Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Redox Biol ; 71: 103104, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38430683

RESUMO

AIMS: Spinal cord injury (SCI) is a devastating neurological disease that often results in tremendous loss of motor function. Increasing evidence demonstrates that diabetes worsens outcomes for patients with SCI due to the higher levels of neuronal oxidative stress. Mammalian sterile 20-like kinase (MST1) is a key mediator of oxidative stress in the central nervous system; however, the mechanism of its action in SCI is still not clear. Here, we investigated the role of MST1 activation in induced neuronal oxidative stress in patients with both SCI and diabetes. METHODS: Diabetes was established in mice by diet induction combined with intraperitoneal injection of streptozotocin (STZ). SCI was performed at T10 level through weight dropping. Advanced glycation end products (AGEs) were applied to mimic diabetic conditions in PC12 cell line in vitro. We employed HE, Nissl staining, footprint assessment and Basso mouse scale to evaluate functional recovery after SCI. Moreover, immunoblotting, qPCR, immunofluorescence and protein-protein docking analysis were used to detect the mechanism. RESULTS: Regarding in vivo experiments, diabetes resulted in up-regulation of MST1, excessive neuronal apoptosis and weakened motor function in SCI mice. Furthermore, diabetes impeded NRF2-mediated antioxidant defense of neurons in the damaged spinal cord. Treatment with AAV-siMST1 could restore antioxidant properties of neurons to facilitate reactive oxygen species (ROS) clearance, which subsequently promoted neuronal survival to improve locomotor function recovery. In vitro model found that AGEs worsened mitochondrial dysfunction and increased cellular oxidative stress. While MST1 inhibition through the chemical inhibitor XMU-MP-1 or MST1-shRNA infection restored NRF2 nuclear accumulation and its transcription of downstream antioxidant enzymes, therefore preventing ROS generation. However, these antioxidant effects were reversed by NRF2 knockdown. Our in-depth studies showed that over-activation of MST1 in diabetes directly hindered the neuroprotective AKT1, and subsequently fostered NRF2 ubiquitination and degradation via the GSK3ß/ß-TrCP pathway. CONCLUSION: MST1 inhibition significantly restores neurological function in SCI mice with preexisting diabetes, which is largely attributed to the activation of antioxidant properties via the GSK3ß(Ser 9)/ß-TrCP/NRF2 pathway. MST1 may be a promising pharmacological target for the effective treatment of spinal cord injury patients with diabetes.


Assuntos
Apoptose , Neurônios , Proteínas Serina-Treonina Quinases , Traumatismos da Medula Espinal , Animais , Camundongos , Ratos , Antioxidantes/farmacologia , Proteínas Contendo Repetições de beta-Transducina/farmacologia , Diabetes Mellitus , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Mamíferos/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Diabetes Mellitus Experimental/metabolismo
2.
Biochem Pharmacol ; 212: 115577, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37137416

RESUMO

Cancer stem cells (CSCs), a subpopulation of tumor cells with the features of self-renewal, tumor initiation, and insensitivity to common physical and chemical agents, are the key to cancer relapses, metastasis, and resistance. Accessible CSCs inhibitory strategies are primarily based on small molecule drugs, yet toxicity limits their application. Here, we report a liposome loaded with low toxicity and high effectiveness of miriplatin, lipo-miriplatin (LMPt) with high miriplatin loading, and robust stability, exhibiting a superior inhibitory effect on CSCs and non-CSCs. LMPt predominantly inhibits the survival of oxaliplatin-resistant (OXA-resistant) cells composed of CSCs. Furthermore, LMPt directly blocks stemness features of self-renewal, tumor initiation, unlimited proliferation, metastasis, and insensitivity. In mechanistic exploration, RNA sequencing (RNA-seq) revealed that LMPt downregulates the levels of pro-stemness proteins and that the ß-catenin-mediated stemness pathway is enriched. Further research shows that either in adherent cells or 3D-spheres, the ß-catenin-OCT4/NANOG axis, the vital pathway to maintain stemness, is depressed by LMPt. The consecutive activation of the ß-catenin pathway induced by mutant ß-catenin (S33Y) and OCT4/NANOG overexpression restores LMPt's anti-CSCs effect, elucidating the key role of the ß-catenin-OCT4/NANOG axis. Further studies revealed that the strengthened binding of ß-catenin and ß-TrCP initiates ubiquitination and degradation of ß-catenin induced by LMPt. In addition, the ApcMin/+ transgenic mouse model, in which colon tumors are spontaneously formed, demonstrates LMPt's potent anti-non-CSCs activity in vivo.


Assuntos
Neoplasias Colorretais , Proteínas Contendo Repetições de beta-Transducina , Animais , Camundongos , Linhagem Celular Tumoral , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Proteínas Contendo Repetições de beta-Transducina/farmacologia , beta Catenina/metabolismo , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas , Transformação Celular Neoplásica/patologia , Neoplasias Colorretais/metabolismo , Via de Sinalização Wnt , Proliferação de Células
3.
Brain Res ; 1800: 148192, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36463959

RESUMO

BACKGROUND: Hypoxia-ischemic brain damage (HIBD) is a primary cause of morbidity and disability in survivors of preterm infants. We previously discovered that miR-200b-3p plays an important role in HIBD via targeting Slit2. This study was designed to identify novel targets of miR-200b-3p and investigate the relationship between miR-200b-3p and its downstream effectors. METHODS AND RESULTS: Cultured primary rat hippocampal neurons were used in the model of oxygen-glucose deprivation (OGD) and RT-qPCR was utilized to detect the alterations of miR-200b-3p in these cells following the OGD. Our study found that the expression of miR-200b-3p was up-regulated in neurons post OGD. Bioinformatics analysis identified that ß transducin repeat-containing protein (ß-TrCP) is a target gene of miR-200b-3p, and our luciferase reporter gene assay confirmed that miR-200b-3p can interact with ß-TrCP mRNA. Hypoxia-ischemic brain damage was induced in three-day-old SD rats and inhibition of miR-200b-3p by injection of antagomir into bilateral lateral ventricles enhanced ß-TrCP expression at both the mRNA and protein levels in rats' brains. TUNEL staining and CCK-8 assays found that the survival of hippocampal neurons in the miR-200b-3p antagomir group was improved significantly (p<0.05), whereas apoptosis of neurons in the miR-200b-3p antagomir group was significantly decreased (p<0.05), as compared with the OGD group. However, silencing of ß-TrCP by ß-TrCP siRNA impaired the neuroprotective effect of miR-200b-3p antagomir. H&E staining showed that miR-200b-3p attenuated the pathological changes in the hippocampal region of rats with HIBD. CONCLUSION: Our study has demonstrated that ß-TrCP is a target gene of miR-200b-3p and that inhibition of miR-200b-3p by antagomir attenuates hypoxia-ischemic brain damage via ß-TrCP.


Assuntos
Hipóxia-Isquemia Encefálica , MicroRNAs , Recém-Nascido , Humanos , Ratos , Animais , Oxigênio/metabolismo , MicroRNAs/metabolismo , Proteínas Contendo Repetições de beta-Transducina/farmacologia , Antagomirs/farmacologia , Ratos Sprague-Dawley , Glucose/farmacologia , Recém-Nascido Prematuro , Hipóxia-Isquemia Encefálica/patologia , Apoptose , RNA Mensageiro
4.
Chem Biol Interact ; 361: 109978, 2022 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-35561756

RESUMO

Human leukemia U937 cells that were continuously treated with hydroquinone (HQ) were transformed into U937/HQ cells with increased MCL1 and BCL2L1 expression. Compared with their parental cells, U937/HQ cells were less sensitive to ABT-263 (BCL2/BCL2L1 inhibitor)/ABT-199 (BCL2 inhibitor) cytotoxicity. The combination of WEHI-539 (BCL2L1 inhibitor) with either ABT-199 or ABT-263 showed synergistic cytotoxicity to U937 and U937/HQ cells. Therefore, we further investigated the cytotoxic mechanism induced by the combination of WEHI-539 and ABT-199. The combined treatment of WEHI-539 and ABT-199 induced NOX4/ROS/p38 MAPK axis-mediated autophagy, which in turn accelerated ß-TrCP mRNA turnover. Downregulation of ß-TrCP increased Sp1 expression, thereby promoting Sp1-mediated NOXA transcription, which in turn induced NOXA-dependent MCL1 degradation. Enforced expression of MCL1 alleviated the cytotoxicity of WEHI-539 plus ABT-199 to induce the loss of mitochondrial membrane potential and cell viability. WEHI-539 alone induced Sp1/NOXA axis-mediated MCL1 downregulation, while ABT-199 significantly decreased the dose of WEHI-539 by approximately 350- and 50-fold to induce MCL1 suppression in parental and HQ-selected cells, respectively. Furthermore, WEHI-539 sensitized ABT-199-resistant U937 cells to ABT-199 cytotoxicity by inducing NOXA-mediated degradation of MCL1. Collectively, the data in this study indicate that ABT-199 and WEHI-539 cooperatively induce NOXA-dependent MCL1 degradation, and the inhibition of MCL1 mainly explains their combined cytotoxicity in parental, HQ-selected, and ABT-199-resistant U937 cells.


Assuntos
Antineoplásicos , Leucemia , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Proteína bcl-X/metabolismo , Proteínas Contendo Repetições de beta-Transducina/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes , Linhagem Celular Tumoral , Leucemia/tratamento farmacológico , Leucemia/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sulfonamidas
5.
Oncogene ; 39(1): 136-150, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31462707

RESUMO

Hypoxia, or the deficiency of oxygen, in solid tumors is majorly responsible for the progression of cancer and remains unaffected by chemotherapy, but still requires definitive definition of the hypoxia signaling. Hypoxia disrupts the complete folding of mitochondrial proteins, leading to several diseases. The present study confirms that hypoxia activates the Hedgehog pathway in colorectal cancer (CRC), considering its role in cancer epithelial to mesenchymal transition, migration, and invasion. The activity of hypoxia-mediated Gli-1, a Hedgehog signaling factor in hypoxia, was confirmed by in vitro western blotting, immunofluorescence staining, wound-healing assay, and matrigel invasion assay, as well as by in vivo xenograft models (n = 5 per group). The Gli-1 mechanism in hypoxia was analyzed via mass spectrometry. Hypoxia enhanced the interaction of Gli-1 and T-complex protein 1 subunit beta (CCT2), as observed in the mass spectrometric analysis. We observed that reduction in CCT2 inhibits tumor induction by Gli-1. Ubiquitination-mediated Gli-1 degradation by ß-TrCP occurs during incomplete folding of Gli-1 in hypoxia. The human CRC tissues revealed greater CCT2 expression than did the normal colon tissues, indicating that higher CCT2 expression in tumor tissues from CRC patients reduced their survival rate. Moreover, we suggest that CCT2 correlates with Gli-1 expression and is an important determinant of survival in the CRC patients. The results reveal that CCT2 can regulate the folding of Gli-1 in relation to hypoxia in CRC.


Assuntos
Chaperonina com TCP-1/genética , Neoplasias Colorretais/genética , Hipóxia Tumoral/genética , Proteína GLI1 em Dedos de Zinco/genética , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Chaperonina com TCP-1/química , Neoplasias Colorretais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Proteínas Hedgehog/genética , Xenoenxertos , Humanos , Masculino , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Dobramento de Proteína , Proteólise/efeitos dos fármacos , Transdução de Sinais/genética , Taxa de Sobrevida , Ubiquitinação/genética , Proteína GLI1 em Dedos de Zinco/química , Proteínas Contendo Repetições de beta-Transducina/farmacologia
6.
Prostate ; 79(4): 403-413, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30488478

RESUMO

The substantial availability of hypoxia-inducible factor 1 (HIF-1) for pathophysiological states, such as malignancies and ischemia, is primarily regulated post-translationally through the ubiquitin proteolytic system. The balance between degradation and stabilization of HIF-1α protein is determined by specific E3 ligases. In our search for new E3 ligases that might affect HIF-1α protein expression, we studied the effects of beta-transducin repeat-containing protein (ß-TrCP) on the hypoxic pathway in cancer cells. ß-TrCP is overexpressed in many tumors and regulates various cellular processes through mediating the degradation of important targets. Unexpectedly, we found that ß-TrCP overexpression increases HIF-1α protein expression level as well as HIF-1 transcriptional activity by stabilizing HIF-1α protein and preventing its ubiquitination and proteasomal degradation in prostate cancer cells. By using a proteomic approach, we succeeded in demonstrating that ß-TrCP interferes with the association between HIF-1α and HSP70/CHIP, a HIF-1α established E3 ligase complex. Whereas the E3 ligase activity of ß-TrCP is well known, antagonizing another E3 ligase is a new mechanism of action of this important E3. We suggest that destroying or suppressing ß-TrCP and thereby interrupting the HIF-1 pathway, could be valuable antitumor therapy.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias da Próstata/metabolismo , Regulação para Cima/fisiologia , Proteínas Contendo Repetições de beta-Transducina/fisiologia , Linhagem Celular Tumoral , Expressão Gênica , Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico HSP70/química , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Fosfotreonina/metabolismo , Proteômica , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Regulação para Cima/efeitos dos fármacos , Proteínas Contendo Repetições de beta-Transducina/genética , Proteínas Contendo Repetições de beta-Transducina/farmacologia
7.
Cancer Res ; 64(11): 3949-57, 2004 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15173007

RESUMO

Cyclin-dependent kinases (cdks) represent potentially promising molecular targets for cancer therapeutic strategies. To evaluate the antitumor activity of selective cyclin/cdk inhibition, we constructed a chimeric protein composed of a F-box protein (TrCP) fused to a peptide comprising the cyclin/cdk2 binding motif in p21-like cdk inhibitors (TrCP-LFG). We now demonstrate that endogenous cyclin A and its binding substrate, cdk2, can be tethered to beta-TrCP, ubiquitinated, and effectively degraded. Degradation of cdk2 and cyclin A together, but not cdk2 alone, results in massive tumor cell apoptosis in vitro and in vivo in a proteasome-dependent manner with no toxicity to normal tissue. These data demonstrate that cyclin A and/or the cyclin A/cdk2 complex is a promising anticancer target with a high therapeutic index.


Assuntos
Quinases relacionadas a CDC2 e CDC28/antagonistas & inibidores , Proteínas de Transporte/farmacologia , Ciclina A/antagonistas & inibidores , Cisteína Endopeptidases/metabolismo , Complexos Multienzimáticos/metabolismo , Proteínas Contendo Repetições de beta-Transducina/farmacologia , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose , Quinases relacionadas a CDC2 e CDC28/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina , Feminino , Glioma/tratamento farmacológico , Glioma/patologia , Células HeLa , Humanos , Proteínas de Membrana , Camundongos , Camundongos Nus , Complexo de Endopeptidases do Proteassoma , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Contendo Repetições de beta-Transducina/genética , Proteínas Contendo Repetições de beta-Transducina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA