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










Database
Language
Publication year range
1.
J Nutr Biochem ; 69: 73-86, 2019 07.
Article in English | MEDLINE | ID: mdl-31063918

ABSTRACT

Microbiota in the gut affect brain physiology via various pathways, and dysbiosis seems to play a role in the pathogenesis of Parkinson's disease (PD). Probiotics showed pleiotropic effects on functions of the central nervous system via microbiota-gut-brain axis. However, no studies displayed the neuroprotective effects of probiotics in the Parkinson's disease. This study aimed to test the neuroprotective effects of probiotics in two different models of PD. We evaluated neuroprotective effects of a probiotic cocktail containing Lactobacillus rhamnosus GG, Bifidobacterium animalis lactis, and Lactobacillus acidophilus in PD models induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or rotenone utilizing behavioral tests, immunohistochemistry and neurochemical analysis. To assure the neuroprotection came from increased production of butyrate, we further determined beneficial effects of butyrate in the MPTP-mediated PD model. The probiotic mixture overtly protected the dopaminergic neurons against MPTP neurotoxicity. However, the probiotics downregulated expression of monoamine oxidase (MAO) B in the striatum, which was accompanied by a lower level of 1-methyl-4-phenylpyridinium (MPP+), the main neurotoxic metabolite of MPTP. Thus, we extended the investigation into the rotenone-induced PD model. Rescuing effects of the probiotics were observed in the setup, which came with increased levels of neurotrophic factors and butyrate in the brain. Lactobacillus rhamnosus GG was identified to be a major contributor to the induction of neurotrophic factors and downregulation of MAO B. Finally, we demonstrated that sodium butyrate attenuated MPTP-induced neuronal loss in the nigrostriatal pathway. Probiotics could ameliorate neurodegeneration at least partially by increasing butyrate level. These data highlight the role of probiotics for brain health, and their potential as a preventive measure for neurodegenerative diseases such as PD.


Subject(s)
Butyrates/metabolism , Dopaminergic Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/drug therapy , Probiotics/pharmacology , Rotenone/toxicity , Acetylation/drug effects , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Dopamine , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Histones/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/metabolism , Male , Mice, Inbred C57BL , Monoamine Oxidase/metabolism , Neuroglia/drug effects , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Parkinson Disease/drug therapy
2.
Neurotoxicology ; 71: 113-121, 2019 03.
Article in English | MEDLINE | ID: mdl-30605763

ABSTRACT

Animal models for Parkinson's disease (PD) are very useful in understanding the pathogenesis of PD and screening for new therapeutic approaches. 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine (MPTP) and rotenone are common neurotoxins used for the development of experimental PD models, and both inhibit complex I of mitochondria; this is thought to be an instrumental mechanism for dopaminergic neurodegeneration in PD. In this study, we treated mice with MPTP (30 mg/kg/day) or rotenone (2.5 mg/kg/day) for 1 week and compared the neurotoxic effects of these toxins. MPTP clearly produced dopaminergic lesions in both the substantia nigra and the striatum as shown by loss of dopaminergic neurons, depletion of striatal dopamine, activation of glial cells in the nigrostriatal pathway and behavioral impairment. In contrast, rotenone treatment did not show any significant neuronal injury in the nigrostriatal pathway, but it caused neurodegeneration and glial activation only in the hippocampus. MPTP showed no such deleterious effects in the hippocampus suggesting the higher susceptibility of the hippocampus to rotenone than to MPTP. Interestingly, rotenone caused upregulation of the neurotrophic factors and their downstream PI3K-Akt pathway along with adenosine monophosphate-activated protein kinase (AMPK) activation. These results suggest that MPTP-induced dopaminergic neurotoxicity is more acute and specific in comparison to rotenone toxicity, and compensatory brain-derived neurotrophic factor (BDNF) induction and AMPK activation in the rotenone-treated brain might suppress the neuronal injury.


Subject(s)
Brain/drug effects , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Neurons/drug effects , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Rotenone/toxicity , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Dopamine/metabolism , Male , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Parkinsonian Disorders/chemically induced
3.
J Chem Neuroanat ; 94: 173-182, 2018 12.
Article in English | MEDLINE | ID: mdl-30040987

ABSTRACT

IL-32 is a proinflammatory cytokine, and involved in various diseases including infection, inflammation, and cancer. However, effects of IL-32 on neuroinflammation remain obscure. Herein, we examined the effects of IL-32ß on systemic LPS-induced neuroinflammation using IL-32ß transgenic (Tg) mice. IL-32ß wild type (WT) and Tg mice received LPS injection (5 mg/kg, i.p.), and then neuroinflammatory responses were evaluated. Systemic LPS caused remarkable gliosis in the brain at 12 h regardless of genotypes. The gliosis in WT mice was sustained by 24 h, whereas it became more severe in Tg mice by 24 h. Proinflammatory cytokines and proteins were increased at 12 h both in WT and Tg brains. The elevated levels of TNFα and VCAM-1were not altered over time, while levels of IL-6, IL-1ß and iNOS were dropped in WT mice. In contrast, elevated levels IL-6, IL-1ß, iNOS and VCAM-1 were sustained, and level of TNFα was augmented in Tg brains by 24 h. Interestingly, level of IL-10 mRNA in Tg mice was remarkably higher than in WT mice at 0 h, which was decreased at 12 h and maintained by 24 h. In WT brain, mRNA level of IL-10 was raised at 12 h after LPS injection, and further increased at 24 h. Activation of NF-κB signaling pathway was detected in glia cells after LPS injection which was exaggerated at 24 h in Tg mice in comparison to WT mice. These results indicate that IL-32ß enhances neuroinflammatory responses caused by systemic LPS, and this might be attributable to prolonged activation of NF-κB signaling pathway.


Subject(s)
Brain/pathology , Gliosis/pathology , Inflammation/pathology , Interleukins/genetics , Lipopolysaccharides , Animals , Brain/metabolism , Cytokines/metabolism , Gliosis/chemically induced , Gliosis/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Interleukins/metabolism , Mice , Mice, Transgenic , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Signal Transduction , Vascular Cell Adhesion Molecule-1/metabolism
4.
Ann N Y Acad Sci ; 1431(1): 58-71, 2018 11.
Article in English | MEDLINE | ID: mdl-29882218

ABSTRACT

The relatively old, yet clinically used, drug methylene blue (MB) is known to possess neuroprotective properties by reducing aggregated proteins, augmenting the antioxidant response, and enhancing mitochondrial function and survival in various models of neurodegenerative diseases. In this study, we aimed to examine the effects of MB in Parkinson's disease (PD) in vivo and in vitro models by using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/1-methyl-4-phenylpyridinium (MPP+ ) with a focus on possible effects on induction of neurotrophic factors. Our results indicate that pretreatment with MB significantly attenuated MPTP-induced loss of dopaminergic neurons, glial cell activation, and depletion of dopamine. We also found that MB upregulated brain-derived neurotrophic factor (BDNF) and activated its downstream signaling pathways, suggesting that BDNF might be a contributor to MB-associated neuroprotection. Specific inhibition of the BDNF receptor or extracellular signal-regulated kinase (Erk) reversed the MB-mediated protection against MPP+ toxicity, thus implying a role for BDNF and the Erk pathway in the neuroprotective effects. Taken together, our data suggest that MB protects neurons from MPTP neurotoxicity via induction of BDNF. Further study to determine whether MB preserves dopaminergic neurons in the brains of PD patients is warranted.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Dopaminergic Neurons/drug effects , MPTP Poisoning/prevention & control , Methylene Blue/pharmacology , Neuroprotective Agents/pharmacology , Up-Regulation/drug effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Disease Models, Animal , Dopaminergic Neurons/metabolism , MPTP Poisoning/metabolism , Male , Methylene Blue/therapeutic use , Mice , Neuroprotective Agents/therapeutic use , Phosphorylation/drug effects , Signal Transduction/drug effects , Tyrosine 3-Monooxygenase/metabolism
5.
Sci Rep ; 8(1): 7799, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29773813

ABSTRACT

CD4+ T cells are the central for the mammalian adaptive immune system. Naïve CD4+ T cells mainly differentiate in to pro-inflammatory Th1, Th2 and Th17 cells upon antigenic stimulation. IFN-γ secreting Th1 cells and IL-17 secreting Th17 cells are found to play key roles in autoimmune diseases like multiple sclerosis (MS) and ulcerative colitis (UC). In this study we found NTG-A-009, 6-aminopyridin-3-ol, has great inhibitory effect on in vitro differentiation of Th1 and Th17 cells without affecting regulatory T cells. Moreover, NTG-A-009 had no effect on CD4+ T cell proliferation and viability. In vivo treatment has shown that NTG-A-009 has ameliorated experimental autoimmune encephalomyelitis (EAE) and dextran sulfate sodium (DSS) induced colitis through the inhibition of Th1 and Th17 cells differentiation. Mechanistically, NTG-A-009 suppressed Th1 and Th17 cells differentiation via the modulation of JAK/STAT signaling pathway. Thus, our data demonstrated that NTG-A-009 ameliorated inflammation through the inhibition of Th1 and Th17 cells generation making it a potential therapeutic candidate for the treatment of inflammatory diseases.


Subject(s)
Aminopyridines/pharmacology , Cell Differentiation/drug effects , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Th1 Cells/drug effects , Th17 Cells/drug effects , Animals , Colitis/drug therapy , Colitis/prevention & control , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Mice , Signal Transduction , Th1 Cells/cytology , Th17 Cells/cytology
6.
Mol Neurobiol ; 55(1): 554-566, 2018 01.
Article in English | MEDLINE | ID: mdl-27975170

ABSTRACT

Neurotrophic factors are essential for neuronal survival, plasticity, and development and have been implicated in the action mechanism of antidepressants. In this study, we assessed the neurotrophic factor-inducing and neuroprotective properties of antidepressants. In the first part of the study, we found that fluoxetine, imipramine, and milnacipran (i.p., 20 mg/kg/day for 1 week or 3 weeks) upregulated brain-derived neurotrophic factor in the striatum and substantia nigra both at 1 week and 3 weeks. In contrast, an increase in the glial-derived neurotrophic factor was more obvious at 3 weeks after the antidepressants treatment. Specifically, it was found that fluoxetine and imipramine are more potent in raising the levels of neurotrophic factors than milnacipran. Furthermore, antidepressants elevated the phosphorylation of extracellular signal-regulated-protein kinase (ERK1/2) and the serine/threonine kinase Akt. In the second part of the study, we compared the neuroprotective effects of fluoxetine, imipramine, and milnacipran in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of Parkinson's disease. Pretreament with fluoxetine, imipramine or milnacipran for 3 weeks reduced MPTP-induced dopaminergic neurodegeneration and microglial activation in the nigrostriatal pathway. Neurochemical analysis by HPLC exhibited that antidepressants attenuated the depletion of striatal dopamine. In consistent, beam test showed that behavioral impairment was ameliorated by antidepressants. Neuroprotective effects were more prominent in the fluoxetine or imipramine treatment group than in milnacipran treatment group. Finally, we found that neuroprotection of the antidepressants against 1-methyl-4-phenylpyridinium neurotoxicity in SH-SY5Y cells was attenuated by ERK or Akt inhibitor. These results indicate that neuroprotection by antidepressants might be associated with the induction of neurotrophic factors, and antidepressant could be a potential therapeutic intervention for treatment of Parkinson's disease.


Subject(s)
Antidepressive Agents/therapeutic use , Nerve Growth Factors/metabolism , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Up-Regulation , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Antidepressive Agents/pharmacology , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/enzymology , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Substantia Nigra/drug effects , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Up-Regulation/drug effects
7.
Neuropharmacology ; 125: 396-407, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28807678

ABSTRACT

In spite of the massive research for the identification of neurorestorative or neuroprotective intervention for curing Parkinson's disease (PD), there is still lack of clinically proven neuroprotective agents. Metformin, a common anti-hyperglycemic drug has been known to possess neuroprotective properties. However, specific mechanisms by which metformin protects neurons from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxicity remain to be elucidated. In this study, we assessed the neuroprotective effects of metformin in the subchronic MPTP model of PD, and explored its feasible mechanisms for neuroprotection. Animals received saline or MPTP injection (30 mg/kg/day) for the first 7 days, and then saline or metformin (200 mg/kg/day) for the next 7 days. Immunohistochemical stainings showed that metformin rescued the tyrosine hydroxylase-positive neurons and attenuated astroglial activation in the nigrostriatal pathway. In parallel, metformin restored dopamine depletion and behavioral impairments exerted by MPTP. Western blot analysis revealed that metformin ameliorated MPTP-induced α-synuclein phosphorylation which was accompanied by increased methylation of protein phosphatase 2A (PP2A), a phosphatase related to α-synuclein dephosphorylation. Moreover, the metformin regimen significantly increased the level of brain derived neurotrophic factor in the substantia nigra, and activated signaling pathways related to cell survival. Proof of concept study revealed that inhibition of PP2A or tropomyosin receptor kinase B reversed neuroprotective property of metformin in SH-SY5Y cells. Our results indicate that metformin provides neuroprotection against MPTP neurotoxicity, which might be mediated by inhibition of α-synuclein phosphorylation and induction of neurotrophic factors.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/metabolism , Metformin/pharmacology , Neuroprotective Agents/pharmacology , alpha-Synuclein/metabolism , Animals , Antiparkinson Agents/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , MPTP Poisoning/pathology , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Phosphorylation/drug effects , Proof of Concept Study , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/metabolism , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
8.
Neurochem Int ; 102: 79-88, 2017 01.
Article in English | MEDLINE | ID: mdl-27956238

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by prominent loss of the nigral dopaminergic neurons and motor symptoms, such as resting tremor and bradykinesia. Evidence suggests that neuroinflammation may play a critical role in PD pathogenesis. Interleukin (IL)-32 is a newly-identified proinflammatory cytokine, which regulates innate and adaptive immune responses by activating p38 MAPK and NF-κB signaling pathways. The cytokine has been implicated in cancers and autoimmune, inflammatory, and infectious diseases. In this study, we attempted to identify the effects of IL-32ß on dopaminergic neurotoxicity induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), using IL-32ß transgenic mice. Male wild type and IL-32ß transgenic mice received intraperitoneal injections of vehicle or MPTP (15 mg/kg × 4). Immunohistochemistry showed that overexpression of IL-32ß significantly increased MPTP-mediated loss of dopaminergic neurons in the substantia nigra and deletion of tyrosine hydroxylase-positive fibers in the striatum. Dopamine depletion in the striatum and deficit in locomotor activity were enhanced in IL-32ß transgenic mice. These results were accompanied by higher neuroinflammatory responses in the brains of transgenic mice. Finally, we found that IL-32ß exaggerated MPTP-mediated activation of p38 MAPK and JNK pathways, which have been shown to be involved in MPTP neurotoxicity. These results suggest that IL-32ß exacerbates MPTP neurotoxicity through enhanced neuroinflammatory responses.


Subject(s)
Corpus Striatum/drug effects , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Interleukins/genetics , MPTP Poisoning , Animals , Corpus Striatum/metabolism , Disease Models, Animal , MPTP Poisoning/metabolism , Mice, Transgenic , Substantia Nigra/drug effects , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...