Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 295(50): 16906-16919, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33060198

ABSTRACT

Kinases are critical components of intracellular signaling pathways and have been extensively investigated with regard to their roles in cancer. p21-activated kinase-1 (PAK1) is a serine/threonine kinase that has been previously implicated in numerous biological processes, such as cell migration, cell cycle progression, cell motility, invasion, and angiogenesis, in glioma and other cancers. However, the signaling network linked to PAK1 is not fully defined. We previously reported a large-scale yeast genetic interaction screen using toxicity as a readout to identify candidate PAK1 genetic interactions. En masse transformation of the PAK1 gene into 4,653 homozygous diploid Saccharomyces cerevisiae yeast deletion mutants identified ∼400 candidates that suppressed yeast toxicity. Here we selected 19 candidate PAK1 genetic interactions that had human orthologs and were expressed in glioma for further examination in mammalian cells, brain slice cultures, and orthotopic glioma models. RNAi and pharmacological inhibition of potential PAK1 interactors confirmed that DPP4, KIF11, mTOR, PKM2, SGPP1, TTK, and YWHAE regulate PAK1-induced cell migration and revealed the importance of genes related to the mitotic spindle, proteolysis, autophagy, and metabolism in PAK1-mediated glioma cell migration, drug resistance, and proliferation. AKT1 was further identified as a downstream mediator of the PAK1-TTK genetic interaction. Taken together, these data provide a global view of PAK1-mediated signal transduction pathways and point to potential new drug targets for glioma therapy.


Subject(s)
Cell Movement , Glioma/pathology , Saccharomyces cerevisiae/growth & development , Signal Transduction , Spindle Apparatus/genetics , p21-Activated Kinases/genetics , Animals , Cell Line , Cell Proliferation , Cell Survival , Disease Models, Animal , Epistasis, Genetic , Female , Glioma/genetics , Glioma/metabolism , Humans , Mice , Mice, Inbred C57BL , Mitosis , Protein Kinase Inhibitors/pharmacology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , p21-Activated Kinases/metabolism
2.
Int J Mol Sci ; 22(6)2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33802760

ABSTRACT

Neurotrophic factors (NTFs) are essential for cell growth, survival, synaptic plasticity, and maintenance of specific neuronal population in the central nervous system. Multiple studies have demonstrated that alterations in the levels and activities of NTFs are related to the pathology and symptoms of neurodegenerative disorders, such as Parkinson's disease (PD), Alzheimer's disease (AD), and Huntington's disease. Hence, the key molecule that can regulate the expression of NTFs is an important target for gene therapy coupling adeno-associated virus vector (AAV) gene. We have previously reported that the Ras homolog protein enriched in brain (Rheb)-mammalian target of rapamycin complex 1 (mTORC1) axis plays a vital role in preventing neuronal death in the brain of AD and PD patients. AAV transduction using a constitutively active form of Rheb exerts a neuroprotective effect through the upregulation of NTFs, thereby promoting the neurotrophic interaction between astrocytes and neurons in AD conditions. These findings suggest the role of Rheb as an important regulator of the regulatory system of NTFs to treat neurodegenerative diseases. In this review, we present an overview of the role of Rheb in neurodegenerative diseases and summarize the therapeutic potential of AAV serotype 1 (AAV1)-Rheb(S16H) transduction in the treatment of neurodegenerative disorders, focusing on diseases, such as AD and PD.


Subject(s)
Neurodegenerative Diseases/therapy , Parvovirinae/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Transduction, Genetic , Animals , Dependovirus , Humans , Models, Biological , Nerve Growth Factors/metabolism
3.
Int J Mol Sci ; 22(9)2021 May 03.
Article in English | MEDLINE | ID: mdl-34063708

ABSTRACT

Alzheimer's disease (AD) is the most frequent cause of age-related neurodegeneration and cognitive impairment, and there are currently no broadly effective therapies. The underlying pathogenesis is complex, but a growing body of evidence implicates mitochondrial dysfunction as a common pathomechanism involved in many of the hallmark features of the AD brain, such as formation of amyloid-beta (Aß) aggregates (amyloid plaques), neurofibrillary tangles, cholinergic system dysfunction, impaired synaptic transmission and plasticity, oxidative stress, and neuroinflammation, that lead to neurodegeneration and cognitive dysfunction. Indeed, mitochondrial dysfunction concomitant with progressive accumulation of mitochondrial Aß is an early event in AD pathogenesis. Healthy mitochondria are critical for providing sufficient energy to maintain endogenous neuroprotective and reparative mechanisms, while disturbances in mitochondrial function, motility, fission, and fusion lead to neuronal malfunction and degeneration associated with excess free radical production and reduced intracellular calcium buffering. In addition, mitochondrial dysfunction can contribute to amyloid-ß precursor protein (APP) expression and misprocessing to produce pathogenic fragments (e.g., Aß1-40). Given this background, we present an overview of the importance of mitochondria for maintenance of neuronal function and how mitochondrial dysfunction acts as a driver of cognitive impairment in AD. Additionally, we provide a brief summary of possible treatments targeting mitochondrial dysfunction as therapeutic approaches for AD.


Subject(s)
Alzheimer Disease/genetics , Cognitive Dysfunction/genetics , Oxidative Stress/genetics , Plaque, Amyloid/genetics , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Cognitive Dysfunction/pathology , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
4.
Medicina (Kaunas) ; 57(4)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33915966

ABSTRACT

To date, there is no curable treatment option for non-hereditary degenerative cerebellar ataxia. Here we report the case of a patient with sporadic adult-onset ataxia (SAOA) who underwent allogeneic bone marrow-derived mesenchymal stem cell (MSC) therapy via the intrathecal route. A 60-year-old male patient visited our clinic complaining of progressive gait disturbance that commenced two years ago. Upon neurologic examination, the patient exhibited limb dysmetria and gait ataxia. Brain magnetic resonance imaging (MRI) revealed cerebellar atrophy whereas the autonomic function test was normal. The patient was diagnosed with SAOA. The medications that were initially prescribed had no significant effects on the course of this disease and the symptoms deteriorated progressively. At the age of 64, the patient was treated with allogeneic bone marrow-derived MSC therapy. The subsequent K-SARA (Korean version of the Scale for the Assessment and Rating of Ataxia) scores demonstrated a distinct improvement up until 10 months post-administration. No adverse events were reported. The improved post-treatment K-SARA scores may suggest that the MSC therapy can have a neuroprotective effect and that stem cell therapy may serve as a potential therapeutic option for degenerative cerebellar ataxia.


Subject(s)
Cerebellar Ataxia , Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Adult , Bone Marrow , Cerebellar Ataxia/therapy , Humans , Male , Middle Aged
5.
Biochem Biophys Res Commun ; 529(1): 70-76, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32560822

ABSTRACT

Pathogenic bacteria acquire the acquisition of iron from the host to ensure their survival. Salmonella spp. utilizes siderophores, including salmochelin, for high affinity aggressive import of iron. Although the iroBCDEN operon is reportedly responsible for the production and the transport of salmochelin, the molecular mechanisms underlying the regulation of its gene expression have not yet been characterized. Here, we analyzed the expression pattern of iroB using the lacZY transcriptional reporter system and determined the transcription start site in response to iron availability using primer extension analysis. We further examined the regulation of iroB expression by the ferric uptake regulator (Fur), a key regulatory protein involved in the maintenance of iron homeostasis in various bacteria, including Salmonella. Using sequence analysis followed by a gel shift assay, we verified that the Fur box lies within the promoter region of iroBCDE. The Fur box contained the consensus sequence (GATATTGGTAATTATTATC) and overlapped with the -10-element region. The expression of iroB was repressed by Fur in the presence of iron, as determined using an in vitro transcription assay. Therefore, we found that the iron acquisition system is regulated in a Fur-dependent manner in Salmonella.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Enterobactin/analogs & derivatives , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Bacterial Proteins/chemistry , Base Sequence , Biobehavioral Sciences , Consensus Sequence , DNA, Bacterial/genetics , Enterobactin/metabolism , Gene Expression Regulation, Bacterial , Humans , Iron/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Multimerization , Repressor Proteins/chemistry , Siderophores/metabolism , Transcription, Genetic
6.
J Neuroinflammation ; 17(1): 299, 2020 Oct 14.
Article in English | MEDLINE | ID: mdl-33054766

ABSTRACT

BACKGROUND: Cytoplasmic inclusions of transactive response DNA binding protein of 43 kDa (TDP-43) in neurons and astrocytes are a feature of some neurodegenerative diseases, such as frontotemporal lobar degeneration with TDP-43 (FTLD-TDP) and amyotrophic lateral sclerosis (ALS). However, the role of TDP-43 in astrocyte pathology remains largely unknown. METHODS: To investigate whether TDP-43 overexpression in primary astrocytes could induce inflammation, we transfected primary astrocytes with plasmids encoding Gfp or TDP-43-Gfp. The inflammatory response and upregulation of PTP1B in transfected cells were examined using quantitative RT-PCR and immunoblot analysis. Neurotoxicity was analysed in a transwell coculture system of primary cortical neurons with astrocytes and cultured neurons treated with astrocyte-conditioned medium (ACM). We also examined the lifespan, performed climbing assays and analysed immunohistochemical data in pan-glial TDP-43-expressing flies in the presence or absence of a Ptp61f RNAi transgene. RESULTS: PTP1B inhibition suppressed TDP-43-induced secretion of inflammatory cytokines (interleukin 1 beta (IL-1ß), interleukin 6 (IL-6) and tumour necrosis factor alpha (TNF-α)) in primary astrocytes. Using a neuron-astrocyte coculture system and astrocyte-conditioned media treatment, we demonstrated that PTP1B inhibition attenuated neuronal death and mitochondrial dysfunction caused by overexpression of TDP-43 in astrocytes. In addition, neuromuscular junction (NMJ) defects, a shortened lifespan, inflammation and climbing defects caused by pan-glial overexpression of TDP-43 were significantly rescued by downregulation of ptp61f (the Drosophila homologue of PTP1B) in flies. CONCLUSIONS: These results indicate that PTP1B inhibition mitigates the neuronal toxicity caused by TDP-43-induced inflammation in mammalian astrocytes and Drosophila glial cells.


Subject(s)
Astrocytes/metabolism , DNA-Binding Proteins/biosynthesis , Inflammation Mediators/metabolism , Nerve Degeneration/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/biosynthesis , Animals , Animals, Genetically Modified , Astrocytes/pathology , Cells, Cultured , DNA-Binding Proteins/genetics , Drosophila , Gene Expression , Inflammation Mediators/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics
7.
Int J Mol Sci ; 21(5)2020 Mar 09.
Article in English | MEDLINE | ID: mdl-32182914

ABSTRACT

To test the hypothesis that myricitrin (MYR) improves type 2 diabetes, we examined the effect of MYR on hyperglycemia, glucose intolerance, hepatic steatosis, and inflammation in high-fat diet (HFD) and streptozotocin (STZ)-induced type 2 diabetic mice. Male C57BL/6J mice were randomly divided into three groups: non-diabetic, diabetic control, and MYR (0.005%, w/w)-supplemented diabetic groups. Diabetes was induced by HFD and STZ, and MYR was administered orally for 5 weeks. Myricitrin exerted no significant effects on food intake, body weight, fat weight, or plasma lipids levels. However, MYR significantly decreased fasting blood glucose levels, improved glucose intolerance, and increased pancreatic ß-cell mass compared to the diabetic control group. Myricitrin administration also markedly increased glucokinase mRNA expression and activity as well as lowered glucose-6-phosphatase and phosphoenolpyruvate carboxykinase mRNA expression and activity in the liver. In addition, liver weight, hepatic triglyceride content, and lipid droplet accumulation were markedly decreased following MYR administration. These changes were seemingly attributable to the suppression of the hepatic lipogenic enzymes-fatty acid synthase and phosphatidate phosphohydrolase. Myricitrin also significantly lowered plasma MCP-1 and TNF-α levels and the mRNA expression of hepatic pro-inflammatory genes. These results suggest that MYR has anti-diabetic potential.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Fatty Liver/drug therapy , Flavonoids/pharmacology , Glucose Intolerance/drug therapy , Hyperglycemia/drug therapy , Inflammation/drug therapy , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/drug therapy , Diet, High-Fat/adverse effects , Fatty Liver/metabolism , Glucokinase/metabolism , Glucose Intolerance/metabolism , Hyperglycemia/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Insulin Resistance , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Streptozocin/pharmacology , Triglycerides/metabolism
8.
Int J Mol Sci ; 21(6)2020 Mar 16.
Article in English | MEDLINE | ID: mdl-32188096

ABSTRACT

Ras homolog protein enriched in brain (Rheb) is a key activator of mammalian target of rapamycin complex 1 (mTORC1). The activation of mTORC1 by Rheb is associated with various processes such as protein synthesis, neuronal growth, differentiation, axonal regeneration, energy homeostasis, autophagy, and amino acid uptake. In addition, Rheb-mTORC1 signaling plays a crucial role in preventing the neurodegeneration of hippocampal neurons in the adult brain. Increasing evidence suggests that the constitutive activation of Rheb has beneficial effects against neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD). Our recent studies revealed that adeno-associated virus serotype 1 (AAV1) transduction with Rheb(S16H), a constitutively active form of Rheb, exhibits neuroprotective properties through the induction of various neurotrophic factors, promoting neurotrophic interactions between neurons and astrocytes in the hippocampus of the adult brain. This review provides compelling evidence for the therapeutic potential of AAV1-Rheb(S16H) transduction in the hippocampus of the adult brain by exploring its neuroprotective effects and mechanisms.


Subject(s)
Brain/metabolism , Hippocampus/metabolism , Neuroprotective Agents/metabolism , Ras Homolog Enriched in Brain Protein/genetics , Ras Homolog Enriched in Brain Protein/metabolism , Alzheimer Disease/metabolism , Animals , Astrocytes/drug effects , Brain/drug effects , Dependovirus , Hippocampus/drug effects , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease/metabolism , Parvovirinae , Ras Homolog Enriched in Brain Protein/pharmacology , Ras Homolog Enriched in Brain Protein/therapeutic use , Signal Transduction , Up-Regulation
9.
Biochem Biophys Res Commun ; 513(4): 925-932, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31005259

ABSTRACT

The most prominent hallmarks of many neurodegenerative diseases are the accumulation of misfolded protein aggregates and the death of certain neuronal populations. Autophagy is the major intracellular mechanism that degrades protein aggregates and damaged cellular components. Many studies have reported that the dysfunction of autophagy is associated with several neurodegenerative diseases, such as Alzheimer's disease, amyotrophic lateral sclerosis (ALS), and Parkinson's disease. Here, we identified a novel mechanism of autophagy regulation. Inhibition of MEK5 reduced the level of p62 and increased the ratio of LC3-II to LC3-I, which is a marker for the activation of the autophagy-lysosome pathway (ALP). One of the most well-known regulators of the ALP is mTOR, and previous studies have reported that the major substrate of MEK5 is ERK5. However, we found that MEK5 modulates the autophagy-lysosome pathway in an mTOR- and ERK5-independent manner. Moreover, MEK5 inhibition alleviated the mislocalization of TDP-43 (an ALS-associated protein) and cell death in TDP-43-GFP-expressing neuronal cells. Taken together, these findings suggest that MEK5 is a novel autophagy modulator and that this kinase could be a therapeutic target for neurodegenerative diseases such as amyotrophic lateral sclerosis.


Subject(s)
Autophagy , DNA-Binding Proteins/toxicity , Lysosomes/metabolism , MAP Kinase Kinase 5/antagonists & inhibitors , Metabolic Networks and Pathways/physiology , Neurons/cytology , Animals , Humans , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , RNA-Binding Proteins/metabolism , TOR Serine-Threonine Kinases/physiology
10.
Int J Mol Sci ; 21(1)2019 Dec 20.
Article in English | MEDLINE | ID: mdl-31861806

ABSTRACT

Developmentally regulated GTP-binding protein 2 (DRG2) was first identified in the central nervous system of mice. However, the physiological function of DRG2 in the brain remains largely unknown. Here, we demonstrated that knocking out DRG2 impairs the function of dopamine neurons in mice. DRG2 was strongly expressed in the neurons of the dopaminergic system such as those in the striatum (Str), ventral tegmental area (VTA), and substantia nigra (SN), and on neuronal cell bodies in high-density regions such as the hippocampus (HIP), cerebellum, and cerebral cortex in the mouse brain. DRG2 knockout (KO) mice displayed defects in motor function in motor coordination and rotarod tests and increased anxiety. However, unexpectedly, DRG2 depletion did not affect the dopamine (DA) neuron population in the SN, Str, or VTA region or dopamine synthesis in the Str region. We further demonstrated that dopamine release was significantly diminished in the Str region of DRG2 KO mice and that treatment of DRG2 KO mice with l-3,4-dihydroxyphenylalanine (L-DOPA), a dopamine precursor, rescued the behavioral motor deficiency in DRG2 KO mice as observed with the rotarod test. This is the first report to identify DRG2 as a key regulator of dopamine release from dopamine neurons in the mouse brain.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , GTP-Binding Proteins/genetics , Motor Disorders/genetics , Animals , Anxiety/genetics , Anxiety/metabolism , Corpus Striatum/cytology , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , GTP-Binding Proteins/analysis , GTP-Binding Proteins/metabolism , Gene Deletion , Mice , Mice, Knockout , Motor Disorders/metabolism
11.
J Neurosci ; 36(20): 5608-22, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27194339

ABSTRACT

UNLABELLED: Lipocalin-2 (LCN2) is a member of the highly heterogeneous secretory protein family of lipocalins and increases in its levels can contribute to neurodegeneration in the adult brain. However, there are no reports on the role of LCN2 in Parkinson's disease (PD). Here, we report for the first time that LCN2 expression is increased in the substantia nigra (SN) of patients with PD. In mouse brains, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment for a neurotoxin model of PD significantly upregulated LCN2 expression, mainly in reactive astrocytes in both the SN and striatum. The increased LCN2 levels contributed to neurotoxicity and neuroinflammation, resulting in disruption of the nigrostriatal dopaminergic (DA) projection and abnormal locomotor behaviors, which were ameliorated in LCN2-deficient mice. Similar to the effects of MPTP treatment, LCN2-induced neurotoxicity was also observed in the 6-hydroxydopamine (6-OHDA)-treated animal model of PD. Moreover, treatment with the iron donor ferric citrate (FC) and the iron chelator deferoxamine mesylate (DFO) increased and decreased, respectively, the LCN2-induced neurotoxicity in vivo In addition to the in vivo results, 1-methyl-4-phenylpyridinium (MPP(+))-induced neurotoxicity in cocultures of mesencephalic neurons and astrocytes was reduced by LCN2 gene deficiency in the astrocytes and conditioned media derived from MPP(+)-treated SH-SY5Y neuronal enhanced glial expression of LCN2 in vitro Therefore, our results demonstrate that astrocytic LCN2 upregulation in the lesioned DA system may play a role as a potential pathogenic factor in PD and suggest that inhibition of LCN2 expression or activity may be useful in protecting the nigrostriatal DA system in the adult brain. SIGNIFICANCE STATEMENT: Lipocalin-2 (LCN2), a member of the highly heterogeneous secretory protein family of lipocalins, may contribute to neuroinflammation and neurotoxicity in the brain. However, LCN2 expression and its role in Parkinson's disease (PD) are largely unknown. Here, we report that LCN2 is upregulated in the substantia nigra of patients with PD and neurotoxin-treated animal models of PD. Our results suggest that LCN2 upregulation might be a potential pathogenic mechanism of PD, which would result in disruption of the nigrostriatal dopaminergic system through neurotoxic iron accumulation and neuroinflammation. Therefore, inhibition of LCN2 expression or activity may be useful in protecting the nigrostriatal dopaminergic projection in PD.


Subject(s)
Lipocalin-2/metabolism , Neuroglia/metabolism , Parkinson Disease/metabolism , Up-Regulation , Aged , Aged, 80 and over , Animals , Case-Control Studies , Dopaminergic Neurons/metabolism , Female , Humans , Lipocalin-2/genetics , MPTP Poisoning/metabolism , Male , Mice , Mice, Inbred C57BL , Parkinson Disease/pathology , Substantia Nigra/cytology , Substantia Nigra/metabolism
12.
Mol Ther ; 23(3): 445-55, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25502903

ABSTRACT

Recent evidence has shown that Ras homolog enriched in brain (Rheb) is dysregulated in Alzheimer's disease (AD) brains. However, it is still unclear whether Rheb activation contributes to the survival and protection of hippocampal neurons in the adult brain. To assess the effects of active Rheb in hippocampal neurons in vivo, we transfected neurons in the cornu ammonis 1 (CA1) region in normal adult rats with an adeno-associated virus containing the constitutively active human Rheb (hRheb(S16H)) and evaluated the effects on thrombin-induced neurotoxicity. Transduction with hRheb(S16H) significantly induced neurotrophic effects in hippocampal neurons through activation of mammalian target of rapamycin complex 1 (mTORC1) without side effects such as long-term potentiation impairment and seizures from the alteration of cytoarchitecture, and the expression of hRheb(S16H) prevented thrombin-induced neurodegeneration in vivo, an effect that was diminished by treatment with specific neutralizing antibodies against brain-derived neurotrophic factor (BDNF). In addition, our results showed that the basal mTORC1 activity might be insufficient to mediate the level of BDNF expression, but hRheb(S16H)-activated mTORC1 stimulated BDNF production in hippocampal neurons. These results suggest that viral vector transduction with hRheb(S16H) may have therapeutic value in the treatment of neurodegenerative diseases such as AD.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , CA1 Region, Hippocampal/metabolism , Monomeric GTP-Binding Proteins/genetics , Neurons/metabolism , Neuropeptides/genetics , Transduction, Genetic/methods , Animals , Antibodies, Neutralizing/pharmacology , Brain-Derived Neurotrophic Factor/agonists , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/genetics , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , Dependovirus/genetics , Dependovirus/metabolism , Gene Expression , Genetic Vectors/administration & dosage , Humans , Mechanistic Target of Rapamycin Complex 1 , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes/agonists , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neurons/cytology , Neurons/drug effects , Neuropeptides/metabolism , Ras Homolog Enriched in Brain Protein , Rats , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Thrombin/antagonists & inhibitors , Thrombin/toxicity
14.
Br J Nutr ; 113(6): 867-77, 2015 Mar 28.
Article in English | MEDLINE | ID: mdl-25744306

ABSTRACT

The aim of the present study was to identify the genes differentially expressed in the visceral adipose tissue in a well-characterised mouse model of high-fat diet (HFD)-induced obesity. Male C57BL/6J mice (n 20) were fed either HFD (189 % of energy from fat) or low-fat diet (LFD, 42 % of energy from fat) for 16 weeks. HFD-fed mice exhibited obesity, insulin resistance, dyslipidaemia and adipose collagen accumulation, along with higher levels of plasma leptin, resistin and plasminogen activator inhibitor type 1, although there were no significant differences in plasma cytokine levels. Energy intake was similar in the two diet groups owing to lower food intake in the HFD group; however, energy expenditure was also lower in the HFD group than in the LFD group. Microarray analysis revealed that genes related to lipolysis, fatty acid metabolism, mitochondrial energy transduction, oxidation-reduction, insulin sensitivity and skeletal system development were down-regulated in HFD-fed mice, and genes associated with extracellular matrix (ECM) components, ECM remodelling and inflammation were up-regulated. The top ten up- or down-regulated genes include Acsm3, mt-Nd6, Fam13a, Cyp2e1, Rgs1 and Gpnmb, whose roles in the deterioration of obesity-associated adipose tissue are poorly understood. In conclusion, the genes identified here provide new therapeutic opportunities for prevention and treatment of diet-induced obesity.


Subject(s)
Adipose Tissue, White/metabolism , Coenzyme A Ligases/metabolism , Cytochrome P-450 CYP2E1/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation, Enzymologic , NADH Dehydrogenase/metabolism , Obesity/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/pathology , Adiposity , Animals , Biomarkers/blood , Biomarkers/metabolism , Coenzyme A Ligases/genetics , Cytochrome P-450 CYP2E1/genetics , Diet, High-Fat/adverse effects , Energy Metabolism , Extracellular Matrix/immunology , Extracellular Matrix/pathology , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Insulin Resistance , Male , Mice, Inbred C57BL , Mitochondria/enzymology , Mitochondria/immunology , Mitochondria/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , NADH Dehydrogenase/genetics , Obesity/etiology , Obesity/immunology , Obesity/pathology
15.
Biomolecules ; 14(3)2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38540798

ABSTRACT

Although granule cell dispersion (GCD) in the hippocampus is known to be an important feature associated with epileptic seizures in temporal lobe epilepsy (TLE), the endogenous molecules that regulate GCD are largely unknown. In the present study, we have examined whether there is any change in AEG-1 expression in the hippocampus of a kainic acid (KA)-induced mouse model of TLE. In addition, we have investigated whether the modulation of astrocyte elevated gene-1 (AEG-1) expression in the dentate gyrus (DG) by intracranial injection of adeno-associated virus 1 (AAV1) influences pathological phenotypes such as GCD formation and seizure susceptibility in a KA-treated mouse. We have identified that the protein expression of AEG-1 is upregulated in the DG of a KA-induced mouse model of TLE. We further demonstrated that AEG-1 upregulation by AAV1 delivery in the DG-induced anticonvulsant activities such as the delay of seizure onset and inhibition of spontaneous recurrent seizures (SRS) through GCD suppression in the mouse model of TLE, while the inhibition of AEG-1 expression increased susceptibility to seizures. The present observations suggest that AEG-1 is a potent regulator of GCD formation and seizure development associated with TLE, and the significant induction of AEG-1 in the DG may have therapeutic potential against epilepsy.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Animals , Mice , Astrocytes/metabolism , Dentate Gyrus/metabolism , Epilepsy/metabolism , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/drug therapy , Hippocampus/metabolism , Kainic Acid/adverse effects , Kainic Acid/metabolism , Seizures/chemically induced , Seizures/genetics , Seizures/metabolism
16.
Nanoscale ; 16(2): 833-847, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38093712

ABSTRACT

Astrocytes are highly activated following brain injuries, and their activation influences neuronal survival. Additionally, SOX9 expression is known to increase in reactive astrocytes. However, the role of SOX9 in activated astrocytes following ischemic brain damage has not been clearly elucidated yet. Therefore, in the present study, we investigated the role of SOX9 in reactive astrocytes using a poly-lactic-co-glycolic acid (PLGA) nanoparticle plasmid delivery system in a photothrombotic stroke animal model. We designed PLGA nanoparticles to exclusively enhance SOX9 gene expression in glial fibrillary acidic protein (GFAP)-immunoreactive astrocytes. Our observations indicate that PLGA nanoparticles encapsulated with GFAP:SOX9:tdTOM reduce ischemia-induced neurological deficits and infarct volume through the prostaglandin D2 pathway. Thus, the astrocyte-targeting PLGA nanoparticle plasmid delivery system provides a potential opportunity for stroke treatment. Since the only effective treatment currently available is reinstating the blood supply, cell-specific gene therapy using PLGA nanoparticles will open a new therapeutic paradigm for brain injury patients in the future.


Subject(s)
Brain Injuries , Nanoparticles , Stroke , Humans , Animals , Astrocytes/metabolism , Stroke/therapy , Stroke/genetics , Stroke/metabolism , Brain Injuries/metabolism , Peptides/pharmacology , Brain/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/pharmacology
17.
Mol Ther ; 20(2): 275-86, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22008911

ABSTRACT

There are currently no therapies that provide either protection or restoration of neuronal function for adult-onset neurodegenerative diseases such as Parkinson's disease (PD). Many clinical efforts to provide such benefits by infusion of neurotrophic factors have failed, in spite of robust effects in preclinical assessments. One important reason for these failures is the difficulty, due to diffusion limits, of providing these protein molecules in sufficient amounts to the intended cellular targets in the central nervous system. This challenge suggests an alternative approach, that of viral vector transduction to directly activate the intracellular signaling pathways that mediate neurotrophic effects. To this end we have investigated the ability of a constitutively active form of the GTPase Rheb, an important activator of mammalian target of rapamycin (mTor) signaling, to mediate neurotrophic effects in dopamine neurons of the substantia nigra (SN), a population of neurons affected in PD. We find that constitutively active hRheb(S16H) induces many neurotrophic effects in mice, including abilities to both preserve and restore the nigrostriatal dopaminergic axonal projections in a highly destructive neurotoxin model. We conclude that direct viral vector transduction of vulnerable neuronal populations to activate intracellular neurotrophic signaling pathways offers promise for the treatment of neurodegenerative disease.


Subject(s)
Axons/metabolism , Dependovirus/genetics , Dopaminergic Neurons/metabolism , Genetic Vectors/genetics , Monomeric GTP-Binding Proteins/genetics , Neuropeptides/genetics , Parkinson Disease/prevention & control , Transduction, Genetic , Adaptor Proteins, Signal Transducing , Animals , Axons/drug effects , Carrier Proteins/metabolism , Cell Cycle Proteins , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Eukaryotic Initiation Factors , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Male , Mice , Mice, Inbred C57BL , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/metabolism , Oxidopamine/adverse effects , Parkinson Disease/therapy , Phosphoproteins/metabolism , Phosphorylation , Ras Homolog Enriched in Brain Protein , Signal Transduction , Substantia Nigra/drug effects , Substantia Nigra/metabolism
18.
Biomedicines ; 11(5)2023 May 07.
Article in English | MEDLINE | ID: mdl-37239054

ABSTRACT

The blood-brain barrier (BBB) restricts entry of neurotoxic plasma components, blood cells, and pathogens into the brain, leading to proper neuronal functioning. BBB impairment leads to blood-borne protein infiltration such as prothrombin, thrombin, prothrombin kringle-2, fibrinogen, fibrin, and other harmful substances. Thus, microglial activation and release of pro-inflammatory mediators commence, resulting in neuronal damage and leading to impaired cognition via neuroinflammatory responses, which are important features observed in the brain of Alzheimer's disease (AD) patients. Moreover, these blood-borne proteins cluster with the amyloid beta plaque in the brain, exacerbating microglial activation, neuroinflammation, tau phosphorylation, and oxidative stress. These mechanisms work in concert and reinforce each other, contributing to the typical pathological changes in AD in the brain. Therefore, the identification of blood-borne proteins and the mechanisms involved in microglial activation and neuroinflammatory damage can be a promising therapeutic strategy for AD prevention. In this article, we review the current knowledge regarding the mechanisms of microglial activation-mediated neuroinflammation caused by the influx of blood-borne proteins into the brain via BBB disruption. Subsequently, the mechanisms of drugs that inhibit blood-borne proteins, as a potential therapeutic approach for AD, along with the limitations and potential challenges of these approaches, are also summarized.

19.
Brain Behav Immun Health ; 28: 100593, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36798617

ABSTRACT

We recently demonstrated that prothrombin kringle-2 (pKr-2) derived from blood-brain barrier (BBB) disruption could induce hippocampal neurodegeneration and object recognition impairment through neurotoxic inflammatory responses in the five familial Alzheimer's disease mutation (5XFAD) mice. In the present study, we aimed to determine whether pKr-2 induces microglial activation by stimulating toll-like receptor 4 (TLR4) upregulation and examine whether this response contributes to pKr-2-induced neuroinflammatory damage in the hippocampi of mice models. We observed that inflammatory responses induced by pKr-2 administration in the hippocampi of wild-type mice were significantly abrogated in TLR4-deficient mice (TLR4-/-), and caffeine supply or rivaroxaban treatment that inhibits the overexpression of hippocampal pKr-2 reduced TLR4 upregulation in 5XFAD mice, resulting in the inhibition of neuroinflammatory responses. Similar to the expression patterns of pKr-2, TLR4, and the TLR4 transcription factors, PU.1 and p-c-Jun, seen in the postmortem hippocampal tissues of Alzheimer's disease (AD) patients, our results additionally showed the influence of transcriptional regulation on TLR4 expression following pKr-2 expression in triggering the production of neurotoxic inflammatory mediators. Therefore, we conclude that pKr-2 may play a role in initiating upregulation of microglial TLR4, consequently inducing hippocampal neurodegeneration. Furthermore, the control of pKr-2-induced microglial TLR4 could be a useful therapeutic strategy against hippocampal neurodegeneration in AD.

20.
Brain Struct Funct ; 228(7): 1629-1641, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37421418

ABSTRACT

Astrocyte elevated gene-1 (AEG-1) is a well-known oncogene implicated in various types of human cancers, including brain tumors. Recently, AEG-1 has also been reported to play pivotal roles in glioma-associated neurodegeneration and neurodegenerative diseases like Parkinson's disease and amyotrophic lateral sclerosis. However, the normal physiological functions and expression patterns of AEG-1 in the brain are not well understood. In this study, we investigated the expression patterns of AEG-1 in the normal mouse brain and found that AEG-1 is widely expressed in neurons and neuronal precursor cells, but little in glial cells. We observed differential expression levels of AEG-1 in various brain regions, and its expression was mainly localized in the cell body of neurons rather than the nucleus. Additionally, AEG-1 was expressed in the cytoplasm of Purkinje cells in both the mouse and human cerebellum, suggesting its potential role in this brain region. These findings suggest that AEG-1 may have important functions in normal brain physiology and warrant further investigation. Our results may also shed light on the differential expression patterns of AEG-1 in normal and pathological brains, providing insights into its roles in various neurological disorders.


Subject(s)
Brain , Membrane Proteins , Animals , Humans , Mice , Brain/metabolism , Brain Neoplasms/pathology , Cell Adhesion Molecules/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL