Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Neuroradiol J ; : 19714009241242642, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38565221

ABSTRACT

BACKGROUND AND PURPOSE: Perivascular spaces (PVS) are interstitial fluid-filled spaces surrounding blood vessels traversing the deep gray nuclei and white matter of the brain. These are commonly encountered on CT and MR imaging and are generally asymptomatic and of no clinical significance. However, occasional changes in the size of focal PVS, for example, when enlarging, may mimic pathologies including neoplasms and infections, hence potentially confounding radiological interpretation. Given these potential diagnostic issues, we sought to better characterize common clinical and imaging features of focal PVS demonstrating size fluctuations. MATERIALS AND METHODS: Upon institutional approval, we retrospectively identified 4 cases demonstrating PVS with size changes at our institution. To supplement our cases, we also performed a literature review, which identified an additional 14 cases. Their clinical and imaging data were analyzed to identify characteristic features. RESULTS: Of the 18 total cases (including the 4 institutional cases), 10 cases increased and 8 decreased in size. These focal PVS ranged from 0.4-4.5 cm in size. Whereas a decrease in size did not represent a diagnostic issue, focal increase in size of PVS led to concerning differential diagnoses in at least 30% of the radiology reports. These enlarging PVS were most found in the basal ganglia and temporal lobe, and in patients with previous brain radiation treatment. CONCLUSION: Focal size change of PVS can occur, especially years after brain radiation treatment. Being cognizant of this benign finding is important to consider in the differential diagnosis to avoid undue patient anxiety or unnecessary medical intervention.

2.
Article in English | MEDLINE | ID: mdl-38521092

ABSTRACT

BACKGROUND AND PURPOSE: Interest in artificial intelligence (AI) and machine learning (ML) has been growing in neuroradiology, but there is limited knowledge on how this interest has manifested into research and specifically, its qualities and characteristics. This study aims to characterize the emergence and evolution of AI/ML articles within neuroradiology and provide a comprehensive overview of the trends, challenges, and future directions of the field. MATERIALS AND METHODS: We performed a bibliometric analysis of the American Journal of Neuroradiology (AJNR): the journal was queried for original research articles published since inception (Jan. 1, 1980) to Dec. 3, 2022 that contained any of the following key terms: "machine learning", "artificial intelligence", "radiomics", "deep learning", "neural network", "generative adversarial network", "object detection", or "natural language processing". Articles were screened by two independent reviewers, and categorized into Statistical Modelling (Type 1), AI/ML Development (Type 2), both representing developmental research work but without a direct clinical integration, or End-user Application (Type 3) which is the closest surrogate of potential AI/ML integration into day-to-day practice. To better understand the limiting factors to Type 3 articles being published, we analyzed Type 2 articles as they should represent the precursor work leading to Type 3. RESULTS: A total of 182 articles were identified with 79% being non-integration focused (Type 1 n = 53, Type 2 n = 90) and 21% (n = 39) being Type 3. The total number of articles published grew roughly five-fold in the last five years, with the non-integration focused articles mainly driving this growth. Additionally, a minority of Type 2 articles addressed bias (22%) and explainability (16%). These articles were primarily led by radiologists (63%), with most of them (60%) having additional postgraduate degrees. CONCLUSIONS: AI/ML publications have been rapidly increasing in neuroradiology with only a minority of this growth being attributable to end-user application. Areas identified for improvement include enhancing the quality of Type 2 articles, namely external validation, and addressing both bias and explainability. These results ultimately provide authors, editors, clinicians, and policymakers important insights to promote a shift towards integrating practical AI/ML solutions in neuroradiology. ABBREVIATIONS: AI = artificial intelligence; ML = machine learning.

4.
Mol Psychiatry ; 28(10): 4474-4484, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37648779

ABSTRACT

Mitochondrial dysfunction has been implicated in Parkinson's Disease (PD) progression; however, the mitochondrial factors underlying the development of PD symptoms remain unclear. One candidate is CR6-interacting factor1 (CRIF1), which controls translation and membrane insertion of 13 mitochondrial proteins involved in oxidative phosphorylation. Here, we found that CRIF1 mRNA and protein expression were significantly reduced in postmortem brains of elderly PD patients compared to normal controls. To evaluate the effect of Crif1 deficiency, we produced mice lacking the Crif1 gene in dopaminergic neurons (DAT-CRIF1-KO mice). From 5 weeks of age, DAT-CRIF1-KO mice began to show decreased dopamine production with progressive neuronal degeneration in the nigral area. At ~10 weeks of age, they developed PD-like behavioral deficits, including gait abnormalities, rigidity, and resting tremor. L-DOPA, a medication used to treat PD, ameliorated these defects at an early stage, although it was ineffective in older mice. Taken together, the observation that CRIF1 expression is reduced in human PD brains and deletion of CRIF1 in dopaminergic neurons leads to early-onset PD with stepwise PD progression support the conclusion that CRIF1-mediated mitochondrial function is important for the survival of dopaminergic neurons.


Subject(s)
Dopaminergic Neurons , Parkinson Disease , Humans , Mice , Animals , Aged , Dopaminergic Neurons/metabolism , Parkinson Disease/genetics , Levodopa/pharmacology , Dopamine/metabolism , Brain/metabolism , Cell Cycle Proteins/genetics
5.
Stem Cell Res Ther ; 14(1): 178, 2023 07 19.
Article in English | MEDLINE | ID: mdl-37468918

ABSTRACT

BACKGROUND: Preclinical studies showed that mesenchymal stem cells (MSCs) ameliorate tau phosphorylation, amyloid-beta accumulation, and inflammation in Alzheimer's disease (AD) mouse models via secretion of neurotrophic factors and cytokines. We aimed to identify CSF biomarkers that can be used to predict or monitor the response to MSCs in patients with AD. METHODS: AD patients were injected with human umbilical cord blood-MSCs (n = 22) or placebo (n = 12). The cerebrospinal fluid (CSF) samples were collected at baseline, one day after the first injection, and one day after the third injection. The patients injected with MSCs were classified into good responder (GR) or poor responder (PR) groups based on the rate of changes in the ratio of total-tau and phosphorylated-tau in the CSF. We selected three typical participants in each group, and their CSF protein levels were analyzed using liquid chromatography/tandem mass spectrometry (LC-MS/MS). RESULTS: In the LC-MS/MS analysis, 1,667 proteins were identified. Eleven proteins showed significant differences between the typical GR and PR at baseline. Based on their significance level and known functions, two proteins, reticulocalbin-3 (RCN3) and follistatin-related protein 3 (FSTL3), were selected as potential biomarkers to predict MSC response. A total of 173 proteins showed significant change one day after the third injection compared to the baseline in typical GR. We excluded 45 proteins that showed significant change after the third injection compared to the baseline in the typical PR. Based on their significance level and known function, four proteins, scrapie-responsive protein 1 (SCRG1), neural proliferation differentiation and control protein (NPDC1), apolipoprotein E (ApoE), and cystatin C (CysC), were selected as potential biomarker to monitor MSC response. Additionally, functional analysis revealed that the increased CSF proteins after the third injection compared to the baseline in the typical GR were associated with synaptogenesis. CONCLUSIONS: This study identified two proteins (RCN3 and FSTL3) that may be potential biomarkers for predicting MSC response and four proteins (SCRG1, NPDC1, ApoE, CysC) that may be potential biomarkers for monitoring MSC response in patients with AD. Further studies are needed to validate our results. Trial registration Clinical Trials.gov, NCT02054208. Registered on 4 February 2014. Samsung Medical Center IRB File No.2017-04-025. Registered on 20 June 2017.


Subject(s)
Alzheimer Disease , Animals , Mice , Humans , Alzheimer Disease/therapy , Chromatography, Liquid , tau Proteins/genetics , tau Proteins/metabolism , Tandem Mass Spectrometry , Amyloid beta-Peptides , Apolipoproteins E/metabolism , Biomarkers , Peptide Fragments , Calcium-Binding Proteins
6.
Biomedicines ; 10(8)2022 Aug 04.
Article in English | MEDLINE | ID: mdl-36009433

ABSTRACT

We have recently reported on how transplantation of human mesenchymal stem cells (MSCs) into the mouse parenchyma generated immune responses. To facilitate the clinical translation of MSC-based AD therapy, the safety and efficacy of human derived MSCs (hMSCs) must be confirmed in the pre-clinical stage. Thus, it is imperative to investigate measures to reduce immune responses exerted via xenotransplantation. In this study, immunosuppressants were co-administered to mice that had received injections of hMSCs into the parenchyma. Prior to performing experiments using transgenic AD mice (5xFAD), varying immunosuppressant regimens were tested in wild-type (WT) mice and the combination of dexamethasone and tofacitinib (DexaTofa) revealed to be effective in enhancing the persistence of hMSCs. According to transcriptome sequencing and immunohistochemical analyses, administration of DexaTofa reduced immune responses generated via transplantation of hMSCs in the parenchyma of 5xFAD mice. Significant mitigation of amyloid burden, however, was not noted following transplantation of hMSCs alone or hMSCs with DexaTofa. The efficacy of the immunosuppressant regimen should be tested in multiple AD mouse models to promote its successful application and use in AD stem cell therapy.

7.
Mol Cells ; 45(3): 134-147, 2022 Mar 31.
Article in English | MEDLINE | ID: mdl-34887364

ABSTRACT

The anti-oxidant enzyme heme oxygenase-1 (HO-1) is known to exert anti-inflammatory effects. From a library of pyrazolo[3,4-d]pyrimidines, we identified a novel compound KKC080096 that upregulated HO-1 at the mRNA and protein levels in microglial BV-2 cells. KKC080096 exhibited anti-inflammatory effects via suppressing nitric oxide, interleukin-1ß (IL-1ß), and iNOS production in lipopolysaccharide (LPS)-challenged cells. It inhibited the phosphorylation of IKK and MAP kinases (p38, JNK, ERK), which trigger inflammatory signaling, and whose activities are inhibited by HO-1. Further, KKC080096 upregulated anti-inflammatory marker (Arg1, YM1, CD206, IL-10, transforming growth factor-ß [TGF-ß]) expression. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice, KKC080096 lowered microglial activation, protected the nigral dopaminergic neurons, and nigral damage-associated motor deficits. Next, we elucidated the mechanisms by which KKC080096 upregulated HO-1. KKC080096 induced the phosphorylation of AMPK and its known upstream kinases LKB1 and CaMKKbeta, and pharmacological inhibition of AMPK activity reduced the effects of KKC080096 on HO-1 expression and LPS-induced NO generation, suggesting that KKC080096-induced HO-1 upregulation involves LKB1/AMPK and CaMKKbeta/AMPK pathway activation. Further, KKC080096 caused an increase in cellular Nrf2 level, bound to Keap1 (Nrf2 inhibitor protein) with high affinity, and blocked Keap1-Nrf2 interaction. This Nrf2 activation resulted in concurrent induction of HO-1 and other Nrf2-targeted antioxidant enzymes in BV-2 and in dopaminergic CATH.a cells. These results indicate that KKC080096 is a potential therapeutic for oxidative stress- and inflammation-related neurodegenerative disorders such as Parkinson's disease.


Subject(s)
Heme Oxygenase-1 , Neuroprotective Agents , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Heme Oxygenase-1/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Lipopolysaccharides/pharmacology , Mice , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Pyrimidines/pharmacology
8.
Cell Transplant ; 30: 9636897211019025, 2021.
Article in English | MEDLINE | ID: mdl-34044601

ABSTRACT

It has been widely accepted that mesenchymal stem cells (MSCs) can evade the immune surveillance of the recipient. However, emerging research cast doubt on whether MSCs are intrinsically immune-privileged. Previously, we observed that the transplantation of human MSCs (hMSCs) into the mouse parenchyma attracted a high infiltration of leukocytes into the injection tract. Thus, in order to reduce the immune responses generated by hMSCs, the aim of this study was to assess which immunosuppressant condition (dexamethasone only, tacrolimus only, or dexamethasone and tacrolimus together) would not only reduce the overall immune response but also enhance the persistence of MSCs engrafted into the caudate putamen of wild-type C57BL/6 mice. According to immunohistochemical analysis, compared to the hMSC only group, the administration of immunosuppressants (for all three conditions) reduced the infiltration of CD45-positive leukocytes and neutrophils at the site of injection. The highest hMSC persistence was detected from the group that received combinatorial administrations of dexamethasone and tacrolimus. Moreover, compared to the immunocompetent WT mouse, higher MSC engraftment was observed from the immunodeficient BALB/c mice. The results of this study support the use of immunosuppressants to tackle MSC-mediated immune responses and to possibly prolong the engraftment of transplanted MSCs.


Subject(s)
Immunity/drug effects , Immunosuppressive Agents/therapeutic use , Mesenchymal Stem Cell Transplantation/methods , Parenchymal Tissue/transplantation , Animals , Immunosuppressive Agents/pharmacology , Mice
9.
Int J Mol Sci ; 21(19)2020 Sep 23.
Article in English | MEDLINE | ID: mdl-32977637

ABSTRACT

Mesenchymal stem cells (MSCs) are a useful source for cell-based therapy of a variety of immune-mediated diseases, including neurodegenerative disorders. However, poor migration ability and survival rate of MSCs after brain transplantation hinder the therapeutic effects in the disease microenvironment. Therefore, we attempted to use a preconditioning strategy with pharmacological agents to improve the cell proliferation and migration of MSCs. In this study, we identified ethionamide via the screening of a drug library, which enhanced the proliferation of MSCs. Preconditioning with ethionamide promoted the proliferation of Wharton's jelly-derived MSCs (WJ-MSCs) by activating phosphatidylinositol 3-kinase (PI3K)/Akt and mitogen-activated protein kinase/extracellular signal-regulated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK)1/2 signaling. Preconditioning with ethionamide also enhanced the migration ability of MSCs by upregulating expression of genes associated with migration, such as C-X-C motif chemokine receptor 4 (CXCR4) and C-X-C motif chemokine ligand 12 (CXCL12). Furthermore, preconditioning with ethionamide stimulated the secretion of paracrine factors, including neurotrophic and growth factors in MSCs. Compared to naïve MSCs, ethionamide-preconditioned MSCs (ETH-MSCs) were found to survive longer in the brain after transplantation. These results suggested that enhancing the biological process of MSCs induced by ethionamide preconditioning presents itself as a promising strategy for enhancing the effectiveness of MSCs-based therapies.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Ethionamide/pharmacology , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Mesenchymal Stem Cells/metabolism , Animals , Brain/cytology , Brain/metabolism , Heterografts , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mice
10.
Exp Neurol ; 332: 113387, 2020 10.
Article in English | MEDLINE | ID: mdl-32580013

ABSTRACT

The transcription factor nuclear factor-erythroid 2-related factor-2 (Nrf2) is known to induce neuroprotective and anti-inflammatory effects and is considered to be an excellent molecular target for drugs related to neurodegenerative disease therapy. Nrf2 activators previously tested in clinical trials were electrophilic, causing adverse effects due to non-selective and covalent modification of cellular thiols. In order to circumvent this issue, we constructed and screened a chemical library consisting of 241 pyrazolo [3,4-d] pyrimidine derivatives and discovered a novel, non-electrophilic compound: 1-benzyl-6-(methylthio)-N-(1-phenylethyl)-1H-pyrazolo[3,4-d]pyrimidine-4-amine (KKC080106). KKC080106 was able to activate Nrf2 signaling as it increases the cellular levels of Nrf2, binds to the Nrf2 inhibitor protein Keap1, and causes the accumulation of nuclear Nrf2. We also observed an increase in the expression levels of Nrf2-dependent genes for antioxidative/neuroprotective enzymes in dopaminergic neuronal cells. In addition, in lipopolysaccharide-activated microglia, KKC080106 suppressed the generation of the proinflammatory markers, such as IL-1ß, TNF-α, cyclooxygenase-2, inducible nitric oxide synthase, and nitric oxide, and inhibited the phosphorylation of kinases known to be involved in inflammatory signaling, such as IκB kinase, p38, JNK, and ERK. As a drug, KKC080106 exhibited excellent stability against plasma enzymes and a good safety profile, evidenced by no mortality after the administration of 2000 mg/kg body weight, and minimal inhibition of the hERG channel activity. Pharmacokinetic analysis revealed that KKC080106 has good bioavailability and enters the brain after oral and intravenous administration, in both rats and mice. In MPTP-treated mice that received KKC080106 orally, the compound blocked microglial activation, protected the nigral dopaminergic neurons from degeneration, and prevented development of the dopamine deficiency-related motor deficits. These results suggest that KKC080106 has therapeutic potential for neurodegenerative disorders such as Parkinson's disease.


Subject(s)
Dopaminergic Neurons/drug effects , NF-E2-Related Factor 2/agonists , Neuroprotective Agents/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sambucus nigra/cytology , Sambucus nigra/drug effects , Animals , Antioxidants , Brain/metabolism , Cytokines/metabolism , Inflammation/genetics , Kelch-Like ECH-Associated Protein 1/drug effects , Kelch-Like ECH-Associated Protein 1/genetics , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Neuroprotective Agents/pharmacokinetics , Nitric Oxide/metabolism , Phosphorylation/drug effects , Protein Kinases/metabolism , Signal Transduction/drug effects
11.
Int J Mol Sci ; 21(9)2020 Apr 26.
Article in English | MEDLINE | ID: mdl-32357509

ABSTRACT

Due to their multifactorial aspects, mesenchymal stem cells (MSCs) have been widely established as an attractive and potential candidate for the treatment of a multitude of diseases. A substantial number of studies advocate that MSCs are poorly immunogenic. In several studies, however, immune responses were observed following injections of xenogeneic donor MSCs. In this study, the aim was to examine differences in immune responses exerted based on transplantations of xenogeneic, syngeneic, and allogeneic MSCs in the wild-type mouse brain. Xenogeneic, allogeneic, and syngeneic MSCs were intracerebrally injected into C57BL/6 mice. Mice were sacrificed one week following transplantation. Based on immunohistochemical (IHC) analysis, leukocytes and neutrophils were expressed at the injection sites in the following order (highest to lowest) xenogeneic, allogeneic, and syngeneic. In contrast, microglia and macrophages were expressed in the following order (highest to lowest): syngeneic, allogeneic, and xenogeneic. Residual human MSCs in the mouse brain were barely detected after seven days. Although the discrepancy between leukocytes versus macrophages/microglia infiltration should be resolved, our results overall argue against the previous notions that MSCs are poorly immunogenic and that modulation of immune responses is a prerequisite for preclinical and clinical studies in MSC therapy of central nervous system diseases.


Subject(s)
Leukocytes/metabolism , Macrophages/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Microglia/metabolism , Neutrophils/metabolism , Animals , Cells, Cultured , Female , Humans , Immunity , Mice , Mice, Inbred C57BL , Transplantation, Heterologous/methods , Transplantation, Homologous/methods , Transplantation, Isogeneic/methods
12.
Neurochem Int ; 112: 96-107, 2018 01.
Article in English | MEDLINE | ID: mdl-29158022

ABSTRACT

The transcription factor Nrf2 is known to induce gene expression of antioxidant enzymes and proteasome subunits. Because both oxidative stress and protein aggregation have damaging effects on neurons, activation of the Nrf2 signaling should be beneficial against neurodegeneration. In this study, we report a novel synthetic morpholine-containing chalcone KMS99220 that confers neuroprotection. It showed high binding affinity to the Nrf2 inhibitory protein Keap-1 and increased nuclear translocation of Nrf2 and gene expression of the antioxidant enzymes heme oxygenase-1, NAD(P)H:quinone oxidoreductase-1, and the catalytic and modifier subunits of glutamate-cysteine ligase in dopaminergic CATH.a cells. KMS99220 also increased expression of the proteasome subunits PSMB5, PSMB7, PSMB8 and PSMA1, and the respective chymotrypsin and trypsin-like proteasomal enzyme activities, and reduced α-synuclein aggregate in GFP-α-syn A53T-overexpressing cells. KMS99220 exhibited a favorable pharmacokinetic profile with excellent bioavailability and metabolic stability, did not interfere with activities of the cytochrome p450 isotypes, and showed no apparent in vivo toxicity when administered up to 2000 mg/kg. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice, oral administration of KMS99220 prevented degeneration of the nigral dopaminergic neurons, induced the Nrf2 target genes, and effectively prevented the associated motor deficits. These results suggest KMS99220 as a potential candidate for therapy against Parkinson's disease.


Subject(s)
Dopaminergic Neurons/metabolism , Morpholines/pharmacology , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Substantia Nigra/metabolism , Animals , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Dose-Response Relationship, Drug , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , MPTP Poisoning/prevention & control , Male , Mice , Mice, Inbred C57BL , Morpholines/chemistry , Morpholines/therapeutic use , Neuroprotection/drug effects , Neuroprotection/physiology , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Substantia Nigra/drug effects
13.
Sci Rep ; 7(1): 11368, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900300

ABSTRACT

Kohlschutter-Tönz syndrome (KTS) is a rare genetic disorder with neurological dysfunctions including seizure and intellectual impairment. Mutations at the Rogdi locus have been linked to development of KTS, yet the underlying mechanisms remain elusive. Here we demonstrate that a Drosophila homolog of Rogdi acts as a novel sleep-promoting factor by supporting a specific subset of gamma-aminobutyric acid (GABA) transmission. Rogdi mutant flies displayed insomnia-like behaviors accompanied by sleep fragmentation and delay in sleep initiation. The sleep suppression phenotypes were rescued by sustaining GABAergic transmission primarily via metabotropic GABA receptors or by blocking wake-promoting dopaminergic pathways. Transgenic rescue further mapped GABAergic neurons as a cell-autonomous locus important for Rogdi-dependent sleep, implying metabotropic GABA transmission upstream of the dopaminergic inhibition of sleep. Consistently, an agonist specific to metabotropic but not ionotropic GABA receptors titrated the wake-promoting effects of dopaminergic neuron excitation. Taken together, these data provide the first genetic evidence that implicates Rogdi in sleep regulation via GABAergic control of dopaminergic signaling. Given the strong relevance of GABA to epilepsy, we propose that similar mechanisms might underlie the neural pathogenesis of Rogdi-associated KTS.


Subject(s)
Dopamine/metabolism , Drosophila/physiology , Nuclear Proteins/genetics , Signal Transduction , Sleep/genetics , Wakefulness/genetics , gamma-Aminobutyric Acid/metabolism , Alleles , Animals , Animals, Genetically Modified , Anticonvulsants/pharmacology , Brain/drug effects , Brain/metabolism , Circadian Rhythm/genetics , Female , GABAergic Neurons/metabolism , Loss of Function Mutation , Models, Biological , Mutation , Nuclear Proteins/metabolism , Receptors, GABA/metabolism , Signal Transduction/drug effects
14.
Free Radic Res ; 51(6): 633-645, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28770670

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterised by selective degeneration of the nigral dopaminergic neurons, and neuroinflammation and oxidative stress are believed to be involved in its pathogenesis. In the present study, we provide data that the synthetic steroid exemestane, which is currently being used to treat breast cancer, may be useful for PD therapy. In BV-2 microglial cells, exemestane activated the transcription factor Nrf2 and induced expression of the Nrf2-dependent genes that encode the antioxidant enzymes NAD(P)H: quinone oxidoreductase 1, haem oxygenase-1, and glutamylcysteine ligase. It also downregulated gene expression of inducible nitric oxide (NO) synthase, lowered the levels of NO and reactive oxygen species, interleukin-1ß and tumour necrosis factor-α in lipopolysaccharide-activated microglial cells. In CATH.a dopaminergic neuronal cells, exemestane also induced the same set of Nrf2-dependent antioxidant enzyme genes and provided neuroprotection against oxidative damage. In vivo, the drug protected the nigral dopaminergic neurons, decreased microglial activation, and prevented motor deficits in C57Bl/6 male mice that had been administered with the dopaminergic neurotoxin MPTP. Taken together, the results suggested a utility of repositioning exemestane towards disease-modifying therapy for PD.


Subject(s)
Androstadienes/pharmacology , Antiparkinson Agents/pharmacology , Dopaminergic Neurons/drug effects , Drug Repositioning , NF-E2-Related Factor 2/genetics , Parkinsonian Disorders/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Antineoplastic Agents/pharmacology , Cell Line , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Gene Expression Regulation , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hindlimb Suspension , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lipopolysaccharides , Male , Membrane Proteins/agonists , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Rotarod Performance Test , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Neurobiol Aging ; 50: 119-133, 2017 02.
Article in English | MEDLINE | ID: mdl-27960106

ABSTRACT

Fungicide exposure causes degeneration of dopaminergic neurons and contributes to Parkinson's disease (PD). Benomyl inhibits enzymes responsible for detoxifying the reactive dopamine metabolite 3,4-dihydroxyphenylacetaldehyde. Aldose reductase (AR) is known as tetrahydrobiopterin (BH4) reductase that generates BH4, a cofactor for tyrosine hydroxylase (TH) involved in dopamine synthesis. AR also acts as an aldehyde reductase involved in detoxifying 3,4-dihydroxyphenylacetaldehyde. In PD patients, the level of AR is significantly lower in the cerebellum. To determine if AR deficiency contributes to PD, AR wild-type (AR+/+) and knockout (AR-/-) mice were administrated with 1-methyl-4-phenyl -1,2,3,6- tetrahydropyridine (MPTP). The MPTP-treated AR-/- mice showed more severe behavioral deficits and brain damage than that of AR+/+ mice. Contrary to expectation, under normal or MPTP-treated condition, AR-/- mice showed a significant elevation of BH4 and dopamine in the midbrain, suggesting that either AR does not contribute to BH4 production, or other BH4 synthetic pathways are induced. The AR-/- brain showed upregulation of peroxynitrite, inducible nitric oxide synthase and downregulation of antioxidant enzymes, Cu/Zn superoxide dismutase (SOD) and peroxiredoxin 2 (Prx2), which indicate an increase in oxidative stress. In line with the animal data, pretreating the SH-SY5Y cells with AR inhibitors (Fidarestat or Epalrestat) before MPP+ treatment, increased severe cell death and mitochondrial fragmentation with downregulation of SOD were observed when compared to the MPP+ treatment alone. Cycloxygenase 2 (COX2), which can lead to the oxidation of dopamine, was upregulated in AR-/- brains. Autophagic proteins, beclin-1 and LC3B were also downregulated. The loss of dopaminergic neurons was associated with activation of p-ERK1/2. These findings suggest that AR plays an important role in protecting dopaminergic neuron against neurotoxic metabolites in PD.


Subject(s)
Aldehyde Reductase/deficiency , Autophagy , Dopaminergic Neurons/pathology , Oxidative Stress/physiology , Parkinson Disease/etiology , Parkinson Disease/pathology , Animals , Cells, Cultured , Disease Models, Animal , Male , Mice, Inbred C57BL , Mice, Transgenic , Parkinson Disease/physiopathology
16.
Free Radic Res ; 50(11): 1188-1199, 2016.
Article in English | MEDLINE | ID: mdl-27598306

ABSTRACT

The degenerative process of the nigral dopamine(DA)rgic neurons in Parkinson's disease (PD) involves both oxidative stress and neuroinflammation. In the present study, we aimed at developing a novel antioxidant and anti-inflammatory agent for PD therapy. Toward this end, we screened a novel focused library of isothiocyanate derivatives that we have generated for an anti-inflammatory property. We obtained a novel compound ITC-57 and found that ITC-57 effectively induced gene expression of the antioxidant enzymes NAD(P)H quinone oxidoreductase-1, the catalytic and modulatory subunits of glutamylcysteine ligase, and HO-1 in DAergic neuronal CATH.a cells and protected CATH.a cells from oxidative damages. The compound also induced the same antioxidant enzymes in microglial BV-2 cells and suppressed the production of the proinflammatory molecules nitric oxide, interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) in lipopolysaccharide-activated BV-2 cells. In the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-elicited mouse model of PD, ITC-57 protected the DAergic neurons from degeneration, induced HO-1, lowered TNF-α, and suppressed microglial activation in the nigra. Furthermore, ITC-57 prevented the PD-associated motor deficits from occurring. Taken together, ITC-57 would be useful toward development of a disease-modifying therapy for PD.


Subject(s)
Isothiocyanates/pharmacology , Parkinson Disease/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Cell Death , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Oxidative Stress
17.
Eur J Pharmacol ; 771: 152-61, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26687634

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by degeneration of dopamine(DA)ergic neurons. Neuroinflammation caused by microglial activation is believed to be involved in the pathogenesis of neurodegenerative diseases including PD. In the present study, we tested the effects of a novel compound 2-acetyl-7-hydroxy-6-methoxy-1-methyl-1,2,3,4,-tetarhydroisoquinoline (AMTIQ) on neuroinflammatory response and DAergic neurodegeneration. In lipopolysaccharide-activated BV-2 microglial cells, AMTIQ lowered nitric oxide and tetrahydrobiopterin levels and downregulated gene expression of inducible nitric oxide synthase and GTP cyclohydrolase I. AMTIQ also repressed gene expression of the proinflammatory cytokines IL-1ß and TNF-α, and attenuated nuclear translocation of NF-κB. AMTIQ was stable against liver microsomal enzymes from human and mouse and did not interfere with activities of the cytochrome p450 enzymes 1A2, 2D6, 2C9, 2C19 and 3A4. Pharmacokinetic studies revealed the brain to plasma ratio of AMTIQ to be 45%, suggesting it can penetrate the blood brain barrier. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mouse PD model, AMTIQ led to decreased microglial activation, increased survival of DAergic neurons and their fibers, and improved behavioral scores on rotarod and vertical grid tests. Taken together, these results suggest that AMTIQ might serve as a candidate preventive-therapeutic agent for neurodegenerative diseases such as PD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antiparkinson Agents/pharmacology , Dopaminergic Neurons/drug effects , Isoquinolines/pharmacology , Neuroprotective Agents/pharmacology , Substantia Nigra/cytology , Substantia Nigra/drug effects , Animals , Anti-Inflammatory Agents/pharmacokinetics , Antiparkinson Agents/pharmacokinetics , Blood-Brain Barrier/metabolism , Brain/metabolism , Cell Line , Cytokines/biosynthesis , Cytokines/genetics , Humans , In Vitro Techniques , Isoquinolines/pharmacokinetics , Macrophage Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Microglia/drug effects , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Neuroprotective Agents/pharmacokinetics , Nitric Oxide/metabolism
18.
Neurotox Res ; 28(4): 332-45, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26233727

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder accompanied by a selective loss of the dopamine(DA)ergic neurons residing in the substantia nigra. There is ample evidence that neuroinflammation and oxidative stress are involved in the pathogenesis of PD. In the present study, we aimed at protecting the DAergic neurons by suppressing these cellular events and generated a novel synthetic isothiocyanate ITC-3. The compound led to elevation of nuclear and total levels of the transcription factor Nrf2 and interacted with its binding protein Keap1 with high affinity, suggesting Nrf2 activation. ITC-3 was able to suppress production of the proinflammatory mediators in lipopolysaccharide-activated BV-2 microglial cells. It also increased mRNA and protein levels of the Nrf2-dependent antioxidant enzymes NAD(P)H quinone oxidoreductase, heme oxygenase-1, and glutamylcysteine ligase in both BV-2 and DAergic neuronal CATH.a cells. The compound protected the DAergic cells against oxidative stress. In vivo, ITC-3 attenuated the loss of tyrosine hydroxylase-immunopositive nigrostriatal DAergic neurons, suppressed microglial activation, and abolished PD-associated motor deficits in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-elicited animal model of PD. Taken together, ITC-3 may be useful toward development of neuroprotective therapy for PD.


Subject(s)
Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Isothiocyanates/pharmacology , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Parkinson Disease/metabolism , Animals , Antioxidants/metabolism , Cells, Cultured , Disease Models, Animal , Heme Oxygenase-1/metabolism , Inflammation Mediators/metabolism , Isothiocyanates/administration & dosage , Isothiocyanates/chemical synthesis , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Motor Activity/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemical synthesis , Nuclear Proteins/metabolism , Parkinson Disease/prevention & control , Signal Transduction/drug effects
19.
J Mol Neurosci ; 56(2): 263-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25702135

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder associated with a selective loss of the neurons containing dopamine (DA) in the substantia nigra pars compacta. Lines of evidence suggest that oxidative stress is a major factor contributing to the vulnerability of DA cells and that the enzyme NAD(P)H quinone oxidoreductase (NQO1) provides protection in these cells. In the present study, we report the synthesis of a novel compound KMS04014 and show that it induces NQO1 gene expression and protects DAergic neuronal cells in both cell culture and animal models of PD. In vitro, KMS04014 increased both mRNA and protein levels of NQO1 and induced nuclear translocation of Nrf2 in the DAergic neuronal cell line CATH.a. It also protected the cells against oxidative stress generated by tetrahydrobiopterin, 1-methyl-4-phenylpyridinium (MPP(+)), and H2O2. In vivo, KMS04014 attenuated the loss of tyrosine hydroxylase-immunopositive DAergic neurons in the substantia nigra and reduced degeneration of the nigral neurons and striatal fibers in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice, an animal model of PD. Taken together, KMS04014 may be utilized toward development of neuroprotective therapy for PD.


Subject(s)
Antioxidants/therapeutic use , Coumaric Acids/pharmacology , MPTP Poisoning/drug therapy , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neuroprotective Agents/therapeutic use , Animals , Antioxidants/chemical synthesis , Antioxidants/pharmacology , Cell Line, Tumor , Coumaric Acids/chemical synthesis , Coumaric Acids/therapeutic use , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Male , Mice , Mice, Inbred C57BL , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substantia Nigra/cytology , Substantia Nigra/drug effects , Substantia Nigra/metabolism
20.
Br J Pharmacol ; 172(4): 1087-100, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25297649

ABSTRACT

BACKGROUND AND PURPOSE: Neuroinflammation through microglial activation is involved in the pathogenesis of neurodegenerative diseases including Parkinson's disease (PD), a major neurodegenerative disorder characterized by dopaminergic neuronal death in the substantia nigra. We examined our novel synthetic compound VSC2 for its anti-inflammatory properties towards development of a PD therapy. EXPERIMENTAL APPROACH: We tested the effects of VSC2 on production of various NF-κB-dependent proinflammatory molecules and Nrf2-dependent antioxidant enzymes in BV-2 microglia and in vivo. KEY RESULTS: The vinyl sulfone compound, VSC2, most effectively suppressed the production of NO in LPS-activated microglia. It also down-regulated expression of inducible NOS (iNOS), COX-2, IL-1ß and TNF-α and inhibited nuclear translocalization and transcriptional activity of NF-κB. VSC2 increased total and nuclear Nrf2 levels, induced Nrf2 transcriptional activity and was bound to Keap1 with high affinity. Expression of the Nrf2-regulated antioxidant enzyme genes NAD(P)H quinone oxidoreducase-1 (NQO-1), haem oxygenase-1 (HO-1) and glutamylcysteine ligase (GCL) were up-regulated by VSC2. In the MPTP mouse model of PD, oral administration of VSC2 decreased the number of activated microglia in the substantia nigra, lowered the levels of iNOS, COX-2 and IL-1ß, and protected the dopaminergic neurons. VSC2 also elevated the levels of NQO1, HO-1, GCL and Nrf2 in the nigrostriatal area. CONCLUSIONS AND IMPLICATIONS: VSC2 has both anti-inflammatory and antioxidant properties and prevented neuroinflammation in microglia and in an animal model of PD. This suggests VSC2 as a potential candidate for PD therapy.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Microglia/drug effects , Parkinson Disease/drug therapy , Sulfones , Vinyl Compounds , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Adaptor Proteins, Signal Transducing/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cell Line , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Kelch-Like ECH-Associated Protein 1 , Male , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Parkinson Disease/pathology , Sulfones/pharmacology , Sulfones/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Vinyl Compounds/pharmacology , Vinyl Compounds/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...