Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Clin Exp Nephrol ; 28(5): 375-390, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38238499

RESUMO

BACKGROUND: NLRP3 inflammasome activation is significantly associated with sepsis-induced acute kidney injury (S-AKI). Cytosolic DNA derived from damaged mitochondria has been reported to activate NLRP3 inflammasome via upregulating the cyclic GMP-AMP synthase (cGAS)-the stimulator of interferon genes (STING) axis in nucleus pulposus cell and cardiomyocytes. However, the regulatory effect of mitochondria DNA (mtDNA)-cGAS-STING axis on the NLRP3 inflammasome in S-AKI remains unclear. METHODS: In the current study, we established an in vivo model of S-AKI by intraperitoneally injecting male C57BL/6 J mice with lipopolysaccharide (LPS). Next, selective cGAS inhibitor RU.521, and STING agonist DMXAA were intraperitoneally injected in the mice; then, blood urea nitrogen (BUN), serum creatinine (CRE), urinary kidney injury molecular-1 (KIM-1), pathological changes, and infiltrated neutrophils were detected to assess kidney injury. We also performed western blot and immunofluorescence assays to evaluate STING, cGAS, TBK-1, p-TBK-1, IRF3, p-IRF3, NF-kB, p-NF-kB, NLRP3, cleaved caspase-1, caspase-1, GSDMD-N, and GSDMD expression levels in kidney tissues. IL-18 and IL-1ß in renal tissue were identified by ELISA. In vitro, we treated HK-2 cells with LPS to establish a cell model of S-AKI. Furthermore, ethidium bromide (EtBr) was administered to deplete mitochondria DNA (mtDNA). LPS-induced cytotoxicity was evaluated by LDH release assay. Protein expression of cGAS, STING, and NLRP3 in was quantified by western blot. Cytosolic mtDNA was detected by immunofluorescence and q-PCR. Released IL-1ß and IL-18 in HK-2 supernatants were detected by ELISA. RESULTS: LPS injection induced S-AKI in mice, as evidenced by neutrophil infiltration, tubular vacuolation, and increased levels of serum creatinine (CRE), blood urea nitrogen (BUN), and urinary KIM-1. In addition, LPS activated the cGAS-STING axis and NLRP3 inflammasome in vivo, illustrated by increased phosphorylation levels of TBK-1, IRF3, and NF-kB protein, increased ratio of cleaved caspase-1 to caspase-1 and GSDMD-N to GSDMD, and increased IL-1ß and IL-18 levels. Moreover, the cGAS inhibitor RU.521 effectively attenuated NLRP3 inflammasome and S-AKI; however, these effects were abolished by treatment with the STING agonist DMXAA. Furthermore, cytosolic release of mtDNA and activation of the cGAS-STING-NLRP3 axis were observed in LPS-treated HK-2 cells. Inhibiting mtDNA replication by Ethidium Bromide (EtBr) treatment reduced cytosolic mtDNA accumulation and downregulated the cGAS-STING-NLRP3 axis, ameliorating the cytotoxicity induced by LPS. CONCLUSION: This study demonstrated that the cGAS-STING axis was triggered by cytosolic mtDNA and participated in the development of S-AKI by activating NLRP3 inflammasome. Reducing cytosolic mtDNA accumulation or inhibiting the cGAS-STING axis may be potential therapeutic targets for S-AKI.


Assuntos
Injúria Renal Aguda , DNA Mitocondrial , Inflamassomos , Proteínas de Membrana , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR , Nucleotidiltransferases , Sepse , Animais , Masculino , Camundongos , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/etiologia , Citosol/metabolismo , Modelos Animais de Doenças , DNA Mitocondrial/metabolismo , Inflamassomos/metabolismo , Lipopolissacarídeos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Nucleotidiltransferases/metabolismo , Sepse/complicações , Sepse/metabolismo , Transdução de Sinais
2.
Neurosci Lett ; 809: 137308, 2023 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-37244447

RESUMO

Bupivacaine (BUP) has previously been shown to trigger neurotoxicity after spinal anesthesia. Further, ferroptosis has been implicated in the pathological processes associated with various central nervous system diseases. Although the impact of ferroptosis on BUP-induced neurotoxicity in the spinal cord has not been fully understood, this research aims to investigate this relationship in rats. Additionally, this study aims to determine whether ferrostatin-1 (Fer-1), a potent inhibitor of ferroptosis, can provide protection against BUP-induced spinal neurotoxicity. The experimental model for BUP-induced spinal neurotoxicity involved the administration of 5% bupivacaine through intrathecal injection. Then, the rats were randomized into the Control, BUP, BUP + Fer-1, and Fer-1 groups. BBB scores, %MPE of TFL, and H&E and Nissl stainings showed that intrathecal Fer-1 administration improved functional recovery, histological outcomes, and neural survival in BUP-treated rats. Moreover, Fer-1 has been found to alleviate the BUP-induced alterations related to ferroptosis, such as mitochondrial shrinkage and disruption of cristae, while also reducing the levels of malondialdehyde (MDA), iron, and 4-hydroxynonenal (4HNE). Fer-1 also inhibits the accumulation of reactive oxygen species (ROS) and restores the normal levels of glutathione peroxidase 4 (GPX4), cystine/glutamate transporter (xCT), and glutathione (GSH). Furthermore, double-immunofluorescence staining revealed that GPX4 is primarily localized in the neurons instead of microglia or astroglia in the spinal cord. In summary, we demonstrated that ferroptosis play a pivotal role in mediating BUP-induced spinal neurotoxicity, and Fer-1 ameliorated BUP-induced spinal neurotoxicity by reversing the underlying ferroptosis-related changes in rats.


Assuntos
Ferroptose , Síndromes Neurotóxicas , Animais , Ratos , Medula Espinal , Bupivacaína , Glutationa
3.
Neurosci Lett ; 772: 136472, 2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-35065245

RESUMO

Bupivacaine is a common local anesthetic that causes neurotoxicity when used at clinical concentrations. Melatonin (MT), is a potent neuroprotective molecule. The study aimed to characterize the neuroprotective effects of MT on spinal neurotoxicity induced by bupivacaine in rats. It showed that bupivacaine, by intrathecal injection, induced spinal injury, and that the protein levels of Nod-like receptor protein 3 (NLRP3), cleaved caspase-1, and the N-terminal region of gasdermin D (GSDMD-N) were significantly increased. NLRP3 was expressed mainly in neurons and microglia. MT treatment ameliorated bupivacaine-induced spinal cord injury in rats by suppressing activation of neuronal NLRP3 inflammasomes.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Melatonina/uso terapêutico , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Síndromes Neurotóxicas/tratamento farmacológico , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Bupivacaína/toxicidade , Masculino , Melatonina/farmacologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Ratos , Ratos Sprague-Dawley , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
4.
Drug Des Devel Ther ; 13: 707-718, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30858700

RESUMO

BACKGROUND: Local anesthetics in spinal anesthesia have neurotoxic effects, resulting in severe neurological complications. Intrathecal monosialoganglioside (GM1) administration has a therapeutic effect on bupivacaine-induced neurotoxicity. The aim of this study was to determine the underlying mechanisms of bupivacaine-induced neurotoxicity and the potential neuroprotective role of GM1. MATERIALS AND METHODS: A rat spinal cord neurotoxicity model was established by injecting bupivacaine (5%, 0.12 µL/g) intrathecally. The protective effect of GM1 (30 mg/kg) was evaluated by pretreating the animals with it prior to the bupivacaine regimen. The neurological and locomotor functions were assessed using standard tests. The histomorphological changes, neuron degeneration and apoptosis, and endoplasmic reticulum stress (ERS) relevant markers were analyzed using immunofluorescence, quantitative real-time PCR, and Western blotting. RESULTS: Bupivacaine resulted in significant neurotoxicity in the form of aberrant neurolocomoter functions and spinal cord histomorphology and neuronal apoptosis. Furthermore, the ERS specific markers were significantly upregulated during bupivacaine-induced neurotoxicity. These neurotoxic effects were ameliorated by GM1. CONCLUSION: Pretreatment with GM1 protects against bupivacaine-induced neurotoxicity via the inhibition of the GRP78/PERK/eIF2α/ATF4-mediated ERS.


Assuntos
Bupivacaína/antagonistas & inibidores , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Gangliosídeos/farmacologia , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/tratamento farmacológico , Animais , Bupivacaína/toxicidade , Gangliosídeos/química , Masculino , Fármacos Neuroprotetores/química , Ratos , Ratos Sprague-Dawley
5.
Cell Biochem Funct ; 34(6): 455-62, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27558076

RESUMO

OBJECTIVE: Studies indicate that bupivacaine-induced neurotoxicity results from apoptosis. Gangliosides have been shown to promote neuronal repair and recovery of neurological function after spinal cord injury. Previously, we confirmed that in vivo administration of the ganglioside GM-1 attenuated bupivacaine-induced neurotoxicity in various animal models; however, the underlying mechanism remains unclear. METHODS: Cells of the neuroblastoma line N2a (Neuro2a cells) were divided into three experimental groups: control, bupivacaine-treated, and bupivacaine-treated with GM-1 pretreatment. Cell viability and apoptosis were assessed through CCK-8 assays, Hoechst staining, and flow cytometry analysis of Annexin-V/propidium iodide double labeling. Real-time polymerase chain reaction and western blotting assessed the expression of caspase-3, caspase-8, and caspase-9. RESULTS: Bupivacaine-induced apoptosis worsened with increasing dose and exposure time. Bupivacaine induced increased expression of caspase-3 and caspase-9, but not caspase-8, indicating that the mitochondrial pathway but not the death receptor apoptosis pathway was activated. GM-1 pretreatment inhibited bupivacaine-induced apoptosis and the expression of caspase-3 and caspase-9 in a dose-dependent manner. CONCLUSION: Bupivacaine induced neurotoxicity by activating apoptosis via the mitochondrial pathway, and this was inhibited by GM-1 pretreatment.


Assuntos
Bupivacaína/toxicidade , Gangliosídeo G(M1)/farmacologia , Neuroblastoma/patologia , Neurotoxinas/toxicidade , Animais , Apoptose/efeitos dos fármacos , Bisbenzimidazol/metabolismo , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citometria de Fluxo , Camundongos , Neuroproteção/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
6.
Biomed Pharmacother ; 69: 311-6, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25661376

RESUMO

BACKGROUND: Bupivacaine causes neuronal and axonal degeneration, leading to cauda equina syndrome or permanent nerve damage. Our previous studies have shown that intrathecal or intravenous gangliosides monosialogangliosides (GM-1s) have therapeutic effects against bupivacaine-induced neurotoxicity, but we do not know what are the differences between the two methods. METHODS AND RESULTS: Bupivacaine-induced neurotoxicity was induced in rats by three times injection of 5% bupivacaine (0.24µl/g) to the L3 spinal cord. We observed by H&E staining that bupivacaine caused obvious neuronal injuries in the spinal cord, such as edema, vacuolation of myelin sheaths, and neuronal degeneration. Electron microscopy revealed similar pathohistological changes. Neural functions, evaluated by tail-flicking test and locomotor scaling, were also impaired. Treatment with GM-1s (30mg/kg) repaired the neural lesions and gradually improved the neural functions. By days 14 and 28 post GM-1s, the pathohistological changes in the posterior root and posterior column had significantly recovered but not completely. Compared with intravenous routes, intrathecal application of GM-1s demonstrated faster and greater efficacies in regeneration of neural damages and in improvement of neural dysfunctions. Caspase-3, a marker of cellular apoptosis, was shown by immunohistochemistry to be suppressed in protein transcription by GM-1s application and intrathecal GM-1s had potentiated a greater reduction in caspase-3 protein than intravenous GM-1s. CONCLUSIONS: Treatment with GM-1s in intrathecal routes more effectively reverses bupivacaine-induced neural injuries and improves the neural dysfunctions than intravenous routes. This may be partly attributed to that GM-1 inhibits the expression of cellular apoptosis factor caspase-3 protein.


Assuntos
Bupivacaína/efeitos adversos , Gangliosídeos/uso terapêutico , Síndromes Neurotóxicas/tratamento farmacológico , Medula Espinal/patologia , Animais , Axônios/efeitos dos fármacos , Axônios/ultraestrutura , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Caspase 3/metabolismo , Gangliosídeos/administração & dosagem , Gangliosídeos/farmacologia , Temperatura Alta , Imuno-Histoquímica , Injeções Intravenosas , Injeções Espinhais , Locomoção/efeitos dos fármacos , Masculino , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/ultraestrutura , Neurônios/efeitos dos fármacos , Neurônios/patologia , Síndromes Neurotóxicas/patologia , Síndromes Neurotóxicas/fisiopatologia , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos , Medula Espinal/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA