Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Cerebellum ; 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38472628

ABSTRACT

Responding to burst stimulation of parallel fibers (PFs), cerebellar Purkinje neurons (PNs) generate a convolved synaptic response displaying a fast excitatory postsynaptic current (EPSCFast) followed by a slow EPSC (EPSCSlow). The latter is companied with a rise of intracellular Ca2+ and critical for motor coordination. The genesis of EPSCSlow in PNs results from activation of metabotropic type 1 glutamate receptor (mGluR1), oligomerization of stromal interaction molecule 1 (STIM1) on the membrane of endoplasmic reticulum (ER) and opening of transient receptor potential canonical 3 (TRPC3) channels on the plasma membrane. Neuronal nitric oxide synthase (nNOS) is abundantly expressed in PFs and granule neurons (GNs), catalyzing the production of nitric oxide (NO) hence regulating PF-PN synaptic function. We recently found that nNOS/NO regulates the morphological development of PNs through mGluR1-regulated Ca2+-dependent mechanism. This study investigated the role of nNOS/NO in regulating EPSCSlow. Electrophysiological analyses showed that EPSCSlow in cerebellar slices of nNOS knockout (nNOS-/-) mice was significantly larger than that in wildtype (WT) mice. Activation of mGluR1 in cultured PNs from nNOS-/- mice evoked larger TRPC3-channel mediated currents and intracellular Ca2+ rise than that in PNs from WT mice. In addition, nNOS inhibitor and NO-donor increased and decreased, respectively, the TRPC3-current and Ca2+ rise in PNs. Moreover, the NO-donor effectively decreased TRPC3 currents in HEK293 cells expressing WT STIM1, but not cells expressing a STIM1 with cysteine mutants. These novel findings indicate that nNOS/NO inhibits TRPC3-containig channel mediated cation influx during EPSCSlow, at least in part, by S-nitrosylation of STIM1.

2.
Virus Res ; 344: 199366, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38548137

ABSTRACT

Gamma-aminobutyric acid (GABA) signals in various non-neuronal cells including hepatocytes and some immune cells. Studies, including ours, show that type A GABA receptors (GABAARs)-mediated signaling occurs in macrophages regulating tissue-specific functions. Our recent study reveals that activation of GABAARs in liver macrophages promotes their M2-like polarization and increases HBV replication in mice. This short article briefly summarizes the GABA signaling system in macrophages and discusses potential mechanisms by which GABA signaling promotes HBV replication.


Subject(s)
Hepatitis B , Macrophages , Receptors, GABA-A , Signal Transduction , Virus Replication , gamma-Aminobutyric Acid , Animals , Humans , Mice , Disease Models, Animal , gamma-Aminobutyric Acid/metabolism , Hepatitis B/virology , Hepatitis B/metabolism , Hepatitis B virus/physiology , Hepatitis B virus/genetics , Liver/virology , Liver/metabolism , Macrophages/virology , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics
3.
Antiviral Res ; 217: 105680, 2023 09.
Article in English | MEDLINE | ID: mdl-37494980

ABSTRACT

Macrophages display functional phenotypic plasticity. Hepatitis B virus (HBV) infection induces polarizations of liver macrophages either to M1-like pro-inflammatory phenotype or to M2-like anti-inflammatory phenotype. Gamma-aminobutyric acid (GABA) signaling exists in various non-neuronal cells including hepatocytes and some immune cells. Here we report that macrophages express functional GABAergic signaling components and activation of type A GABA receptors (GABAARs) promotes M2-polarization thus advancing HBV replication. Notably, intraperitoneal injection of GABA or the GABAAR agonist muscimol increased HBV replication in HBV-carrier mice that were generated by hydrodynamical injection of adeno-associated virus/HBV1.2 plasmids (pAAV/HBV1.2). The GABA-augmented HBV replication in HBV-carrier mice was significantly reduced by the GABAAR inhibitor picrotoxin although picrotoxin had no significant effect on serum HBsAg levels in control HBV-carrier mice. Depletion of liver macrophages by liposomal clodronate treatment also significantly reduced the GABA-augmented HBV replication. Yet adoptive transfer of liver macrophages isolated from GABA-treated donor HBV-carrier mice into the liposomal clodronate-pretreated recipient HBV-carrier mice restored HBV replication. Moreover, GABA or muscimol treatment increased the expression of "M2" cytokines in macrophages, but had no direct effect on HBV replication in the HepG2.2.15 cells, HBV1.3-transfected Huh7, HepG2, or HepaRG cells, or HBV-infected Huh7-NTCP cells. Taken together, these results suggest that increasing GABA signaling in the liver promotes HBV replication in HBV-carrier mice by suppressing the immunity of liver macrophages, but not by increasing the susceptibility of hepatocytes to HBV infection. Our study shows that a previously unknown GABAergic system in liver macrophage has an essential role in HBV replication.


Subject(s)
Hepatitis B virus , Hepatitis B , Mice , Animals , Hepatitis B virus/genetics , Muscimol/pharmacology , Clodronic Acid/pharmacology , Picrotoxin/pharmacology , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology , Macrophages/metabolism , Virus Replication
4.
Int J Med Sci ; 20(6): 702-708, 2023.
Article in English | MEDLINE | ID: mdl-37213671

ABSTRACT

This study aimed to investigate the possible association between nasopharyngeal carcinoma (NPC) and following open angle glaucoma (OAG). A retrospective research applying the National Health Insurance Research Database (NHIRD) of Taiwan was conducted with a follow up period from January 1, 2000 to December 31, 2016. There were 4184 and 16736 participants that selected and categorized into the NPC and non-NPC groups after exclusion. The major outcome of our study was the development of OAG according to diagnostic codes, exam and managements. The Cox proportional hazard regression was employed to estimate the adjusted hazard ratio (aHR) and 95% confidence interval (CI) of OAG between the two groups. In this study, a numbers of 151 and 513 OAG episodes occurred in the NPC and non-NPC groups and the NPC population showed a significantly higher incidence of OAG compared to the non-NPC population in multivariable analysis (aHR: 1.293, 95% CI: 1.077-1.551, p = 0.0057). Besides, the cumulative probability of OAG was significantly higher in the NPC group than that in the non-NPC population (p = 0.0041). About other risk factor for OAG, age older than 40 years old, diabetes mellitus and persistent steroid usage were related to OAG occurrence (all p < 0.05). In conclusion, the NPC may be an independent risk factor of following OAG development.


Subject(s)
Glaucoma, Open-Angle , Nasopharyngeal Neoplasms , Humans , Adult , Cohort Studies , Retrospective Studies , Glaucoma, Open-Angle/epidemiology , Glaucoma, Open-Angle/etiology , Glaucoma, Open-Angle/diagnosis , Nasopharyngeal Carcinoma/epidemiology , Risk Factors , Incidence , Nasopharyngeal Neoplasms/epidemiology
5.
Cerebellum ; 22(6): 1200-1215, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36402869

ABSTRACT

The cerebellum is a major site of endocannabinoid (eCB) production and signaling. The predominant eCB within the cerebellum, 2-arachidonoylglycerol (2-AG), is produced by a metabotropic glutamate receptor type 1 (mGluR1)-initiated signaling cascade within Purkinje neurons (PNs). 2-AG retrogradely stimulates cannabinoid 1 receptors (CB1Rs) located on presynaptic terminals. The activated CB1R decreases neurotransmitter release and leads to the production of nitric oxide (NO), a gaseous molecule. Recently, our group discovered that during development in mice lacking neuronal nitric oxide synthase (nNOS-/-), PNs display an excitotoxic phenotype associated with overactivated mGluR1. Considering the importance of mGluR1 in 2-AG synthesis, the present study explored the role of nNOS-derived NO in regulating the eCB pathway within the cerebella of wildtype (WT) and nNOS-/- mice at postnatal day 7 (PD7), 2 weeks (2 W), and 7 weeks (7 W). Our analysis showed that diacylglycerol lipase α, the enzyme that catalyzes 2-AG production, was elevated at early postnatal ages, and followed by elevated levels of 2-AG in nNOS-/- cerebella compared to WT. CB1R expression in nNOS-/- cerebella was upregulated at PD7 but decreased at 2 W and 7 W when compared to age-matched WT mice cerebella. Importantly, treating organotypic nNOS-/- cerebellar slice cultures with an NO-donor-attenuated CB1R levels after 7 days in vitro. In addition, expression of the eCB hydrolases fatty acid amide hydrolase and monoacylglycerol lipase were significantly downregulated in nNOS-/- cerebella compared to WT cerebella at 7 W. Together, these results reveal a novel role for nNOS/NO in regulating eCB signaling in the cerebellum.


Subject(s)
Cerebellum , Endocannabinoids , Mice , Animals , Endocannabinoids/metabolism , Nitric Oxide Synthase Type I/metabolism , Cerebellum/metabolism , Purkinje Cells/physiology , Synaptic Transmission/physiology , Nitric Oxide
6.
Sci Rep ; 12(1): 711, 2022 01 13.
Article in English | MEDLINE | ID: mdl-35027613

ABSTRACT

The purpose of this retrospective interventional case series is to compare the functional and anatomical outcomes in eyes with diabetic macular edema (DME) and proliferative diabetic retinopathy (PDR) treated intravitreally with aflibercept or ranibizumab under the Taiwan National Insurance Bureau reimbursement policy. 84 eyes were collected and all eyes were imaged with spectral-domain optical coherence tomography (SD-OCT), color fundus photographs (CFPs), and fluorescein angiography (FA). At 24 months after therapy initiation, the logMAR BCVA improved from 0.58 ± 0.33 to 0.47 ± 0.38 (p < 0.01), the CRT decreased from 423.92 ± 135.84 to 316.36 ± 90.02 (p < 0.01), and the number of microaneurysms decreased from 142.14 ± 57.23 to 75.32 ± 43.86 (p < 0.01). The mean injection count was 11.74 ± 5.44. There was no intergroup difference in logMAR BCVA (p = 0.96), CRT (p = 0.69), or injection count (p = 0.81). However, the mean number of microaneurysms was marginally reduced (p = 0.06) in eyes treated with aflibercept at the end of the follow-up, and the incidence rates of supplementary panretinal photocoagulation (PRP) (p = 0.04) and subthreshold micropulse laser (SMPL) therapy sessions (p = 0.01) were also reduced. Multivariate analysis revealed that only initial logMAR BCVA influenced the final VA improvements (odds ratio (OR) 0.49, 95% confidence interval (CI) 0.21 ~ 0.93, p < 0.01); in contrast, age (OR - 0.38, 95% CI - 6.97 ~ - 1.85, p < 0.01) and initial CRT (OR 0.56, 95% CI 0.34 ~ 0.84, p < 0.01) both influenced the final CRT reduction at 24 months. To sum up, both aflibercept and ranibizumab are effective in managing DME with PDR in terms of VA, CRT and MA count. Eyes receiving aflibercept required less supplementary PRP and SMPL treatment than those receiving ranibizumab. The initial VA influenced the final VA improvements at 24 months, while age and initial CRT were prognostic predictors of 24-month CRT reduction.


Subject(s)
Diabetes Complications , Diabetic Retinopathy/therapy , Insurance, Health, Reimbursement , Macular Edema/therapy , National Health Programs , Ranibizumab/therapeutic use , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Aged , Diabetic Retinopathy/diagnostic imaging , Female , Humans , Laser Coagulation , Light Coagulation , Macular Edema/diagnostic imaging , Macular Edema/etiology , Male , Middle Aged , Prognosis , Ranibizumab/administration & dosage , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Retrospective Studies , Taiwan , Time Factors , Treatment Outcome
7.
Glia ; 70(5): 858-874, 2022 05.
Article in English | MEDLINE | ID: mdl-35006609

ABSTRACT

Bergmann glia (BG) predominantly use glutamate/aspartate transporters (GLAST) for glutamate uptake in the cerebellum. Recently, nitric oxide (NO) treatment has been shown to upregulate GLAST function and increase glutamate uptake in vitro. We previously discovered that neuronal nitric oxide synthase knockout (nNOS-/- ) mice displayed structural and functional neuronal abnormalities in the cerebellum during development, in addition to previously reported motor deficits. Although these developmental deficits have been identified in the nNOS-/- cerebellum, it is unknown whether BG morphology and GLAST expression are also affected in the absence of nNOS in vivo. This study is the first to characterize BG morphology and GLAST expression during development in nNOS-/- mice using immunohistochemistry and western blotting across postnatal development. Results showed that BG in nNOS-/- mice exhibited abnormal morphology and decreased GLAST expression compared with wildtype (WT) mice across postnatal development. Treating ex vivo WT cerebellar slices with the NOS inhibitor L-NAME decreased GLAST expression while treating nNOS-/- slices with the slow-release NO-donor NOC-18 increased GLAST expression when compared with their respective controls. In addition, treating primary BG isolated from WT mice with the selective nNOS inhibitor 7N decreased the membrane expression of GLAST and influx of Ca2+ /Na+ , while treating nNOS-/- BG with SNAP increased the membrane expression of GLAST and Ca2+ /Na+ influx. Moreover, the effects of SNAP on GLAST expression and Ca2+ /Na+ influx in nNOS-/- BG were significantly reduced by a PKG inhibitor. Together, these results reveal a novel role for nNOS/NO signaling in BG development, regulated by a PKG-mediated mechanism.


Subject(s)
Amino Acid Transport System X-AG , Glutamic Acid , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Aspartic Acid , Cerebellum/metabolism , Glutamic Acid/metabolism , Mice , Mice, Knockout , Neuroglia/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism
8.
Thyroid ; 32(2): 128-137, 2022 02.
Article in English | MEDLINE | ID: mdl-34652970

ABSTRACT

Background: XB130 (actin filament-associated protein 1-like 2, AFAP1L2) is a thyroid-abundant adaptor/scaffold protein. Xb130-/- mice exhibit transient growth retardation postnatally due to congenital hypothyroidism with diminished thyroglobulin iodination and release at both embryonic and early postnatal stages due to disorganized thyroid apical membrane structure and function. We hypothesized that XB130 is crucial for polarity and folliculogenesis by mediating proper cytoskeletal structure and function in thyrocytes. Methods: Primary thyrocytes isolated from thyroid glands of Xb130-/- mice and their wild-type littermates at postnatal week 2 were cultured in 10% Matrigel for different time periods. Folliculogenesis was studied with immunofluorescence staining, followed by confocal microscopy. Cells were also transfected to express human XB130 fused Green Fluorescent Protein (XB130-GFP) or Green Fluorescent Protein (GFP) only before morphological analysis. Cytoskeletal structures from embryo and postnatal thyroid glands were also studied. Results: In three-dimensional cultures of thyrocytes, XB130, aligned with actin filaments, participated in defining the site of apical membrane formation and coalescence to form a thyroid follicle lumen. Xb130-/- thyrocytes displayed delayed folliculogenesis, reduced recruitment of a microtubule (MT)-associated proteins, and disorganized acetylated tubulin under the apical membrane, resulting in delayed folliculogenesis with reduced efficiency in formation of the thyroid follicle lumen. Conclusions: XB130 critically regulates thyrocyte polarization by functioning as a link between the actin filament cortex and MT network at the apical membrane of thyrocytes. Defects of adaptor scaffold proteins may affect cellular polarity and cytoskeletal structure and function and result in disorders of epithelial function, such as congenital hypothyroidism.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Microfilament Proteins/metabolism , Ovarian Follicle/metabolism , Thyroid Gland/metabolism , Animals , Disease Models, Animal , Female , Hypothyroidism/physiopathology , Mice
9.
Healthcare (Basel) ; 9(11)2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34828558

ABSTRACT

The objective was to investigate different comorbidities developed in normal-tension glaucoma (NTG) and primary open-angle glaucoma (POAG) patients. This was a case-control study, with 1489 people in the NTG group and 5120 people in the POAG group. Patient data were obtained from the Longitudinal Health Insurance Database 2010 (LHID2010) of Taiwan for the 2008-2013 period. The chi-square test was used to compare categorical variables, such as gender, income and urbanisation level, between NTG and POAG patients, and the two-tailed t test was used to compare continuity between the two groups. We use a multivariate logic regression model to assess the risk of each participant. The results are expressed in terms of odds ratio (OR) and 95% confidence intervals (CI). Patients with NTG had significantly higher proportions of hypotension (adjusted OR, 1.984; 95% CI, 1.128-3.490), sleep disturbances (adjusted OR, 1.323; 95% CI, 1.146-1.528), peptic ulcers (adjusted OR, 1.383; 95% CI, 1.188-1.609) and allergic rhinitis (adjusted OR, 1.484; 95% CI, 1.290-1.707) than those with POAG. Conversely, arterial hypertension (adjusted OR, 0.767; 95% CI, 0.660-0.893), diabetes (adjusted OR, 0.850; 95% CI, 0.728-0.993) and atopic dermatitis (adjusted OR, 0.869; 95% CI, 0.763-0.990) had a lower risk in the NTG group than in the POAG group. We found that comorbidities such a hypotension, sleep disturbances and peptic ulcer and allergic rhinitis are more highly associated to NTG than POAG.

10.
Thyroid ; 31(11): 1650-1661, 2021 11.
Article in English | MEDLINE | ID: mdl-34470464

ABSTRACT

Background: Congenital hypothyroidism is often caused by genetic mutations that impair thyroid hormone (TH) production, resulting in growth and development defects. XB130 (actin filament associated protein 1 like 2) is an adaptor/scaffold protein that plays important roles in cell proliferation, migration, intracellular signal transduction, and tumorigenesis. It is highly expressed in thyrocytes, however, its function in the thyroid remains largely unexplored. Methods:Xb130-/- mice and their littermates were studied. Postnatal growth and growth hormone levels were measured, and responses to low or high-iodine diet, and levothyroxine treatment were examined. TH and thyrotropin in the serum and TH in the thyroid glands were quantified. Structure and function of thyrocytes in embryos and postnatal life were studied with histology, immunohistochemistry, immunofluorescence staining, Western blotting, and quantitative reverse transcription polymerase chain reaction. Results:Xb130-/- mice exhibited transient growth retardation postnatally, due to congenital hypothyroidism with reduced TH synthesis and secretion, which could be rescued by exogenous thyroxine supplementation. The thyroid glands of Xb130-/- mice displayed diminished thyroglobulin iodination and release at both embryonic and early postnatal stages. XB130 was found mainly on the apical membrane of thyroid follicles. Thyroid glands of embryonic and postnatal Xb130-/- mice exhibited disorganized apical membrane structure, delayed folliculogenesis, and abnormal formation of thyroid follicle lumina. Conclusion: XB130 critically regulates folliculogenesis by maintaining apical membrane structure and function of thyrocytes, and its deficiency leads to congenital hypothyroidism.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Congenital Hypothyroidism/genetics , Microfilament Proteins/deficiency , Thyroid Epithelial Cells/metabolism , Animals , Iodine/administration & dosage , Mice , Thyroid Hormones/blood , Thyroxine/administration & dosage , Thyroxine/pharmacology
11.
Cell Cycle ; 20(4): 417-433, 2021 02.
Article in English | MEDLINE | ID: mdl-33530820

ABSTRACT

Microglia proliferation is critical for proper development and function of the central nervous system (CNS), while dysregulation of proliferation contributes to pathology. We recently reported that male inducible nitric oxide synthase knockout (iNOS-/-) mice displayed significantly more proliferating microglia in their postnatal cortex than age-matched wildtype (WT) male mice. Moreover, nitric oxide (NO) signaling in mouse microglia greatly upregulates calcium entry through transient receptor potential vanilloid type 2 (TRPV2) channels. Considering that TRPV2 activity restricts astrocytic proliferation within glioma tissues, we investigated the roles of iNOS/NO signaling and TRPV2 expression in the regulation of microglial proliferation in vitro using assays of calcium imaging, immunocytochemistry, western blot, and polymerase chain reaction. Results showed that non-dividing microglia exhibited substantially higher expression of TRPV2 on the plasma membrane and significantly larger calcium influx through TRPV2 channels in comparison to dividing microglia. Additionally, non-dividing WT microglia exhibited significantly more NO production than dividing WT microglia. Furthermore, the NO-donor NOC18 increased the nuclear translocation of nuclear factor of activated T-cells cytoplasmic 2 (NFATC2) and the mRNA of the cyclin-dependent kinase inhibitor p21 and decreased the percentage of dividing WT and iNOS-/- microglia in culture. Importantly, the presence of the TRPV2 inhibitor tranilast abolished these effects of NOC18. Together, results from this study indicated that iNOS/NO signaling inhibits microglial proliferation through TRPV2-mediated calcium influx, nuclear translocation of the transcription factor NFATC2, and p21 expression. [Figure: see text].


Subject(s)
Calcium Channels/biosynthesis , Calcium Signaling/physiology , Microglia/metabolism , NFATC Transcription Factors/biosynthesis , Nitric Oxide/biosynthesis , TRPV Cation Channels/biosynthesis , p21-Activated Kinases/biosynthesis , Animals , Calcium Channels/genetics , Calcium Signaling/drug effects , Cell Line , Cell Proliferation/physiology , Cells, Cultured , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , NFATC Transcription Factors/genetics , Nitric Oxide/genetics , TRPV Cation Channels/genetics , Transcription, Genetic/physiology , p21-Activated Kinases/genetics
12.
Nitric Oxide ; 108: 28-39, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33418057

ABSTRACT

Calcium is a critical secondary messenger in microglia. In response to inflammation, microglia mobilize intracellular calcium and increase the expression of inducible nitric oxide synthase (iNOS), which produces nitric oxide (NO). This study set to explore whether NO regulates intracellular calcium dynamics through transient receptor potential (TRP) channels in primary wildtype (WT) and iNOS knockout (iNOS-/-) microglia, and the BV2 microglial cell line using calcium imaging and voltage-clamp recordings. Our results demonstrated that application of the NO-donor SNAP induced a biphasic calcium response in naïve murine microglia. Specifically, phase I was characterized by a rapid decline in calcium influx that was attenuated by pretreatment of the store operated calcium channel (SOCC) inhibitor 2APB, while phase II presented as a slow calcium influx that was abolished by pretreatment with the TRP vanilloid type 2 (TRPV2) channel inhibitor tranilast. Importantly, in the presence of a protein kinase G (PKG) inhibitor, the SNAP-mediated calcium decline in phase I persisted while the calcium influx in phase II was abolished. Application of thapsigargin to activate SOCCs caused a calcium influx through a nonselective cation conductance in BV2 microglia, which was abruptly attenuated by SNAP. Importantly, iNOS-/- microglia displayed a significantly larger calcium influx though SOCCs while expressing less stromal interaction molecule 1, Orai1, and TRP canonical type 1 and 3 mRNA, when compared to WT microglia. Together, these results demonstrate that NO signaling restricts calcium influx through SOCCs independent of PKG signaling and increases calcium influx through TRPV2 channels in a PKG-dependent mechanism in microglia.


Subject(s)
Calcium/metabolism , Microglia/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , S-Nitroso-N-Acetylpenicillamine/pharmacology , Animals , Calcium Channels/metabolism , Cell Line , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , TRPV Cation Channels/metabolism
13.
Brain Behav Immun ; 88: 791-801, 2020 08.
Article in English | MEDLINE | ID: mdl-32434046

ABSTRACT

Microglia express muscarinic G protein-coupled receptors (GPCRs) that sense cholinergic activity and are activated by acetylcholine to potentially regulate microglial functions. Knowledge about how distinct types of muscarinic GPCR signaling regulate microglia function in vivo is still poor, partly due to the fact that some of these receptors are also present in astrocytes and neurons. We generated mice expressing the hM3Dq Designer Receptor Exclusively Activated by Designer Drugs (DREADD) selectively in microglia to investigate the role of muscarinic M3Gq-linked signaling. We show that activation of hM3Dq using clozapine N-oxide (CNO) elevated intracellular calcium levels and increased phagocytosis of FluoSpheres by microglia in vitro. Interestingly, whereas acute treatment with CNO increased synthesis of cytokine mRNA, chronic treatment attenuated LPS-induced cytokine mRNA changes in the brain. No effect of CNO on cytokine expression was observed in DREADD-negative mice. Interestingly, CNO activation of M3Dq in microglia was able to attenuate LPS-mediated decrease in social interactions. These results suggest that chronic activation of M3 muscarinic receptors (the hM3Dq progenitor) in microglia, and potentially other Gq-coupled GPCRs, can trigger an inflammatory-like response that preconditions microglia to decrease their response to further immunological challenges. Our results indicate that hM3Dq can be a useful tool to modulate neuroinflammation and study microglial immunological memory in vivo, which may be applicable for manipulations of neuroinflammation in neurodegenerative and psychiatric diseases.


Subject(s)
Clozapine , Microglia , Acetylcholine , Animals , Clozapine/pharmacology , Male , Mice , Neurons , Receptors, G-Protein-Coupled , Signal Transduction
14.
J Ophthalmol ; 2020: 4919154, 2020.
Article in English | MEDLINE | ID: mdl-32454988

ABSTRACT

PURPOSE: To compare the patterns of relative peripheral refractions of myopic children who were currently on atropine treatment for myopia control and myopic children who did not use atropine. METHODS: Chinese children (n = 209) aged 7 to 12 years participated in the study, 106 used atropine and 103 did not. Participants were also classified into three groups: emmetropes (SE: +0.50 to -0.50 D), low myopes (SE: -0.50 to -3.00 D), and moderate myopes (SE: -3.00 to -6.00 D). The central and peripheral refractions along the horizontal meridians (for both nasal and temporal fields) were measured in 10-degree steps to 30 degrees. RESULTS: There were no statistically significant differences in spherical equivalent and astigmatism of the three refractive groups in either the nasal or temporal retina. The atropine group showed a significant relative myopia in the temporal 30° field in spherical equivalent compared to the emmetropic group (t 49 = 3.36, P=0.02). In eyes with low myopia, the atropine group had significant relative myopia in the nasal 30° and temporal 30° fields (t 118 = 2.59, P=0.01; t 118 = 2.06, P=0.04), and it is also observed at 20° and 30° of the nasal field for the moderate myopic group (t 36 = 2.37, P=0.02; t 2.84 = 2.84, P=0.01). CONCLUSION: Significant differences in relative peripheral refraction were found between the atropine group and its controls. The findings suggested that the eyes that received atropine may have a less prolate shape and thus explain why using atropine is effective in controlling myopia progression.

15.
Cerebellum ; 19(4): 510-526, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32270464

ABSTRACT

Nitric oxide (NO), specifically derived from neuronal nitric oxide synthase (nNOS), is a well-established regulator of synaptic transmission in Purkinje neurons (PNs), governing fundamental processes such as motor learning and coordination. Previous phenotypic analyses showed similar cerebellar structures between neuronal nitric oxide null (nNOS-/-) and wild-type (WT) adult male mice, despite prominent ataxic behavior within nNOS-/- mice. However, a study has yet to characterize PN molecular structure and their excitatory inputs during development in nNOS-/- mice. This study is the first to explore morphological abnormalities within the cerebellum of nNOS-/- mice, using immunohistochemistry and immunoblotting. This study sought to examine PN dendritic morphology and the expression of metabotropic glutamate receptor type 1 (mGluR1), vesicular glutamate transporter type 1 and 2 (vGluT1 and vGluT2), stromal interaction molecule 1 (STIM1), and calpain-1 within PNs of WT and nNOS-/- mice at postnatal day 7 (PD7), 2 weeks (2W), and 7 weeks (7W) of age. Results showed a decrease in PN dendritic branching at PD7 in nNOS-/- cerebella, while aberrant dendritic spine formation was noted in adult ages. Total protein expression of mGluR1 was decreased in nNOS-/- cerebella across development, while vGluT2, STIM1, and calpain-1 were significantly increased. Ex vivo treatment of WT slices with NOS inhibitor L-NAME increased calpain-1 expression, whereas treating nNOS-/- cerebellar slices with NO donor NOC-18 decreased calpain-1. Moreover, mGluR1 agonist DHPG increased calpain-1 in WT, but not in nNOS-/- slices. Together, these results indicate a novel role for nNOS/NO signaling in PN development, particularly by regulating an mGluR1-initiated calcium signaling mechanism.


Subject(s)
Dendrites/metabolism , Neurogenesis/physiology , Nitric Oxide/metabolism , Purkinje Cells/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Cerebellum/cytology , Cerebellum/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type I/metabolism , Purkinje Cells/cytology , Signal Transduction/physiology
17.
Nitric Oxide ; 94: 125-134, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31759970

ABSTRACT

Microglia population is primarily determined by a finely-regulated proliferation process during early development of the central nervous system (CNS). Nitric oxide (NO) is known to inhibit proliferation in numerous cell types. However, how NO signaling regulates microglia proliferation remains elusive. Using wildtype (WT) and inducible nitric oxide synthase knockout (iNOS-/-) mice, this study investigated the role and underlying mechanisms of iNOS/NO signaling in microglia proliferation. Here we reported that iNOS-/- mice displayed significantly more BrdU-labeled proliferating microglia in the cortex than that in WT mice at postnatal day 10. Compared to microglia isolated from WT mouse cortex, significantly more iNOS-/- microglia displayed the specific cell-cycle markers Ki67 and phospho-histone H3 (pH3) in their nuclei. In addition, treating WT microglia with the NOS inhibitor LNAME drastically increased the percentage of cells expressing Ki67 and pH3, whereas treating iNOS-/- microglia with NOC18, a slow-release NO-donor, significantly decreased the percentage of microglia expressing the two cell-cycle markers. Moreover, inhibition of protein kinase-G (PKG) in WT microglia increased the proportion of microglia expressing Ki67 and pH3, whereas activation of PKG signaling using 8Br-cGMP in iNOS-/- microglia significantly decreased the fraction of microglia displaying Ki67 and pH3. Interestingly, in the presence of a PKG inhibitor, NOC18 increased the quantity of iNOS-/- microglia expressing Ki67 and pH3. Together, these results indicate that basal activity of iNOS/NO signaling impedes microglial cell-cycle progression and attenuates proliferation through activation of the cGMP-PKG pathway. However, NO increases microglia cell-cycle progression in the absence of cGMP-PKG signaling.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Microglia/metabolism , Nitric Oxide/metabolism , Animals , Cell Proliferation , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/metabolism , Signal Transduction
18.
Article in English | MEDLINE | ID: mdl-31523359

ABSTRACT

The growth of neurites underlies the axonal pathfinding and synaptic formation during neuronal development and regeneration. Neurite growth is regulated by specific interactions between growth cone receptors and their ligands that function as molecular cues existing in microenvironments. Neurexins (NRXNs) are concentrated on growth cones and they may function to constrain axonal branches of invertebrate neurons. The present study explored the role of NRXN-1α in regulating neurite growth of mammalian neurons. Results showed that transfecting an effective NRXN-1α siRNA to cultured rat hippocampal neurons significantly increased neurite length. Adding NRXN-1α ligands including neuroligin (NLGN) peptide and/or α-latrotoxin (α-LTX) to the culture media largely decreased neurite growth of naïve neurons in a Ca2+-dependent manner, but had no effect on neurite growth of neurons transfected with NRXN-1α siRNA. Our results suggest that NRXN-1α regulates neurite development of mammalian neurons.

19.
Glia ; 67(12): 2294-2311, 2019 12.
Article in English | MEDLINE | ID: mdl-31453646

ABSTRACT

Microglia phagocytosis is critical for central nervous system development, and dysregulation of phagocytosis may contribute to a variety of neurological disorders. During initial stages of phagocytosis, microglia display increased nitric oxide (NO) production via inducible nitric oxide synthase (iNOS) activity and amplified calcium entry through transient receptor potential vanilloid type 2 (TRPV2) channels. The present study investigated the regulatory role of iNOS/NO signaling in microglial phagocytosis and TRPV2 channel activation using phagocytosis assay, calcium imaging, patch clamp electrophysiology, immunocytochemistry, and immunoblot assays. Results showed that primary microglia from iNOS-knockout (iNOS-/- ) mice exhibited substantial deficits in phagocytic capacity and TRPV2 channel activity relative to wild-type (WT) controls. Specifically, iNOS-/- microglia displayed a lower level of TRPV2 protein localized on the plasma membrane (PM) without any significant change in the mRNA levels of Fc-gamma receptors and TRPV2. In addition, iNOS-/- microglia, unlike their WT controls, failed to elicit a calcium influx in response to application of the TRPV2-agonist 2-aminoethoxydiphenyl borate (2APB). Importantly, the phagocytic capacity and the PM expression and activity of TRPV2 in iNOS-/- microglia were largely corrected by pretreatment with NO-donors. Accordingly, the 2APB-evoked calcium influx and the PM expression of TRPV2 in WT microglia were significantly decreased by selective inhibition of iNOS, protein kinase-G (PKG), or phosphoinositide-3-kinase (PI3K), respectively. Together, results from this study indicated that iNOS/NO signaling upregulates microglial phagocytosis and increases TRPV2 trafficking to the PM via PKG/PI3K dependent pathway(s).


Subject(s)
Calcium Channels/biosynthesis , Cell Membrane/metabolism , Microglia/metabolism , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide/metabolism , Phagocytosis/physiology , TRPV Cation Channels/biosynthesis , Animals , Calcium Channels/genetics , Cell Membrane/genetics , Cells, Cultured , Male , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , TRPV Cation Channels/genetics , Up-Regulation/physiology
20.
Cell Immunol ; 332: 7-23, 2018 10.
Article in English | MEDLINE | ID: mdl-30017085

ABSTRACT

In response to micro-environmental cues such as microbial infections or T-helper 1 and 2 (TH1 and TH2) cytokines, macrophages (Mϕs) develop into M1- or M2-like phenotypes. Phenotypic polarization/activation of Mϕs are also essentially regulated by autocrine signals. Type-A γ-aminobutyric acid receptor (GABAAR)-mediated autocrine signaling is critical for phenotypic differentiation and transformation of various cell types. The present study explored whether GABAAR signaling regulates lung Mϕ (LMϕ) phenotypic activation under M1/TH1 and M2/TH2 environments. Results showed that GABAAR subunits were expressed by primary LMϕ of mice and the mouse Mϕ cell line RAW264.7. The expression levels of GABAAR subunits in mouse LMϕs and RAW264.7 cells decreased or increased concurrently with classical (M1) or alternative (M2) activation, respectively. Moreover, activation or blockade of GABAARs distinctively influenced the phenotypic characteristics of Mϕ. These results suggested that microenvironments leading to LMϕ phenotypic polarization concurrently modulates autocrine GABA signaling and its role in Mϕ activation.


Subject(s)
Autocrine Communication/physiology , Macrophage Activation/physiology , Macrophages, Alveolar/metabolism , Signal Transduction/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Cell Line , Cytokines/metabolism , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , RAW 264.7 Cells , Receptors, GABA/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...