Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Life Sci ; 332: 122072, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37704067

ABSTRACT

AIMS: Bladder function is regulated by clock genes and dysregulation of circadian bladder function can cause nocturia. The blood concentration of palmitoylethanolamide (PEA), a fatty acid metabolite, changes with circadian rhythm. Clock gene abnormalities demonstrate the highest PEA levels during the sleep phase. PEA is a GPR55 agonist that influences urination; therefore, increased PEA during the sleep phase may cause nocturia. Herein, we investigated the function of GPR55 to evaluate the relationship between GPR55 and nocturia that evoked higher PEA during the sleep phase in patients with circadian rhythm disorders. MAIN METHODS: Male C57BL/6 mice were used. GPR55 localization was evaluated by immunofluorescence staining, qRT-PCR, and western blotting. Variations in PEA-induced intracellular Ca2+ concentrations were measured in primary cultured mouse urothelial cells (UCs) using Ca2+ imaging. PEA-induced NGF and PGI2 release in UCs was measured by ELISA. The micturition reflex pathway after PEA administration was evaluated using immunofluorescence staining. KEY FINDINGS: GPR55 was predominant in the UC layer. PEA induced release of Ca2+ from the endoplasmic reticulum into the UC cytoplasm. ELISA and immunofluorescence staining revealed that NGF and PGI2 were released from bladder UCs, stimulated the pontine micturition center in mice, and induced nocturia. SIGNIFICANCE: The loss of regular circadian metabolizing rhythm in fatty acids causes higher blood PEA levels during the sleep phase. Binding of PEA to GPR55 in UC may activate the downstream processes of the micturition reflex, leading to nocturia. These findings suggest a new mechanism for nocturia and its potential as a therapeutic target.

2.
Sci Adv ; 8(44): eabq1081, 2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36332025

ABSTRACT

Astrocyte abnormalities have received great attention for their association with various diseases in the brain but not so much in the eye. Recent independent genome-wide association studies of glaucoma, optic neuropathy characterized by retinal ganglion cell (RGC) degeneration, and vision loss found that single-nucleotide polymorphisms near the ABCA1 locus were common risk factors. Here, we show that Abca1 loss in retinal astrocytes causes glaucoma-like optic neuropathy in aged mice. ABCA1 was highly expressed in retinal astrocytes in mice. Thus, we generated macroglia-specific Abca1-deficient mice (Glia-KO) and found that aged Glia-KO mice had RGC degeneration and ocular dysfunction without affected intraocular pressure, a conventional risk factor for glaucoma. Single-cell RNA sequencing revealed that Abca1 deficiency in aged Glia-KO mice caused astrocyte-triggered inflammation and increased the susceptibility of certain RGC clusters to excitotoxicity. Together, astrocytes play a pivotal role in eye diseases, and loss of ABCA1 in astrocytes causes glaucoma-like neuropathy.

3.
J Exp Med ; 219(4)2022 04 04.
Article in English | MEDLINE | ID: mdl-35319723

ABSTRACT

Activation of astrocytes has a profound effect on brain plasticity and is critical for the pathophysiology of several neurological disorders including neuropathic pain. Here, we show that metabotropic glutamate receptor 5 (mGluR5), which reemerges in astrocytes in a restricted time frame, is essential for these functions. Although mGluR5 is absent in healthy adult astrocytes, it transiently reemerges in astrocytes of the somatosensory cortex (S1). During a limited spatiotemporal time frame, astrocytic mGluR5 drives Ca2+ signals; upregulates multiple synaptogenic molecules such as Thrombospondin-1, Glypican-4, and Hevin; causes excess excitatory synaptogenesis; and produces persistent alteration of S1 neuronal activity, leading to mechanical allodynia. All of these events were abolished by the astrocyte-specific deletion of mGluR5. Astrocytes dynamically control synaptic plasticity by turning on and off a single molecule, mGluR5, which defines subsequent persistent brain functions, especially under pathological conditions.


Subject(s)
Astrocytes , Chronic Pain , Animals , Astrocytes/metabolism , Chronic Pain/pathology , Mice , Neuronal Plasticity , Neurons/metabolism , Receptor, Metabotropic Glutamate 5/metabolism
4.
Transl Vis Sci Technol ; 11(1): 25, 2022 01 03.
Article in English | MEDLINE | ID: mdl-35040915

ABSTRACT

Purpose: Because the importance of glia in regulating brain functions has been demonstrated, genetic technologies that manipulate glial cell-specific gene expression in the brain have become essential and have made great progress. However, it is unknown whether the same strategy that is used in the brain can be applied to the retina because retinal glia differs from glia in the brain. Here, we aimed to find a method for selective gene expression in Müller cells (characteristic glial cells in the retina) and identified Mlc1 as a specific promoter of Müller cells. Methods: Mlc1-tTA::Yellow-Cameleon-NanotetO/tetO (YC-Nano) mice were used as a reporter line. YC-Nano, a fluorescent protein, was ectopically expressed in the cell type controlled by the Mlc1 promotor. Immunofluorescence staining was used to identify the cell type expressing YC-Nano protein. Results: YC-Nano-positive (+) signals were observed as vertical stalks in the sliced retina and spanned from the nerve fiber layer through the outer nuclear layer. The density of YC-Nano+ cells was higher around the optic nerve head and lower in the peripheral retina. The YC-Nano+ signals colocalized with vimentin, a marker of Müller cells, but not with the cell markers for blood vessels, microglia, neurons, or astrocytes. Conclusions: The Mlc1 promoter allows us to manipulate gene expression in Müller cells without affecting astrocytes in the retina. Translational Relevance: Gene manipulation under control of Mlc1 promoter offers novel technique to investigate the role of Müller cells.


Subject(s)
Ependymoglial Cells , Retina , Animals , Astrocytes , Gene Expression , Membrane Proteins , Mice , Neuroglia
5.
Glia ; 69(11): 2546-2558, 2021 11.
Article in English | MEDLINE | ID: mdl-34339538

ABSTRACT

Metabotropic glutamate receptor 5 (mGluR5) in astrocytes is a key molecule for controlling synapse remodeling. Although mGluR5 is abundant in neonatal astrocytes, its level is gradually down-regulated during development and is almost absent in the adult. However, in several pathological conditions, mGluR5 re-emerges in adult astrocytes and contributes to disease pathogenesis by forming uncontrolled synapses. Thus, controlling mGluR5 expression in astrocyte is critical for several diseases, but the mechanism that regulates mGluR5 expression remains unknown. Here, we show that adenosine triphosphate (ATP)/adenosine-mediated signals down-regulate mGluR5 in astrocytes. First, in situ Ca2+ imaging of astrocytes in acute cerebral slices from post-natal day (P)7-P28 mice showed that Ca2+ responses evoked by (S)-3,5-dihydroxyphenylglycine (DHPG), a mGluR5 agonist, decreased during development, whereas those evoked by ATP or its metabolite, adenosine, increased. Second, ATP and adenosine suppressed expression of the mGluR5 gene, Grm5, in cultured astrocytes. Third, the decrease in the DHPG-evoked Ca2+ responses was associated with down-regulation of Grm5. Interestingly, among several adenosine (P1) receptor and ATP (P2) receptor genes, only the adenosine A2B receptor gene, Adora2b, was up-regulated in the course of development. Indeed, we observed that down-regulation of Grm5 was suppressed in Adora2b knockout astrocytes at P14 and in situ Ca2+ imaging from Adora2b knockout mice indicated that the A2B receptor inhibits mGluR5 expression in astrocytes. Furthermore, deletion of A2B receptor increased the number of excitatory synapse in developmental stage. Taken together, the A2B receptor is critical for down-regulation of mGluR5 in astrocytes, which would contribute to terminate excess synaptogenesis during development.


Subject(s)
Astrocytes , Receptor, Adenosine A2B , Receptor, Metabotropic Glutamate 5 , Adenosine/metabolism , Adenosine/pharmacology , Animals , Astrocytes/metabolism , Carrier Proteins/metabolism , Mice , Receptor, Adenosine A2B/metabolism , Receptor, Metabotropic Glutamate 5/metabolism
6.
J Neurochem ; 151(1): 64-78, 2019 10.
Article in English | MEDLINE | ID: mdl-31278875

ABSTRACT

Methylmercury (MeHg), an environmental pollutant, causes serious damage to many organs. Effects on the CNS were initially thought to arise from MeHg acting directly on neurons, but it also has significant effects on non-neuronal cells such as microglia. Microglia, which are very sensitive to changes in the brain environment, show various phenotypes. We previously reported that upon short exposure to MeHg (MeHgshort ) at low concentration, microglia exhibited a neuroprotective phenotype; whereas, long-term exposure (MeHglong ) induced a neurotoxic phenotype of microglia. However, contributions of microglia to MeHg-induced CNS damage remain unknown. Even at very low concentrations, MeHglong but not MeHgshort caused significant neuronal damage associated with an increased number of reactive microglia in cortical slices from wild-type (WT) mice. Two-photon imaging of cortical slices from Iba1-GFP mice revealed that microglia in control conditions exhibited elongated and complex processes with high motility. MeHglong caused a significant reduction in process motility, retraction of processes, and hypertrophic cell bodies, indicating activated microglia. Moreover, MeHglong -treated microglia upregulated pro-inflammatory molecues, suggesting a change into a neurotoxic phenotype of microglia. As a molecular target, Rho-kinase (ROCK) was found to be key for controlling microglial reactivity and neurotoxicity. Expression level of ROCK was increased by MeHglong in WT slices, which was abolished by minocycline or Y-27632. We confirmed that MeHg directly activates microglial ROCK pathways prepared from WT mice. In addition, MeHg-evoked damage of primary neurons was significantly enhanced by the presence of microglia from WT mice, but offset by minocycline or Y-27632. Taken together, our data demonstrate that MeHg causes neurodegeneration by inducing a neurotoxic microglia phenotype via a ROCK-mediated mechanism.


Subject(s)
Methylmercury Compounds/toxicity , Microglia/metabolism , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Neurons/drug effects , rho-Associated Kinases/metabolism , Animals , Mice , Mice, Inbred C57BL , Microglia/drug effects , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...