RT-qPCR were used to analyse protein expression and transcription levels in the tissues. Immunofluorescence was used to determine the morphology and function of glial cells and the expression of matrix metalloproteinase-9 in the central canal of the spinal cord. Finally, peripheral serum cytokine levels were determined by enzyme-linked immunosorbent assay. RESULTS: Compared with the SCI group, the SCI+KD group exhibited reduced inflammatory infiltration, preserved myelin, and functional recovery. Specifically, the early arrest of α-synuclein inhibited the pro-inflammatory factors IL-1ß, TNF-α, and IL-2 and increased the expression of the anti-inflammatory factors IL-10, TGF-ß, and IL-4. The neuroinflammatory response was regulated by reduced proliferation of Iba1+ microglia/macrophages and promotion of the shift of M1-polarized Iba1+/iNOS+ microglia/macrophages to M2-polarized Iba1+/Arg1+ microglia/macrophages after injury. In addition, compared with the SCI group, the SCI+KD group also exhibited a smaller microglia/astrocyte (Iba1/GFAP) immunostaining area in the central canal, lower MMP-9 expression, and improved cerebrospinal barrier function. CONCLUSION: Lentivirus-mediated downregulation of α-synuclein reduces neuroinflammation, improves blood-cerebrospinal barrier function, promotes functional recovery, reduces microglial activation, and promotes the polarization of M1 microglia/macrophages to an M2 phenotype to confer a neuroprotective immune microenvironment in rats with SCI.


Subject(s)
Recovery of Function , Spinal Cord Injuries/immunology , Spinal Cord Injuries/metabolism , alpha-Synuclein/antagonists & inhibitors , Animals , Down-Regulation , Genetic Vectors , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lentivirus , Male , RNA, Small Interfering/administration & dosage , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology
6.
Neurosci Lett ; 683: 13-18, 2018 09 14.
Article in English | MEDLINE | ID: mdl-29894768

ABSTRACT

Oxidative stress and inflammation are two key secondary pathological mechanisms following spinal cord injury (SCI). Ketogenic diet (KD) and its metabolite ß-hydroxybutyrate have been found to exhibit anti-oxidative and anti-inflammatory properties both in rats with SCI and in healthy rats; however, the underlying mechanisms are not yet fully understood. We investigated the effects of KD on the suppression of oxidative stress and inflammation, activation of nuclear factor-E2 related factor 2 (Nrf2), and inhibition of the nuclear factor-κB (NF-κB) signaling pathway in rats with SCI. We assessed functional recovery and evaluated the status of oxidative stress and inflammation using tests of superoxide dismutase and myeloperoxidase activity. We further assessed the presence of the proinflammatory cytokines tumor necrosis factor α (TNF-α), interleukin 1ß (IL-1ß), and interferon γ (IFN-γ) by ELISA. Western blotting was used to detect Nrf2 and NF-κB pathway-associated proteins in spinal cord tissue. Finally, we measured the levels of the NF-κB downstream genes TNF-α, IL-1ß, and IFN-γ by western blotting and real-time quantitative PCR. Following SCI, KD improved functional recovery, attenuated oxidative stress and inflammation, and induced Nrf2 activation. In addition, KD suppressed the NF-κB pathway and the expression of TNF-α, IL-1ß, and IFN-γ. Together, these findings provide new insight into the underlying regulatory mechanisms of KD.


Subject(s)
Diet, Ketogenic/methods , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Oxidative Stress/physiology , Spinal Cord Injuries/diet therapy , Spinal Cord Injuries/metabolism , Animals , Diet, Ketogenic/trends , Inflammation/diet therapy , Inflammation/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , NF-kappa B/antagonists & inhibitors , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...