Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Glia ; 71(9): 2266-2284, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37300531

RESUMO

Synucleinopathies refer to a range of neurodegenerative diseases caused by abnormal α-synuclein (α-Syn) deposition, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). Their pathogenesis is strongly linked to microglial dysfunction and neuroinflammation, which involves the leucine-rich-repeat kinase 2 (LRRK2)-regulated nuclear factor of activated T-cells (NFAT). Of the NFAT family, NFATc1 has been found to be increasingly translocated into the nucleus in α-syn stimulation. However, the specific role of NFATc1-mediated intracellular signaling in PD remains elusive in regulating microglial functions. In the current study, we crossbred LRRK2 or NFATc1 conditional knockout mice with Lyz2Cre mice to generate mice with microglia-specific deletion of LRRK2 or NFATc1, and by stereotactic injection of fibrillary α-Syn, we generated PD models in these mice. We found that LRRK2 deficiency enhanced microglial phagocytosis in the mice after α-Syn exposure and that genetic inhibition of NFATc1 markedly diminished phagocytosis and α-Syn elimination. We further demonstrated that LRRK2 negatively regulated NFATc1 in α-Syn-treated microglia, in which microglial LRRK2-deficiency facilitated NFATc1 nuclear translocation, CX3CR1 upregulation, and microglia migration. Additionally, NFATc1 translocation upregulated the expression of Rab7 and promoted the formation of late lysosomes, resulting in α-Syn degradation. In contrast, the microglial NFATc1 deficiency impaired CX3CR1 upregulation and the formation of Rab7-mediated late lysosomes. These findings highlight the critical role of NFATc1 in modulating microglial migration and phagocytosis, in which the LRRK2-NFATc1 signaling pathway regulates the expression of microglial CX3CR1 and endocytic degradative Rab7 to attenuate α-synuclein immunotoxicity.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Animais , Camundongos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Lisossomos/metabolismo , Camundongos Knockout , Microglia/metabolismo , Doença de Parkinson/genética , Fagocitose/genética
2.
Biochem Biophys Res Commun ; 638: 192-199, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36462493

RESUMO

Neuropathic pain (NP) is a chronic disease caused by damage to the peripheral or central nervous system. Connexin 43 (Cx43), the primary connexin expressed by astrocytes, has been reported to be significantly increased in NP. However, the roles and mechanisms of Cx43 in the development and maintenance of NP remain largely unknown, while microglia activation has been commonly regarded as a key factor of NP. In the present study, we found that Cx43 deletion significantly ameliorated spared nerve injury (SNI)-induced NP and suppressed SNI induced c-Fos expression in the spinal cord. Notably, Cx43 deletion led to much less SNI-induced microglia activation in the spinal cord. These results suggest that astrocyte Cx43 may play a significant role in regulating microglial activation and NP.


Assuntos
Astrócitos , Conexina 43 , Neuralgia , Astrócitos/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Hiperalgesia/metabolismo , Microglia/metabolismo , Neuralgia/genética , Neuralgia/patologia , Medula Espinal/metabolismo , Animais , Camundongos
3.
J Neuroinflammation ; 20(1): 261, 2023 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-37953259

RESUMO

BACKGROUND: Subarachnoid hemorrhage (SAH) causes significant long-term neurocognitive dysfunction, which is associated with hippocampal neuroinflammation. Growing evidences have shown that astrocytes played a significant role in mediating neuroinflammation. Recently, in vivo reprogramming of astrocytes to neurons by NeuroD1 or PTBP1 administration has generated a lot of interests and controversies. While the debates centered on the source of neurogenesis, no attention has been paid to the changes of the astrocytes-mediated neuroinflammation and its impact on endogenous neurogenesis after NeuroD1 administration. METHODS: 80 adult male C57BL/6 mice were used in this study. SAH was established by pre-chiasmatic injection of 100 µl blood. AAV-NeuroD1-GFP virus was injected to the hippocampus 3 day post-SAH. Neurocognitive function, brain water content, in vivo electrophysiology, Golgi staining, western blot and immunofluorescent staining were assessed at day 14 post-virus injection. RESULTS: NeuroD1 administration markedly attenuated reactive astrocytes-mediated neuroinflammation by reversing neurotoxic A1 astrocytes transformation, decreasing the secretion of neuroinflammatory cytokines, and reducing the activation of harmful microglia. NeuroD1 treatment significantly reversed the brain-blood barrier impairment and promoted the release of neurotrophic factors pleiotrophin (PTN), all of which contributed to the improvement of cellular microenvironment and made it more suitable for neurogenesis. Interestingly, besides neurogenesis in the hippocampus from cells transfected with NeuroD1 at the early phase of SAH, NeuroD1 administration significantly boosted the endogenous neurogenesis at the late phase of SAH, which likely benefited from the improvement of the neuroinflammatory microenvironment. Functionally, NeuroD1 treatment significantly alleviated neurocognitive dysfunction impaired by SAH. CONCLUSIONS: NeuroD1 significantly promoted neurofunctional recovery by attenuating reactive astrocytes-mediated neuroinflammation and boosting neurogenesis decimated by SAH. Specifically, NeuroD1 efficiently converted transfected cells, most likely astrocytes, to neurons at the early phase of SAH, suppressed astrocytes-mediated neuroinflammation and boosted endogenous neurogenesis at the late phase of SAH.


Assuntos
Doenças Neuroinflamatórias , Hemorragia Subaracnóidea , Camundongos , Animais , Masculino , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/tratamento farmacológico , Camundongos Endogâmicos C57BL , Encéfalo , Neurogênese/fisiologia
4.
J Neurochem ; 160(3): 376-391, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34757653

RESUMO

Trigeminal neuralgia (TN) is a type of severe paroxysmal neuropathic pain commonly triggered by mild mechanical stimulation in the orofacial area. Piezo2, a mechanically gated ion channel that mediates tactile allodynia in neuropathic pain, can be potentiated by a cyclic adenosine monophosphate (cAMP)-dependent signaling pathway that involves the exchange protein directly activated by cAMP 1 (Epac1). To study whether Piezo2-mediated mechanotransduction contributes to peripheral sensitization in a rat model of TN after trigeminal nerve compression injury, the expression of Piezo2 and activation of cAMP signal-related molecules in the trigeminal ganglion (TG) were detected. Changes in purinergic P2 receptors in the TG were also studied by RNA-seq. The expression of Piezo2, cAMP, and Epac1 in the TG of the TN animals increased after chronic compression of the trigeminal nerve root (CCT) for 21 days, but Piezo2 knockdown by shRNA in the TG attenuated orofacial mechanical allodynia. Purinergic P2 receptors P2X4, P2X7, P2Y1, and P2Y2 were significantly up-regulated after CCT injury. In vitro, Piezo2 expression in TG neurons was significantly increased by exogenous adenosine 5'-triphosphate (ATP) and Ca2+ ionophore ionomycin. ATP pre-treated TG neurons displayed elevated [Ca2+ ]i and faster increase in responding to blockage of Na+ /Ca2+ exchanger by KB-R7943. Furthermore, mechanical stimulation of cultured TG neurons led to sustained elevation in [Ca2+ ]i in ATP pre-treated TG neurons, which is much less in naïve TG neurons, or is significantly reduced by Piezo2 inhibitor GsMTx4. These results indicated a pivotal role of Piezo2 in peripheral mechanical allodynia in the rat CCT model. Extracellular ATP, Ca2+ influx, and the cAMP-to-Epac1 signaling pathway synergistically contribute to the pathogenesis and the persistence of mechanical allodynia.


Assuntos
Trifosfato de Adenosina/metabolismo , AMP Cíclico/metabolismo , Espaço Extracelular/metabolismo , Hiperalgesia/fisiopatologia , Canais Iônicos/genética , Transdução de Sinais , Traumatismos do Nervo Trigêmeo/fisiopatologia , Animais , Sinalização do Cálcio , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Canais Iônicos/antagonistas & inibidores , Masculino , Síndromes de Compressão Nervosa/metabolismo , Síndromes de Compressão Nervosa/fisiopatologia , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2/efeitos dos fármacos , Trocador de Sódio e Cálcio/antagonistas & inibidores , Traumatismos do Nervo Trigêmeo/metabolismo , Neuralgia do Trigêmeo
5.
Neurochem Res ; 42(9): 2551-2559, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28401401

RESUMO

Anion channels and connexin hemichannels are permeable to amino acid neurotransmitters. It is hypothesized that these conductive pathways release GABA, thereby influencing ambient GABA levels and tonic GABAergic inhibition. To investigate this, we measured the effects of anion channel/hemichannel antagonists on tonic GABA currents of rat hippocampal neurons. In contrast to predictions, blockade of anion channels and hemichannels with NPPB potentiated tonic GABA currents of neurons in culture and acute hippocampal slices. In contrast, the anion channel/hemichannel antagonist carbenoxolone (CBX) inhibited tonic currents. These findings could result from alterations of ambient GABA concentration or direct effects on GABAA receptors. To test for effects on GABAA receptors, we measured currents evoked by exogenous GABA. Coapplication of NPPB with GABA potentiated GABA-evoked currents. CBX dose-dependently inhibited GABA-evoked currents. These results are consistent with direct effects of NPPB and CBX on GABAA receptors. GABA release from hippocampal cell cultures was directly measured using HPLC. Inhibition of anion channels with NPPB or CBX did not affect GABA release from cultured hippocampal neurons. NPPB reduced GABA release from pure astrocytic cultures by 21%, but the total GABA release from astrocytes was small compared to that of mixed cultures. These data indicate that drugs commonly used to antagonize anion channels and connexin hemichannels may affect tonic currents via direct effects on GABAA receptors and have negligible effects on ambient GABA concentrations. Interpretation of experiments using NPPB or CBX should include consideration of their effects on tonic GABA currents.


Assuntos
Conexinas/antagonistas & inibidores , Conexinas/fisiologia , Antagonistas de Receptores de GABA-A/farmacologia , Receptores de GABA-A/fisiologia , Canais de Ânion Dependentes de Voltagem/antagonistas & inibidores , Canais de Ânion Dependentes de Voltagem/fisiologia , Aminobenzoatos/farmacologia , Animais , Animais Recém-Nascidos , Carbenoxolona/farmacologia , Células Cultivadas , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Masculino , Nitrobenzoatos/farmacologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley , Ácido gama-Aminobutírico/farmacologia
6.
J Neurosci ; 35(47): 15599-611, 2015 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-26609155

RESUMO

Ischemic preconditioning (IPC) is a robust neuroprotective phenomenon whereby brief ischemic exposure confers tolerance to a subsequent ischemic challenge. IPC has not been studied selectively in CNS white matter (WM), although stroke frequently involves WM. We determined whether IPC is present in WM and, if so, its mechanism. We delivered a brief in vivo preconditioning ischemic insult (unilateral common carotid artery ligation) to 12- to 14-week-old mice and determined WM ischemic vulnerability [oxygen-glucose deprivation (OGD)] 72 h later, using acutely isolated optic nerves (CNS WM tracts) from the preconditioned (ipsilateral) and control (contralateral) hemispheres. Functional and structural recovery was assessed by quantitative measurement of compound action potentials (CAPs) and immunofluorescent microscopy. Preconditioned mouse optic nerves (MONs) showed better functional recovery after OGD than the non-preconditioned MONs (31 ± 3 vs 17 ± 3% normalized CAP area, p < 0.01). Preconditioned MONs also showed improved axon integrity and reduced oligodendrocyte injury compared with non-preconditioned MONs. Toll-like receptor-4 (TLR4) and type 1 interferon receptor (IFNAR1), key receptors in innate immune response, are implicated in gray matter preconditioning. Strikingly, IPC-mediated WM protection was abolished in both TLR4(-/-) and IFNAR1(-/-) mice. In addition, IPC-mediated protection in WM was also abolished in IFNAR1(fl/fl) LysM(cre), but not in IFNAR1(fl/fl) control, mice. These findings demonstrated for the first time that IPC was robust in WM, the phenomenon being intrinsic to WM itself. Furthermore, WM IPC was dependent on innate immune cell signaling pathways. Finally, these data demonstrated that microglial-specific expression of IFNAR1 plays an indispensable role in WM IPC. SIGNIFICANCE STATEMENT: Ischemic preconditioning (IPC) has been studied predominantly in gray matter, but stroke in humans frequently involves white matter (WM) as well. Here we describe a novel, combined in vivo/ex vivo mouse model to determine whether IPC occurs in WM. It does. Using genetically altered mice, we identified two innate immune cell receptors, Toll-like receptor 4 and type 1 interferon receptor (IFNAR1), that are required for IPC-mediated protection in WM. Furthermore, using microglia-targeted IFNAR1 knockdown, we demonstrate that interferon signaling specifically in microglia is essential for this protection. The discovery of IPC as an intrinsic capability of WM is novel and important. This is also the first in vivo demonstration that cell-type-specific expression of an individual gene plays an indispensable role in IPC-mediated protection.


Assuntos
Isquemia Encefálica/metabolismo , Precondicionamento Isquêmico/métodos , Receptor de Interferon alfa e beta/biossíntese , Receptor 4 Toll-Like/biossíntese , Substância Branca/metabolismo , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Receptor 4 Toll-Like/genética , Substância Branca/patologia
7.
J Biol Chem ; 289(38): 26058-26073, 2014 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-25086040

RESUMO

Astrocytes and neurons express several large pore (hemi)channels that may open in response to various stimuli, allowing fluorescent dyes, ions, and cytoplasmic molecules such as ATP and glutamate to permeate. Several of these large pore (hemi)channels have similar characteristics with regard to activation, permeability, and inhibitor sensitivity. Consequently, their behaviors and roles in astrocytic and neuronal (patho)physiology remain undefined. We took advantage of the Xenopus laevis expression system to determine the individual characteristics of several large pore channels in isolation. Expression of connexins Cx26, Cx30, Cx36, or Cx43, the pannexins Px1 or Px2, or the purinergic receptor P2X7 yielded functional (hemi)channels with isoform-specific characteristics. Connexin hemichannels had distinct sensitivity to alterations of extracellular Ca(2+) and their permeability to dyes and small atomic ions (conductance) were not proportional. Px1 and Px2 exhibited conductance at positive membrane potentials, but only Px1 displayed detectable fluorescent dye uptake. P2X7, in the absence of Px1, was permeable to fluorescent dyes in an agonist-dependent manner. The large pore channels displayed overlapping sensitivity to the inhibitors Brilliant Blue, gadolinium, and carbenoxolone. These results demonstrated isoform-specific characteristics among the large pore membrane channels; an open (hemi)channel is not a nonselective channel. With these isoform-specific properties in mind, we characterized the divalent cation-sensitive permeation pathway in primary cultured astrocytes. We observed no activation of membrane conductance or Cx43-mediated dye uptake in astrocytes nor in Cx43-expressing C6 cells. Our data underscore that although Cx43-mediated transport is observed in overexpressing cell systems, such transport may not be detectable in native cells under comparable experimental conditions.


Assuntos
Astrócitos/metabolismo , Conexina 43/metabolismo , Neurônios/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Benzenossulfonatos/farmacologia , Carbenoxolona/farmacologia , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Etídio/metabolismo , Corantes Fluorescentes/metabolismo , Gadolínio/farmacologia , Junções Comunicantes/metabolismo , Potenciais da Membrana , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Ratos Sprague-Dawley , Receptores Purinérgicos P2X7/metabolismo , Xenopus laevis
8.
Glia ; 63(12): 2208-19, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26200696

RESUMO

Oxidative stress plays an important role in the progression of Alzheimer's disease (AD) and other neurodegenerative conditions. Glutathione (GSH), the major antioxidant in the central nervous system, is primarily synthesized and released by astrocytes. We determined if ß-amyloid (Aß42), crucially involved in Alzheimer's disease, affected GSH release. Monomeric Aß (mAß) stimulated GSH release from cultured cortical astrocytes more effectively than oligomeric Aß (oAß) or fibrillary Aß (fAß). Monomeric Aß increased the expression of the transporter ABCC1 (also referred to as MRP1) that is the main pathway for GSH release. GSH release from astrocytes, with or without mAß stimulation, was reduced by pharmacological inhibition of ABCC1. Astrocytes robustly express connexin proteins, especially connexin43 (Cx43), and mAß also stimulated Cx43 hemichannel-mediated glutamate and GSH release. Aß-stimulation facilitated hemichannel opening in the presence of normal extracellular calcium by reducing astrocyte cholesterol level. Aß treatment did not alter the intracellular concentration of reduced or oxidized glutathione. Using a mouse model of AD with early onset Aß deposition (5xFAD), we found that cortical ABCC1 was significantly increased in temporal register with the surge of Aß levels in these mice. ABCC1 levels remained elevated from 1.5 to 3.5 months of age in 5xFAD mice, before plunging to subcontrol levels when amyloid plaques appeared. Similarly, in cultured astrocytes, prolonged incubation with aggregated Aß, but not mAß, reduced induction of ABCC1 expression. These results support the hypothesis that in the early stage of AD pathogenesis, less aggregated Aß increases GSH release from astrocytes (via ABCC1 transporters and Cx43 hemichannels) providing temporary protection from oxidative stress which promotes AD development.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Glutationa/metabolismo , Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Cálcio/metabolismo , Células Cultivadas , Córtex Cerebral/metabolismo , Colesterol/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/antagonistas & inibidores , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Oxirredução , Placa Amiloide/metabolismo
9.
Ann Neurol ; 75(4): 492-507, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24242287

RESUMO

OBJECTIVE: Hypoglycemia is a common adverse event and can injure central nervous system (CNS) white matter (WM). We determined whether glutamate receptors were involved in hypoglycemic WM injury. METHODS: Mouse optic nerves (MON), CNS WM tracts, were maintained at 37°C with oxygenated artificial cerebrospinal fluid (ACSF) containing 10mM glucose. Aglycemia was produced by switching to 0 glucose ACSF. Supramaximal compound action potentials (CAPs) were elicited using suction electrodes, and axon function was quantified as the area under the CAP. Amino acid release was measured using high-performance liquid chromatography. Extracellular lactate concentration ([lactate(-)]o) was measured using an enzyme electrode. RESULTS: About 50% of MON axons were injured after 60 minutes of aglycemia (90% after 90 minutes); injury extent was not affected by animal age. Blockade of N-methyl-D-aspartate (NMDA)-type glutamate receptors improved recovery after 90 minutes of aglycemia by 250%. Aglycemic injury was increased by reducing [Mg(2+)]o or increasing [glycine]o , and decreased by lowering pHo , expected results for NMDA receptor-mediated injury. pHo increased during aglycemia due to a drop in [lactate(-)]o. Aglycemic injury was dramatically reduced in the absence of [Ca(2+)]o. Extracellular aspartate, a selective NMDA receptor agonist, increased during aglycemia ([glutamate]o fell). INTERPRETATION: Aglycemia injured WM by a unique excitotoxic mechanism involving NMDA receptors (located primarily on oligodendrocytes). During WM aglycemia, the selective NMDA agonist aspartate is released, probably from astrocytes. Injury is mediated by Ca(2+) influx through aspartate-activated NMDA receptors made permeable by an accompanying alkaline shift in pHo caused by a fall in [lactate(-)]o. These insights have important clinical implications.


Assuntos
Leucoencefalopatias/etiologia , Traumatismos do Nervo Óptico/etiologia , Traumatismos do Nervo Óptico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Ácido Aspártico/metabolismo , Encéfalo/efeitos dos fármacos , Cálcio/metabolismo , Modelos Animais de Doenças , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Ácido Glutâmico/metabolismo , Glicina/metabolismo , Glicogênio/metabolismo , Concentração de Íons de Hidrogênio , Hipoglicemia/complicações , Ácido Cinurênico/análogos & derivados , Ácido Cinurênico/uso terapêutico , Ácido Láctico/metabolismo , Leucoencefalopatias/tratamento farmacológico , Leucoencefalopatias/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Traumatismos do Nervo Óptico/tratamento farmacológico , Quinoxalinas/uso terapêutico , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
10.
Biochem Biophys Res Commun ; 450(1): 295-9, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24944014

RESUMO

Glioma glutamate release has been shown to promote the growth of glioma cells and induce neuronal injuries from epilepsy to neuronal death. However, potential counteractions from normal astrocytes against glioma glutamate release have not been fully evaluated. In this study, we investigated the glutamate/glutamine cycling between glioma cells and astrocytes and their impact on neuronal function. Co-cultures of glioma cells with astrocytes (CGA) in direct contact were established under different mix ratio of astrocyte/glioma. Culture medium conditioned in these CGAs were sampled for HPLC measurement, for neuronal ratiometric calcium imaging, and for neuronal survival assay. We found: (1) High levels of glutaminase expression in glioma cells, but not in astrocytes, glutaminase enables glioma cells to release large amount of glutamate in the presence of glutamine. (2) Glutamate levels in CGAs were directly determined by the astrocyte/glioma ratios, indicating a balance between glioma glutamate release and astrocyte glutamate uptake. (3) Culture media from CGAs of higher glioma/astrocyte ratios induced stronger neuronal Ca(2+) response and more severe neuronal death. (4) Co-culturing with astrocytes significantly reduced the growth rate of glioma cells. These results indicate that normal astrocytes in the brain play pivotal roles in glioma growth inhibition and in reducing neuronal injuries from glioma glutamate release. However, as tumor growth, the protective role of astrocytes gradually succumb to glioma cells.


Assuntos
Astrócitos/metabolismo , Astrócitos/patologia , Comunicação Celular , Glioma/metabolismo , Glioma/patologia , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica
11.
Exp Neurol ; 359: 114230, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36162511

RESUMO

The disruption of nucleus accumbens (NAc) function impacts mood and learning behavior in α-Synucleinopathy, in which microglial synaptic pruning plays a pivotal role in modulating the neuropathologic progression. Available literature documents that in microglia, the activation of cannabinoid receptor 2 (CB2R) decreases inflammation, but it remains obscured regarding the roles of CB2R in microglia-mediated synaptic pruning in the NAc during the neuropathological progression of α-Synucleinopathy. We adopted the fibrillar α-Synuclein (α-Syn) treatment to characterize the effect of genetic CB2R deletion on microglial function and the signaling pathway. CB2R knockout (CB2-/-) mice and wild-type (CB2+/+) mice were divided into the α-Syn or saline treatment groups. Biochemical and microscopy approaches, including immunofluorescence, real-time PCR, and western blotting, were employed to assess the changes in homeostasis of synaptic pruning in NAc under the α-Syn-induced microglia. Moreover, the underlying mechanisms of CB2R on α-Syn induced microglial activity was assessed in vitro. After the injection of α-Syn into the NAc, distinct microglial morphological changes and M1 phenotype transformation were observed between CB2-/- and CB2+/+ mice. Meanwhile, after the α-Syn treatment, CB2-/- mice showed an increased upregulation of CD68 protein and IL-1ß mRNA but decreased brain-derived neurotrophic factor (BDNF) and TGF-ß mRNA compared with CB2+/+ mice. Additionally, CB2-/- microglia after the treatment showed a highly enriched complement 3a receptor (C3aR) producing excessive pruning of cholinergic synapses but less engulfment of dopaminergic synapses. Mechanistically, the loss of CB2R function in the α-Syn stimulation triggered c-Fos activation in microglia, but not in neurons. Further inhibition of microglial CB2R functions under α-Syn stimulation activated the phosphorylated cAMP-response element-binding protein (pCREB)-c-Fos, which was closely related to the C3aR upregulation. Our results reveal a critical and mechanistic role of CB2R in altering the microglial function and its value in the homeostasis of synaptic circuits in the NAc under the α-Syn pathology.


Assuntos
Microglia , Sinucleinopatias , Animais , Camundongos , Microglia/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Núcleo Accumbens/metabolismo , Transdução de Sinais , Plasticidade Neuronal , RNA Mensageiro/metabolismo , Receptores de Canabinoides/metabolismo
12.
J Mol Neurosci ; 72(3): 527-543, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34409578

RESUMO

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most frequent cause of autosomal dominant Parkinson's disease (PD), producing psychiatric and motor symptoms. We conducted this study to explore whether microglial dopaminergic (DAergic) fiber refinement and synaptic pruning are involved in the abnormal behavioral phenotypes of carriers of the LRRK2 G2019S mutation, by employing young and middle-aged PD model mice. The results revealed a characteristic late-onset hyperactivity and a progressive decline in the motor coordination of the LRRK2 G2019S mutation mice. LRRK2 G2019S mutation-induced aberrant microglial morphogenesis, with more branches and junctions per cell, resulted in excessive microglial refinement of dopaminergic (DAergic) fibers. Moreover, aberrant synaptic pruning distinctly impacted the prefrontal cortex (PFC) and dorsal striatum (DS), with significantly higher spine density in the PFC but the opposite effects in the DS region. Furthermore, LRRK2 G2019S mutation remodeled the inflammatory transcription landscape of microglia, rendering certain cerebral areas highly susceptible to microglial immune response. These findings indicate that LRRK2 G2019S mutation induces the production of inflammatory cytokines and mediates abnormal microglial morphogenesis and activity, resulting in abnormal phagocytosis, synaptic pruning and loss of DAergic fibers during aging, and, eventually, PD-related behavioral abnormalities.


Assuntos
Dopamina , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Microglia , Animais , Comportamento Animal , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Camundongos , Mutação , Plasticidade Neuronal
13.
Exp Neurol ; 358: 114228, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36108713

RESUMO

BACKGROUND AND PURPOSE: Enhancing phagocytosis can facilitate the removal of inflammatory molecules, limit the toxicity of dead cells and debris, and promote recovery after brain injury. In this study, we aimed to explore the role of bexarotene (Bex), a retinoid X receptor (RXR) agonist, in promoting astrocyte phagocytosis and neurobehavioral recovery after subarachnoid hemorrhage (SAH). METHODS: Mice SAH model was induced by pre-chiasmatic injection of blood. Modified Garcia score, novel object recognition, rotarod test, and Morris water maze were performed to assess neurological function. Immunofluorescence and electron microscopy were used to evaluate astrocyte phagocytosis in vivo. In addition, ABCA1/MEGF10&GULP1, the primary astrocyte phagocytosis pathway, were stimulated by Bex or suppressed by HX531 (a RXR antagonist) to evaluate their impacts on astrocyte phagocytosis and neurological recovery. RESULTS: Astrocytes phagocytosis of blood components were observed in mice after SAH induction, which is further increased by Bex treatment. Bex dramatically attenuated neuroinflammation, reduced brain edema, improved early neurological performance and promoted neurocognitive recovery. Meanwhile, Bex decreased neurotoxic reactive astrocytes and preserved neurogenesis after SAH. Bex increased the expression of astrocyte phagocytosis-related proteins ABCA1, MEGF10, and GULP1. Bex also increased the lysosomal processing of engulfed blood components in astrocytes. Moreover, Bex significantly promoted astrocytes to phagocytize debris in vitro by increasing the expression of ABCA1, MEGF10 and GULP1, while HX531 inhibited astrocyte phagocytosis and decreased these protein levels. CONCLUSIONS: Bex enhanced astrocyte phagocytosis through the ABCA1-mediated pathways, and promoted neurobehavior recovery in mice after SAH induction.


Assuntos
Hemorragia Subaracnóidea , Transportador 1 de Cassete de Ligação de ATP , Animais , Astrócitos/metabolismo , Benzoatos , Bexaroteno/farmacologia , Bexaroteno/uso terapêutico , Compostos de Bifenilo , Modelos Animais de Doenças , Proteínas de Membrana/metabolismo , Camundongos , Fagocitose , Receptores X de Retinoides/agonistas , Hemorragia Subaracnóidea/tratamento farmacológico
14.
Front Pharmacol ; 13: 859978, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35652041

RESUMO

Mitochondria are essential organelles that perform important roles in cell biologies such as ATP synthesis, metabolic regulation, immunomodulatory, and apoptosis. Parkinson's disease (PD) is connected with mitochondrial neuronal damage related to mitochondrial unfolded protein response (mtUPR). Rosmarinic acid (RA) is a naturally occurring hydroxylated polyphenolic chemical found in the Boraginaceae and the Labiatae subfamily Nepetoideae. This study looked into RA's protective effect against mitochondrial loss in the substantia nigra (SN) caused by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), the underlying mechanism associated with the mtUPR. Pretreatment with RA reduced motor impairments and dopaminergic neuronal degeneration in the SN of a mouse model injected with MPTP. Pretreatment of SH-SY5Y cells from cell viability loss, morphological damage, and oxidative stress. Furthermore, RA pre-injection suppressed MPTP-induced mtUPR, lowered the expression of HSPA9, HSPE1, CLPP, LONP1, and SIRT 4, and protected the MPTP-mice and SH-SY5Y cells from mitochondrial failure. These findings imply that RA can prevent Parkinson's disease by preventing mitochondrial damage in dopaminergic neurons in Parkinson's disease via alleviating mitochondrial unfolded protein response.

15.
Neuro Oncol ; 23(6): 905-919, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33249476

RESUMO

BACKGROUND: Failure of glioblastoma (GBM) therapy is often ascribed to different types of glioblastoma stem-like cell (GSLC) niche; in particular, a hypoxic perivascular niche (HPVN) is involved in GBM progression. However, the cells responsible for HPVNs remain unclear. METHODS: Immunostaining was performed to determine the cells involved in HPVNs. A hypoxic chamber and 3-dimensional (3D) microfluidic chips were designed to simulate a HPVN based on the pathological features of GBM. The phenotype of GSLCs was evaluated by fluorescence scanning in real time and proliferation and apoptotic assays. The expression of JAG1, DLL4, and Hes1 was determined by immunostaining, ELISA, Western blotting, and quantitative PCR. Their clinical prognostic significance in GBM HPVNs and total tumor tissues were verified by clinical data and The Cancer Genome Atlas databases. RESULTS: Nestin+/CD31+ cells and pericytes constitute the major part of microvessels in the HPVN, and the high ratio of nestin+/CD31+ cells rather than pericytes are responsible for the poor prognosis of GBM. A more real HPVN was simulated by a hypoxic coculture system in vitro, which consisted of 3D microfluidic chips and a hypoxic chamber. Nestin+/CD31+ cells in the HPVN were derived from GSLC transdifferentiation and promoted GSLC chemoresistance by providing more JAG1 and DLL4 to induce downstream Hes1 overexpression. Poor GBM prognosis correlated with Hes1 expression of tumor cells in the GBM HPVN, and not with total Hes1 expression in GBM tissues. CONCLUSIONS: These results highlight the critical role of nestin+/CD31+ cells in HPVNs that acts in GBM chemoresistance and reveal the distinctive prognostic value of these molecular markers in HPVNs.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Ligação ao Cálcio , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Humanos , Hipóxia , Proteína Jagged-1 , Nestina/genética
16.
Am J Pathol ; 175(1): 54-65, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19498008

RESUMO

Parkinson disease (PD) typically affects the cortical regions during the later stages of disease, with neuronal loss, gliosis, and formation of diffuse cortical Lewy bodies in a significant portion of patients with dementia. To identify novel proteins involved in PD progression, we prepared synaptosomal fractions from the frontal cortices of pathologically verified PD patients at different stages along with age-matched controls. Protein expression profiles were compared using a robust quantitative proteomic technique. Approximately 100 proteins displayed significant differences in their relative abundances between PD patients at various stages and controls; three of these proteins were validated using independent techniques. One of the confirmed proteins, glutathione S-transferase Pi, was further investigated in cellular models of PD, demonstrating that its level was intimately associated with several critical cellular processes that are directly related to neurodegeneration in PD. These results have, for the first time, suggested that the levels of glutathione S-transferase Pi may play an important role in modulating the progression of PD.


Assuntos
Química Encefálica , Glutationa S-Transferase pi/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Animais , Western Blotting , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , MAP Quinase Quinase 4/metabolismo , Masculino , Camundongos , Reação em Cadeia da Polimerase , Transdução de Sinais/fisiologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Transfecção
17.
J Neurosci ; 28(6): 1479-89, 2008 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-18256269

RESUMO

Stroke incidence increases with age and this has been attributed to vascular factors. We show here that CNS white matter (WM) is intrinsically more vulnerable to ischemic injury in older animals and that the mechanisms of WM injury change as a function of age. The mouse optic nerve was used to study WM function. WM function in older animals (12 months) was not protected from ischemic injury by removal of extracellular Ca2+ or by blockade of reverse Na+/Ca2+ exchange, as is the case with young adults. Ischemic WM injury in older mice is predominately mediated by glutamate release and activation of AMPA/kainate-type glutamate receptors. Glutamate release, attributable to reverse glutamate transport, occurs earlier and is more robust in older mice that show greater expression of the glutamate transporter. The observation that WM vulnerability to ischemic injury is age dependent has possible implications for the pathogenesis of other age-related CNS conditions.


Assuntos
Envelhecimento/patologia , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Fibras Nervosas Mielinizadas/metabolismo , Neuropatia Óptica Isquêmica/fisiopatologia , Envelhecimento/efeitos dos fármacos , Envelhecimento/metabolismo , Animais , Cálcio/metabolismo , Ácido Glutâmico/metabolismo , Masculino , Camundongos , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Neuropatia Óptica Isquêmica/metabolismo
18.
Glia ; 57(3): 258-69, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18837047

RESUMO

The study of ion channels has relied heavily on the use of pharmacological blocking agents. However, many of these agents have multiple effects, which may compromise interpretation of results when the affected mechanisms/pathways mediate similar functions. Volume regulated anion channels (VRAC) and connexin hemichannels can both mediate the release of glutamate and taurine, although these channels have distinct activation stimuli and hemichannels, but not VRAC, are permeable to Lucifer Yellow (LY). It has been reported that some anion channel blockers may inhibit connexin hemichannels. We further examined the effects of classic gap junction/hemichannel blockers and anion channel blockers on these channels. The typical VRAC blockers, NPPB, IAA-94, and tamoxifen blocked low divalent cation-induced glutamate and taurine release and LY loading, presumed due to hemichannel opening. The blocking action of these compounds on hemichannels was concentration dependent and fell within the same range where the drugs classically block VRACs. Conversely, carbenoxolone (CBX), the most widely used gap junction/hemichannel blocker, was an effective blocker of VRAC-mediated glutamate and taurine release, and blocked these channels at similar concentrations at which it blocked hemichannels. The CBX effect on VRACs was verified using astrocytes from connexin 43 knock out (Cx43 KO) animals. In these cells, the hypotonic induced amino acid flux was retained whereas the low divalent cation solution-induced flux was lost. These results extend our knowledge about "cross-inhibition" of VRACs and gap junctions/hemichannels by certain pharmacological agents. Given the overlap in function of these two types of channels, great care must be exerted in using pharmacological blockers to identify one channel from the other.


Assuntos
Astrócitos/efeitos dos fármacos , Tamanho Celular , Conexina 43/fisiologia , Ativação do Canal Iônico/fisiologia , Canais Iônicos/fisiologia , Aminoácidos/metabolismo , Análise de Variância , Inibidores da Angiogênese/farmacologia , Animais , Animais Recém-Nascidos , Carbenoxolona/farmacologia , Tamanho Celular/efeitos dos fármacos , Células Cultivadas , Cromatografia Líquida de Alta Pressão/métodos , Conexina 43/antagonistas & inibidores , Conexina 43/deficiência , Conexina 43/efeitos dos fármacos , Ácido Glicirretínico/análogos & derivados , Ácido Glicirretínico/farmacologia , Hipocampo/citologia , Soluções Hipotônicas/farmacologia , Indanos/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/genética , Canais Iônicos/efeitos dos fármacos , Isoquinolinas/metabolismo , Camundongos , Camundongos Knockout , Nitrobenzoatos/farmacologia , Ratos , Tamoxifeno/farmacologia
20.
J Cereb Blood Flow Metab ; 27(9): 1540-52, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17299453

RESUMO

Axonal injury and dysfunction in white matter (WM) are caused by many neurologic diseases including ischemia. We characterized ischemic injury and the role of glutamate-mediated excitotoxicity in a purely myelinated WM tract, the mouse optic nerve (MON). For the first time, excitotoxic WM injury was directly correlated with glutamate release. Oxygen and glucose deprivation (OGD) caused duration-dependent loss of axon function in optic nerves from young adult mice. Protection of axon function required blockade of both alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) and kainate receptors, or removal of extracellular Ca(2+). Blockade of N-methyl-D-aspartate receptors did not preserve axon function. Curiously, even extended periods of direct exposure to glutamate or kainate or AMPA failed to induce axon dysfunction. Brief periods of OGD, however, caused glutamate receptor agonist exposure to become toxic, suggesting that ionic disruption enabled excitotoxic injury. Glutamate release, directly measured using quantitative high-performance liquid chromatography, occurred late during a 60-mins period of OGD and was due to reversal of the glutamate transporter. Brief periods of OGD (i.e., 15 mins) did not cause glutamate release and produced minimal injury. These results suggested that toxic glutamate accumulation during OGD followed the initial ionic changes mediating early loss of excitability. The onset of glutamate release was an important threshold event for irreversible ischemic injury. Regional differences appear to exist in the specific glutamate receptors that mediate WM ischemic injury. Therapy for ischemic WM injury must be designed accordingly.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Ácido Glutâmico/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Animais , Cálcio/metabolismo , Cromatografia Líquida de Alta Pressão , Eletrofisiologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Masculino , Camundongos , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Nervo Óptico/efeitos dos fármacos , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Técnicas de Cultura de Órgãos , Receptores de AMPA/efeitos dos fármacos , Receptores de AMPA/metabolismo , Receptores de Glutamato/efeitos dos fármacos , Receptores de Glutamato/metabolismo , Receptores de Ácido Caínico/efeitos dos fármacos , Receptores de Ácido Caínico/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa