Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Cancer Res ; 12(11): 5049-5061, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36504903

RESUMO

Recurrent and/or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) represents an advanced stage of the disease and frequently shows resistance to these current treatments, including platinum chemotherapy, cetuximab plus chemotherapy, and checkpoint inhibitors. EGFR overexpression and TP53 mutation are the most frequent genetic changes in patients with HNSCC. On the basis of this genetic feature, we proposed a combinatorial treatment using the EGFR tyrosine kinase inhibitor osimertinib (AZD) and arsenic trioxide (ATO) for compassionate use. The patient obtained treatment response and progression-free survival for about six months. In vitro mechanical verifications showed that ATO and AZD combination (ATO/AZD) significantly increased intracellular ROS levels and DNA damage. Additionally, ATO/AZD decreases the expression and activity of breast cancer type 1 susceptibility protein (BRCA1) and polo-like kinase 1 (PLK1), thereby impairing Rad51 recruitment to DNA double-strand lesion for repair and may ultimately cause tumor cell death. In conclusion, this study provides a concrete experience and an alternate strategy of ATO/AZD therapy for patients with R/M HNSCC.

2.
JCI Insight ; 7(23)2022 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-36264639

RESUMO

Docetaxel (DTX) combined with cisplatin and 5-fluorouracil has been used as induction chemotherapy for head and neck squamous cell carcinoma (HNSCC). However, the development of acquired resistance remains a major obstacle to treatment response. Tumor-associated macrophages are associated with chemotherapeutic resistance. In the present study, increased infiltration of macrophages into the tumor microenvironment (TME) was significantly associated with shorter overall survival and increased resistance to chemotherapeutic drugs, particularly DTX, in patients with HNSCC. Macrophage coculture induced expression of intercellular adhesion molecule 1 (ICAM1), which promotes stemness and the formation of polyploid giant cancer cells, thereby reducing the efficacy of DTX. Both genetic silencing and pharmacological inhibition of ICAM1 sensitized HNSCC to DTX. Macrophage secretion of IL-1ß was found to induce tumor expression of ICAM1. IL-1ß neutralization and IL-1 receptor blockade reversed DTX resistance induced by macrophage coculture. IL-1ß activated superoxide dismutase 2 and inhibited catalase, thereby modulating intracellular levels of ROS and inducing ICAM1 expression. Arsenic trioxide (ATO) reduced macrophage infiltration into the TME and impaired IL-1ß secretion by macrophages. The combinatorial use of ATO enhanced the in vivo efficacy of DTX in a mouse model, which may provide a revolutionary approach to overcoming acquired therapeutic resistance in HNSCC.


Assuntos
Docetaxel , Neoplasias de Cabeça e Pescoço , Molécula 1 de Adesão Intercelular , Interleucina-1beta , Carcinoma de Células Escamosas de Cabeça e Pescoço , Animais , Camundongos , Docetaxel/farmacologia , Docetaxel/uso terapêutico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Molécula 1 de Adesão Intercelular/genética , Macrófagos , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Microambiente Tumoral , Humanos , Interleucina-1beta/metabolismo , Transdução de Sinais
3.
Int J Mol Sci ; 23(13)2022 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-35806005

RESUMO

Nerve injury of the central nervous system and the peripheral nervous system still poses a major challenge in modern clinics. Understanding the roles of neurotrophic factors and their molecular mechanisms on neuro-regeneration will not only benefit patients with neural damage but could potentially treat neurodegenerative disorders, such as amyotrophic lateral sclerosis. In this study, we showed that human IL12 p40-p40 homodimer (hIL12p80) within PLA and PLGA conduits improved sciatic nerve regeneration in mice. As such, the group of conduits with NSCs and hIL12p80 (CNI) showed the best recovery among the groups in the sciatic functional index (SFI), compound muscle action potential (CMAP), and Rotarod performance analyses. In addition, the CNI group had a faster recovery and outperformed the other groups in SFI and Rotarod performance tests beginning in the fourth week post-surgery. Immunohistochemistry showed that the CNI group increased the diameter of the newly regenerated nerve by two-fold (p < 0.01). In vitro studies showed that hIL12p80 stimulated differentiation of mouse NSCs to oligodendrocyte lineages through phosphorylation of Stat3 at Y705 and S727. Furthermore, implantation using PLGA conduits (C2.0 and C2.1) showed better recovery in the Rotarod test and CMAP than using PLA conduits in FVB mice. In B6 mice, the group with C2.1 + NSCs + hIL12p80 (C2.1NI) not only promoted sciatic functional recovery but also reduced the rate of experimental autotomy. These results suggested that hIL12p80, combined with NSCs, enhanced the functional recovery and accelerated the regeneration of damaged nerves in the sciatic nerve injury mice. Our findings could further shed light on IL12's application not only in damaged nerves but also in rectifying the oligodendrocytes' defects in neurodegenerative diseases, such as amyotrophic lateral sclerosis and multiple sclerosis.


Assuntos
Esclerose Lateral Amiotrófica , Interleucina-12 , Traumatismos dos Nervos Periféricos , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/terapia , Animais , Humanos , Interleucina-12/metabolismo , Camundongos , Regeneração Nervosa/fisiologia , Oligodendroglia , Traumatismos dos Nervos Periféricos/metabolismo , Traumatismos dos Nervos Periféricos/patologia , Traumatismos dos Nervos Periféricos/terapia , Nervo Isquiático/lesões
4.
Mol Cell Neurosci ; 79: 1-11, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27865767

RESUMO

Regeneration of injured peripheral nerves is a slow, complicated process that could be improved by implantation of neural stem cells (NSCs) or nerve conduit. Implantation of NSCs along with conduits promotes the regeneration of damaged nerve, likely because (i) conduit supports and guides axonal growth from one nerve stump to the other, while preventing fibrous tissue ingrowth and retaining neurotrophic factors; and (ii) implanted NSCs differentiate into Schwann cells and maintain a growth factor enriched microenvironment, which promotes nerve regeneration. In this study, we identified IL12p80 (homodimer of IL12p40) in the cell extracts of implanted nerve conduit combined with NSCs by using protein antibody array and Western blotting. Levels of IL12p80 in these conduits are 1.6-fold higher than those in conduits without NSCs. In the sciatic nerve injury mouse model, implantation of NSCs combined with nerve conduit and IL12p80 improves motor recovery and increases the diameter up to 4.5-fold, at the medial site of the regenerated nerve. In vitro study further revealed that IL12p80 stimulates the Schwann cell differentiation of mouse NSCs through the phosphorylation of signal transducer and activator of transcription 3 (Stat3). These results suggest that IL12p80 can trigger Schwann cell differentiation of mouse NSCs through Stat3 phosphorylation and enhance the functional recovery and the diameter of regenerated nerves in a mouse sciatic nerve injury model.


Assuntos
Interleucina-12/metabolismo , Regeneração Nervosa , Células-Tronco Neurais/transplante , Neurogênese , Traumatismos dos Nervos Periféricos/terapia , Células de Schwann/citologia , Nervo Isquiático/fisiologia , Animais , Células Cultivadas , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Fator de Transcrição STAT3/metabolismo , Transplante de Células-Tronco
5.
Food Chem Toxicol ; 82: 27-35, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25959841

RESUMO

This study explores human neuroblastoma (SH-SY5Y) and human glioblastoma (U-1240 MG) cellular differentiation changes under exposure to acrylamide (ACR). Differentiation of SH-SY5Y and U-1240 MG cells were induced by retinoic acid (RA) and butyric acid (BA), respectively. Morphological observations and MTT assay showed that the induced cellular differentiation and cell proliferation were inhibited by ACR in a time- and dose-dependent manner. ACR co-treatment with RA attenuated SH-SY5Y expressions of neurofilament protein-L (NF-L), microtubule-associated protein 1b (MAP1b; 1.2 to 0.7, p < 0.001), MAP2c (2.2 to 0.8, p < 0.05), and Janus kinase1 (JAK1; 1.9 to 0.6, p < 0.001), while ACR co-treatment with BA attenuated U-1240 MG expressions of glial fibrillary acidic protein (GFAP), MAP1b (1.2 to 0.6, p < 0.001), MAP2c (1.5 to 0.7, p < 0.01), and JAK1 (2.1 to 0.5, p < 0.001), respectively. ACR also decreased the phosphorylation of extracellular-signal-regulated kinases (ERK) and c-Jun N-terminal kinases (JNK) in U-1240 MG cells, while caffeine reversed this suppression of ERK and JNK phosphorylation caused by ACR treatment. These results showed that RA-induced neurogenesis of SH-SY5Y and BA-induced astrogliogenesis of U-1240 MG cells were attenuated by ACR and were associated with down-regulation of MAPs expression and JAK-STAT signaling.


Assuntos
Acrilamida/toxicidade , Diferenciação Celular/efeitos dos fármacos , Glioblastoma/patologia , Neuroblastoma/patologia , Ácido Butírico/farmacologia , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Humanos , Janus Quinases/metabolismo , Neuritos/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/metabolismo , Fosforilação/efeitos dos fármacos , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tretinoína/farmacologia
6.
Cell Transplant ; 24(5): 779-96, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24380568

RESUMO

Previous studies showed that transplantation of cultured neural stem/progenitor cells (NSPCs) could improve functional recovery for various neurological diseases. This study aims to develop a stem cell-based model for predictive toxicology of development in the neurological system after acrylamide exposure. Treatment of mouse (KT98/F1B-GFP) and human (U-1240 MG/F1B-GFP) NSPCs with 0.5 mM acrylamide resulted in the inhibition of neurosphere formation (definition of self-renewal ability in NSPCs), but not inhibition of cell proliferation. Apoptosis and differentiation of KT98 (a precursor of KT98/F1B-GFP) and KT98/F1B-GFP are not observed in acrylamide-treated neurospheres. Analysis of secondary neurosphere formation and differentiation of neurons and glia illustrated that acrylamide-treated KT98 and KT98/F1B-GFP neurospheres retain the NSPC properties, such as self-renewal and differentiation capacity. Correlation of acrylamide-inhibited neurosphere formation with cell-cell adhesion was observed in mouse NSPCs by live cell image analysis and the presence of acrylamide. Protein expression levels of cell adhesion molecules [neural cell adhesion molecule (NCAM) and N-cadherin] and extracellular signal-regulated kinases (ERK) in acrylamide-treated KT98/F1B-GFP and U-1240 MG/F1B-GFP neurospheres demonstrated that NCAM decreased and phospho-ERK (pERK) increased, whereas expression of N-cadherin remained unchanged. Analysis of AKT (protein kinase B, PKB)/ß-catenin pathway showed decrease in phospho-AKT (p-AKT) and cyclin D1 expression in acrylamide-treated neurospheres of KT98/F1B-GFP. Furthermore, PD98059, an ERK phosphorylation inhibitor, attenuated acrylamide-induced ERK phosphorylation, indicating that pERK contributed to the cell proliferation, but not in neurosphere formation in mouse NSPCs. Coimmunoprecipitation results of KT98/F1B-GFP cell lysates showed that the complex of NCAM and fibroblast growth factor receptor 1 (FGFR1) is present in the neurosphere, and the amount of this complex decreases after acrylamide treatment. Our results reveal that acrylamide inhibits neurosphere formation through the disruption of the neurosphere architecture in NSPCs. The downregulation of cell-cell adhesion resulted from decreasing the levels of NCAM as well as the formation of NCAM/FGFR complex.


Assuntos
Acrilamida/farmacologia , Células-Tronco Neurais/metabolismo , Esferoides Celulares/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Técnicas de Cultura de Células , Células Cultivadas , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Esferoides Celulares/citologia
7.
Arch Toxicol ; 88(3): 769-80, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24318646

RESUMO

Acrylamide is a neurological and reproductive toxicant in humans and laboratory animals; however, the neuron developmental toxicity of acrylamide remains unclear. The aims of this study are to investigate the cytotoxicity and neurite outgrowth inhibition of acrylamide in nerve growth factor (NGF)- or fibroblast growth factor 1 (FGF1)-mediated neural development of PC12 cells. MTS assay showed that acrylamide treatment suppresses NGF- or FGF1-induced PC12 cell proliferation in a time- and dose-dependent manner. Quantification of neurite outgrowth demonstrated that 0.5 mM acrylamide treatment resulted in significant decrease in differentiation of NGF- or FGF1-stimulated PC12 cells. This decrease is accompanied with the reduced expression of growth-associated protein-43, a neuronal marker. Moreover, relative levels of pERK, pAKT, pSTAT3 and pCREB were increased within 5-10 min when PC12 cells were treated with NGF or FGF1. Acrylamide (0.5 mM) decreases the NGF-induced activation of AKT-CREB but not ERK-STAT3 within 20 min. Similarly, acrylamide (0.5 mM) decreases the FGF1-induced activation of AKT-CREB within 20 min. In contrast to the NGF treatment, the ERK-STAT3 activation that was induced by FGF1 was slightly reduced by 0.5 mM acrylamide. We further showed that PI3K inhibitor (LY294002), but not MEK inhibitor (U0126), could synergize with acrylamide (0.5 mM) to reduce the cell viability and neurite outgrowth in NGF- or FGF1-stimulated PC12 cells. Moreover, acrylamide (0.5 mM) increased reactive oxygen species (ROS) activities in NGF- or FGF1-stimulated PC12 cells. This increase was reversed by Trolox (an ROS scavenging agent) co-treatment. Together, our findings reveal that NGF- or FGF1-stimulation of the neuronal differentiation of PC12 cells is attenuated by acrylamide through the inhibition of PI3K-AKT-CREB signaling, along with the production of ROS.


Assuntos
Acrilamida/toxicidade , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fator de Crescimento Neural/farmacologia , Células PC12/efeitos dos fármacos , Animais , Butadienos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Cromonas/farmacologia , Morfolinas/farmacologia , Neuritos/efeitos dos fármacos , Nitrilas/farmacologia , Células PC12/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
Food Chem Toxicol ; 51: 446-52, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23127597

RESUMO

Acrylamide (ACR) can be produced during food processing and has neurotoxic effects in humans. This study aims to determine ACR induced apoptotic responses in human astrocytoma U-1240 MG cells to realize the incurred toxic mechanisms. Under 1 and 2mM ACR exposure, cell viability decreased as time increased. The increments in sub-G(1) phase were 87.5-fold, and pro-caspase 3 and PARP protein expressions decreased 35% and 54.5% respectively relative to the control after 2mM ACR treatment. Molecular evidence of Bax/bcl-2 ratio and cytochrome c expression increased 8.86-fold and 6.81-fold as well as pro-caspase 9 decreased 67.8% relative to the control respectively under 2mM ACR exposure. Trolox, an ROS scavenging agent, attenuated cell death and induced ROS production by 2mM ACR. The ultrastructure alterations of mitochondria showed marked vesicular matrix compartmentalization and cytoplasmic vacuole formation after 2mM ACR was treated for 48h, whereas those treated for 72h showed chromatin condensation, pyknosis, and swelling. These results indicate long-term exposure to ACR induced mitochondria collapse and finally led to apoptosis. Although 2mM ACR is higher than average daily intake dosage, workers in chemical industries may be exposed to sufficient doses to entail health risks.


Assuntos
Acrilamida/toxicidade , Apoptose/efeitos dos fármacos , Astrocitoma/patologia , Mitocôndrias/efeitos dos fármacos , Apoptose/fisiologia , Astrocitoma/induzido quimicamente , Astrocitoma/metabolismo , Caspase 3/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cromanos/farmacologia , Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Fase G1/efeitos dos fármacos , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Poli(ADP-Ribose) Polimerases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Testes de Toxicidade/métodos , Proteína X Associada a bcl-2/metabolismo
9.
Chem Res Toxicol ; 23(9): 1449-58, 2010 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-20734998

RESUMO

Acrylamide (ACR) has been recognized as a neurological and reproductive toxin in humans and laboratory animals. This study aimed to determine the effects of ACR-induced DNA damage on cell cycle regulation in human astrocytoma cell lines. Treatment of U-1240 MG cells with 2 mM ACR for 48 h resulted in a significant inhibition of cell proliferation as evaluated by Ki-67 protein expression and MTT assay. The analysis of DNA damage with the comet assay showed that treatment of the cells with 0.5, 1, and 2 mM ACR for 48 h caused significant increases in DNA damage by 3.5-, 4-, and 14-fold, respectively. Meanwhile, analysis of cell-cycle arrest with flow cytometry revealed that the ACR treatments resulted in significant increases in the G(0)/G(1)-arrested cells in a time- and dose-dependent manner. Expression of DNA damage-associated/checkpoint-related signaling molecules, including phosphorylated-p53 (pp53), p53, p21, p27, Cdk2, and cyclin D(1), in three human astrocytoma cell lines (U-1240 MG, U-251 MG, and U-87 MG) was also analyzed by immunoblotting. Treatment of the three cell lines with 2 mM ACR for 48 h caused marked increases in pp53 and Cdk2, as well as decreases in cyclin D(1) and p27. Moreover, increases in p53 and p21 were detected in both U-1240 and U-87 MG cells, whereas no marked change in p53 and a decrease in p21 were observed in U-251 MG cells. To address the involvement of ataxia telangiectasia mutated/ATM-Rad3-related (ATM/ATR) kinase in the signaling of ACR-induced G(0)/G(1) arrest, caffeine was used to block the ATM/ATR pathway in U-1240 MG cells. Caffeine significantly attenuated the ACR-induced G(0)/G(1) arrest as well as the expression of DNA damage-associated/checkpoint-related signaling molecules in a dose-dependent manner. This in vitro study clearly demonstrates the critical role of ATM/ATR in the signaling of ACR-induced cell-cycle arrest in astrocytoma cells.


Assuntos
Acrilamida/toxicidade , Astrócitos/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Dano ao DNA , Acrilamida/química , Astrocitoma , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaio Cometa , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fase G1/efeitos dos fármacos , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo
10.
Toxicol In Vitro ; 23(5): 855-61, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19426797

RESUMO

This study was to clarify whether acrylamide (ACR) will induce apoptosis and astrogliosis in an astrocytic cell line in vitro. Different time- and dose-dependent cytotoxic studies were conducted upon neuronal (SH-SY5Y) and glial cell lines (U-1240 MG) under exposure to ACR up to 72h. We showed that SH-SY5Y cells were more sensitive in cytotoxic assays than U-1240 MG cells, and significantly decreased cell viability was observed at concentrations higher than 1mM with increased lactate dehydrogenase leakage observed only at 5 and 10mM in U-1240 MG cells. The ACR-induced apoptotic responses and phosphorylation of p53 protein at Ser15 for U-1240 MG cells were identified at 48h. The increase of glial fibrillary acidic protein (GFAP) as a chemical-induced astrogliotic response was found to be associated with different ACR concentrations and exposure times, particularly at >or=48h of >or=2mM. In addition, immunocytochemical staining at 36h of 5 and 10mM treatments had significantly higher density of GFAP than the control. Thus, ACR-induced effects can be seen in neuronal and astrocytic cells. These results suggest that ACR exposure may lead to apoptotic and astrogliotic effects in human astrocytoma cells in vitro in a time- and dose-dependent manner.


Assuntos
Acrilamida/toxicidade , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Neuroblastoma/patologia , Acrilamida/administração & dosagem , Astrócitos/patologia , Astrocitoma/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Proteína Glial Fibrilar Ácida/efeitos dos fármacos , Humanos , L-Lactato Desidrogenase/metabolismo , Fosforilação/efeitos dos fármacos , Fatores de Tempo , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...