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.
STAR Protoc ; 5(1): 102775, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-38085640

RESUMO

The fluorescent non-canonical amino acid tagging (FUNCAT) technique has been used to visualize newly synthesized proteins in cell lines and tissues. Here, we present a protocol for measuring protein synthesis in specific cell types in the mouse brain using in vivo FUNCAT. We describe steps for metabolically labeling newly synthesized proteins with azidohomoalanine, which introduces an azide group into the polypeptide. We then detail procedures for binding a fluorophore-conjugated alkyne to the azide group to allow its visualization. For complete details on the use and execution of this protocol, please refer to tom Dieck et al. (2012)1 and Hooshmandi et al. (2023).2.

2.
Sci Adv ; 9(44): eadh9603, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37922363

RESUMO

Activation of the mechanistic target of rapamycin complex 1 (mTORC1) contributes to the development of chronic pain. However, the specific mechanisms by which mTORC1 causes hypersensitivity remain elusive. The eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) is a key mTORC1 downstream effector that represses translation initiation. Here, we show that nociceptor-specific deletion of 4E-BP1, mimicking activation of mTORC1-dependent translation, is sufficient to cause mechanical hypersensitivity. Using translating ribosome affinity purification in nociceptors lacking 4E-BP1, we identified a pronounced translational up-regulation of tripartite motif-containing protein 32 (TRIM32), an E3 ubiquitin ligase that promotes interferon signaling. Down-regulation of TRIM32 in nociceptors or blocking type I interferon signaling reversed the mechanical hypersensitivity in mice lacking 4E-BP1. Furthermore, nociceptor-specific ablation of TRIM32 alleviated mechanical hypersensitivity caused by tissue inflammation. These results show that mTORC1 in nociceptors promotes hypersensitivity via 4E-BP1-dependent up-regulation of TRIM32/interferon signaling and identify TRIM32 as a therapeutic target in inflammatory pain.


Assuntos
Interferon Tipo I , Nociceptores , Camundongos , Animais , Nociceptores/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Fosfoproteínas/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Interferon Tipo I/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(49): e2308671120, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-38015848

RESUMO

Activation of neuronal protein synthesis upon learning is critical for the formation of long-term memory. Here, we report that learning in the contextual fear conditioning paradigm engenders a decrease in eIF2α (eukaryotic translation initiation factor 2) phosphorylation in astrocytes in the hippocampal CA1 region, which promotes protein synthesis. Genetic reduction of eIF2α phosphorylation in hippocampal astrocytes enhanced contextual and spatial memory and lowered the threshold for the induction of long-lasting plasticity by modulating synaptic transmission. Thus, learning-induced dephosphorylation of eIF2α in astrocytes bolsters hippocampal synaptic plasticity and consolidation of long-term memories.


Assuntos
Astrócitos , Potenciação de Longa Duração , Potenciação de Longa Duração/fisiologia , Plasticidade Neuronal/genética , Hipocampo/fisiologia , Biossíntese de Proteínas , Região CA1 Hipocampal , Memória de Longo Prazo/fisiologia
4.
Neuron ; 111(19): 3028-3040.e6, 2023 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-37473758

RESUMO

Dysregulation of protein synthesis is one of the key mechanisms underlying autism spectrum disorder (ASD). However, the role of a major pathway controlling protein synthesis, the integrated stress response (ISR), in ASD remains poorly understood. Here, we demonstrate that the main arm of the ISR, eIF2α phosphorylation (p-eIF2α), is suppressed in excitatory, but not inhibitory, neurons in a mouse model of fragile X syndrome (FXS; Fmr1-/y). We further show that the decrease in p-eIF2α is mediated via activation of mTORC1. Genetic reduction of p-eIF2α only in excitatory neurons is sufficient to increase general protein synthesis and cause autism-like behavior. In Fmr1-/y mice, restoration of p-eIF2α solely in excitatory neurons reverses elevated protein synthesis and rescues autism-related phenotypes. Thus, we reveal a previously unknown causal relationship between excitatory neuron-specific translational control via the ISR pathway, general protein synthesis, and core phenotypes reminiscent of autism in a mouse model of FXS.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Síndrome do Cromossomo X Frágil , Animais , Camundongos , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/metabolismo , Proteína do X Frágil de Retardo Mental/genética , Neurônios/metabolismo , Fenótipo , Camundongos Knockout , Modelos Animais de Doenças
5.
Brain ; 146(5): 2175-2190, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-36315645

RESUMO

MAPK interacting protein kinases 1 and 2 (Mnk1/2) regulate a plethora of functions, presumably via phosphorylation of their best characterized substrate, eukaryotic translation initiation factor 4E (eIF4E) on Ser209. Here, we show that, whereas deletion of Mnk1/2 (Mnk double knockout) impairs synaptic plasticity and memory in mice, ablation of phospho-eIF4E (Ser209) does not affect these processes, suggesting that Mnk1/2 possess additional downstream effectors in the brain. Translational profiling revealed only a small overlap between the Mnk1/2- and phospho-eIF4E(Ser209)-regulated translatome. We identified the synaptic Ras GTPase activating protein 1 (Syngap1), encoded by a syndromic autism gene, as a downstream target of Mnk1 because Syngap1 immunoprecipitated with Mnk1 and showed reduced phosphorylation (S788) in Mnk double knockout mice. Knockdown of Syngap1 reversed memory deficits in Mnk double knockout mice and pharmacological inhibition of Mnks rescued autism-related phenotypes in Syngap1+/- mice. Thus, Syngap1 is a downstream effector of Mnk1, and the Mnks-Syngap1 axis regulates memory formation and autism-related behaviours.


Assuntos
Transtorno Autístico , Fator de Iniciação 4E em Eucariotos , Animais , Camundongos , Fator de Iniciação 4E em Eucariotos/genética , Camundongos Knockout , Fosforilação , Proteínas Ativadoras de ras GTPase/metabolismo
6.
J Clin Invest ; 133(2)2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36394958

RESUMO

Repeated or prolonged, but not short-term, general anesthesia during the early postnatal period causes long-lasting impairments in memory formation in various species. The mechanisms underlying long-lasting impairment in cognitive function are poorly understood. Here, we show that repeated general anesthesia in postnatal mice induces preferential apoptosis and subsequent loss of parvalbumin-positive inhibitory interneurons in the hippocampus. Each parvalbumin interneuron controls the activity of multiple pyramidal excitatory neurons, thereby regulating neuronal circuits and memory consolidation. Preventing the loss of parvalbumin neurons by deleting a proapoptotic protein, mitochondrial anchored protein ligase (MAPL), selectively in parvalbumin neurons rescued anesthesia-induced deficits in pyramidal cell inhibition and hippocampus-dependent long-term memory. Conversely, partial depletion of parvalbumin neurons in neonates was sufficient to engender long-lasting memory impairment. Thus, loss of parvalbumin interneurons in postnatal mice following repeated general anesthesia critically contributes to memory deficits in adulthood.


Assuntos
Anestesia , Parvalbuminas , Camundongos , Animais , Parvalbuminas/genética , Parvalbuminas/metabolismo , Interneurônios/metabolismo , Neurônios/metabolismo , Células Piramidais/metabolismo , Hipocampo/metabolismo , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/genética , Transtornos da Memória/metabolismo
7.
J Clin Invest ; 132(15)2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35579957

RESUMO

The encoding of noxious stimuli into action potential firing is largely mediated by nociceptive free nerve endings. Tissue inflammation, by changing the intrinsic properties of the nociceptive endings, leads to nociceptive hyperexcitability and thus to the development of inflammatory pain. Here, we showed that tissue inflammation-induced activation of the mammalian target of rapamycin complex 2 (mTORC2) triggers changes in the architecture of nociceptive terminals and leads to inflammatory pain. Pharmacological activation of mTORC2 induced elongation and branching of nociceptor peripheral endings and caused long-lasting pain hypersensitivity. Conversely, nociceptor-specific deletion of the mTORC2 regulatory protein rapamycin-insensitive companion of mTOR (Rictor) prevented inflammation-induced elongation and branching of cutaneous nociceptive fibers and attenuated inflammatory pain hypersensitivity. Computational modeling demonstrated that mTORC2-mediated structural changes in the nociceptive terminal tree are sufficient to increase the excitability of nociceptors. Targeting mTORC2 using a single injection of antisense oligonucleotide against Rictor provided long-lasting alleviation of inflammatory pain hypersensitivity. Collectively, we showed that tissue inflammation-induced activation of mTORC2 causes structural plasticity of nociceptive free nerve endings in the epidermis and inflammatory hyperalgesia, representing a therapeutic target for inflammatory pain.


Assuntos
Dor Crônica , Nociceptores , Humanos , Hiperalgesia/genética , Hiperalgesia/metabolismo , Inflamação/induzido quimicamente , Inflamação/genética , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Nociceptores/fisiologia , Proteína Companheira de mTOR Insensível à Rapamicina/genética , Proteína Companheira de mTOR Insensível à Rapamicina/metabolismo , Sirolimo
8.
Sci Rep ; 11(1): 15490, 2021 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-34326413

RESUMO

Long-lasting cognitive impairment in juveniles undergoing repeated general anesthesia has been observed in numerous preclinical and clinical studies, yet, the underlying mechanisms remain unknown and no preventive treatment is available. We found that daily intranasal insulin administration to juvenile mice for 7 days prior to repeated isoflurane anesthesia rescues deficits in hippocampus-dependent memory and synaptic plasticity in adulthood. Moreover, intranasal insulin prevented anesthesia-induced apoptosis of hippocampal cells, which is thought to underlie cognitive impairment. Inhibition of the mechanistic target of rapamycin complex 1 (mTORC1), a major intracellular effector of insulin receptor, blocked the beneficial effects of intranasal insulin on anesthesia-induced apoptosis. Consistent with this finding, mice lacking mTORC1 downstream translational repressor 4E-BP2 showed no induction of repeated anesthesia-induced apoptosis. Our study demonstrates that intranasal insulin prevents general anesthesia-induced apoptosis of hippocampal cells, and deficits in synaptic plasticity and memory, and suggests that the rescue effect is mediated via mTORC1/4E-BP2 signaling.


Assuntos
Anestesia/efeitos adversos , Insulina/administração & dosagem , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/fisiologia , Memória/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Administração Intranasal , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Fatores de Iniciação em Eucariotos/metabolismo , Medo , Feminino , Hipocampo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Neurológicos , Transdução de Sinais
9.
Cell Rep ; 35(4): 109036, 2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33910008

RESUMO

Recent studies have demonstrated that selective activation of mammalian target of rapamycin complex 1 (mTORC1) in the cerebellum by deletion of the mTORC1 upstream repressors TSC1 or phosphatase and tensin homolog (PTEN) in Purkinje cells (PCs) causes autism-like features and cognitive deficits. However, the molecular mechanisms by which overactivated mTORC1 in the cerebellum engenders these behaviors remain unknown. The eukaryotic translation initiation factor 4E-binding protein 2 (4E-BP2) is a central translational repressor downstream of mTORC1. Here, we show that mice with selective ablation of 4E-BP2 in PCs display a reduced number of PCs, increased regularity of PC action potential firing, and deficits in motor learning. Surprisingly, although spatial memory is impaired in these mice, they exhibit normal social interaction and show no deficits in repetitive behavior. Our data suggest that, downstream of mTORC1/4E-BP2, there are distinct cerebellar mechanisms independently controlling social behavior and memory formation.


Assuntos
Transtorno Autístico/genética , Proteínas de Transporte/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Biossíntese de Proteínas/genética , Células de Purkinje/metabolismo , Memória Espacial/fisiologia , Animais , Humanos , Camundongos
10.
Cell Signal ; 75: 109746, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32858122

RESUMO

Deviations from the optimal level of mRNA translation are linked to disorders with high rates of autism. Loss of function mutations in genes encoding translational repressors such as PTEN, TSC1, TSC2, and FMRP are associated with autism spectrum disorders (ASDs) in humans and their deletion in animals recapitulates many ASD-like phenotypes. Importantly, the activity of key translational control signaling pathways such as PI3K-mTORC1 and ERK is frequently dysregulated in autistic patients and animal models and their normalization rescues many abnormal phenotypes, suggesting a causal relationship. Mutations in several genes encoding proteins not directly involved in translational control have also been shown to mediate ASD phenotypes via altered signaling upstream of translation. This raises the possibility that the dysregulation of translational control signaling is a converging mechanism not only in familiar but also in sporadic forms of autism. Here, we overview the current knowledge on translational signaling in ASD and highlight how correcting the activity of key pathways upstream of translation reverses distinct ASD-like phenotypes.


Assuntos
Transtorno do Espectro Autista/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais , Animais , Regulação da Expressão Gênica , Humanos , PTEN Fosfo-Hidrolase/metabolismo , Serina-Treonina Quinases TOR/metabolismo
11.
Cell Rep ; 29(11): 3620-3635.e7, 2019 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-31825840

RESUMO

The translation initiation repressor 4E-BP2 is deamidated in the brain on asparagines N99/N102 during early postnatal brain development. This post-translational modification enhances 4E-BP2 association with Raptor, a central component of mTORC1 and alters the kinetics of excitatory synaptic transmission. We show that 4E-BP2 deamidation is neuron specific, occurs in the human brain, and changes 4E-BP2 subcellular localization, but not its disordered structure state. We demonstrate that deamidated 4E-BP2 is ubiquitinated more and degrades faster than the unmodified protein. We find that enhanced deamidated 4E-BP2 degradation is dependent on Raptor binding, concomitant with increased association with a Raptor-CUL4B E3 ubiquitin ligase complex. Deamidated 4E-BP2 stability is promoted by inhibiting mTORC1 or glutamate receptors. We further demonstrate that deamidated 4E-BP2 regulates the translation of a distinct pool of mRNAs linked to cerebral development, mitochondria, and NF-κB activity, and thus may be crucial for postnatal brain development in neurodevelopmental disorders, such as ASD.


Assuntos
Fatores de Iniciação em Eucariotos/metabolismo , NF-kappa B/metabolismo , Neurônios/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína Regulatória Associada a mTOR/metabolismo , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Células Cultivadas , Proteínas Culina/metabolismo , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Proteólise
12.
Pharmacol Biochem Behav ; 158: 39-48, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28583577

RESUMO

Herein the effect of hippocampal orexin type-1 receptors (OX1Rs) blockade on morphine withdrawal syndrome was studied. Animals were made dependent by subcutaneous (s.c.) administration of morphine sulfate (10mg/kg) at an interval of 12h for 9 consecutive days. Thereafter, on day 10, naloxone hydrochloride (1.5mg/kg, i.p.) was injected and the somatic signs of withdrawal syndrome were monitored during a 25-min period. Two groups of animals received bilateral microinjection of either SB-334867, a selective OX1Rs antagonist, (0.5µg/0.5µl), or its vehicle into the dorsal hippocampus immediately before each morphine injection. Other groups of animals were made dependent at first and only received a single microinjection of SB-334867 or vehicle on day 10 before naloxone injection. The results showed that intra-hippocampal microinjection of SB-334867 before each morphine treatment, significantly decreased the signs of morphine withdrawal, including teeth chattering (dependent: 18.5±2.3, SB treated: 5±1, p<0.001), diarrhea (dependent: 8.7±0.6, SB treated: 4.1±0.6, p<0.001), ptosis (dependent: 33.8±3.7, SB treated: 11.6±1.1, p<0.001), and chewing (dependent: 40±2.3, SB treated: 29±2.4, p<0.01). SB-334867 did not attenuate withdrawal syndrome, when it was microinjected as a single dose immediately before naloxone injection. The present results suggest a role for orexin in naloxone-precipitated withdrawal and thus possibly morphine dependence and this effect is, at least in part, via OX1Rs in the dorsal hippocampus.


Assuntos
Benzoxazóis/farmacologia , Hipocampo/efeitos dos fármacos , Morfina/efeitos adversos , Naloxona/farmacologia , Antagonistas dos Receptores de Orexina/farmacologia , Receptores de Orexina/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/etiologia , Ureia/análogos & derivados , Animais , Benzoxazóis/administração & dosagem , Hipocampo/metabolismo , Masculino , Microinjeções , Naftiridinas , Antagonistas dos Receptores de Orexina/administração & dosagem , Ratos , Ratos Wistar , Ureia/administração & dosagem , Ureia/farmacologia
13.
Cell J ; 19(1): 102-116, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28367421

RESUMO

OBJECTIVE: Spinal cord injury (SCI) causes inflammation, deformity and cell loss. It has been shown that Melissa officinalis (MO), as herbal medicine, and dexamethasone (DEX) are useful in the prevention of various neurological diseases. The present study evaluated combinational effects of DEX and MO on spinal cord injury. MATERIALS AND METHODS: Thirty six adult male Wistar rats were used in this experimental study. The weight-drop contusion method was employed to induce spinal cord injury in rats. DEX and MO were administrated alone and together in different treatment groups. Intra-muscular injection of DEX (1 mg/kg) was started three hours after injury and continued once a day for seven days after injury. Intra-peritoneal (I.P) injection of MO (150 mg/ kg) was started one day after injury and continued once a day for 14 days. RESULTS: Our results showed motor and sensory functions were improved significantly in the group received a combination of DEX and MO, compared to spinal cord injury group. Mean cavity area was decreased and loss of lower motor neurons and astrogliosis in the ventral horn of spinal cord was significantly prevented in the group received combination of DEX and Melissa officinalis, compared to spinal cord injury group. Furthermore, the findings showed a significant augmentation of electromyography (EMG) recruitment index, increase of myelin diameter, and up-regulation of myelin basic protein in the treated group with combination of DEX and MO. CONCLUSION: Results showed that combination of DEX and MO could be considered as a neuroprotective agent in spinal cord injury.

14.
ASN Neuro ; 8(6)2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27815336

RESUMO

INTRODUCTION: The pathophysiology of spinal cord injury (SCI) has a classically bad prognosis. It has been demonstrated that human umbilical cord blood stem cells (hUCBSCs) and Melissa officinalis (MO) are useful for the prevention of neurological disease. METHODS: Thirty-six adult male rats were randomly divided into intact, sham, control (SCI), MO, hUCBSC, and MO-hUCBSC groups. Intraperitoneal injection of MO (150 mg/kg) was commenced 24 hr post-SCI and continued once a day for 14 days. Intraspinal grafting of hUCBSCs was commenced immediately in the next day. The motor and sensory functions of all animals were evaluated once a week after the commencement of SCI. Electromyography (EMG) was performed in the last day in order to measure the recruitment index. Immunohistochemistry, reverse transcription-polymerase chain reaction, and transmission electron microscopy evaluations were performed to determine the level of astrogliosis and myelination. RESULTS: The results revealed that motor function (MO-hUCBSC: 15 ± 0.3, SCI: 8.2 ± 0.37, p < .001), sensory function (MO-hUCBSC: 3.57 ± 0.19, SCI: 6.38 ± 0.23, p < .001), and EMG recruitment index (MO-hUCBSC: 3.71 ± 0.18, SCI: 1.6 ± 0.1, p < .001) were significantly improved in the MO-hUCBSC group compared with SCI group. Mean cavity area (MO-hUCBSC: 0.03 ± 0.03, SCI: 0.07 ± 0.004, p < .001) was reduced and loss of lower motor neurons (MO-hUCBSC: 7.6 ± 0.43, SCI: 3 ± 0.12, p < .001) and astrogliosis density (MO-hUCBSC: 3.1 ± 0.15, SCI: 6.25 ± 1.42, p < 0.001) in the ventral horn of spinal cord were prevented in MO-hUCBSC group compared with SCI group. CONCLUSION: The results revealed that the combination of MO and hUCBSCs in comparison with the control group has neuroprotective effects in SCI.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical , Melissa/química , Fármacos Neuroprotetores/uso terapêutico , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/cirurgia , Animais , Antígenos CD/metabolismo , Bromodesoxiuridina/metabolismo , Modelos Animais de Doenças , Eletromiografia , Potencial Evocado Motor/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Masculino , Melissa/fisiologia , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , Exame Neurológico , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Wistar , Medula Espinal/metabolismo , Medula Espinal/patologia , Medula Espinal/ultraestrutura , Fatores de Tempo
15.
Data Brief ; 9: 338-44, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27672673

RESUMO

In this dataset, we analyzed galvanic-evoked head movements (GEHMs) in the spatial planes of yaw, and roll in normal and unilaterally labyrinthectomized (UL) Wistar rats. The rats were assigned in 4 groups of 10: control, sham, right-UL and left-UL. Bilateral galvanic vestibular stimulation (GVS) was presented by our "ring-shaped electrode" design (see "Short-term galvanic vestibular stimulation promotes functional recovery and neurogenesis in unilaterally labyrinthectomized rats" (M. Shaabani et al., 2016) [1]). Required data were collected through video recording of GEHMs followed by image processing and statistical analysis.

16.
Brain Res ; 1648(Pt A): 152-162, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27444558

RESUMO

Current experimental research on the therapeutic effects of galvanic vestibular stimulation (GVS) has mainly focused on neurodegenerative disorders. However, it primarily stimulates the vestibular nuclei and could be potentially effective in modulating imbalance between them in the case of unilateral labyrinthectomy (UL). Fifty male Wistar rats (180-220g) were used in 5 groups of 10: intact, sham, right-UL (RUL; without intervention), and two other right-UL groups with GVS intervention [one group treated with low rate GVS (GVS.LF; 6-7Hz), and the other treated with high rate GVS (GVS.HF; 17-18Hz)]. The UL models were prepared by intratympanic injection of sodium arsanilate. GVS protocols were implemented 30min/day and continued for 14 days via ring-shaped copper electrodes inserted subcutaneously over each mastoid. Functional recovery was assessed by several postural tests including support surface area, landing and air-righting reflexes, and rotarod procedure. Immunohistochemical investigations were performed on ipsi- and contra-lesional medial vestibular nuclei (MVN) using bromodeoxyuridine (BrdU) and Ki67, as markers of cell proliferation. Behavioral evaluations showed significant functional recovery of GVS-treated groups compared to RUL group. The percent of marked cells with BrdU and Ki67 were significantly higher in the ipsilesional MVN of both GVS-treated groups compared with other groups. Our findings confirmed the effectiveness of GVS-intervention in accelerating static and dynamic vestibular compensation. This could be explained by the cell proliferation in ipsilesional MVN cells and rapid rebalancing of the VNs and the modulation of their motor outputs. Therefore, GVS could be promising for rehabilitating patients with unilateral vestibular weakness.


Assuntos
Estimulação Elétrica/métodos , Vestíbulo do Labirinto/fisiologia , Animais , Bromodesoxiuridina/farmacologia , Masculino , Neurogênese/fisiologia , Ratos , Ratos Wistar , Recuperação de Função Fisiológica/fisiologia , Sensação , Núcleos Vestibulares/fisiologia , Vestíbulo do Labirinto/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...