Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Brain ; 147(2): 698-716, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37955589

RESUMEN

Alexander disease (AxD) is an intractable neurodegenerative disorder caused by GFAP mutations. It is a primary astrocyte disease with a pathological hallmark of Rosenthal fibres within astrocytes. AxD astrocytes show several abnormal phenotypes. Our previous study showed that AxD astrocytes in model mice exhibit aberrant Ca2+ signals that induce AxD aetiology. Here, we show that microglia have unique phenotypes with morphological and functional alterations, which are related to the pathogenesis of AxD. Immunohistochemical studies of 60TM mice (AxD model) showed that AxD microglia exhibited highly ramified morphology. Functional changes in microglia were assessed by Ca2+ imaging using hippocampal brain slices from Iba1-GCaMP6-60TM mice and two-photon microscopy. We found that AxD microglia showed aberrant Ca2+ signals, with high frequency Ca2+ signals in both the processes and cell bodies. These microglial Ca2+ signals were inhibited by pharmacological blockade or genetic knockdown of P2Y12 receptors but not by tetrodotoxin, indicating that these signals are independent of neuronal activity but dependent on extracellular ATP from non-neuronal cells. Our single-cell RNA sequencing data showed that the expression level of Entpd2, an astrocyte-specific gene encoding the ATP-degrading enzyme NTPDase2, was lower in AxD astrocytes than in wild-type astrocytes. In situ ATP imaging using the adeno-associated virus vector GfaABC1D ATP1.0 showed that exogenously applied ATP was present longer in 60TM mice than in wild-type mice. Thus, the increased ATP level caused by the decrease in its metabolizing enzyme in astrocytes could be responsible for the enhancement of microglial Ca2+ signals. To determine whether these P2Y12 receptor-mediated Ca2+ signals in AxD microglia play a significant role in the pathological mechanism, a P2Y12 receptor antagonist, clopidogrel, was administered. Clopidogrel significantly exacerbated pathological markers in AxD model mice and attenuated the morphological features of microglia, suggesting that microglia play a protective role against AxD pathology via P2Y12 receptors. Taken together, we demonstrated that microglia sense AxD astrocyte dysfunction via P2Y12 receptors as an increase in extracellular ATP and alter their morphology and Ca2+ signalling, thereby protecting against AxD pathology. Although AxD is a primary astrocyte disease, our study may facilitate understanding of the role of microglia as a disease modifier, which may contribute to the clinical diversity of AxD.


Asunto(s)
Enfermedad de Alexander , Ratones , Animales , Enfermedad de Alexander/metabolismo , Enfermedad de Alexander/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Astrocitos/metabolismo , Microglía/metabolismo , Clopidogrel/metabolismo , Calcio/metabolismo , Progresión de la Enfermedad , Adenosina Trifosfato/metabolismo
2.
J Pharmacol Sci ; 154(4): 326-333, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38485351

RESUMEN

PURPOSE: To determine whether combination of topical ripasudil and brimonidine has more effective neuroprotection on retinal ganglion cells (RGCs) following injury to axons composing the optic nerve. METHODS: Topical ripasudil, brimonidine, or mixture of both drugs were administered to adult mice after optic nerve injury (ONI). The influence of drug conditions on RGC health were evaluated by the quantifications of surviving RGCs, phosphorylated p38 mitogen-activated protein kinase (phospho-p38), and expressions of trophic factors and proinflammatory mediators in the retina. RESULTS: Topical ripasudil and brimonidine suppressed ONI-induced RGC death respectively, and mixture of both drugs further stimulated RGC survival. Topical ripasudil and brimonidine suppressed ONI-induced phospho-p38 in the whole retina. In addition, topical ripasudil suppressed expression levels of TNFα, IL-1ß and monocyte chemotactic protein-1 (MCP-1), whereas topical brimonidine increased the expression level of basic fibroblast growth factor (bFGF). CONCLUSIONS: Combination of topical ripasudil and brimonidine may enhance RGC protection by modulating multiple signaling pathways in the retina.


Asunto(s)
Isoquinolinas , Traumatismos del Nervio Óptico , Sulfonamidas , Ratones , Animales , Tartrato de Brimonidina , Traumatismos del Nervio Óptico/tratamiento farmacológico , Traumatismos del Nervio Óptico/metabolismo , Neuroprotección , Combinación de Medicamentos
3.
Int J Mol Sci ; 24(3)2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36769067

RESUMEN

Astrocytes, a non-neuronal glial cell type in the nervous system, are essential for regulating physiological functions of the central nervous system. In various injuries and diseases of the central nervous system, astrocytes often change their phenotypes into neurotoxic ones that participate in pro-inflammatory responses (hereafter referred to as "immune functions"). Such astrocytic immune functions are not only limited to brain diseases but are also found in ocular neurodegenerative diseases such as glaucoma, a retinal neurodegenerative disease that is the leading cause of blindness worldwide. The eye has two astrocyte-lineage cells: astrocytes and Müller cells. They maintain the physiological environment of the retina and optic nerve, thereby controlling visual function. Dysfunction of astrocyte-lineage cells may be involved in the onset and progression of glaucoma. These cells become reactive in glaucoma patients, and animal studies have suggested that their immune responses may be linked to glaucoma-related events: tissue remodeling, neuronal death, and infiltration of peripheral immune cells. In this review, we discuss the role of the immune functions of astrocyte-lineage cells in the pathogenesis of glaucoma.


Asunto(s)
Glaucoma , Enfermedades Neurodegenerativas , Animales , Astrocitos/metabolismo , Células Ganglionares de la Retina/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Glaucoma/metabolismo , Inmunidad
4.
Int J Mol Sci ; 24(24)2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38139424

RESUMEN

Peripheral infection induces inflammation in peripheral tissues and the brain, impacting brain function. Glial cells are key players in this process. However, the effects of peripheral infection on glial activation and brain function remain unknown. Here, we showed that varying degrees of peripheral infection had different effects on the regulation of brain functions by microglia-dependent and -independent mechanisms. Acute mild infection (one-day LPS challenge: 1LPS) exacerbated middle cerebral artery occlusion (MCAO) injury, and severe infection (four-day LPS challenge: 4LPS) for one week suppressed it. MCAO injury was assessed by triphenyltetrazolium chloride staining. We observed early activation of microglia in the 1LPS and 4LPS groups. Depleting microglia with a colony-stimulating factor-1 receptor (CSF1R) antagonist had no effect on 1LPS-induced brain injury exacerbation but abolished 4LPS-induced protection, indicating microglial independence and dependence, respectively. Microglia-independent exacerbation caused by 1LPS involved peripheral immune cells including macrophages. RNA sequencing analysis of 4LPS-treated microglia revealed increased factors related to anti-inflammatory and neuronal tissue repair, suggesting their association with the protective effect. In conclusion, varying degrees of peripheral inflammation had contradictory effects (exacerbation vs. protection) on MCAO, which may be attributed to microglial dependence. Our findings highlight the significant impact of peripheral infection on brain function, particularly in relation to glial cells.


Asunto(s)
Lipopolisacáridos , Microglía , Ratones , Animales , Lipopolisacáridos/toxicidad , Macrófagos , Encéfalo , Infarto de la Arteria Cerebral Media , Inflamación
5.
Glia ; 69(11): 2546-2558, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34339538

RESUMEN

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.


Asunto(s)
Astrocitos , Receptor de Adenosina A2B , Receptor del Glutamato Metabotropico 5 , Adenosina/metabolismo , Adenosina/farmacología , Animales , Astrocitos/metabolismo , Proteínas Portadoras/metabolismo , Ratones , Receptor de Adenosina A2B/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo
6.
Glia ; 69(10): 2332-2348, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34309082

RESUMEN

Microglia are the resident immune cells of the brain, and play essential roles in neuronal development, homeostatic function, and neurodegenerative disease. Human microglia are relatively different from mouse microglia. However, most research on human microglia is performed in vitro, which does not accurately represent microglia characteristics under in vivo conditions. To elucidate the in vivo characteristics of human microglia, methods have been developed to generate and transplant induced pluripotent or embryonic stem cell-derived human microglia into neonatal or adult mouse brains. However, its widespread use remains limited by the technical difficulties of generating human microglia, as well as the need to use immune-deficient mice and conduct invasive surgeries. To address these issues, we developed a simplified method to generate induced pluripotent stem cell-derived human microglia and transplant them into the brain via a transnasal route in immunocompetent mice, in combination with a colony stimulating factor 1 receptor antagonist. We found that human microglia were able to migrate through the cribriform plate to different regions of the brain, proliferate, and become the dominant microglia in a region-specific manner by occupying the vacant niche when exogenous human cytokine is administered, for at least 60 days.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedades Neurodegenerativas , Trasplante de Células Madre , Animales , Encéfalo/fisiología , Diferenciación Celular/fisiología , Humanos , Ratones , Microglía , Nariz , Trasplante de Células Madre/métodos
7.
J Pharmacol Sci ; 145(3): 262-267, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33602506

RESUMEN

Glaucoma, a progressive optic neuropathy and the leading cause of blindness, is characterized by impairment or degeneration of retinal ganglion cells (RGCs), which transmit visual information to the brain. Currently, 70 million people worldwide are affected by glaucoma. Elevated intraocular pressure (IOP), a major risk factor of glaucoma, directly damages RGCs. However, a substantial proportion of glaucoma patients have a normal IOP level. In particular, over 90% of Japanese glaucoma patients are reported to have normal IOP levels. Thus, a new focus for glaucoma pathology has emerged. Glial cells contribute to tissue homeostasis. Under pathological conditions, glial cells become reactive, lose their homeostatic functions, and gain neurotoxic functions, which trigger neurodegeneration in several diseases including glaucoma. Reactive glial cells have been identified in the eyes of glaucoma patients. In a glaucoma animal model, reactive glial cells are observed at early stages of the disease when RGCs are intact, indicating the possible role of glial cells in the pathogenesis of glaucoma. In this review, we introduce potential roles of glial cells in the pathogenesis of glaucoma. We focus on the roles of the ocular macroglial cells such as astrocytes and Müller cells, and discuss their roles in the pathogenesis of glaucoma.


Asunto(s)
Astrocitos/patología , Astrocitos/fisiología , Células Ependimogliales/patología , Células Ependimogliales/fisiología , Glaucoma/etiología , Glaucoma/patología , Retina/citología , Retina/patología , Complemento C3 , Gliosis , Ácido Glutámico/metabolismo , Humanos , Presión Intraocular , Factores de Crecimiento Nervioso , Disco Óptico/patología , Factores de Riesgo , Factor de Transcripción STAT3
8.
J Neurochem ; 151(1): 64-78, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31278875

RESUMEN

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.


Asunto(s)
Compuestos de Metilmercurio/toxicidad , Microglía/metabolismo , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/metabolismo , Neuronas/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Neuronas/metabolismo
9.
Glia ; 66(11): 2366-2384, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30375063

RESUMEN

Excitotoxicity is well known in the neuronal death in the brain and is also linked to neuronal damages in the retina. Recent accumulating evidence show that microglia greatly affect excitotoxicity in the brain, but their roles in retina have received only limited attention. Here, we report that retinal excitotoxicity is mediated by microglia. To this end, we employed three discrete methods, that is, pharmacological inhibition of microglia by minocycline, pharmacological ablation by an antagonist for colony stimulating factor 1 receptor (PLX5622), and genetic ablation of microglia using Iba1-tTA::DTAtetO/tetO mice. Intravitreal injection of NMDA increased the number of apoptotic retinal ganglion cells (RGCs) followed by reduction in the number of RGCs. Although microglia did not respond to NMDA directly, they became reactive earlier than RGC damages. Inhibition or ablation of microglia protected RGCs against NMDA. We found up-regulation of proinflammatory cytokine genes including Il1b, Il6 and Tnfa, among which Tnfa was selectively blocked by minocycline. PLX5622 also suppressed Tnfa expression. Tumor necrosis factor α (TNFα) signals were restricted in microglia at very early followed by spreading into other cell types. TNFα up-regulation in microglia and other cells were significantly attenuated by minocycline and PLX5622, suggesting a central role of microglia for TNFα induction. Both inhibition of TNFα and knockdown of TNF receptor type 1 by siRNA protected RGCs against NMDA. Taken together, our data demonstrate that a phenotypic change of microglia into a neurotoxic one is a critical event for the NMDA-induced degeneration of RGCs, suggesting an importance of non-cell-autonomous mechanism in the retinal neuronal excitotoxicity.


Asunto(s)
Muerte Celular/fisiología , Citocinas/metabolismo , Microglía/fisiología , Células Ganglionares de la Retina/fisiología , Aminopiridinas/farmacología , Animales , Animales Recién Nacidos , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Citocinas/genética , Agonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/ultraestructura , N-Metilaspartato/farmacología , Degeneración Nerviosa/inducido químicamente , Traumatismos del Nervio Óptico/inducido químicamente , Compuestos Orgánicos/farmacología , Pirroles/farmacología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/ultraestructura , Transducción de Señal/efectos de los fármacos , Factor de Transcripción Brn-3A/genética , Factor de Transcripción Brn-3A/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
Neurourol Urodyn ; 37(8): 2535-2543, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30106187

RESUMEN

AIMS: The sensation of bladder fullness (SBF) is triggered by the release of ATP. Therefore, the aim of this study was to investigate whether time-dependent changes in the levels of stretch-released ATP in mouse primary-cultured urothelial cells (MPCUCs) is regulated by circadian rhythm via clock genes. METHODS: MPCUCs were derived from wild-type and Clock mutant mice (ClockΔ19/Δ19 ), presenting a nocturia phenotype. They were cultured in elastic silicone chambers. Stretch-released ATP was quantified every 4 h by ATP photon count. An experiment was also performed to determine whether ATP release correlated with the rhythm of the expression of Piezo1, TRPV4, VNUT, and Connexin26 (Cx26) in MPCUCs regulated by clock genes with circadian rhythms. MPCUCs were treated with carbenoxolone, an inhibitor of gap junction protein; were derived from VNUT-KO mice; or treated with Piezo1-siRNA, TRPV4-siRNA, and Cx26-siRNA. RESULTS: Stretch-released ATP showed time-dependent changes in wild-type mice and correlated with the rhythm of the expression of Piezo1, TRPV4, VNUT, and Cx26. However, these rhythms were disrupted in ClockΔ19/Δ19 mice. Carbenoxolone eliminated the rhythmicity of ATP release in wild-type mice. However, time-dependent ATP release changes were maintained when a single gene was deficient such as VNUT-KO, Piezo1-, TRPV4-, and Cx26-siRNA. CONCLUSIONS: ATP release in the bladder urothelium induces SBF and may have a circadian rhythm regulated by the clock genes. In the bladder urothelium, clock gene abnormalities may disrupt circadian ATP release by inducing Piezo1, TRPV4, VNUT, and Cx26. All these genes can trigger nocturia.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proteínas CLOCK/genética , Proteínas CLOCK/fisiología , Urotelio/metabolismo , Animales , Carbenoxolona/farmacología , Ritmo Circadiano/efectos de los fármacos , Ritmo Circadiano/genética , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Nocturia/genética , Proteínas de Transporte de Nucleótidos/genética , Cultivo Primario de Células , Urotelio/citología
11.
J Neurochem ; 136(4): 741-751, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26560804

RESUMEN

Müller cells, the primary macroglia of the retina, support various functions of retinal ganglion cells (RGCs). Here, we demonstrate a nucleotide-mediated communication between these two types of cells, by which Müller cells control neurite outgrowth of RGCs by activation of P2 receptors such as P2Y6 . Cultured mouse RGCs had significantly enhanced neurite outgrowth when cultured with either cultured mouse Müller cells or conditioned medium derived from Müller cells, and this was completely inhibited by the nucleotide-degrading enzyme, apyrase. This increase in outgrowth was mimicked by exogenously applied nucleotides such as ATP, uridine triphosphate, and uridine diphosphate. Pharmacological and genetic analysis revealed that P2Y6 receptor in RGCs was responsible for the increased neurite outgrowth. P2Y6 receptor was expressed in the ganglion cell layer of the retina and in RGC primary cultures. High performance liquid chromatography has revealed that Müller cells constitutively release uridine triphosphate, which is immediately metabolized into uridine diphosphate, an endogenous agonist for P2Y6 receptor. In the in vitro ocular hypertension model (i.e., glaucoma model), neurite outgrowth in RGCs was significantly reduced, which was associated with a decrease in P2Y6 receptors. Taken together, Müller cells control neurite outgrowth of RGCs by activating P2 receptors such as P2Y6 receptor, and the receptor expression level might be down-regulated in glaucoma. Müller cells support various functions of retina including those of retinal ganglion cells (RGCs). Here, we report an importance of nucleotide-mediated communication between these two types of cells. Müller cells were found to release uridine diphosphate (UTD), uridine triphosphate (UTP), and activate P2Y6 receptors in RGCs, which was essential for neurite outgrowth of RGCs. In addition, P2Y6 receptors in RGCs were reduced in a glaucoma model in vitro, suggesting an involvement of their dysfunction in the pathogenesis of glaucoma.

12.
Glia ; 61(8): 1320-30, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23832620

RESUMEN

Microglia survey the brain environment by sensing several types of diffusible molecules, among which extracellular nucleotides released/leaked from damaged cells have central roles. Microglia sense ATP or other nucleotides by multiple P2 receptors, after which they change into several different phenotypes. However, so far, it is largely unknown whether microglia themselves release ATP and, if so, by what mechanism. Here we show that exocytosis is the mechanism by which microglia release ATP. When we stimulated microglia with ionomycin, they released ATP and the release was dependent on Ca²âº, vesicular H⁺-ATPase, or SNAREs but independent of connexin/pannexin hemichannels. VNUT was found to be expressed in microglia and exhibited no colocalization with lysosome. We also visualized the exocytosis of ATP by a quinacrine-based fluorescent time-lapse imaging. Moreover, we found that lipopolysaccharide increased the ionomycin-induced release of ATP, which was dependent on the increase in VNUT. Taken together, our data suggested that exocytosis is the mechanism of ATP release from microglia. When activated, they would release ATP by increasing VNUT-dependent exocytotic mechanisms.


Asunto(s)
Adenosina Trifosfato/metabolismo , Exocitosis/fisiología , Microglía/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Microglía/citología , Ratas , Ratas Wistar
13.
Neuropharmacology ; 222: 109302, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36341810

RESUMEN

Adenosine triphosphate (ATP), an energy source currency in cells, is released or leaked to the extracellular space under both physiological and pathological conditions. Extracellular ATP functions as an intercellular signaling molecule through activation of purinergic P2 receptors. Ocular tissue and cells release ATP in response to physiological stimuli such as intraocular pressure (IOP), and P2 receptor activation regulates IOP elevation or reduction. Dysregulated purinergic signaling may cause abnormally elevated IOP, which is one of the major risk factors for glaucoma. Glaucoma, a leading cause of blindness worldwide, is characterized by progressive degeneration of optic nerves and retinal ganglion cells (RGCs), which are essential retinal neurons that transduce visual information to the brain. An elevation in IOP may stress RGCs and increase the risk for glaucoma pathogenesis. In the aqueous humor of human patients with glaucoma, the ATP level is significantly elevated. Such excess amount of ATP may directly cause RGC death via a specific subtype of P2 receptors. Dysregulated purinergic signaling may also trigger inflammation, oxidative stress, and excitotoxicity via activating non-neuronal cell types such as glial cells. In this review, we discussed the physiological roles of extracellular nucleotides in the ocular tissue and their potential role in the pathogenesis of glaucoma. This article is part of the Special Issue on 'Purinergic Signaling: 50 years'.


Asunto(s)
Glaucoma , Humanos , Transducción de Señal , Células Ganglionares de la Retina , Nervio Óptico , Adenosina Trifosfato
14.
Life Sci ; 332: 122072, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37704067

RESUMEN

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.

15.
Cell Death Discov ; 9(1): 166, 2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-37188749

RESUMEN

Dedicator of cytokinesis 3 (DOCK3) is an atypical member of the guanine nucleotide exchange factors (GEFs) and plays important roles in neurite outgrowth. DOCK3 forms a complex with Engulfment and cell motility protein 1 (Elmo1) and effectively activates Rac1 and actin dynamics. In this study, we screened 462,169 low-molecular-weight compounds and identified the hit compounds that stimulate the interaction between DOCK3 and Elmo1, and neurite outgrowth in vitro. Some of the derivatives from the hit compound stimulated neuroprotection and axon regeneration in a mouse model of optic nerve injury. Our findings suggest that the low-molecular-weight DOCK3 activators could be a potential therapeutic candidate for treating axonal injury and neurodegenerative diseases including glaucoma.

16.
PLoS Biol ; 7(5): e1000103, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19419241

RESUMEN

The ATP-gated P2X(4) receptor is a cation channel, which is important in various pathophysiological events. The architecture of the P2X(4) receptor in the activated state and how to change its structure in response to ATP binding are not fully understood. Here, we analyze the architecture and ATP-induced structural changes in P2X(4) receptors using fast-scanning atomic force microscopy (AFM). AFM images of the membrane-dissociated and membrane-inserted forms of P2X(4) receptors and a functional analysis revealed that P2X(4) receptors have an upward orientation on mica but lean to one side. Time-lapse imaging of the ATP-induced structural changes in P2X(4) receptors revealed two different forms of activated structures under 0 Ca(2+) conditions, namely a trimer structure and a pore dilation-like tripartite structure. A dye uptake measurement demonstrated that ATP-activated P2X(4) receptors display pore dilation in the absence of Ca(2+). With Ca(2+), the P2X(4) receptors exhibited only a disengaged trimer and no dye uptake was observed. Thus our data provide a new insight into ATP-induced structural changes in P2X(4) receptors that correlate with pore dynamics.


Asunto(s)
Adenosina Trifosfato/química , Calcio/química , Receptores Purinérgicos P2X4/química , Adenosina Trifosfato/metabolismo , Silicatos de Aluminio/química , Animales , Calcio/metabolismo , Línea Celular Tumoral , Colorantes Fluorescentes , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/genética , Proteínas Inmovilizadas/metabolismo , Transporte Iónico , Microscopía de Fuerza Atómica , Modelos Moleculares , Polilisina/química , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Receptores Purinérgicos P2X4/genética , Receptores Purinérgicos P2X4/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Imagen de Lapso de Tiempo
17.
J Vis Exp ; (183)2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35723460

RESUMEN

Microglia are the specialized population of macrophage-like cells of the brain. They play essential roles in both physiological and pathological brain functions. Most of our current understanding of microglia is based on experiments performed in the mouse. Human microglia differ from mouse microglia, and thus response and characteristics of mouse microglia may not always represent that of human microglia. Further, due to ethical and technical difficulties, research on human microglia is restricted to in vitro culture system, which does not capitulate in vivo characteristics of microglia. To overcome these issues, a simplified method to non-invasively transplant induced pluripotent stem cell-derived human microglia (iPSMG) into the immunocompetent mice brain via a transnasal route in combination with pharmacological depletion of endogenous microglia using a colony-stimulating factor 1 receptor (CSF1R) antagonist is developed. This protocol provides a way to non-invasively transplant cells into the mouse brain and may therefore be valuable for evaluating the in vivo role of human microglia in physiological and pathological brain functions.


Asunto(s)
Células Madre Pluripotentes Inducidas , Microglía , Animales , Encéfalo/cirugía , Humanos , Macrófagos , Ratones
18.
Transl Vis Sci Technol ; 11(1): 25, 2022 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-35040915

RESUMEN

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.


Asunto(s)
Células Ependimogliales , Retina , Animales , Astrocitos , Expresión Génica , Proteínas de la Membrana , Ratones , Neuroglía
19.
J Exp Med ; 219(4)2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35319723

RESUMEN

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.


Asunto(s)
Astrocitos , Dolor Crónico , Animales , Astrocitos/metabolismo , Dolor Crónico/patología , Ratones , Plasticidad Neuronal , Neuronas/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo
20.
Sci Adv ; 8(44): eabq1081, 2022 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-36332025

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA