Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Glia ; 71(4): 1057-1080, 2023 04.
Article in English | MEDLINE | ID: mdl-36573349

ABSTRACT

Chronic kidney disease (CKD)-associated mental disorders have been attributed to the excessive accumulation of hemodialysis-resistant indoxyl-3-sulfate (I3S) in the brain. I3S not only induces oxidative stress but is also a potent endogenous agonist of the aryl hydrocarbon receptor (AhR). Here, we investigated the role of AhR in CKD-induced brain disorders using a 5/6 nephrectomy-induced CKD mouse model, which showed increased I3S concentration in both blood and brain, anxiety and impaired novelty recognition, and AhR activation in the anterior cortex. GFAP+ reactive astrocytes were increased accompanied with the reduction of glutamate transporter 1 (GLT1) on perineuronal astrocytic processes (PAPs) in the anterior cingulate cortex (ACC) in CKD mice, and these alterations were attenuated in both neural lineage-specific and astrocyte-specific Ahr conditional knockout mice (nAhrCKO and aAhrCKO). By using chronic I3S treatment in primary astrocytes and glia-neuron (GN) mix cultures to mimic the CKD brain microenvironment, we also found significant reduction of GLT1 expression and activity in an AhR-dependent manner. Chronic I3S treatment induced AhR-dependent pro-oxidant Nox1 and AhR-independent anti-oxidant HO-1 expressions. Notably, AhR mediates chronic I3S-induced neuronal activity enhancement and synaptotoxicity in GN mix, not neuron-enriched cortical culture. In CKD mice, neuronal activity enhancement was observed in ACC and hippocampal CA1, and these responses were abrogated by both nAhrCKO and aAhrCKO. Finally, intranasal AhR antagonist CH-223191 administration significantly ameliorated the GLT1/PAPs reduction, increase in c-Fos+ neurons, and memory impairment in the CKD mice. Thus, astrocytic AhR plays a crucial role in the CKD-induced disturbance of neuron-astrocyte interaction and mental disorders.


Subject(s)
Mental Disorders , Receptors, Aryl Hydrocarbon , Renal Insufficiency, Chronic , Animals , Mice , Astrocytes/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Hippocampus/metabolism , Indican/metabolism , Mental Disorders/etiology , Mental Disorders/metabolism , Mice, Knockout , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism
2.
Life Sci ; 306: 120833, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-35882273

ABSTRACT

Maintaining glutamate homeostasis through astrocyte-enriched glutamate transporter 1 (GLT-1) is critical for neuronal survival, but it is often disrupted after brain injury. Hericium erinaceus (HE), an edible mushroom, was reported to be anti-inflammatory and neuroprotective against brain ischemia, but its effect on glutamate homeostasis was unknown. Here we investigated the neuroprotective effect of erinacine A (EA), an active component of HE, with special focus on the GLT-1 function in the in vitro and in vivo cerebral ischemia mouse models. By using oxygen-glucose deprivation (OGD) to challenge mouse glia-neuron (GN) mixed culture as the in vitro model, we found that EA treatment significantly improved neuronal/astroglial survival and attenuated OGD-induced proinflammatory NFκB and AKT signaling activations. Notably, EA attenuated OGD-induced GLT-1 downregulation, and a selective GLT-1 inhibitor WAY-213613 reversed these EA-mediated neuroprotection. EA also ameliorated glutamate excitotoxicity effectively. In a transient hypoxia-ischemia (tHI) brain injury mouse model, we examined an EA treatment strategy by performing a pre-tHI daily oral gavage of EA (oEA) for 7 days followed by a post-tHI intranasal injection of EA (nEA) for 3 days, and found that this treatment significantly protected sensorimotor cortex and improved the post-tHI forepaw grip strength. Western blotting results further revealed that EA treatment also preserved astrocyte-enriched glutamate and aspartate transporter (GLAST) as well as a GLT-1 function-associated potassium channel Kir4.1 in the cerebral cortex and striatum after tHI. These results suggest that EA is effective for preserving GLT-1 and glutamate clearance machinery to protect against excitotoxicity after ischemic brain injury.


Subject(s)
Brain Injuries , Brain Ischemia , Animals , Astrocytes/metabolism , Brain Ischemia/drug therapy , Diterpenes , Down-Regulation , Excitatory Amino Acid Transporter 2/metabolism , Glucose/pharmacology , Glutamic Acid/metabolism , Mice
3.
J Neuroinflammation ; 19(1): 152, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35705957

ABSTRACT

BACKGROUND: Inflammation is a potential risk factor of mental disturbance. FKBP5 that encodes FK506-binding protein 51 (FKBP51), a negative cochaperone of glucocorticoid receptor (GR), is a stress-inducible gene and has been linked to psychiatric disorders. Yet, the role of FKBP51 in the inflammatory stress-associated mental disturbance remained unclear. METHODS: Fkbp5-deficient (Fkbp5-KO) mice were used to study inflammatory stress by a single intraperitoneal injection of lipopolysaccharide (LPS). The anxiety-like behaviors, neuroimaging, immunofluorescence staining, immunohistochemistry, protein and mRNA expression analysis of inflammation- and neurotransmission-related mediators were evaluated. A dexamethasone drinking model was also applied to examine the effect of Fkbp5-KO in glucocorticoid-induced stress. RESULTS: LPS administration induced FKBP51 elevation in the liver and hippocampus accompanied with transient sickness. Notably, Fkbp5-KO but not wild-type (WT) mice showed anxiety-like behaviors 7 days after LPS injection (LPS-D7). LPS challenge rapidly increased peripheral and central immune responses and hippocampal microglial activation followed by a delayed GR upregulation on LPS-D7, and these effects were attenuated in Fkbp5-KO mice. Whole-brain [18F]-FEPPA neuroimaging, which target translocator protein (TSPO) to indicate neuroinflammation, showed that Fkbp5-KO reduced LPS-induced neuroinflammation in various brain regions including hippocampus. Interestingly, LPS elevated glutamic acid decarboxylase 65 (GAD65), the membrane-associated GABA-synthesizing enzyme, in the hippocampus of WT but not Fkbp5-KO mice on LPS-D7. This FKBP51-dependent GAD65 upregulation was observed in the ventral hippocampal CA1 accompanied by the reduction of c-Fos-indicated neuronal activity, whereas both GAD65 and neuronal activity were reduced in dorsal CA1 in a FKBP51-independent manner. GC-induced anxiety was also examined, which was attenuated in Fkbp5-KO and hippocampal GAD65 expression was unaffected. CONCLUSIONS: These results suggest that FKBP51/FKBP5 is involved in the systemic inflammation-induced neuroinflammation and hippocampal GR activation, which may contribute to the enhancement of GAD65 expression for GABA synthesis in the ventral hippocampus, thereby facilitating resilience to inflammation-induced anxiety.


Subject(s)
Anxiety/metabolism , Glutamate Decarboxylase/metabolism , Lipopolysaccharides , Tacrolimus Binding Proteins/metabolism , Animals , Anxiety/pathology , Glucocorticoids/pharmacology , Glutamate Decarboxylase/genetics , Hippocampus/metabolism , Humans , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/toxicity , Mice , Receptors, GABA/metabolism , Receptors, Glucocorticoid/metabolism , Tacrolimus Binding Proteins/genetics , gamma-Aminobutyric Acid/metabolism
4.
Int J Mol Sci ; 22(15)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34360989

ABSTRACT

Increasing evidence suggests that elderly people with dementia are vulnerable to the development of severe coronavirus disease 2019 (COVID-19). In Alzheimer's disease (AD), the major form of dementia, ß-amyloid (Aß) levels in the blood are increased; however, the impact of elevated Aß levels on the progression of COVID-19 remains largely unknown. Here, our findings demonstrate that Aß1-42, but not Aß1-40, bound to various viral proteins with a preferentially high affinity for the spike protein S1 subunit (S1) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the viral receptor, angiotensin-converting enzyme 2 (ACE2). These bindings were mainly through the C-terminal residues of Aß1-42. Furthermore, Aß1-42 strengthened the binding of the S1 of SARS-CoV-2 to ACE2 and increased the viral entry and production of IL-6 in a SARS-CoV-2 pseudovirus infection model. Intriguingly, data from a surrogate mouse model with intravenous inoculation of Aß1-42 show that the clearance of Aß1-42 in the blood was dampened in the presence of the extracellular domain of the spike protein trimers of SARS-CoV-2, whose effects can be prevented by a novel anti-Aß antibody. In conclusion, these findings suggest that the binding of Aß1-42 to the S1 of SARS-CoV-2 and ACE2 may have a negative impact on the course and severity of SARS-CoV-2 infection. Further investigations are warranted to elucidate the underlying mechanisms and examine whether reducing the level of Aß1-42 in the blood is beneficial to the fight against COVID-19 and AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Peptide Fragments/metabolism , SARS-CoV-2/enzymology , Spike Glycoprotein, Coronavirus/metabolism , A549 Cells , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Animals , COVID-19/complications , COVID-19/metabolism , Chlorocebus aethiops , Humans , Interleukin-6/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/chemistry , Protein Subunits/chemistry , Protein Subunits/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Vero Cells , Virus Internalization
5.
Mol Neurobiol ; 56(12): 8475-8476, 2019 12.
Article in English | MEDLINE | ID: mdl-31317492

ABSTRACT

The original version of this article unfortunately contained a mistake. The authors observed inadvertent error in Fig. 7d, in which the image of the GFAP/DAPI in the WT saline treated mice was rotated left 90-degree by mistake. The corrected representative image is given below.

6.
Mol Neurobiol ; 56(12): 8451-8474, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31257558

ABSTRACT

Astrocytes play pivotal roles in regulating glutamate homeostasis at tripartite synapses. Inhibition of soluble epoxide hydrolase (sEHi) provides neuroprotection by blocking the degradation of 14,15-epoxyeicosatrienoic acid (14,15-EET), a lipid mediator whose synthesis can be activated downstream from group 1 metabotropic glutamate receptor (mGluR) signaling in astrocytes. However, it is unclear how sEHi regulates glutamate excitotoxicity. Here, we used three primary rat cortical culture systems, neuron-enriched (NE), astrocyte-enriched glia-neuron mix (GN), and purified astrocytes, to delineate the underlying mechanism by which sEHi and 14,15-EET attenuate excitotoxicity. We found that sEH inhibitor 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA) and 14,15-EET both attenuated N-methyl-D-aspartate (NMDA)-induced neurite damage and cell death in GN, not NE, cortical cultures. The anti-excitotoxic effects of 14,15-EET and AUDA were both blocked by the group 1 mGluR5 antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP), as were their protective effects against NMDA-disrupted perineuronal astrocyte processes expressing glutamate transporter-1 (GLT-1) and subsequent glutamate uptake. Knockdown of sEH expression also attenuated NMDA neurotoxicity in mGluR5- and GLT-1-dependent manners. The 14,15-EET/AUDA-preserved astroglial integrity was confirmed in glutamate-stimulated primary astrocytes along with the reduction of the c-Jun N-terminal kinase 1 phosphorylation, in which the 14,15-EET effect is mGluR5-dependent. In vivo studies validated that sEHi and genetic deletion of sEH (Ephx2-KO) ameliorated excitotoxic kainic acid-induced seizure, memory impairment, and neuronal loss while preserving GLT-1-expressing perineuronal astrocytes in hippocampal CA3 subregions. These results suggest that 14,15-EET mediates mGluR5-dependent anti-excitotoxicity by protecting astrocytes to maintain glutamate homeostasis, which may account for the beneficial effect of sEH inhibition in excitotoxic brain injury and diseases.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Astrocytes/pathology , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Glutamic Acid/metabolism , Homeostasis , Neuronal Plasticity/drug effects , Neurotoxins/toxicity , 8,11,14-Eicosatrienoic Acid/pharmacology , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Survival/drug effects , Cells, Cultured , Epoxide Hydrolases/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Hippocampus/metabolism , Kainic Acid , Lauric Acids/pharmacology , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/metabolism , Models, Biological , N-Methylaspartate , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Receptor, Metabotropic Glutamate 5/metabolism , Solubility
7.
J Neuroinflammation ; 16(1): 123, 2019 Jun 08.
Article in English | MEDLINE | ID: mdl-31176371

ABSTRACT

BACKGROUND: Astrocyte activation is a common pathological feature in many brain diseases with neuroinflammation, and revealing the underlying mechanisms might shed light on the regulatory processes of the diseases. Recently, soluble epoxide hydrolase (sEH) has been proposed to affect neuroinflammation in brain injuries. However, the roles of astrocytic sEH in brains with neurodegeneration remain unclear. METHODS: The expression of astrocytic sEH in the brains of APPswe/PSEN1dE9 (APP/PS1) mice developing Alzheimer's disease (AD)-like pathology was evaluated by confocal imaging. LPS-activated primary astrocytes with mRNA silencing or overexpression of sEH were used to investigate its regulatory roles in astrocyte activation and the induction of pro-inflammatory markers. Primary astrocytes isolated from a sEH knockout (sEH-/-) background were also applied. RESULTS: The immunoreactivity of sEH was increased in activated astrocytes in parallel with the progression of AD in APP/PS1 mice. Our data from primary astrocyte cultures further demonstrate that the overexpression of sEH ameliorated, while the silencing of sEH mRNA enhanced, the lipopolysaccharides (LPS)-induced expression of pro-inflammatory markers, such as inducible nitric oxide, cyclooxygenase 2 (COX-2), and pro-inflammatory cytokines. These findings suggest that sEH negatively regulates astrocyte immune responses. Enhanced immune responses found in LPS-activated sEH-/- astrocytes also support the notion that the expression of sEH could suppress the immune responses during astrocyte activation. Similarly, sEH-/- mice that received intraperitoneal injection of LPS showed exacerbated astrocyte activation in the brain, as observed by the elevated expression of glial fibrillary acidic protein (GFAP) and pro-inflammatory markers. Moreover, our data show that the phosphorylation of the signal transducer and activator of transcription 3 (STAT3) was upregulated in activated astrocytes from sEH mouse brains, and the pharmacological blockade of STAT3 activity alleviated the pro-inflammatory effects of sEH deletion in LPS-activated primary astrocytes. CONCLUSIONS: Our results provide evidence, for the first time, showing that sEH negatively regulates astrocytic immune responses and GFAP expression, while the underlying mechanism at least partly involves the downregulation of STAT3 phosphorylation. The discovery of a novel function for sEH in the negative control of astrocytic immune responses involving STAT3 activation confers further insights into the regulatory machinery of astrocyte activation during the development of neurodegeneration.


Subject(s)
Astrocytes/immunology , Epoxide Hydrolases/immunology , STAT3 Transcription Factor/immunology , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Animals , Astrocytes/metabolism , Epoxide Hydrolases/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Knockout , Mice, Transgenic , STAT3 Transcription Factor/metabolism
8.
Mol Neurobiol ; 54(2): 866-873, 2017 03.
Article in English | MEDLINE | ID: mdl-26780456

ABSTRACT

Protein phosphorylation plays an important role in regulating soluble L-glutamic acid decarboxylase (GAD) and membrane-associated GAD activity. Previously, we reported the effect of phosphorylation on the two well-defined GAD isoforms, namely, GAD65 and GAD67, using highly purified preparations of recombinant human brain GAD65 (hGAD65) and GAD67. GAD65 was activated by phosphorylation, while GAD67 was inhibited by phosphorylation. The effect of phosphorylation on GAD65 and GAD67 could be reversed by treatment with protein phosphatases. We further demonstrated that protein kinase A (PKA) and protein kinase C isoform ε were the protein kinases responsible for phosphorylation and regulation of GAD67 and GAD65, respectively. In the current study, using MALDI-TOF, a total of four potential phosphorylation sites were identified in GAD65, two of which (threonine-95 (T-95) and Ser-417) were not reported previously. We have identified one specific phosphorylation site, (T95), in hGAD65 that can be phosphorylated by kinase C ε (PKCε) using MALDITOF. When T95 is mutated to alanine, hGAD65 could no longer be phosphorylated by PKCε, and the effect of PKC-mediated activation on hGAD65 is abolished. However, when T95 is mutated to glutamic acid, which mimics the phosphorylation status of hGAD65, the activity was greatly increased. An increase of GAD65 activity by 55 % compared to the wild type hGAD65 was observed indicating that mutation of T95 to glutamic acid mimics the effect of phosphorylation. A model depicting the role of phosphorylation of GAD65 in regulation of GABA neurotransmission is presented.


Subject(s)
Brain/enzymology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Threonine/genetics , Threonine/metabolism , Animals , Brain/pathology , Enzyme Activation/physiology , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley
9.
J Neurosci ; 36(6): 2027-43, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26865625

ABSTRACT

Growth-associated protein 43 (GAP43), a protein kinase C (PKC)-activated phosphoprotein, is often implicated in axonal plasticity and regeneration. In this study, we found that GAP43 can be induced by the endotoxin lipopolysaccharide (LPS) in rat brain astrocytes both in vivo and in vitro. The LPS-induced astrocytic GAP43 expression was mediated by Toll-like receptor 4 and nuclear factor-κB (NF-κB)- and interleukin-6/signal transducer and activator of transcription 3 (STAT3)-dependent transcriptional activation. The overexpression of the PKC phosphorylation-mimicking GAP43(S41D) (constitutive active GAP43) in astrocytes mimicked LPS-induced process arborization and elongation, while application of a NF-κB inhibitory peptide TAT-NBD or GAP43(S41A) (dominant-negative GAP43) or knockdown of GAP43 all inhibited astrogliosis responses. Moreover, GAP43 knockdown aggravated astrogliosis-induced microglial activation and expression of proinflammatory cytokines. We also show that astrogliosis-conditioned medium from GAP43 knock-down astrocytes inhibited GAP43 phosphorylation and axonal growth, and increased neuronal damage in cultured rat cortical neurons. These proneurotoxic effects of astrocytic GAP43 knockdown were accompanied by attenuated glutamate uptake and expression of the glutamate transporter excitatory amino acid transporter 2 (EAAT2) in LPS-treated astrocytes. The regulation of EAAT2 expression involves actin polymerization-dependent activation of the transcriptional coactivator megakaryoblastic leukemia 1 (MKL1), which targets the serum response elements in the promoter of rat Slc1a2 gene encoding EAAT2. In sum, the present study suggests that astrocytic GAP43 mediates glial plasticity during astrogliosis, and provides beneficial effects for neuronal plasticity and survival and attenuation of microglial activation. SIGNIFICANCE STATEMENT: Astrogliosis is a complex state in which injury-stimulated astrocytes exert both protective and harmful effects on neuronal survival and plasticity. In this study, we demonstrated for the first time that growth-associated protein 43 (GAP43), a well known growth cone protein that promotes axonal regeneration, can be induced in rat brain astrocytes by the proinflammatory endotoxin lipopolysaccharide via both nuclear factor-κB and signal transducer and activator of transcription 3-mediated transcriptional activation. Importantly, LPS-induced GAP43 mediates plastic changes of astrocytes while attenuating astrogliosis-induced microglial activation and neurotoxicity. Hence, astrocytic GAP43 upregulation may serve to indicate beneficial astrogliosis after CNS injury.


Subject(s)
Astrocytes/drug effects , GAP-43 Protein/biosynthesis , GAP-43 Protein/genetics , Gliosis/genetics , Microglia/drug effects , NF-kappa B/genetics , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/pathology , STAT3 Transcription Factor/genetics , Toll-Like Receptor 4/genetics , Animals , Cytokines/biosynthesis , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/genetics , Macrophage Activation/drug effects , Neurons , Phosphorylation , Rats , Rats, Sprague-Dawley , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transcription Factors
10.
Mol Cell Biol ; 35(10): 1712-26, 2015 May.
Article in English | MEDLINE | ID: mdl-25755278

ABSTRACT

Growth-associated protein 43 (GAP43) is known to regulate axon growth, but whether it also plays a role in synaptogenesis remains unclear. Here, we found that GAP43 regulates the aggregation of gephyrin, a pivotal protein for clustering postsynaptic GABA(A) receptors (GABA(A)Rs), in developing cortical neurons. Pharmacological blockade of either protein kinase C (PKC) or neuronal activity increased both GAP43-gephyrin association and gephyrin misfolding-induced aggregation, suggesting the importance of PKC-dependent regulation of GABAergic synapses. Furthermore, we found that PKC phosphorylation-resistant GAP43(S41A), but not PKC phosphorylation-mimicking GAP43(S41D), interacted with cytosolic gephyrin to trigger gephyrin misfolding and its sequestration into aggresomes. In contrast, GAP43(S41D), but not GAP43(S41A), inhibited the physiological aggregation/clustering of gephyrin, reduced surface GABA(A)Rs under physiological conditions, and attenuated gephyrin misfolding under transient oxygen-glucose deprivation (tOGD) that mimics pathological neonatal hypoxia. Calcineurin-mediated GAP43 dephosphorylation that accompanied tOGD also led to GAP43-gephyrin association and gephyrin misfolding. Thus, PKC-dependent phosphorylation of GAP43 plays a critical role in regulating postsynaptic gephyrin aggregation in developing GABAergic synapses.


Subject(s)
Carrier Proteins/metabolism , GABAergic Neurons/metabolism , GAP-43 Protein/metabolism , Membrane Proteins/metabolism , Protein Kinase C/metabolism , Animals , Carrier Proteins/chemistry , Cells, Cultured , Female , Flavonoids/pharmacology , GABAergic Neurons/cytology , GAP-43 Protein/chemistry , HEK293 Cells , Humans , Indoles/pharmacology , Membrane Proteins/chemistry , Phosphorylation , Pregnancy , Protein Folding/drug effects , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
11.
Glia ; 63(7): 1138-54, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25690886

ABSTRACT

The aryl hydrocarbon receptor (AhR) regulates peripheral immunity; but its role in microglia-mediated neuroinflammation in the brain remains unknown. Here, we demonstrate that AhR mediates both anti-inflammatory and proinflammatory effects in lipopolysaccharide (LPS)-activated microglia. Activation of AhR by its ligands, formylindolo[3,2-b]carbazole (FICZ) or 3-methylcholanthrene (3MC), attenuated LPS-induced microglial immune responses. AhR also showed proinflammatory effects, as evidenced by the findings that genetic silence of AhR ameliorated the LPS-induced microglial immune responses and LPS-activated microglia-mediated neurotoxicity. Similarly, LPS-induced expressions of tumor necrosis factor α (TNFα) and inducible nitric oxide synthase (iNOS) were reduced in the cerebral cortex of AhR-deficient mice. Intriguingly, LPS upregulated and activated AhR in the absence of AhR ligands via the MEK1/2 signaling pathway, which effects were associated with a transient inhibition of cytochrome P450 1A1 (CYP1A1). Although AhR ligands synergistically enhance LPS-induced AhR activation, leading to suppression of LPS-induced microglial immune responses, they cannot do so on their own in microglia. Chromatin immunoprecipitation results further revealed that LPS-FICZ co-treatment, but not LPS alone, not only resulted in co-recruitment of both AhR and NFκB onto the κB site of TNFα gene promoter but also reduced LPS-induced AhR binding to the DRE site of iNOS gene promoter. Together, we provide evidence showing that microglial AhR, which can be activated by LPS, exerts bi-directional effects on the regulation of LPS-induced neuroinflammation, depending on the availability of external AhR ligands. These findings confer further insights into the potential link between environmental factors and the inflammatory brain disorders.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Microglia/physiology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Death/physiology , Cell Line , Cells, Cultured , Cerebral Cortex/immunology , Chromatin/metabolism , Cytochrome P-450 CYP1A1/metabolism , Gene Knockdown Techniques , Lipopolysaccharides , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Mice, Inbred BALB C , Mice, Knockout , Neurons/physiology , Nitric Oxide Synthase Type II/metabolism , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
12.
J Ethnopharmacol ; 156: 358-64, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25178949

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Our previous study indicated that the traditional Chinese medicine (TCM) formula Liu-Wei-Di-Huang-Wan, which consists of six type of herbs, namely Rehmannia glutinosa (Gaertn.) DC., root, dried; Cornus officinalis Siebold & Zucc., fructus, dried; Dioscorea oppositifolia L., root, dried; Alisma plantago-aquatica subsp. orientale (Sam.) Sam., tuber, dried; Paeonia × suffruticosa Andrews, bark, dried; Poria cocos (Fr.) Wolf., sclerotium, dried, is the most frequently prescribed herbal formula used to treat type 2 diabetes patients. The aim of the study was to evaluate the integration of TCM into diabetes care in terms of how it reduces the risk of developing kidney failure. MATERIALS AND METHODS: The Taiwan׳s National Health Insurance Research Database (NHIRD) provided detailed information of health care services for each patient and covers 98% of all Taiwan residents as of 2007. Case and control subjects were selected from the NHIRD. Two multivariable logistic regression models were constructed in order to explore two types of exposure assessments including prescription of TCMs (model 1) and prescription of different estimated dosages of Liu-Wei-Di-Huang-Wan (model 2). RESULTS: Using logistic regression model 1, having used TCMs was independently associated with a decreased risk of kidney failure by multivariable analysis (OR=0.69, 95% CI: 0.61-0.77). Using logistic regression model 2, there was no difference between non-Liu-Wei-Di-Huang-Wan TCM users and Liu-Wei-Di-Huang-Wan TCM users in terms of the risk of developing kidney failure. Furthermore, there was also no linear dose-response trend when we used exposure to prescribed Liu-Wei-Di-Huang-Wan as a continuous variable (for non-Liu-Wei-Di-Huang-Wan TCM users, OR=0.68, 95% CI: 0.60-0.77; for TCM users consuming 1-30 g of Liu-Wei-Di-Huang-Wan, OR=0.69, 95% CI: 0.54-0.87; for >30 g of Liu-Wei-Di-Huang-Wan, OR=0.84, 95% CI: 0.49-1.44). CONCLUSIONS: Integrating TCM healthcare into diabetes care was found to be associated with a decreased risk of developing kidney failure. Having recognized the use of TCM, exploring any potential interactions and adverse effects, and integrating both technologies into a holistic treatment system may be beneficial to the relief of diabetic nephropathy on patients with type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drugs, Chinese Herbal/adverse effects , Drugs, Chinese Herbal/therapeutic use , Medicine, Chinese Traditional/adverse effects , Renal Insufficiency/chemically induced , Adult , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged , Risk , Young Adult
13.
Chem Biol Interact ; 188(1): 228-36, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20637185

ABSTRACT

Increasing evidence indicates that microglial activation plays an important role in the pathogenesis of Alzheimer's disease (AD). In AD, activated microglia may facilitate the clearance of beta-amyloid (Abeta), a neurotoxic component in AD pathogenesis. However, microglial activation comes at the cost of triggering neuro-inflammation, which contributes to cerebral dysfunction. Thus, pharmacological approaches that can achieve a favorable combination of a reduced microglia-mediated neuro-inflammation, and an enhanced Abeta clearance may be beneficial for preventing the progression of the disease. Here, we show that some newly synthesized compounds may exert such a combination of functions. Using mouse primary microglia and RAW264.7 cells, we found that some thiourea derivatives significantly enhanced microglial Abeta phagocytosis and suppressed microglial immune responses, as evidenced by the reduced expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2). Of note, some commercially available inhibitors for iNOS and/or COX-2, such as ibuprofen, dextromethorphan, and N(G)-methyl-l-arginine (l-NMA), show negligible effects on microglial Abeta phagocytosis. Among the thiourea derivatives, our data show that a lead compound, designated as compound #326, (1-Naphthalen-1-yl-3-[5-(3-thioureido-phenoxy)-pentyl]-thiourea) appears to be the most potent in promoting Abeta phagocytosis and in inhibiting the LPS-induced expression of iNOS and COX-2 (when used at concentrations in the low muM range). The potency of compound #326 may have beneficial effects on modulating microglial activation in AD. The structure-activity relationship indicates that the thiourea group, alkyl linker, and the hydrophobic aryl group largely influence the dual functions of the compounds. These findings may indicate a structural basis for the improved design of future drug therapies for AD.


Subject(s)
Microglia/drug effects , Thiourea/pharmacology , Animals , Blotting, Western , Cell Line , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Inbred BALB C , Microglia/immunology
14.
Curr Med Chem ; 16(6): 643-51, 2009.
Article in English | MEDLINE | ID: mdl-19199928

ABSTRACT

Alzheimer's disease (AD) is the leading cause of dementia in the elderly. Although the etiology of AD remains unclear, microglia-mediated neuroinflammation is believed to play an important role in its pathogenesis. Microglial activation occurs in AD and is characterized by apparent phagocytic activity and by increased production and secretion of several cytokines, chemokines, reactive oxygen and nitrogen species, prostaglandin (PG)E2, and neurotrophic factors. Microglial activation can be neuroprotective through the release of neurotrophic factors and by phagocytosing Abeta, a critical neurotoxic component in AD brain. Concurrently, microglial activation causes elevated inflammatory responses that lead to paracrine damage to neurons. Therefore, a well-controlled microglial activation that diminishes microglial-mediated oxidative damage while promoting neuronal protection may be the key for AD therapy. Peroxisome proliferator-activated receptor gamma (PPARgamma) has recently gained increasing attention in AD due to its function as a molecular target for non-steroidal anti-inflammatory drugs (NSAIDs). In this review, we will discuss the role of PPARgamma in microglial innate immunity in AD and how pharmacological manipulation of microglial activation using PPARgamma ligands might facilitate the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Microglia/immunology , PPAR gamma/immunology , Alzheimer Disease/pathology , Animals , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Ligands , Microglia/drug effects
15.
Eur J Neurosci ; 18(11): 2981-98, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14656294

ABSTRACT

In this study we have characterized the membrane properties and morphology of interneurons which lie between the caudal pole of the trigeminal motor nucleus and the rostral border of the facial motor nucleus. Previous studies suggest that many of these interneurons may participate in the genesis of rhythmical jaw movements. Saggital brainstem slices were taken from rats aged 5-8 days. Interneurons lying caudal to the trigeminal motor nucleus were visualized using near-infrared differential interference contrast (DIC) microscopy, and were recorded from using patch pipettes filled with a K-gluconate- and biocytin-based solution. The 127 neurons recorded could be categorized into three subtypes on the basis of their responses to injection of depolarizing current pulses, namely tonic firing (type I), burst firing (type II) and spike-adaptive (type III) neurons. Type I interneurons had a higher input resistance and a lower rheobase than type II neurons. All three neuron subtypes showed 'sag' of the voltage response to injection of large-amplitude hyperpolarizing current pulses, and, in addition, also showed rectification of the voltage response to injection of depolarizing current pulses, with type II neurons showing significantly greater rectification than type I neurons. The axonal arborizations were reconstructed for 44 of 63 neurons labelled with tracer. Neurons of each subtype were found to issue axon collaterals terminating in the brainstem nuclei, including the parvocellular reticular nucleus (PCRt), the trigeminal motor nucleus (Vmot), the supratrigeminal nucleus or the trigeminal mesencephalic nucleus. Twenty-five of the 43 neurons issued collaterals which terminated in the Vmot and the other brainstem nuclei. When viewed under 100x magnification, the collaterals of some interneurons were seen to give off varicosities and end-terminations which passed close to the somata of unidentified neurons in the trigeminal motor nucleus and in the area close to the interneuron soma itself. This suggests that the interneurons may make synaptic contacts both on motoneurons and also on nearby interneurons. These results provide data on the membrane properties of trigeminal interneurons and evidence for their synaptic connections both with nearby interneurons and also with motoneurons. Thus, the interneurons examined could play roles in the shaping, and possibly also in the generation, of rhythmical signals to trigeminal motoneurons.


Subject(s)
Interneurons/physiology , Lysine/analogs & derivatives , Motor Neurons/physiology , Trigeminal Nuclei/physiology , Anesthetics, Local/pharmacology , Animals , Animals, Newborn , Axons/metabolism , Brain Stem/cytology , Brain Stem/physiology , Calcium/physiology , Electric Conductivity , Electric Stimulation , In Vitro Techniques , Interneurons/classification , Interneurons/cytology , Interneurons/drug effects , Lysine/metabolism , Membrane Potentials/physiology , Motor Neurons/classification , Motor Neurons/cytology , Motor Neurons/drug effects , Neural Conduction , Patch-Clamp Techniques , Rats , Rats, Wistar , Tetrodotoxin/pharmacology , Trigeminal Nuclei/cytology , Trigeminal Nuclei/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...