Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(24)2022 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-36555516

RESUMO

Autophagy facilitates the degradation of organelles and cytoplasmic proteins in a lysosome-dependent manner. It also plays a crucial role in cell damage. Whether loganin affects autophagy in chronic constriction injury (CCI)-induced neuropathic pain remains unclear. We investigated the neuroprotective effect of loganin on the autophagic-lysosomal pathway in the rat CCI model. Sprague-Dawley rats were divided into sham, CCI, sham + loganin, and CCI + loganin. Loganin (5 mg/kg/day) was intraperitoneally injected once daily, and rats were sacrificed on day 7 after CCI. This study focused on the mechanism by which loganin modulates autophagic flux after CCI. CCI enhanced the autophagic marker LC3B-II in the ipsilateral spinal cord. The ubiquitin-binding protein p62 binds to LC3B-II and integrates into autophagosomes, which are degraded by autophagy. CCI caused the accumulation of p62, indicating the interruption of autophagosome turnover. Loganin significantly attenuated the expression of Beclin-1, LC3B-II, and p62. Double immunofluorescence staining was used to confirm that LC3B-II and p62 were reduced by loganin in the spinal microglia and astrocytes. Loganin also lessened the CCI-increased colocalization of both proteins. Enhanced lysosome-associated membrane protein 2 (LAMP2) and pro-cathepsin D (pro-CTSD) in CCI rats were also attenuated by loganin, suggesting that loganin improves impaired lysosomal function and autophagic flux. Loganin also attenuated the CCI-increased apoptosis protein Bax and cleaved caspase-3. Loganin prevents CCI-induced neuropathic pain, which could be attributed to the regulation of neuroinflammation, neuronal autophagy, and associated cell death. These data suggest autophagy could be a potential target for preventing neuropathic pain.


Assuntos
Glicosídeos Cardíacos , Neuralgia , Animais , Ratos , Autofagia , Constrição , Hiperalgesia/etiologia , Hiperalgesia/complicações , Glicosídeos Iridoides , Neuralgia/tratamento farmacológico , Neuralgia/etiologia , Neuralgia/metabolismo , Ratos Sprague-Dawley
2.
J Chin Med Assoc ; 85(1): 102-108, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34380992

RESUMO

BACKGROUND: Recombinant tissue plasminogen activator (rtPA) is currently the most standard treatment for patients with acute ischemic stroke (AIS). However, rtPA treatment may further enhance the immune response poststroke. This study is to investigate the clinical utility of white blood-based inflammatory biomarkers in predicting neurologic outcomes among AIS patients receiving rtPA. METHODS: A retrospective observational cohort study of 100 patients with AIS treated with intravenous rtPA was conducted in an urban tertiary hospital in Taiwan. Favorable neurological outcome defined as modified Rankin Scale (mRS) score 0 to 2 in poststroke follow-up was the primary outcome measure. Baseline and post-rtPA neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) were investigated for potential predictors. RESULTS: These patients had a mean age of 71.3 ± 13.7 years and the average of initial National Institute of Health Stroke Scale was 12.7 ± 6.5. Using multiple regression analysis, PLR was not an independent factor; however, both baseline and post-rtPA NLR were independent factors predicting favorable neurological outcome at 3, 6, 12 months after a stroke. The area under the receiver operating characteristic curve for baseline and post-rtPA NLR were 0.645 (95% confidence interval [CI], 0.537-0.753) and 0.769 (95% CI, 0.676-0.862) (Z score = 2.086) in 3-month, 0.645 (95% CI, 0.537-0.752) and 0.791 (95% CI, 0.701-0.880) (Z score = 2.471) in 6-month, and 0.646 (95% CI, 0.538-0.754) and 0.813 (95% CI, 0.728-0.898) (Z score = 2.857) in 12-month poststroke follow-up. CONCLUSION: For AIS patients treated with rtPA, both lower baseline and post-rtPA NLR levels were independently associated with a favorable neurologic outcome in serial mid- and long-term follow-up. Post-rtPA NLR was superior to baseline NLR in discriminative performance for neurologic prognosis.


Assuntos
AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/prevenção & controle , Linfócitos , Neutrófilos , Terapia Trombolítica , Ativador de Plasminogênio Tecidual/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Feminino , Previsões , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Taiwan , Resultado do Tratamento
3.
Biomed Pharmacother ; 145: 112447, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34808553

RESUMO

Eugenosedin-A (Eu-A) has been shown to protect against hyperglycemia- and hyperlipidemia-induced metabolic syndrome. We investigated the relationship of KATP channel activities and insulin secretion by Eu-A in vitro in pancreatic ß-cells, and examined the effect of Eu-A on streptozotocin (STZ)/nicotinamide (NA)-induced type 2 diabetes mellitus (T2DM) in vivo. We isolated pancreatic islets from adult male Wistar rats (250-350 g) and identified pancreatic ß-cells by the cell size, capacitance and membrane potential. Perforated patch-clamp and inside-out recordings were used to monitor the membrane potential (current-clamp mode) and channel activity (voltage-clamp mode) of ß-cells. The membrane potential of ß-cells was raised by Eu-A and reversed by the KATP channel activator diazoxide. Eu-A inhibited the KATP channel activity measured at - 60 mV and increased the intracellular calcium concentration ([Ca2+]i), resulting in enhanced insulin secretion. Eu-A also reduced Kir6.2 protein on the cell membrane and scattered in the cytosol under normal glucose conditions (5.6 mM). In our animal study, rats were divided into normal and STZ/NA-induced T2DM groups. Normal rats fed with regular chow were divided into control and control+Eu-A (5 mg/kg/day, i.p.) groups. The STZ/NA-induced diabetic rats fed with a high-fat diet (HFD) were divided into three groups: T2DM, T2DM+Eu-A (5 mg/kg/day, i.p.), and T2DM+glibenclamide (0.5 mg/kg/day, i.p.; a KATP channel inhibitor). Both Eu-A and glibenclamide decreased the rats' blood glucose, prevented weight gain, and enhanced insulin secretion. We found that Eu-A blocked pancreatic ß-cell KATP channels, caused membrane potential depolarization, and stimulated Ca2+ influx, thus increasing insulin secretion. Furthermore, Eu-A decreased blood glucose and increased insulin levels in T2DM rats. These results suggested that Eu-A might have clinical benefits for the control of T2DM and its complications.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hiperglicemia/tratamento farmacológico , Piperazinas/farmacologia , Animais , Glicemia/efeitos dos fármacos , Dieta Hiperlipídica , Glibureto/farmacologia , Hiperglicemia/etiologia , Hipoglicemiantes/farmacologia , Secreção de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Canais KATP/metabolismo , Masculino , Obesidade/complicações , Ratos , Ratos Wistar
4.
Phytomedicine ; 92: 153734, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34536822

RESUMO

BACKGROUND: Neuropathic pain has been shown to be modulated by the activation of the chemokine C-X-C motif ligand 12 (CXCL12)/chemokine CXC receptor 4 (CXCR4) dependent nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome. Loganin, an iridoid glycoside, was proven to prevent neuropathic pain, but its underlying mechanisms related to NLRP3 activation are still unknown. PURPOSE: This study investigated the underlying mechanisms of loganin's effect on chronic constriction injury (CCI)-induced NLRP3 inflammasome activation in the spinal cord. METHODS: Sprague-Dawley rats were randomly divided into four groups: sham, CCI, sham + loganin, and CCI + loganin. Loganin (5 mg/kg/day) was administered intraperitoneally starting the day after surgery. Paw withdrawal threshold (PWT) and latency (PWL) were assessed before CCI and on days 1, 3, 7 and 14 after CCI. Spinal cords were collected for western blots and immunofluorescence studies. RESULTS: Loganin prevented CCI-attenuated PWT and PWL, suggesting improved mechanical allodynia and thermal hyperalgesia. The expression of CXCL12, CXCR4, thioredoxin-interacting protein (TXNIP), NLRP3 inflammasome (NLRP3, ASC, and caspase-1), IL-1ß, and IL-18 were enhanced on day 7 after CCI, and all were reduced after loganin treatment. Dual immunofluorescence also showed that increased CXCL12, CXCR4, and NLRP3 were colocalized with NeuN (neuronal marker), GFAP (astrocyte marker), and Iba1 (microglial marker) on day 7 in the ipsilateral spinal dorsal horn (SDH). These immunoreactivities were attenuated in loganin-treated rats. Moreover, loganin decreased the assembly of NLRP3/ASC inflammasome after CCI in the ipsilateral SDH. Loganin appears to attenuate CCI-induced neuropathic pain by suppressing CXCL12/CXCR4-mediated NLRP3 inflammasome. CONCLUSION: Our findings suggest that loganin might be a suitable candidate for managing CCI-provoked neuropathic pain.


Assuntos
Inflamassomos , Neuralgia , Animais , Proteínas de Ciclo Celular , Hiperalgesia/tratamento farmacológico , Iridoides , Proteína 3 que Contém Domínio de Pirina da Família NLR , Neuralgia/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Receptores CXCR4 , Receptores de Quimiocinas , Medula Espinal
5.
Cells ; 9(9)2020 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-32842536

RESUMO

Diabetic peripheral neuropathy (DPN) is caused by hyperglycemia, which induces oxidative stress and inflammatory responses that damage nerve tissue. Excessive generation of reactive oxygen species (ROS) and NOD-like receptor protein 3 (NLRP3) inflammasome activation trigger the inflammation and pyroptosis in diabetes. Schwann cell dysfunction further promotes DPN progression. Loganin has been shown to have antioxidant and anti-inflammatory neuroprotective activities. This study evaluated the neuroprotective effect of loganin on high-glucose (25 mM)-induced rat Schwann cell line RSC96 injury, a recognized in vitro cell model of DPN. RSC96 cells were pretreated with loganin (0.1, 1, 10, 25, 50 µM) before exposure to high glucose. Loganin's effects were examined by CCK-8 assay, ROS assay, cell death assay, immunofluorescence staining, quantitative RT-PCR and western blot. High-glucose-treated RSC96 cells sustained cell viability loss, ROS generation, NF-κB nuclear translocation, P2 × 7 purinergic receptor and TXNIP (thioredoxin-interacting protein) expression, NLRP3 inflammasome (NLRP3, ASC, caspase-1) activation, IL-1ß and IL-18 maturation and gasdermin D cleavage. Those effects were reduced by loganin pretreatment. In conclusion, we found that loganin's antioxidant effects prevent RSC96 Schwann cell pyroptosis by inhibiting ROS generation and suppressing NLRP3 inflammasome activation.


Assuntos
Glucose/efeitos adversos , Inflamassomos/metabolismo , Iridoides/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose/efeitos dos fármacos , Células de Schwann/metabolismo , Animais , Humanos , Ratos
6.
Phytomedicine ; 67: 153166, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31955133

RESUMO

BACKGROUND: Peripheral nerve injury can produce chronic and ultimately neuropathic pain. The chronic constriction injury (CCI) model has provided a deeper understanding of nociception and chronic pain. Loganin is a well-known herbal medicine with glucose-lowering action and neuroprotective activity. PURPOSE: This study investigated the molecular mechanisms by which loganin reduced CCI-induced neuropathic pain. METHODS: Sprague-Dawley rats were randomly divided into four groups: sham, sham+loganin, CCI and CCI+loganin. Loganin (1 or 5 mg/kg/day) was injected intraperitoneally once daily for 14 days, starting the day after CCI. For behavioral testing, mechanical and thermal responses were assessed before surgery and on d1, d3, d7 and d14 after surgery. Sciatic nerves (SNs) were collected to measure proinflammatory cytokines. Proximal and distal SNs were collected separately for Western blotting and immunofluorescence studies. RESULTS: Thermal hyperalgesia and mechanical allodynia were reduced in the loganin-treated group as compared to the CCI group. Loganin (5 mg/kg/day) prevented CCI from inducing proinflammatory cytokines (TNF-α, IL-1ß), inflammatory proteins (TNF-α, IL-1ß, pNFκB, pIκB/IκB, iNOS) and receptor (TNFR1, IL-1R), adaptor protein (TRAF2) of TNF-α, and Schwann cell demyelination and axonal damage. Loganin also blocked IκB phosphorylation (p-IκB). Double immunofluorescent staining further demonstrated that pNFκB/pIκB protein was reduced by loganin in Schwann cells on d7 after CCI. In the distal stumps of injured SN, Schwann cell demyelination was correlated with pain behaviors in CCI rats. CONCLUSION: Our findings indicate that loganin improves CCI-induced neuroinflammation and pain behavior by downregulating TNF-α/IL-1ß-dependent NF-κB activation.


Assuntos
Analgésicos não Narcóticos/farmacologia , Iridoides/farmacologia , NF-kappa B/metabolismo , Neuralgia/tratamento farmacológico , Células de Schwann/efeitos dos fármacos , Animais , Dor Crônica/tratamento farmacológico , Dor Crônica/metabolismo , Dor Crônica/patologia , Constrição , Citocinas/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Interleucina-1beta/metabolismo , Masculino , Neuralgia/metabolismo , Neuralgia/patologia , Ratos Sprague-Dawley , Células de Schwann/metabolismo , Células de Schwann/patologia , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
Hum Mol Genet ; 26(20): 3909-3921, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-29016849

RESUMO

Impaired clearance of amyloid-ß peptide (Aß) leads to abnormal extracellular accumulation of this neurotoxic protein that drives neurodegeneration in sporadic Alzheimer's disease (AD). Connective tissue growth factor (CTGF/CCN2) expression is elevated in plaque-surrounding astrocytes in AD patients. However, the role of CTGF in AD pathogenesis remains unclear. Here we characterized the neuroprotective activity of CTGF. We found that CTGF facilitated Aß uptake and subsequent degradation within primary glia and neuroblastoma cells. CTGF enhanced extracellular Aß degradation via membrane-bound matrix metalloproteinase-14 (MMP14) in glia and extracellular MMP13 in neurons. In the brain of a Drosophila AD model, glial-expression of CTGF reduced Aß deposits, improved locomotor function, and rescued memory deficits. Neuroprotective potential of CTGF against Aß42-induced photoreceptor degeneration was disrupted through silencing MMPs. Therefore, CTGF may represent a node for potential AD therapeutics as it intervenes in glia-neuron communication via specific MMPs to alleviate Aß neurotoxicity in the central nervous system.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Animais Geneticamente Modificados , Astrócitos/metabolismo , Encéfalo/metabolismo , Fator de Crescimento do Tecido Conjuntivo/fisiologia , Modelos Animais de Doenças , Drosophila , Humanos , Metaloproteinase 14 da Matriz/metabolismo , Camundongos , Neuroglia/metabolismo , Neuroglia/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/metabolismo , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/metabolismo , Ratos
8.
EMBO Rep ; 18(7): 1150-1165, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28507162

RESUMO

Dihydroceramide desaturases are evolutionarily conserved enzymes that convert dihydroceramide (dhCer) to ceramide (Cer). While elevated Cer levels cause neurodegenerative diseases, the neuronal activity of its direct precursor, dhCer, remains unclear. We show that knockout of the fly dhCer desaturase gene, infertile crescent (ifc), results in larval lethality with increased dhCer and decreased Cer levels. Light stimulation leads to ROS increase and apoptotic cell death in ifc-KO photoreceptors, resulting in activity-dependent neurodegeneration. Lipid-containing Atg8/LC3-positive puncta accumulate in ifc-KO photoreceptors, suggesting lipophagy activation. Further enhancing lipophagy reduces lipid droplet accumulation and rescues ifc-KO defects, indicating that lipophagy plays a protective role. Reducing dhCer synthesis prevents photoreceptor degeneration and rescues ifc-KO lethality, while supplementing downstream sphingolipids does not. These results pinpoint that dhCer accumulation is responsible for ifc-KO defects. Human dhCer desaturase rescues ifc-KO larval lethality, and rapamycin reverses defects caused by dhCer accumulation in human neuroblastoma cells, suggesting evolutionarily conserved functions. This study demonstrates a novel requirement for dhCer desaturase in neuronal maintenance in vivo and shows that lipophagy activation prevents activity-dependent degeneration caused by dhCer accumulation.


Assuntos
Autofagia , Ceramidas/metabolismo , Metabolismo dos Lipídeos , Animais , Apoptose , Linhagem Celular Tumoral , Ceramidas/análise , Drosophila , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Ácidos Graxos Dessaturases/genética , Técnicas de Inativação de Genes , Humanos , Luz/efeitos adversos , Lipólise , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Doenças Neurodegenerativas/prevenção & controle , Células Fotorreceptoras de Invertebrados/patologia , Células Fotorreceptoras de Invertebrados/efeitos da radiação , Esfingolipídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA