Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
eNeuro ; 11(3)2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38548335

RESUMO

Neuroprotection after injury or in neurodegenerative disease remains a major goal for basic and translational neuroscience. Retinal ganglion cells (RGCs), the projection neurons of the eye, degenerate in optic neuropathies after axon injury, and there are no clinical therapies to prevent their loss or restore their connectivity to targets in the brain. Here we demonstrate a profound neuroprotective effect of the exogenous expression of various Ca2+/calmodulin-dependent protein kinase II (CaMKII) isoforms in mice. A dramatic increase in RGC survival following the optic nerve trauma was elicited by the expression of constitutively active variants of multiple CaMKII isoforms in RGCs using adeno-associated viral (AAV) vectors across a 100-fold range of AAV dosing in vivo. Despite this neuroprotection, however, short-distance RGC axon sprouting was suppressed by CaMKII, and long-distance axon regeneration elicited by several pro-axon growth treatments was likewise inhibited even as CaMKII further enhanced RGC survival. Notably, in a dose-escalation study, AAV-expressed CaMKII was more potent for axon growth suppression than the promotion of survival. That diffuse overexpression of constitutively active CaMKII strongly promotes RGC survival after axon injury may be clinically valuable for neuroprotection per se. However, the associated strong suppression of the optic nerve axon regeneration demonstrates the need for understanding the intracellular domain- and target-specific CaMKII activities to the development of CaMKII signaling pathway-directed strategies for the treatment of optic neuropathies.


Assuntos
Doenças Neurodegenerativas , Doenças do Nervo Óptico , Traumatismos do Nervo Óptico , Camundongos , Animais , Células Ganglionares da Retina/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Axônios/metabolismo , Doenças Neurodegenerativas/metabolismo , Regeneração Nervosa/fisiologia , Doenças do Nervo Óptico/metabolismo , Isoformas de Proteínas/metabolismo , Sobrevivência Celular/fisiologia
2.
Nature ; 626(7999): 574-582, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38086421

RESUMO

The intrinsic mechanisms that regulate neurotoxic versus neuroprotective astrocyte phenotypes and their effects on central nervous system degeneration and repair remain poorly understood. Here we show that injured white matter astrocytes differentiate into two distinct C3-positive and C3-negative reactive populations, previously simplified as neurotoxic (A1) and neuroprotective (A2)1,2, which can be further subdivided into unique subpopulations defined by proliferation and differential gene expression signatures. We find the balance of neurotoxic versus neuroprotective astrocytes is regulated by discrete pools of compartmented cyclic adenosine monophosphate derived from soluble adenylyl cyclase and show that proliferating neuroprotective astrocytes inhibit microglial activation and downstream neurotoxic astrocyte differentiation to promote retinal ganglion cell survival. Finally, we report a new, therapeutically tractable viral vector to specifically target optic nerve head astrocytes and show that raising nuclear or depleting cytoplasmic cyclic AMP in reactive astrocytes inhibits deleterious microglial or macrophage cell activation and promotes retinal ganglion cell survival after optic nerve injury. Thus, soluble adenylyl cyclase and compartmented, nuclear- and cytoplasmic-localized cyclic adenosine monophosphate in reactive astrocytes act as a molecular switch for neuroprotective astrocyte reactivity that can be targeted to inhibit microglial activation and neurotoxic astrocyte differentiation to therapeutic effect. These data expand on and define new reactive astrocyte subtypes and represent a step towards the development of gliotherapeutics for the treatment of glaucoma and other optic neuropathies.


Assuntos
Astrócitos , Neuroproteção , Adenilil Ciclases/metabolismo , Astrócitos/citologia , Astrócitos/enzimologia , Astrócitos/metabolismo , Diferenciação Celular , Núcleo Celular/metabolismo , Sobrevivência Celular , AMP Cíclico/metabolismo , Citoplasma/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Microglia/metabolismo , Microglia/patologia , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Traumatismos do Nervo Óptico/terapia , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/metabolismo , Substância Branca/metabolismo , Substância Branca/patologia , Glaucoma/patologia , Glaucoma/terapia
3.
Stem Cell Reports ; 17(12): 2690-2703, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36368332

RESUMO

Retinal ganglion cell (RGC) replacement therapy could restore vision in glaucoma and other optic neuropathies. We developed a rapid protocol for directly induced RGC (iRGC) differentiation from human stem cells, leveraging overexpression of NGN2. Neuronal morphology and neurite growth were observed within 1 week of induction; characteristic RGC-specific gene expression confirmed identity. Calcium imaging demonstrated γ-aminobutyric acid (GABA)-induced excitation characteristic of immature RGCs. Single-cell RNA sequencing showed more similarities between iRGCs and early-stage fetal human RGCs than retinal organoid-derived RGCs. Intravitreally transplanted iRGCs survived and migrated into host retinas independent of prior optic nerve trauma, but iRGCs protected host RGCs from neurodegeneration. These data demonstrate rapid iRGC generation in vitro into an immature cell with high similarity to human fetal RGCs and capacity for retinal integration after transplantation and neuroprotective function after optic nerve injury. The simplicity of this system may benefit translational studies on human RGCs.


Assuntos
Glaucoma , Traumatismos do Nervo Óptico , Humanos , Células Ganglionares da Retina , Traumatismos do Nervo Óptico/metabolismo , Retina , Células-Tronco
4.
eNeuro ; 8(1)2021.
Artigo em Inglês | MEDLINE | ID: mdl-33441400

RESUMO

The failure of adult CNS neurons to survive and regenerate their axons after injury or in neurodegenerative disease remains a major target for basic and clinical neuroscience. Recent data demonstrated in the adult mouse that exogenous expression of Sry-related high-mobility-box 11 (Sox11) promotes optic nerve regeneration after optic nerve injury but exacerbates the death of a subset of retinal ganglion cells (RGCs), α-RGCs. During development, Sox11 is required for RGC differentiation from retinal progenitor cells (RPCs), and we found that mutation of a single residue to prevent SUMOylation at lysine 91 (K91) increased Sox11 nuclear localization and RGC differentiation in vitro Here, we explored whether this Sox11 manipulation similarly has stronger effects on RGC survival and optic nerve regeneration. In vitro, we found that non-SUMOylatable Sox11K91A leads to RGC death and suppresses axon outgrowth in primary neurons. We furthermore found that Sox11K91A more strongly promotes axon regeneration but also increases RGC death after optic nerve injury in vivo in the adult mouse. RNA sequence (RNA-seq) data showed that Sox11 and Sox11K91A increase the expression of key signaling pathway genes associated with axon growth and regeneration but downregulated Spp1 and Opn4 expression in RGC cultures, consistent with negatively regulating the survival of α-RGCs and ipRGCs. Thus, Sox11 and its SUMOylation site at K91 regulate gene expression, survival and axon growth in RGCs, and may be explored further as potential regenerative therapies for optic neuropathy.


Assuntos
Doenças Neurodegenerativas , Traumatismos do Nervo Óptico , Animais , Axônios/metabolismo , Sobrevivência Celular , Camundongos , Regeneração Nervosa , Doenças Neurodegenerativas/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Processamento de Proteína Pós-Traducional , Fatores de Transcrição SOXC/genética , Fatores de Transcrição SOXC/metabolismo
5.
RSC Adv ; 11(57): 35796-35805, 2021 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-35492766

RESUMO

Magnetic nanoparticles (MNPs) are widely used in cell sorting, organelle selection, drug delivery, cell delivery, and cell tracking applications. However, organelle manipulation in living cells has been limited due to the endocytic uptake and sequestration of MNPs. Here, we introduce a method for modifying MNPs with fusogenic liposomes that facilitate MNP passage directly into the cytosol. MNPs were enclosed in fusogenic liposomes that exhibit a core-shell structure under a transmission electron microscope (TEM). The lipid-to-MNP ratio was optimized for one layer of liposome coating around each MNP, so that MNPs were delivered to the cytosol without endosomal or liposomal coatings. After incubation with the retinal pigment epithelial cell line ARPE-19, single-layer liposome-coated MNPs exhibited the highest MNP delivery efficiency. Although uncoated MNPs are taken up through endocytosis, less than 15% of the fusogenic liposome-coated MNPs co-localized with early endosomes. MNPs delivered by fusogenic liposomes showed cytosolic localization early on and increased lysosomal localization at later time points. The movement of intracellular MNPs could be manipulated with an external magnet to estimate cytosolic viscosity. Bypassing endocytosis in this way allowed efficient delivery of MNPs to the cytosol, potentially allowing for the targeting of specific organelles and controlling their motion in living cells.

6.
PLoS One ; 15(12): e0242884, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33315889

RESUMO

Loss of retinal ganglion cells (RGCs) in optic neuropathies results in permanent partial or complete blindness. Myocyte enhancer factor 2 (MEF2) transcription factors have been shown to play a pivotal role in neuronal systems, and in particular MEF2A knockout was shown to enhance RGC survival after optic nerve crush injury. Here we expanded these prior data to study bi-allelic, tri-allelic and heterozygous allele deletion. We observed that deletion of all MEF2A, MEF2C, and MEF2D alleles had no effect on RGC survival during development. Our extended experiments suggest that the majority of the neuroprotective effect was conferred by complete deletion of MEF2A but that MEF2D knockout, although not sufficient to increase RGC survival on its own, increased the positive effect of MEF2A knockout. Conversely, MEF2A over-expression in wildtype mice worsened RGC survival after optic nerve crush. Interestingly, MEF2 transcription factors are regulated by post-translational modification, including by calcineurin-catalyzed dephosphorylation of MEF2A Ser-408 known to increase MEF2A-dependent transactivation in neurons. However, neither phospho-mimetic nor phospho-ablative mutation of MEF2A Ser-408 affected the ability of MEF2A to promote RGC death in vivo after optic nerve injury. Together these findings demonstrate that MEF2 gene expression opposes RGC survival following axon injury in a complex hierarchy, and further support the hypothesis that loss of or interference with MEF2A expression might be beneficial for RGC neuroprotection in diseases such as glaucoma and other optic neuropathies.


Assuntos
Fatores de Transcrição MEF2/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Células Ganglionares da Retina/patologia , Alelos , Animais , Contagem de Células , Humanos , Fatores de Transcrição MEF2/deficiência , Fatores de Transcrição MEF2/genética , Camundongos , Traumatismos do Nervo Óptico/genética , Mutação Puntual , Transdução de Sinais
7.
Sci Rep ; 7(1): 6015, 2017 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-28729651

RESUMO

Photoreceptor degeneration is a central pathology of various retinal degenerative diseases which currently lack effective therapies. Antioxidant and anti-inflammatory activities are noted for Panax notoginsenoside saponins (PNS) and related saponin compound(s). However, the photoreceptor protective potentials of PNS or related saponin compound(s) remain unknown. The current study revealed that PNS protected against photoreceptor loss in bright light-exposed BALB/c mice. Combination of ginsenoside Rb1 and Rd, two major saponin compounds of PNS, recapitulated the retinal protection of PNS and attenuated retinal oxidative stress and inflammatory changes. Rb1 or Rd partially alleviated all-trans-Retinal-induced oxidative stress in ARPE19 cells. Rb1 or Rd suppressed lipopolysaccharides (LPS)-induced proinflammatory gene expression in ARPE19 and RAW264.7 cells. Rb1 or Rd also modulated the expression of proinflammatory microRNA, miR-155 and its direct target, anti-inflammatory SHIP1, in LPS-stimulated RAW264.7 cells. The retinal expression of miR-155 and SHIP1 was altered preceding extensive retinal damage, which was maintained at normal level by Rb1 and Rd combination. This work shows for the first time that altered expression of miR-155 and SHIP1 are involved in photoreceptor degeneration. Most importantly, novel retinal protective activities of combination of Rb1 and Rd justify further evaluation for the treatment of related retinal degenerative disorders.


Assuntos
Ginsenosídeos/farmacologia , Luz/efeitos adversos , Substâncias Protetoras/farmacologia , Retina/efeitos dos fármacos , Degeneração Retiniana/etiologia , Degeneração Retiniana/metabolismo , Animais , Antioxidantes/farmacologia , Biomarcadores , Adesão Celular/genética , Expressão Gênica , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Células Fotorreceptoras/efeitos dos fármacos , Células Fotorreceptoras/metabolismo , Células RAW 264.7 , Espécies Reativas de Oxigênio , Retina/metabolismo , Degeneração Retiniana/patologia , Degeneração Retiniana/prevenção & controle , Saponinas/farmacologia
8.
Brain Res ; 1663: 141-150, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28336272

RESUMO

Vision impairment in retinal degenerative diseases such as age-related macular degeneration is primarily associated with photoreceptor degeneration, in which oxidative stress and inflammatory responses are mechanistically involved as central players. Therapies with photoreceptor protective properties remain to be developed. Apigenin-7-diglucuronide (A7DG), a flavonoid glycoside, is present in an assortment of medicinal plants with anti-inflammatory or ant-oxidant activities. However, the pharmacological significance of A7DG remains unknown in vivo. The current study isolated A7DG from Glechoma longituba (Nakai) Kuprian and investigated the retinal protective effect A7DG in mice characterized by bright light-induced photoreceptor degeneration. The results showed that A7DG treatment led to remarkable photoreceptor protection in bright light-exposed BALB/c mice. Moreover, A7DG treatment alleviated photoreceptor apoptosis, mitigated oxidative stress, suppressed reactive gliosis and microglial activation and attenuated the expression of proinflammatory genes in bright light-exposed retinas. The results demonstrated for the first time remarkable photoreceptor protective activities of A7DG in vivo. Inhibition of bright light-induced retinal oxidative stress and retinal inflammatory responses was associated with the retinal protection conferred by A7DG. The work here warrants further evaluation of A7DG as a pharmacological candidate for the treatment of vision-threatening retinal degenerative disorders. Moreover, given the general implication of oxidative stress and inflammation in the pathogenesis of neurodegeneration, A7DG could be further tested for the treatment of other neurodegenerative disorders.


Assuntos
Apigenina/uso terapêutico , Retina/efeitos dos fármacos , Animais , Apigenina/metabolismo , Apigenina/farmacologia , Apoptose/efeitos dos fármacos , Eletrorretinografia/efeitos dos fármacos , Inflamação/patologia , Luz/efeitos adversos , Degeneração Macular/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Células Fotorreceptoras de Vertebrados/fisiologia , Substâncias Protetoras/farmacologia , Lesões Experimentais por Radiação/patologia , Retina/metabolismo , Degeneração Retiniana/patologia
9.
J Neuroinflammation ; 13: 50, 2016 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-26920853

RESUMO

BACKGROUND: Photoreceptor death leads to vision impairment in several retinal degenerative disorders. Therapies protecting photoreceptor from degeneration remain to be developed. Anti-inflammation, anti-oxidative stress, and neuroprotective effects of celastrol have been demonstrated in a variety of disease models. The current study aimed to investigate the photoreceptor protective effect of celastrol. METHODS: Bright light-induced retinal degeneration in BALB/c mice was used, and morphological, functional, and molecular changes of retina were evaluated in the absence and presence of celastrol treatment. RESULTS: Significant morphological and functional protection was observed as a result of celastrol treatment in bright light-exposed BALB/c mice. Celastrol treatment resulted in suppression of cell death in photoreceptor cells, alleviation of oxidative stress in the retinal pigment epithelium and photoreceptors, downregulation of retinal expression of proinflammatory genes, and suppression of microglia activation and gliosis in the retina. Additionally, leukostasis was found to be induced in the retinal vasculature in light-exposed BALB/c mice, which was significantly attenuated by celastrol treatment. In vitro, celastrol attenuated all-trans-retinal-induced oxidative stress in cultured APRE19 cells. Moreover, celastrol treatment significantly suppressed lipopolysaccharides-stimulated expression of proinflammatory genes in both APRE19 and RAW264.7 cells. CONCLUSIONS: The results demonstrated for the first time that celastrol prevents against light-induced retinal degeneration through inhibition of retinal oxidative stress and inflammation.


Assuntos
Inflamação/tratamento farmacológico , Luz/efeitos adversos , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Degeneração Retiniana/prevenção & controle , Triterpenos/uso terapêutico , Animais , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Proteína Glial Fibrilar Ácida/metabolismo , Inflamação/induzido quimicamente , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Fármacos Neuroprotetores/farmacologia , Opsinas/metabolismo , Triterpenos Pentacíclicos , Degeneração Retiniana/etiologia , Degeneração Retiniana/patologia , Rodopsina/metabolismo , Triterpenos/farmacologia
10.
Proc Natl Acad Sci U S A ; 111(47): 16883-8, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25385594

RESUMO

Amyotrophic lateral sclerosis (ALS; Lou Gehrig's disease) affects motor neurons (MNs) in the brain and spinal cord. Understanding the pathophysiology of this condition seems crucial for therapeutic design, yet few electrophysiological studies in actively degenerating animal models have been reported. Here, we report a novel preparation of acute slices from adult mouse spinal cord, allowing visualized whole cell patch-clamp recordings of fluorescent lumbar MN cell bodies from ChAT-eGFP or superoxide dismutase 1-yellow fluorescent protein (SOD1YFP) transgenic animals up to 6 mo of age. We examined 11 intrinsic electrophysiologic properties of adult ChAT-eGFP mouse MNs and classified them into four subtypes based on these parameters. The subtypes could be principally correlated with instantaneous (initial) and steady-state firing rates. We used retrograde tracing using fluorescent dye injected into fast or slow twitch lower extremity muscle with slice recordings from the fluorescent-labeled lumbar MN cell bodies to establish that fast and slow firing MNs are connected with fast and slow twitch muscle, respectively. In a G85R SOD1YFP transgenic mouse model of ALS, which becomes paralyzed by 5-6 mo, where MN cell bodies are fluorescent, enabling the same type of recording from spinal cord tissue slices, we observed that all four MN subtypes were present at 2 mo of age. At 4 mo, by which time substantial neuronal SOD1YFP aggregation and cell loss has occurred and symptoms have developed, one of the fast firing subtypes that innvervates fast twitch muscle was lost. These results begin to describe an order of the pathophysiologic events in ALS.


Assuntos
Esclerose Amiotrófica Lateral/fisiopatologia , Neurônios Motores/fisiologia , Medula Espinal/patologia , Superóxido Dismutase/fisiologia , Esclerose Amiotrófica Lateral/enzimologia , Animais , Camundongos , Neurônios Motores/patologia , Técnicas de Patch-Clamp , Superóxido Dismutase-1
11.
Hum Mol Genet ; 23(4): 1056-72, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24105468

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by a severe decline of memory performance. A widely studied AD mouse model is the APPswe/PSEN1ΔE9 (APP/PS1) strain, as mice exhibit amyloid plaques as well as impaired memory capacities. To test whether restoring synaptic plasticity and decreasing ß-amyloid load by Parkin could represent a potential therapeutic target for AD, we crossed APP/PS1 transgenic mice with transgenic mice overexpressing the ubiquitin ligase Parkin and analyzed offspring properties. Overexpression of Parkin in APP/PS1 transgenic mice restored activity-dependent synaptic plasticity and rescued behavioral abnormalities. Moreover, overexpression of Parkin was associated with down-regulation of APP protein expression, decreased ß-amyloid load and reduced inflammation. Our data suggest that Parkin could be a promising target for AD therapy.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/fisiopatologia , Potenciação de Longa Duração , Ubiquitina-Proteína Ligases/metabolismo , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/genética , Animais , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Feminino , Expressão Gênica , Hipocampo/metabolismo , Humanos , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Sinaptofisina/metabolismo , Transcrição Gênica , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
12.
PLoS One ; 8(10): e75823, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24155871

RESUMO

GABA is the major inhibitory neurotransmitter in the mammalian central nervous system that has been strongly implicated in the regulation of sleep. GABA transporter subtype 1 (GAT1) constructs high affinity reuptake sites for GABA and regulates GABAergic transmission in the brain. However, the role of GAT1 in sleep-wake regulation remains elusive. In the current study, we characterized the spontaneous sleep-wake cycle and responses to sleep deprivation in GAT1 knock-out (KO) mice. GAT1 KO mice exhibited dominant theta-activity and a remarkable reduction of EEG power in low frequencies across all vigilance stages. Under baseline conditions, spontaneous rapid eye movement (REM) sleep of KO mice was elevated both during the light and dark periods, and non-REM (NREM) sleep was reduced during the light period only. KO mice also showed more state transitions from NREM to REM sleep and from REM sleep to wakefulness, as well as more number of REM and NREM sleep bouts than WT mice. During the dark period, KO mice exhibited more REM sleep bouts only. Six hours of sleep deprivation induced rebound increases in NREM and REM sleep in both genotypes. However, slow wave activity, the intensity component of NREM sleep was briefly elevated in WT mice but remained completely unchanged in KO mice, compared with their respective baselines. These results indicate that GAT1 plays a critical role in the regulation of REM sleep and homeostasis of NREM sleep.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Privação do Sono/fisiopatologia , Sono REM/fisiologia , Animais , Eletroencefalografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ritmo Teta , Vigília/fisiologia
13.
PLoS One ; 7(6): e39953, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22792139

RESUMO

Mutations in the parkin gene are currently thought to be the most common cause of recessive familial Parkinsonism. Parkin functions as an E3 ligase to regulate protein turnover, and its function in mitochondrial quality control has been reported recently. Overexpression of parkin has been found to prevent neuronal degeneration under various conditions both in vivo and in vitro. Here, we generated a transgenic mouse model in which expression of wild type parkin was driven by neuron-specific enolase (NSE) promoter. We reported that both young and old parkin transgenic mice exhibited less reduction of striatal TH protein and number of TH positive neurons in the substantia nigra induced by 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine (MPTP), compared to wild type littermates. MPTP-induced mitochondrial impairment in the substantia nigra was improved in young parkin transgenic mice. Decreased striatal α-synuclein was demonstrated in old parkin transgenic mice. These results provide reliable evidence from the transgenic mouse model for parkin that overexpression of parkin may attenuate dopaminergic neurodegeneration induced by MPTP through protection of mitochondria and reduction of α-synuclein in the nigrostriatal pathway.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Doenças Neurodegenerativas/induzido quimicamente , Doenças Neurodegenerativas/metabolismo , Transtornos Parkinsonianos/metabolismo , Ubiquitina-Proteína Ligases/biossíntese , Animais , Dopamina/genética , Neurônios Dopaminérgicos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Transtornos Parkinsonianos/genética , Peroxirredoxinas , Fosfopiruvato Hidratase/genética , Fosfopiruvato Hidratase/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteína Desglicase DJ-1 , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo , Transcrição Gênica/efeitos dos fármacos , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
14.
Acta Biochim Biophys Sin (Shanghai) ; 43(6): 448-54, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21558280

RESUMO

In the present study, we tested the effect of cold water stress (CWS) on dopaminergic neurons in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease (PD) mouse model, and found that CWS pretreatment elicited less MPTP neurotoxicity. To understand the molecular mechanism underlying this phenomenon, we detected the expression of heat shock protein 70 (Hsp70) in the striatum of the experimental mice, and found that CWS pretreatment could significantly increase striatal Hsp70 in MPTP-treated mice. Furthermore, in parallel with the induction of Hsp70, the MPTP-induced increase of striatal α-synuclein was inhibited in the CWS + MPTP-treated mice. CWS pretreatment also significantly inhibited the reduction of anti-apoptotic molecule Bcl-2 expression in the striatum and enhanced Bcl-2 transcription in the substantia nigra of MPTP-treated mice. Taken together, these data indicated that Hsp70 might be an important intermediate for the neuroprotective effect of CWS against MPTP-induced dopaminergic toxicity.


Assuntos
Intoxicação por MPTP/fisiopatologia , Síndromes Neurotóxicas/fisiopatologia , Estresse Fisiológico/fisiologia , Animais , Temperatura Baixa , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Dopamina , Proteínas de Choque Térmico HSP70/biossíntese , Imersão , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/biossíntese
15.
Brain Res ; 1302: 256-64, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-19631617

RESUMO

IL-1beta is a potent pro-inflammatory cytokine that regulates neuroinflammation during brain damage. The expression of IL-1beta has been reported to be elevated in the striatum and substantia nigra of patients with Parkinson's disease (PD). Moreover, greater prevalence of PD in men than in women is described previously. Here, by using a sensitive mice model in which the expression of luciferase reporter gene is under the control of human IL-1beta gene promoter, we examined IL-1beta gene expression pattern in vivo after subacute MPTP toxication in old male and female mice and found that MPTP elicited greater dopaminergic toxicity in old male than in female mice. Old male mice showed dramatically elevated luciferase signals in a flexuous manner at early period of time, meanwhile, the changes in female were more moderate. However, no significant difference in astroglial reaction was observed at later time point between sexes. In conclusion, the present study demonstrated that elevated IL-1beta gene expression at early period of time may be in part responsible for the DA neuron susceptibility to MPTP in old male mice.


Assuntos
Envelhecimento/imunologia , Interleucina-1beta/metabolismo , Degeneração Neural/imunologia , Transtornos Parkinsonianos/imunologia , Substância Negra/imunologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Fatores Etários , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Biomarcadores/análise , Biomarcadores/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Feminino , Regulação da Expressão Gênica/imunologia , Marcadores Genéticos/imunologia , Proteína Glial Fibrilar Ácida/análise , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/induzido quimicamente , Gliose/imunologia , Gliose/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-1beta/genética , Luciferases/genética , Masculino , Camundongos , Camundongos Transgênicos , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/patologia , Neurotoxinas/farmacologia , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/fisiopatologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Caracteres Sexuais , Substância Negra/metabolismo , Substância Negra/fisiopatologia , Regulação para Cima/imunologia
16.
Acta Biochim Biophys Sin (Shanghai) ; 40(6): 505-12, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18535749

RESUMO

The newly discovered Cbl-interacting protein of 85 kDa (CIN85) is involved in many cellular processes, but its functions in the brain and in neurodegenerative diseases remain unclear. In this paper, we investigated the distribution of CIN85 protein in different regions of adult mouse brain using Western blot analysis and immunohistochemistry, and found that CIN85 was ubiquitously expressed in mouse brain. In the striatum and substantia nigra, two regions most deeply affected in Parkinson's disease, the level of CIN85 protein was relatively high. In the MPTP mouse model of Parkinson's disease, the expression of CIN85 in the striatum and substantia nigra was complicated. But in 1-methyl-4-phenyl-pyridinium ion-treated human dopaminergic SH-SY5Y cells, the expression of CIN85 increased dramatically. Knocking down of CIN85 by short hairpin RNA reduced SH-SY5Y cell death. Therefore, CIN85 might play different roles in the dopaminergic cell line and in the nigrostriatum of mouse brain under neurotoxin challenge.


Assuntos
1-Metil-4-fenilpiridínio/administração & dosagem , Apoptose/efeitos dos fármacos , Encéfalo/metabolismo , Modelos Animais de Doenças , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Transtornos Parkinsonianos/metabolismo , Proteínas Proto-Oncogênicas c-cbl/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linhagem Celular , Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/patologia , Transtornos Parkinsonianos/induzido quimicamente , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...