Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Cell Biol Int ; 48(4): 461-472, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38196274

RESUMO

The rapid rise in propofol dependency and abuse has highlighted limited resources for addressing substance abuse-related cognitive impairment, prompting the development of novel therapies. Dysregulated autophagy flow accelerates neuronal cell death, and interventions countering this dysregulation offer an appealing strategy for neuronal protection. Curcumin, a potent natural polyphenol derived from turmeric rhizomes, is renowned for its robust antineurotoxic properties and enhanced cognitive function. Utilizing CCK-8 and Ki67 fluorescent staining, our study revealed that curcumin treatment increased cell viability and proliferative potential in MN9D cells exposed to propofol-induced neurotoxicity. Furthermore, enzyme-linked immunosorbent assay and western blot analysis demonstrated the partial restoration of dopamine synthesis, secretion levels, and TH expression in damaged MN9D cells treated with curcumin. Scanning electrode microscope images displayed reduced autolysosomes and phagosomes in curcumin-treated cells compared to the propofol group. Immunoblotting revealed that curcumin mitigated the degradation of LC3I to LC3II and p62 induced by propofol stimulation, with green fluorescence expression of LC3 postcurcumin treatment resembling that following autophagy inhibitor HCQ treatment, indicating that modulating autophagy flow can alleviate propofol's toxic effects. Moreover, curcumin treatment upregulated the Akt/mTOR/p70S6K signaling pathway, suggesting that curcumin potentially curtails autophagy dysregulation in nerve cells by activating Akt/mTOR/p70S6K. In conclusion, our findings suggest that curcumin can ameliorate propofol abuse-induced neurotoxicity, partially through autophagy regulation and Akt/mTOR/p70S6K signaling activation.


Assuntos
Curcumina , Propofol , Proteínas Proto-Oncogênicas c-akt/metabolismo , Curcumina/farmacologia , Propofol/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Autofagia
2.
Dev Neurosci ; 43(2): 84-94, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34130286

RESUMO

Minichromosome maintenance protein 5 (MCM5), a member of the microchromosomal maintenance protein family, plays an important role in the initiation and extension of DNA replication. However, its role in neural development in zebrafish remains unclear. Here, we used morpholino (MO) and CRISPR/Cas9 to knock down mcm5 and investigated the developmental features of facial motor neurons (FMNs) in the hindbrain of zebrafish. We found that knockdown of mcm5 using mcm5 MO resulted in a small head, small eyes, and a blurred midbrain-hindbrain boundary, while MO injection of mcm5 led to decrease in FMNs and their migration disorder. However, the mutant of mcm5 only resulted in the migration defect of FMNs rather than quantity change. We further investigated the underlying mechanism of mcm5 in the development of hindbrain using in situ hybridization (ISH) and fgfr1a mRNA co-injected with mcm5 MO. Results from ISH showed that the fibroblast growth factor (FGF) signaling pathway was changed when the MCM5 function was lost, with the decrease in fgfr1a and the increase in fgf8, while that of pea3 had opposite trend. FMN development defects were rescued by fgfr1a mRNA co-injected with mcm5 MO. Our results demonstrated that FGF signaling pathway is required for FMN development in zebrafish. Specifically, mcm5 regulates FMN development during zebrafish growing.


Assuntos
Fatores de Crescimento de Fibroblastos , Peixe-Zebra , Animais , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Neurônios Motores/metabolismo , Transdução de Sinais , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
3.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 50(2): 164-170, 2019 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-31106533

RESUMO

OBJECTIVE: To explore the effect and mechanism of human adipose-derived mesenchymal stem cells (hADMSCs) on phenotypic polarization of microglia. METHODS: BV-2 microglia of C57/BL6 mice were co-cultured with hADMSCs+lipopolysaccharide (LPS), or cultured with LPS alone. Cell morphology was observed under an inverted microscope. The effect of hADMSCs on microglial proliferation was evaluated by CCK-8 assay. The impact of hADMSCs on microglia M1/M2 phenotype markers were detected using quantitative real-time PCR (RT-qPCR). The affect of hADMSCs on the proteins expression levels of Toll-like receptor 4 (TLR4)-TIR domain containing adaptor protein inducing interferon ß (TRIF) signaling pathway in BV-2 microglia was detected by using Western blot analysis. RESULTS: As compared with the LPS treatment, hADMSCs treatment had no obvious effect on microglia morphology, whereas showed significant inhibition on microglial proliferation activity (P<0.05). Simultaneously, hADMSCs treatment reduced expression of microglia M1 phenotype markers (P<0.05), and increased microglia M1 phenotype markers in gene levels (P<0.05). At the same time, protein expression levels of TRIF, TLR4, phosphorylated interferon regulatory factor 3 (P-IRF3) and interferon regulatory factor 3 (IRF3) in BV-2 microglia were decreased after hADMSCs treatment. CONCLUSION: hADMSCs can blockade the LPS-induced pro-inflammatory microglia M1 phenotype, whereas induces protective microglial M2 phenotype, which may be related to inhibition of the TLR4-TRIF signaling pathway.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Polaridade Celular , Células-Tronco Mesenquimais/citologia , Microglia/citologia , Transdução de Sinais , Receptor 3 Toll-Like/metabolismo , Tecido Adiposo/citologia , Animais , Células Cultivadas , Humanos , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
4.
J Neuroinflammation ; 15(1): 81, 2018 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-29544518

RESUMO

BACKGROUND: Antigen-specific and MHCII-restricted CD4+ αß T cells have been shown or suggested to play an important role in the transition from acute to chronic mechanical allodynia after peripheral nerve injuries. However, it is still largely unknown where these T cells infiltrate along the somatosensory pathways transmitting mechanical allodynia to initiate the development of chronic mechanical allodynia after nerve injuries. Therefore, the purpose of this study was to ascertain the definite neuroimmune interface for these T cells to initiate the development of chronic mechanical allodynia after peripheral nerve injuries. METHODS: First, we utilized both chromogenic and fluorescent immunohistochemistry (IHC) to map αß T cells along the somatosensory pathways for the transmission of mechanical allodynia after modified spared nerve injuries (mSNIs), i.e., tibial nerve injuries, in adult male Sprague-Dawley rats. We further characterized the molecular identity of these αß T cells selectively infiltrating into the leptomeninges of L4 dorsal roots (DRs). Second, we identified the specific origins in lumbar lymph nodes (LLNs) for CD4+ αß T cells selectively present in the leptomeninges of L4 DRs by two experiments: (1) chromogenic IHC in these lymph nodes for CD4+ αß T cell responses after mSNIs and (2) fluorescent IHC for temporal dynamics of CD4+ αß T cell infiltration into the L4 DR leptomeninges after mSNIs in prior lymphadenectomized or sham-operated animals to LLNs. Finally, following mSNIs, we evaluated the effects of region-specific targeting of these T cells through prior lymphadenectomy to LLNs and chronic intrathecal application of the suppressive anti-αßTCR antibodies on the development of mechanical allodynia by von Frey hair test and spinal glial or neuronal activation by fluorescent IHC. RESULTS: Our results showed that during the sub-acute phase after mSNIs, αß T cells selectively infiltrate into the leptomeninges of the lumbar DRs along the somatosensory pathways responsible for transmitting mechanical allodynia. Almost all these αß T cells are CD4 positive. Moreover, the temporal dynamics of CD4+ αß T cell infiltration into the lumbar DR leptomeninges are specifically determined by LLNs after mSNIs. Prior lymphadenectomy to LLNs specifically reduces the development of mSNI-induced chronic mechanical allodynia. More importantly, intrathecal application of the suppressive anti-αßTCR antibodies reduces the development of mSNI-induced chronic mechanical allodynia. In addition, prior lymphadenectomy to LLNs attenuates mSNI-induced spinal activation of glial cells and PKCγ+ excitatory interneurons. CONCLUSIONS: The noteworthy results here provide the first evidence that CD4+ αß T cells selectively infiltrate into the DR leptomeninges of the somatosensory pathways transmitting mechanical allodynia and contribute to the transition from acute to chronic mechanical allodynia after peripheral nerve injuries.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Hiperalgesia/etiologia , Hiperalgesia/patologia , Meninges/fisiopatologia , Raízes Nervosas Espinhais/patologia , Neuropatia Tibial/complicações , Animais , Movimento Celular , Modelos Animais de Doenças , Região Lombossacral , Masculino , Infiltração de Neutrófilos/fisiologia , Medição da Dor , Limiar da Dor/fisiologia , Fosfopiruvato Hidratase/metabolismo , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Fatores de Tempo
5.
Bioessays ; 36(3): 293-304, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24464475

RESUMO

Disruption of Nodal in the lateral plate mesoderm (LPM) usually leads to left-right (LR) patterning defects in multiple organs. However, whether the LR patterning of organs is always regulated in a coupled way has largely not yet been elucidated. In addition, whether other crucial regulators exist in the LPM that coordinate with Nodal in regulating organ LR patterning is also undetermined. In this paper, after briefly summarizing the common process of LR patterning, the most puzzling question regarding the initiation of asymmetry is considered and the divergent mechanisms underlying the uncoupled LR patterning in different organs are discussed. On the basis of cases in which different organ LR patterning is determined in an uncoupled way via an independent mechanism or at a different time, we propose that there are other critical factors in the LPM that coordinate with Nodal to regulate heart LR asymmetry patterning during early LR patterning.


Assuntos
Padronização Corporal , Organogênese , Animais , Humanos , Proteína Nodal/metabolismo
6.
Front Pharmacol ; 15: 1385330, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38860164

RESUMO

Liver fibrosis occurs in response to chronic damage and inflammation to the liver. Leaving untreated, it can lead to decreased liver function and can eventually progress to cirrhosis, a more advanced and irreversible state of liver damage. Clinical investigations showed that chronic liver disease associated with neurological symptoms including anxiety, depression, and cognitive decline. However, few therapeutic options are available for treating liver and related brain pathologies simultaneously. In this study, we aim to find therapeutic candidates that target the liver-brain axis. Gossypetin, a flavonoid from sedum, shows promising capability in treating liver and brain pathologies in CCl4-induced mouse model. Short term of gossypetin administration is sufficient to ameliorate impaired liver function and pre-existing liver fibrosis, suppress MKK3/6-p38 MAPK and p53 activation, and abolish the activation of hepatic stellate cells and Kupffer cells. Although we observe no neuronal loss in the brain of mice with liver fibrosis, we do observe astrogliosis and microglial activation in certain brain regions, especially the hippocampus. Brief gossypetin administration also shows potential in alleviating neuroinflammation in these regions. These results suggest that gossypetin can target the liver-brain axis and be a promising candidate for treating chronic liver fibrosis patients with neurological symptoms.

7.
J Neuroinflammation ; 9: 39, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22361049

RESUMO

BACKGROUND: TIR-domain-containing adapter-inducing interferon-ß (TRIF) is the sole downstream adaptor of Toll-like receptor (TLR)3, which is one of the major signaling pathways in immune cells leading to neuroinflammation in the central nervous system. Overexpression of TRIF may lead to activation of inflammatory responses, and contribute to pathophysiological progression in both acute and chronic neurodegenerative retinal diseases. In the present study, was aimed to elucidate the contributions of TRIF to optic nerve (ON) regeneration and retinal ganglion cell (RGC) survival following injury to the ON, a widely studied model of central nervous system injury and of degenerative diseases such as glaucoma. METHODS: We used retrograde labeling with a fluorochrome, hydroxystilbamidine (Fluorogold) to evaluate RGC survival, and immunostaining with growth-associated protein-43 to evaluate axon regeneration in an ON crush model. Changes in microglial cytokines following RGC injury was examined with ELISA and real-time PCR. In vivo studies were carried out in wild-type and trif-/- mice. A Transwell co-culture system and migration test were used to mimic the crosstalk between microglia and RGCs. TRIF-associated downstream adaptors were determined by western blotting. RESULTS: Compared with wild-type (WT) mice, TRIF knockout (KO) mice displayed a robust ability to regenerate axons 3 or 7 days after nerve injury. In addition, RGC survival was considerably higher in trif-/- than in WT mice. ON lesion induced less microglial activation in trif-/- than in WT mice. and more WT microglia distorted and migrated toward the foramen opticum. In the transwell system, few trif-/- microglia migrated through the membrane when stimulated by the performed lesion on RGC axons in a transwell system. Inactivation of microglial cells in trif-/- mice was associated with reduced production of inflammatory cytokines, as detected with real-time RT-PCR and ELISA. Furthermore western blot analysis showed that activation of known downstream effectors of TRIF, including TBK1, IKKε and NF-κB, were significantly inhibited by TRIF deficiency. CONCLUSION: Our results indicate that TRIF deficiency promotes ON axon regeneration by attenuating microglial activation and consequently reducing the release of harmful cytokines via NF-κB inactivation.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/deficiência , Microglia/metabolismo , Regeneração Nervosa/genética , Doenças do Nervo Óptico/patologia , Células Ganglionares da Retina/fisiologia , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/métodos , Proteína GAP-43/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regeneração Nervosa/fisiologia , Células Ganglionares da Retina/patologia , Estilbamidinas
8.
J Neuroinflammation ; 9: 46, 2012 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-22394415

RESUMO

BACKGROUND: Inflammatory injury plays a critical role in intracerebral hemorrhage (ICH)-induced neurological deficits; however, the signaling pathways are not apparent by which the upstream cellular events trigger innate immune and inflammatory responses that contribute to neurological impairments. Toll-like receptor 4 (TLR4) plays a role in inflammatory damage caused by brain disorders. METHODS: In this study, we investigate the role of TLR4 signaling in ICH-induced inflammation. In the ICH model, a significant upregulation of TLR4 expression in reactive microglia has been demonstrated using real-time RT-PCR. Activation of microglia was detected by immunohistochemistry, cytokines were measured by ELISA, MyD88, TRIF and NF-κB were measured by Western blot and EMSA, animal behavior was evaluated by animal behavioristics. RESULTS: Compared to WT mice, TLR4(-/-) mice had restrained ICH-induced brain damage showing in reduced cerebral edema and lower neurological deficit scores. Quantification of cytokines including IL-6, TNF-α and IL-1ß and assessment of macrophage infiltration in perihematoma tissues from TLR4(-/-), MyD88(-/-) and TRIF(-/-) mice showed attenuated inflammatory damage after ICH. TLR4(-/-) mice also exhibited reduced MyD88 and TRIF expression which was accompanied by decreased NF-κB activity. This suggests that after ICH both MyD88 and TRIF pathways might be involved in TLR4-mediated inflammatory injury possibly via NF-κB activation. Exogenous hemin administration significantly increased TLR4 expression and microglial activation in cultures and also exacerbated brain injury in WT mice but not in TLR4(-/-) mice. Anti-TLR4 antibody administration suppressed hemin-induced microglial activation in cultures and in the mice model of ICH. CONCLUSIONS: Our findings suggest that heme potentiates microglial activation via TLR4, in turn inducing NF-κB activation via the MyD88/TRIF signaling pathway, and ultimately increasing cytokine expression and inflammatory injury in ICH. Targeting TLR4 signaling may be a promising therapeutic strategy for ICH.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Hemorragia Cerebral/complicações , Heme/farmacologia , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo , Regulação para Cima/efeitos dos fármacos , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais/uso terapêutico , Células Cultivadas , Córtex Cerebral/citologia , Hemorragia Cerebral/tratamento farmacológico , Citocinas/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Inflamação/etiologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/efeitos dos fármacos , Fator 88 de Diferenciação Mieloide/deficiência , NF-kappa B/metabolismo , Exame Neurológico , RNA Mensageiro/metabolismo , Estatísticas não Paramétricas , Fatores de Tempo , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Regulação para Cima/genética , Regulação para Cima/fisiologia
9.
Synapse ; 66(10): 849-57, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22623226

RESUMO

Hippocampus is one of the most important structures that mediates learning and memory, cognition, and mental behaviors and profoundly regulated by sex hormones in a sex-specific manner, but the mechanism of underlying sex differences regulation is still unclear. We have previously reported that in the male and female mice, steroid receptor coactivator-1 (SRC-1) and some key synaptic proteins share similar developmental profile in the hippocampus, but how circulating sex hormones affect hippocampal SRC-1 as well as these synaptic proteins remain unclear. In this study, we examined how gonad sex hormones regulate hippocampal SRC-1, synaptophysin, PSD-95, and AMPA receptor subtype GluR1 by using immunohistochemistry and Western blot. The results showed that in the female mice, ovariectomy affected hippocampal SRC-1 and GluR1 were only detected at 2 weeks post operation, then it recovered to sham level; synaptophysin was unaffected at any timepoint examined; significant decrease of PSD-95 was only detected at 4 weeks post operation. However, in the male hippocampus, SRC-1 and PSD-95 were decreased from one week and lasted to 4 weeks after orchidectomy, GluR1 decreased from 2 weeks after orchidectomy, but synaptophysin remained unchanged as in the females. Correlation analysis showed the profiles of SRC-1 were positively correlated with GluR1 of the females, PSD-95 and GluR1 of the males, respectively. The above results suggested a distinct regulatory mode between female and male gonad hormones in the regulation of hippocampal SRC-1 and synaptic proteins, which may be one of the mechanisms contributing to the dimorphism of hippocampus during development and ageing.


Assuntos
Hormônios Esteroides Gonadais/deficiência , Hipocampo/química , Coativador 1 de Receptor Nuclear/análise , Sinapses/química , Animais , Western Blotting , Proteína 4 Homóloga a Disks-Large , Feminino , Guanilato Quinases/análise , Imuno-Histoquímica , Masculino , Proteínas de Membrana/análise , Camundongos , Camundongos Endogâmicos C57BL , Orquiectomia , Ovariectomia , Receptores de AMPA/análise , Fatores Sexuais , Sinaptofisina/análise
10.
Biomolecules ; 12(2)2022 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-35204787

RESUMO

Minichromosome maintenance protein 5 (MCM5) is a critical cell cycle regulator; its role in DNA replication is well known, but whether it is involved in the regulation of organogenesis in a cell cycle-independent way, is far from clear. In this study, we found that a loss of mcm5 function resulted in a mildly smaller liver, but that mcm5 overexpression led to liver bifida. Further, the data showed that mcm5 overexpression delayed endodermal migration in the ventral-dorsal axis and induced the liver bifida. Cell cycle analysis showed that a loss of mcm5 function, but not overexpression, resulted in cell cycle delay and increased cell apoptosis during gastrulation, implying that liver bifida was not the result of a cell cycle defect. In terms of its mechanism, our data proves that mcm5 represses the expression of cxcr4a, which sequentially causes a decrease in the expression of itgb1b during gastrulation. The downregulation of the cxcr4a-itgb1b cascade leads to an endodermal migration delay during gastrulation, as well as to the subsequent liver bifida during liver morphogenesis. In conclusion, our results suggest that in a cell cycle-independent way, mcm5 works as a gene expression regulator, either partially and directly, or indirectly repressing the expression of cxcr4a and the downstream gene itgb1b, to coordinate endodermal migration during gastrulation and liver location during liver organogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Ciclo Celular , Replicação do DNA , Receptores CXCR4 , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Ciclo Celular , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Divisão Celular , Receptores CXCR4/metabolismo , Transdução de Sinais
11.
Dev Neurosci ; 33(1): 57-63, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21228553

RESUMO

Female steroids such as estrogens and progestins, through their nuclear receptors, play important roles in regulation of the structure and function of the hippocampus. Steroid receptor coactivator-1 (SRC-1) has been detected in embryonic and/or adult hippocampus of rodents, and SRC-1 null mice showed significantly longer escape latency in the Morris maze test, indicating a role of this coactivator in the regulation of hippocampus function. Whether this is regulated by development and circulating ovary hormones remains unclear. In this study, postnatal development and ovariectomy for regulation of hippocampal SRC-1 in female rats were investigated by Western blot and immunohistochemistry. The results showed that SRC-1-immunopositive materials were predominantly detected in the CA1 pyramidal cell layer and dentate gyrus granular cell layer. Very low levels of SRC-1 were detected at postnatal day 0, but they increased with development. The highest levels of SRC-1 were detected at postnatal day 14, then they decreased to adult levels from postnatal day 30; significantly lower levels of SRC-1 were detected in the middle-aged (18-month-old) hippocampus when compared with that of the adult. Western blot and immunohistochemistry demonstrated that hippocampal SRC-1 expression was unchanged after ovariectomy, no significant differences were noticed from day 3 to 8 weeks postsurgery when compared with sham animals. The above results showed that hippocampal SRC-1 is regulated by postnatal development but not ovariectomy, and that the exact role of SRC-1 in the estradiol regulation of hippocampus needs further investigation.


Assuntos
Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Coativador 1 de Receptor Nuclear/metabolismo , Ovariectomia , Animais , Estrogênios/metabolismo , Feminino , Hipocampo/citologia , Camundongos , Ratos , Ratos Sprague-Dawley
12.
J Toxicol Sci ; 45(7): 391-399, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32612007

RESUMO

This study was aimed at examining propofol- (a known anesthetic) induced emotion-related behavioral disorders in mice, and exploring the possible molecular mechanisms. A total of 60 mice were divided into two groups: control and propofol group. Mice were injected with propofol (150 mg/kg, ip) at 8:00 a.m. (once a day, lasting for 30 days). During the 30 days, loss of righting reflex (LORR) and return of righting reflex (RORR) of mice were recorded every day. At the 1st (T1) and 30th (T2) day of drug discontinuance (T2), 15 mice of each group were selected to perform the open field test; then the mice underwent perfusion fixation, and the midbrain and corpus striatum were separated for immunofluorescence assay with anti-tyrosine hydroxylase (Th) and anti- dopamine transporter (DAT) antibodies. Results showed that after propofol injection, LORR and RORR increased and decreased, respectively. Long-term use of propofol resulted in decreased activities of mice (activity trajectory, line crossing, rearing time, scratching times and defecating frequency). Immunofluorescence assay showed long-term use of propofol induced decrease of Th and DAT. Collectively, our present work suggested long-term abuse of propofol induces neuropsychiatric function impairments, and the possible mechanisms are related to dopamine dyssynthesis via down-regulating tyrosine hydroxylase and dopamine transporter.


Assuntos
Anestésicos/toxicidade , Comportamento Animal/efeitos dos fármacos , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Neurônios Dopaminérgicos/patologia , Transtornos Mentais/induzido quimicamente , Propofol/toxicidade , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/patologia , Tirosina 3-Mono-Oxigenase/metabolismo , Área Tegmentar Ventral/metabolismo , Área Tegmentar Ventral/patologia , Anestésicos/efeitos adversos , Animais , Neurônios Dopaminérgicos/metabolismo , Emoções/efeitos dos fármacos , Masculino , Transtornos Mentais/metabolismo , Transtornos Mentais/fisiopatologia , Transtornos Mentais/psicologia , Camundongos Endogâmicos C57BL , Propofol/efeitos adversos , Reflexo de Endireitamento/efeitos dos fármacos
13.
Neurosci Lett ; 450(2): 106-10, 2009 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-19041369

RESUMO

Low dose total-body gamma-irradiation (TBI) was reported to confer neuroprotection against MPTP-induced dopaminergic neurotoxicity. After being pretreated with a single low dose (0.5Gy, 2.0Gy or 3.5Gy) TBI, C57BL/6 mice were administered with MPTP (15mg/kg, four times, 2h apart) intraperitoneally (i.p.). In the group pretreated with 2.0Gy TBI, with lower lymphocytes number, neuroprotection was found by High Performance Liquid Chromatography (HPLC) determination of the striatal dopamine. Contrarily, in the group pretreated with 0.5Gy TBI, with higher lymphocytes number, dopaminergic neuron toxicity was enhanced. So it was probably the decrease of lymphocytes, not the radiation hormesis that rendered the potential neuroprotection. And it was the balance between radiation injury and lymphocytopenia neuroprotection that decided the effect of low dose gamma-irradiation on MPTP-induced dopaminergic neurotoxicity.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/administração & dosagem , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Neurotoxinas/administração & dosagem , Irradiação Corporal Total/métodos , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/efeitos da radiação , Cromatografia Líquida de Alta Pressão/métodos , Dopamina/metabolismo , Relação Dose-Resposta à Radiação , Ácido Homovanílico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Síndromes Neurotóxicas/patologia , Tirosina 3-Mono-Oxigenase/metabolismo
14.
Diagn Pathol ; 14(1): 94, 2019 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-31439011

RESUMO

BACKGROUND: Castleman's disease (CD) is an uncommon type of benign proliferation of the lymphoid tissue, characterized by local or systemic lymphadenopathy that most frequently appears in the mediastinum; involvement of the kidney is uncommon, and proliferation originating from the kidney is extremely rare. Herein, we report a rare case of hyaline vascular Castleman's disease (HV-CD) in a 56-year-old male patient and discuss its morphological characteristics and differential diagnoses including mantle cell lymphoma (MCL), follicular lymphoma (FL), and nodal marginal zone lymphoma (NMZL). CASE PRESENTATION: A right upper-middle renal mass was detected after physical examination in a 56-year-old man without any clinical symptoms and a previous partial resection of the right kidney. Microscopically, the lymphoid follicles were increased in number and had expanded mantle zones and atrophic germinal centers. Vascular proliferation and hyalinization in the interfollicular zones were observed. Immunohistochemical staining showed CD20-positive cells in the mantle zones; CD21 and CD35 were expressed in the dendritic cells, CD3 was positive in a small number of T cells, and CD38 and CD138 were positive in the plasma cells. Additionally, Ki-67 expression was positive in the follicle centers. In contrast, staining for Bcl-2 in the germinal centers and cyclin D1 were negative. The immunohistochemical analysis combined with the morphological results supported the diagnosis of HV-CD. The patient recovered well after surgery. CONCLUSIONS: Primarily renal HV-CD without lymph node hyperplasia or clinical symptoms is extraordinarily rare and different from the multicentric-type CD (MCD) with kidney involvement. Therefore, it is extremely important to improve the awareness of this diagnosis. Attention should be paid to the difference between HV-CD and common lymph node reactive hyperplasia, MCL, FL, NMZL, and so on. To avoid misdiagnosis as a renal malignant tumor requiring radical resection, distinguishing these diseases is crucial.


Assuntos
Hiperplasia do Linfonodo Gigante/patologia , Nefropatias/patologia , Humanos , Hialina , Masculino , Pessoa de Meia-Idade
15.
Neurosci Lett ; 690: 61-68, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30300683

RESUMO

Neural stem cells (NSCs) as sources of new neurons in brain injuries or diseases are required to not only elicit neurons for neuronal repair, but also to enhance neurite outgrowth for neuronal network reestablishment. Various trophic or chemotropic factors have been shown to cooperatively improve NSC neurogenesis. However, effects of combined treatment of all-trans-retinoic acid (RA) with GF (Basic fibroblast growth factor and epidermal growth factor, bFGF/EGF) on neurogenesis of NSCs are poorly understood. To address this question, NSCs were isolated from the forebrains of embryonic mice, and treated with GF and RA either alone or in combination for differentiation in vitro. Neurons and astrocytes differentiated from NSCs were stained for MAP2 and GFAP separately by immunofluorescence. The results indicated that GF displayed superior efficacy in promoting neuronal differentiation, and RA showed better efficacy in advancing neurite outgrowth by increasing both neurite length and number. In addition, higher differentiation efficiency of neurons to astrocytes in RA or GF, or both acted at the early stage. However, more importantly, compared with RA alone, GF and RA in combination enhanced neuronal differentiation. Moreover, the combined use of GF and RA increased the length and number of neurites compared with GF, as well as the relative expression level of Smurf1. In addition, astrocytes induced by GF, RA, or both exhibited a radial glia-like morphology with long processes differing from serum effects, which might in part attribute to the total numbers of neurons. These findings for the first time unveil the roles of combined use of GF and RA on the neurogenesis of NSCs, suggesting that the use of this combination could be a comprehensive strategy for the functional repair of the nervous system through promoting neuronal differentiation, and advancing neurite outgrowth.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Tretinoína/farmacologia , Animais , Astrócitos/metabolismo , Células Cultivadas , Sinergismo Farmacológico , Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/metabolismo , Prosencéfalo , Ubiquitina-Proteína Ligases/metabolismo
16.
Int J Biol Sci ; 15(6): 1225-1239, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31223282

RESUMO

The G protein-coupled receptor APJ/Aplnr has been widely reported to be involved in heart and vascular development and disease, but whether it contributes to organ left-right patterning is largely unknown. Here, we show that in zebrafish, aplnra/b coordinates organ LR patterning in an apela/apln ligand-dependent manner using distinct mechanisms at different stages. During gastrulation and early somitogenesis, aplnra/b loss of function results in heart and liver LR asymmetry defects, accompanied by disturbed KV/cilia morphogenesis and disrupted left-sided Nodal/spaw expression in the LPM. In this process, only aplnra loss of function results in KV/cilia morphogenesis defect. In addition, only apela works as the early endogenous ligand to regulate KV morphogenesis, which then contributes to left-sided Nodal/spaw expression and subsequent organ LR patterning. The aplnra-apela cascade regulates KV morphogenesis by enhancing the expression of foxj1a, but not fgf8 or dnh9, during KV development. At the late somite stage, both aplnra and aplnrb contribute to the expression of lft1 in the trunk midline but do not regulate KV formation, and this role is possibly mediated by both endogenous ligands, apela and apln. In conclusion, our study is the first to identify a role for aplnra/b and their endogenous ligands apela/apln in LR patterning, and it clarifies the distinct roles of aplnra-apela and aplnra/b-apela/apln in orchestrating organ LR patterning.


Assuntos
Receptores de Apelina/fisiologia , Padronização Corporal , Peixe-Zebra/crescimento & desenvolvimento , Animais , Receptores de Apelina/genética , Receptores de Apelina/metabolismo , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/genética , Gastrulação/genética , Ligantes , Ligantes da Sinalização Nodal/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Proteínas de Peixe-Zebra/metabolismo
17.
J Neurochem ; 106(4): 1815-27, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18564367

RESUMO

Recent studies from environmental toxicology and molecular genetics demonstrate that midbrain dopamine (DA) neurons are particularly vulnerable to microtubule depolymerizing agents, indicating the involvement of microtubule dysfunction in the pathogenesis of Parkinson's disease. Here we show that intrastriatal injection of colchicine (COL), a well-known microtubule disruptor, induced degeneration of striatonigral pathway. Microtubule disruption caused by unilateral injection of COL blocked the retrograde axonal transport of fluorogold previously injected into striatum and induced substantial death of striatal and DA neurons in substantia nigra pars compacta. Furthermore, COL-induced pathologic changes were associated with robust glial reaction, which may be conducive to the degeneration of striatonigral pathway. We also found that intrastriatal injection of COL resulted in side bias in spontaneous turning activities and apomorphine-induced rotational behavior. Together, our results provide in vivo data lending support to the concept that microtubule dysfunction may play a significant role in the death of DA neurons, though glial reaction may be involved and contribute to the degenerative process. Moreover, intrastriatal COL may serve as another experimental model of striatonigral degeneration (Parkinson's variant of multiple system atrophy), given the concurrent loss of both striatal and DA neurons.


Assuntos
Colchicina/toxicidade , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Degeneração Estriatonigral/induzido quimicamente , Degeneração Estriatonigral/patologia , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Axônios/patologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Colchicina/administração & dosagem , Corpo Estriado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Estriatonigral/metabolismo
18.
J Mater Chem B ; 6(19): 3163-3180, 2018 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-32254351

RESUMO

The safe and efficient targeted delivery of chemotherapeutic drugs has remained a challenge in metastatic breast cancer therapy. Herein, we report a rational drug delivery strategy of co-administering tumor-penetrating peptide-iRGD with self-assembled amphiphilic block copolymer nanoparticles (HA-PLA) to inhibit tumor growth and lung metastasis in 4T1 breast cancer xenograft bearing mice through increasing drug accumulation in the tumors, inducing receptor-mediated tumor cell targeting without causing severe side effects. In vitro, HA-PLA displayed sustained and pH-sensitive release behavior. The cellular uptake of HA-PLA on high CD44-expressing 4T1 cells was significantly higher than the endocytosis on low CD44-expressing L929 fibroblast cells. In vivo, HA-PLA significantly extended the blood circulation time of DOX, displayed no "accelerated blood clearance (ABC) phenomenon" after repeated injection and decreased the side effects of DOX. When combined with iRGD, the drug distribution and penetration of HA-PLA in tumors were remarkably increased, resulting in better antitumor efficacy and the longest whole survival. In particular, the co-administration of iRGD with HA-PLA greatly increased drug distribution in the lung, which contributed to the effective inhibition of the lung metastasis of breast cancer. Therefore, co-administering iRGD with HA-PLA is a promising approach for metastatic breast cancer therapy.

19.
Exp Ther Med ; 15(2): 1424-1432, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29434727

RESUMO

Corneal integrity, transparency and vision acuity are maintained by corneal epithelial cells (CECs), which are continuously renewed by corneal limbal stem cells (LSCs). Deficiency of CECs and/or LSCs is associated with numerous ocular diseases. Paired box (PAX)6 is an eye development-associated transcription factor that is necessary for cell fate determination and differentiation of LSCs and CECs. In the present study, the PAX6 gene was introduced into adipose-derived rat mesenchymal stem cells (ADMSCs) to investigate whether PAX6-transfected cells were able to transdifferentiate into corneal-like epithelial cells and to further verify whether the cells were suitable as a cell source for corneal transplantation. The ADMSCs were isolated from the bilateral inguinal region of healthy Sprague Dawley rats. The characteristics of ADMSCs were identified using flow cytometric analysis. After subculture, ADMSCs underwent transfection with recombinant plasmid containing either PAX6-enhanced green fluorescent protein (EGFP) complementary (c)DNA or EGFP cDNA (blank plasmid group), followed by selection with G418 and determination of the transfection efficiency. Subsequently, the morphology of the ADMSCs and the expression profiles of corneal-specific markers CK3/12 and epithelial-specific adhesion protein were determined. E-cadherin was detected using immunofluorescence staining and western blot analysis at 21 days following transfection. An MTT cell proliferation and a colony formation assay were performed to assess the proliferative activity and clonogenicity of PAX6-transfected ADMSCs. Finally, the PAX6-expressing ADMSCs were transplanted onto the cornea of a rabbits with limbal stem cell deficiency (LSCD). At 21 days after transfection, the ADMSCs with PAX6 transfection exhibited a characteristic flagstone-like appearance with assembled corneal-like epithelial cells, and concomitant prominent expression of the corneal-specific markers cytokeratin 3/12 and E-cadherin. Furthermore, the proliferation and colony formation ability of PAX6-overexpressing ADMSCs was significantly retarded. The transplantation experiment indicated that PAX6-reprogramed ADMSCs attached to and replenished the damaged cornea via formation of stratified corneal epithelium. Taken together, these results suggested that conversion of ADMSCs into corneal-like epithelium may be driven by PAX6 transfection, which makes ADMSCs a promising cell candidate for the treatment of LSCD.

20.
Brain Res ; 1178: 132-40, 2007 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-17900537

RESUMO

Recent studies have invoked inflammation as a major contributor to the pathogenesis of Parkinson's disease (PD). Emerging evidence indicated that components of complement system may be involved in such disorder and contribute to its development. We thus observed the influence of deficiency of complement 3 (C3), the key component of complement system, on the death of dopaminergic neurons in substantia nigra pars compacta (SNpc) and the loss of dopaminergic fibers in striatum induced by acute or chronic administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Immunohistochemical staining of dopaminergic neurons in SNpc and neurochemical analysis of dopamine and its metabolites in striata revealed that there was no significant difference between the two genotypes. Longer survival time also indicated that C3 might not mediate the spontaneous recovery of dopaminergic fibers in mouse striatum acutely challenged by MPTP. We conclude that, despite growing evidence indicating the involvement of complement system in the pathogenesis of PD, our data do not support a role for C3 in this established model of PD, as indicated by results from HPLC analysis and immunohistochemical staining.


Assuntos
Complemento C3/genética , Complemento C3/fisiologia , Dopamina/fisiologia , Intoxicação por MPTP/genética , Intoxicação por MPTP/patologia , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Complemento C3/deficiência , Dopamina/metabolismo , Eletroquímica , Ácido Homovanílico/metabolismo , Imuno-Histoquímica , Intoxicação por MPTP/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Neostriado/metabolismo , Neostriado/patologia , Regeneração Nervosa , Tirosina 3-Mono-Oxigenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA