Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 35(5): e21601, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33913201

RESUMO

Peritoneal dissemination threatens the survival of patients with gastric cancer (GC). Bufalin is an extract of traditional Chinese medicine, which has been proved to have anticancer effect. The target of bufalin in suppressing gastric cancer peritoneal dissemination (GCPD) and the underlying mechanism are still unclear. In this research, GC cell line MGC-803 and high-potential peritoneal dissemination cell line MKN-45P were treated with bufalin or L-NAME. Malignant biological behavior and protein level of GC cell lines were detected with MTT, wound healing, transwell, adhesion, and western blotting. Bioinformatics analysis and patient tissues were used to verify the role of endothelial nitric oxide synthase (NOS3) in GC. Mice model was used to assess the effect of bufalin and role of NOS3 in vivo. We found that bufalin inhibited the proliferation, invasion, and migration in GC cell lines. NOS3, which was an independent prognostic factor of GC patients, was predicted to be a potential target of bufalin. Further experiments proved that bufalin reduced the phosphorylation of NOS3, thereby inhibiting the mitogen-activated protein kinase (MAPK) signaling pathway, and ultimately suppressed GCPD by inhibiting EMT process. In conclusion, NOS3 was a potential therapeutic target and prognostic biomarker of GC. Bufalin could suppress GCPD through NOS3-MAPK signaling pathway, which provided more evidence support for intraperitoneal perfusion of bufalin to treat GCPD.


Assuntos
Biomarcadores Tumorais/metabolismo , Bufanolídeos/farmacologia , Regulação Neoplásica da Expressão Gênica , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Neoplasias Peritoneais/tratamento farmacológico , Neoplasias Gástricas/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Quinases Ativadas por Mitógeno/genética , Óxido Nítrico Sintase Tipo III/genética , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/secundário , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Ecotoxicol Environ Saf ; 242: 113868, 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-35863215

RESUMO

Environmental pollution has emerged as a major threat to bird populations. Many shorebird populations are declining, although contamination has been documented in some shorebirds, evidence of negative impacts is sparse and this important topic remains understudied. To guide future research and develop effective conservation strategies, we carried out a comprehensive review of environmental pollutants and their consequences on shorebirds. In total, we found 93 relevant articles which examined pollutant contamination in ~37% (79 of 215) of all shorebird species, mostly from the Charadriidae and Scolopacidae families. Studies were geographically biased: the majority were conducted in American flyways, while only 1 was found from Australasia and few were conducted in Asian flyways. The main geographic gap for research includes East Africa, South Asia and Siberian Arctic. The most well-documented pollutants included mercury (Hg, 37 studies), cadmium (33), and lead (Pb, 28); less well studied pollutants were barium (1), calcium (1), strontium (1), dicofols (1), and other newly emerging contaminants, such as plastic debris/microplastics (4) and antibiotics resistance (2). Several pollutants have caused considerable concerns in shorebirds, including embryotoxicity caused by PCBs at non-optimum temperature (laboratory experiments); reduced reproduction performance linked to maternal Hg and paternal Pb (field evidence); and reduced refueling and flight performance related to oil contamination (both field and laboratory evidence). Our results confirm that an in-depth understanding of the local, regional and global factors that influence population trends of shorebirds in light of increasing pollution threats is essential for accurate and effective management and conservation strategies.


Assuntos
Charadriiformes , Poluentes Ambientais , Mercúrio , Animais , Aves , Monitoramento Ambiental , Poluentes Ambientais/análise , Poluentes Ambientais/toxicidade , Humanos , Chumbo , Plásticos
3.
J Pathol ; 249(1): 26-38, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30953361

RESUMO

Mesenchymal glioblastoma (GBM) is the most aggressive subtype of GBM. Our previous study found that neurotrophic factor prosaposin (PSAP) is highly expressed and secreted in glioma and can promote the growth of glioma. The role of PSAP in mesenchymal GBM is still unclear. In this study, bioinformatic analysis, western blotting and RT-qPCR were used to detect the expression of PSAP in different GBM subtypes. Human glioma cell lines and patient-derived glioma stem cells were studied in vitro and in vivo, revealing that mesenchymal GBM expressed and secreted the highest level of PSAP among four subtypes of GBM, and PSAP could promote GBM invasion and epithelial-mesenchymal transition (EMT)-like processes in vivo and in vitro. Bioinformatic analysis and western blotting showed that PSAP mainly played a regulatory role in GBM invasion and EMT-like processes via the TGF-ß1/Smad signaling pathway. In conclusion, the overexpression and secretion of PSAP may be an important factor causing the high invasiveness of mesenchymal GBM. PSAP is therefore a potential target for the treatment of mesenchymal GBM. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Neoplasias Encefálicas/metabolismo , Transição Epitelial-Mesenquimal , Glioblastoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Saposinas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica , Células-Tronco Neoplásicas/patologia , Fosforilação , Saposinas/genética , Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/genética , Células Tumorais Cultivadas
4.
Crit Rev Microbiol ; 45(2): 239-251, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30776938

RESUMO

Epidemiological studies have shown that Helicobacter pylori (HP) infection is a risk factor for gastric cancer (GC). HP infection may induce the release of pro-inflammatory mediators, and abnormally increase the level of reactive oxygen species (ROS), nitric oxide (NO), and cytokines in mucosal epithelial cells of the stomach. However, the specific mechanism underlying the pathogenesis of HP-associated GC is still poorly understood. Recent studies have revealed that abnormal microRNA expression may affect the proliferation, differentiation, and apoptosis of mucosal epithelial cells of the stomach to further influence GC occurrence, development, and metastasis. Herein, we summarize the role of abnormal microRNAs in the regulation of HP-associated GC progression. Abnormal microRNA expression in HP-positive GC may be a biomarker for GC diagnosis, occurrence, and development as well as its targeted treatment and prognosis.


Assuntos
Infecções por Helicobacter/genética , Helicobacter pylori/fisiologia , MicroRNAs/genética , Neoplasias Gástricas/genética , Animais , Apoptose , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/fisiopatologia , Humanos , MicroRNAs/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/fisiopatologia
5.
Cell Biol Int ; 42(10): 1377-1385, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29957841

RESUMO

ß-Elemene, an anti-cancer drug extracted from traditional Chinese medicinal herb, showed anti-tumor effects on gastric cancer cells. Our previous studies reported gastric cancer cells are insensitive to TRAIL. However, whether ß-elemene could enhance anti-cancer effects of TRAIL on gastric cancer cells is unknown. In our present study, ß-elemene prevented gastric cancer cell viability in dose-dependent manner, and when combined with TRAIL, obviously inhibited proliferation and promoted apoptosis in gastric cancer cells. Compared to ß-elemene or TRAIL alone, treatment with ß-elemene and TRAIL obviously promoted DR5 clustering as well as translocation of Caspase-8, DR5 and FADD into lipid rafts. This led to cleavage of Caspase-8 and the formation of death-inducing signaling complex (DISC) in lipid rafts. The cholesterol-sequestering agent nystatin partially reversed DR5 clustering and DISC formation, preventing apoptosis triggered by the combination of ß-elemene and TRAIL. Our results suggest that ß-elemene increases the sensitivity of gastric cancer cells to TRAIL partially by promoting the formation of DISC in lipid rafts.


Assuntos
Sesquiterpenos/metabolismo , Sesquiterpenos/farmacologia , Neoplasias Gástricas/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspase 8/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , China , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/efeitos dos fármacos , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Microdomínios da Membrana , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/patologia , Ligante Indutor de Apoptose Relacionado a TNF/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia
6.
BMC Cancer ; 17(1): 492, 2017 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-28724364

RESUMO

BACKGROUND: Although the efficacy of tamoxifen (TAM) for breast cancer has been attributed to inducing cell cycle arrest and apoptosis by inhibiting estrogen receptor (ER) signaling, recent evidence indicates that TAM also possesses ER-independent antitumor activity through an unclear mechanism. The present study investigated the anti-tumor mechanism of TAM on mesenchymal triple-negative breast cancer (TNBC). METHODS: The inhibitory effect of TAM on tumor migration and metastasis was analyzed by transwell chamber in vitro and by murine xenograft model in vivo. The promoter sequence of miR-200c was predicted by an online CpG island predictor. Relative expression of miR-200c was measured by quantitative real-time PCR. RESULTS: After treatment with TAM, mesenchymal TNBC cells (MCF-7/ADR and MDA-MB-231) morphologically changed from mesenchymal to epithelial types. Meanwhile, cell migration ability was also significantly decreased in ER-positive breast cancer cells after exposure to TAM. Consistent with these in-vitro results, TAM significantly suppressed lung metastasis rate of mesenchymal TNBC cells in murine xenograft tumors. miRNA array analysis of two types of breast cancer cells showed that miR-200c expression was inhibited in mesenchymal TNBC cells, but increased after TAM treatment due to demethylation of miR-200c promoters. CONCLUSIONS: Our results indicate that TAM inhibits cell migration and enhances chemosensitivity of mesenchymal TNBC cells by reversing their EMT-like property; and that this EMT-reversal effect results from upregulation of miR-200c through demethylating its promoter. To our knowledge, this is the first explanation of a non-ER-related mechanism for the effect of TAM on mesenchymal TNBC cells.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , MicroRNAs/efeitos dos fármacos , Tamoxifeno/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Antineoplásicos Hormonais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Desmetilação do DNA , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Camundongos , MicroRNAs/química , MicroRNAs/genética , MicroRNAs/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais , Tamoxifeno/uso terapêutico , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Anticancer Drugs ; 27(1): 9-16, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26398931

RESUMO

Bufalin, a cardiotonic steroid isolated from toad venom, has been shown to kill various types of tumor cells. Our previous study showed that triple-negative breast cancer (TNBC) cells were less sensitive to bufalin than other types of breast cancer cells, but the reason for this lower sensitivity remains unclear. In this study, we showed that bufalin induced apoptosis in MDA-MB-231 and MCF-7/ADR TNBC cell lines, accompanied by increased miR-155-5p expression. Overexpression of miR-155-5p promoted proliferation and reduced bufalin-induced apoptosis of TNBC cells. In contrast, downregulation of miR-155-5p increased sensitivity to bufalin and upregulated the expression of FOXO3A. Bufalin also downregulated DNA methyltransferases 1 and 3a (DNMT1 and DNMT3a), and concurrent inhibition of DNMT1 and DNMT3a significantly increased miR-155-5p expression. These results indicate that miR-155-5p antagonizes bufalin sensitivity in TNBC cells, and that downregulation of DNMT1 and DNMT3a may be responsible for the bufalin-induced upregulation of miR-155-5p.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Bufanolídeos/farmacologia , MicroRNAs/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Humanos
8.
Biochim Biophys Acta ; 1842(4): 635-45, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24365239

RESUMO

Impairment of epithelial barrier is observed in various intestinal disorders including inflammatory bowel diseases (IBD). Numerous factors may cause temporary damage of the intestinal epithelium. A complex network of highly divergent factors regulates healing of the epithelium to prevent inflammatory response. However, the exact repair mechanisms involved in maintaining homeostatic intestinal barrier integrity remain to be clarified. In this study, we demonstrate that activation of M1 muscarinic acetylcholine receptor (mAChR) augments the restitution of epithelial barrier function in T84 cell monolayers after ethanol-induced epithelial injury, via ERK-dependent phosphorylation of focal adhesion kinase (FAK). We have shown that ethanol injury decreased the transepithelial electrical resistance (TER) along with the reduction of ERK and FAK phosphorylation. Carbachol (CCh) increased ERK and FAK phosphorylation with enhanced TER recovery, which was completely blocked by either MT-7 (M1 antagonist) or atropine. The CCh-induced enhancement of TER recovery was also blocked by either U0126 (ERK pathway inhibitor) or PF-228 (FAK inhibitor). Treatment of T84 cell monolayers with interferon-γ (IFN-γ) impaired the barrier function with the reduction of FAK phosphorylation. The CCh-induced ERK and FAK phosphorylation were also attenuated by the IFN-γ treatment. Immunological and binding experiments exhibited a significant reduction of M1 mAChR after IFN-γ treatment. The reduction of M1 mAChR in inflammatory area was also observed in surgical specimens from IBD patients, using immunohistochemical analysis. These findings provide important clues regarding mechanisms by which M1 mAChR participates in the maintenance of intestinal barrier function under not only physiological but also pathological conditions.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Mucosa Intestinal/metabolismo , Receptor Muscarínico M1/fisiologia , Linhagem Celular Tumoral , Impedância Elétrica , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/análise , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/análise , Humanos , Imuno-Histoquímica , Interferon gama/farmacologia , Fosforilação , Receptor Muscarínico M1/análise
9.
Biochim Biophys Acta ; 1832(1): 151-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23064287

RESUMO

Disruption of epithelial barrier function was identified as one of the pathologic mechanisms in inflammatory bowel diseases (IBD). Epithelial barrier consists of various intercellular junctions, in which the tight junction (TJ) is an important component. However, the regulatory mechanism of tight junction is still not clear. Here we examined the role of focal adhesion kinase (FAK) in the epithelial barrier function on Caco-2 monolayers using a specific FAK inhibitor, PF-573, 228 (PF-228). We found that the decrease of transepithelial resistance and the increase of paracellular permeability were accompanied with the inhibition of autophosphorylation of FAK by PF-228 treatment. In addition, PF-228 inhibited the FAK phosphorylation at Y576/577 on activation loop by Src, suggesting Src-dependent regulation of FAK in Caco-2 monolayers. In an ethanol-induced barrier injury model, PF-228 treatment also inhibited the recovery of transepithelial resistance as well as these phosphorylations of FAK. In a sucrose gradient ultracentrifugation, FAK co-localized with claudin-1, an element of the TJ complex, and they co-migrate after ethanol-induced barrier injury. Immunofluorescence imaging analysis revealed that PF-228 inhibited the FAK redistribution to the cell border and reassembly of TJ proteins in the recovery after ethanol-induced barrier injury. Finally, knockdown of FAK by siRNA resulted in the decrease of transepithelial resistance. These findings reveal that activation of FAK is necessary for maintaining and repairing epithelial barrier in Caco-2 cell monolayer via regulating TJ redistribution.


Assuntos
Células Epiteliais/enzimologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Intestinos/enzimologia , Junções Íntimas/enzimologia , Motivos de Aminoácidos , Células CACO-2 , Proteína-Tirosina Quinases de Adesão Focal/química , Proteína-Tirosina Quinases de Adesão Focal/genética , Humanos , Intestinos/citologia , Fosforilação , Junções Íntimas/genética
10.
Mol Cancer ; 13: 136, 2014 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-24885194

RESUMO

BACKGROUND: Insulin-like growth factor I (IGF-I) can induce epithelial mesenchymal transition (EMT) in many epithelial tumors; however, the molecular mechanism by which this occurs is not clearly understood. Additionally, little is known about the involvement of IGF-I in gastric cancer. METHODS: Two gastric cancer cell lines were treated with IGF-I to induce EMT and levels of transcription factor ZEB2 and microRNA-200c (miR-200c) were measured. Cells were treated with Akt/ERK inhibitors to investigate the role of these pathways in IGF-I-mediated EMT. Transfection of shRNA plasmids was used to silence the ubiquitin ligase Cbl-b to assess its involvement in this process. The relationship between IGF-IR and Cbl-b expression, and the effect of IGF-IR and Cbl-b on metastasis were analyzed in primary gastric adenocarcinoma patients. RESULTS: IGF-I-induced gastric cancer cell EMT was accompanied by ZEB2 up-regulation. Furthermore, both Akt/ERK inhibitors and knockdown of Akt/ERK gene reversed IGF-I-induced ZEB2 up-regulation and EMT through up-regulation of miR-200c, suggesting the involvement of an Akt/ERK-miR-200c-ZEB2 axis in IGF-I-induced EMT. The ubiquitin ligase Cbl-b also ubiquitinated and degraded IGF-IR and inhibited the Akt/ERK-miR-200c-ZEB2 axis, leading to the repression of IGF-I-induced EMT. There was a significant negative correlation between the expression of IGF-IR and Cbl-b in gastric cancer patient tissues (r = -0.265, p < 0.05). More of patients with IGF-IR-positive expression and Cbl-b-negative expression were with lymph node metastasis (p < 0.001). CONCLUSIONS: Together, these findings demonstrate that the ubiquitin ligase Cbl-b represses IGF-I-induced EMT, likely through targeting IGF-IR for degradation and further inhibiting the Akt/ERK-miR-200c-ZEB2 axis in gastric cancer cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Adenocarcinoma/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Fator de Crescimento Insulin-Like I/metabolismo , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-cbl/genética , Proteínas Repressoras/genética , Neoplasias Gástricas/genética , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Idoso , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Proteínas de Homeodomínio/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Metástase Linfática , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Estadiamento de Neoplasias , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-cbl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-cbl/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Homeobox 2 de Ligação a E-box com Dedos de Zinco
11.
Tumour Biol ; 35(11): 11129-35, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25099615

RESUMO

Trypsin is a digestive enzyme that is widely used for cell detachment, which is the first stage of tumor metastasis. Recent studies show that adhesion-related kinases are involved in cell detachment. Proline-rich tyrosine kinase 2 (Pyk2) is a crucial kinase in the regulation of cell adhesion and detachment. However, the effect of Pyk2 on cell detachment is controversial. In the present study, we found that Pyk2 expression was rapidly decreased after trypsin treatment in gastric cancer, breast cancer, colon cancer, lung cancer, and human gastric epithelial cells. Knockdown of Pyk2 accelerated cell detachment. Furthermore, lysosome inhibitor NH4CL suppressed cell detachment and increased ubiquitination of Pyk2. Cbl-b is a type of E3 ubiquitin ligase that interacted with Pyk2, reduced the expression of Pyk2, and promoted trypsin-induced degradation of Pyk2. These findings suggest that Cbl-b promoted cell detachment through mono-ubiquitination of Pyk2. Our data provide a new insight into the role of Cbl-b in cell detachment.


Assuntos
Adesão Celular/efeitos dos fármacos , Quinase 2 de Adesão Focal/metabolismo , Neoplasias/patologia , Proteólise/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Tripsina/farmacologia , Ubiquitinação/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Quinase 2 de Adesão Focal/antagonistas & inibidores , Quinase 2 de Adesão Focal/genética , Humanos , Imunoprecipitação , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , Células Tumorais Cultivadas
12.
Anticancer Drugs ; 25(6): 683-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24710190

RESUMO

Studies have shown that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in cancer cells. However, breast cancer cells are generally resistant to TRAIL. In the present study, we explored the effect of bufalin on TRAIL-induced breast cancer cell apoptosis. The results showed that bufalin enhanced TRAIL-induced apoptosis in MCF-7 and MDA-MB-231 breast cancer cells by activating the extrinsic apoptotic pathway. Bufalin also promoted the clustering of death receptor 4 (DR4) and DR5 in aggregated lipid rafts. The cholesterol-sequestering agent methyl-ß-cyclodextrin reversed the DR4 and DR5 clustering and reduced bufalin+TRAIL-induced apoptosis. Overall, these results indicate that bufalin enhanced TRAIL-induced apoptosis in breast cancer cells by the partial redistribution of DRs in lipid rafts.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Bufanolídeos/farmacologia , Microdomínios da Membrana/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Colesterol/metabolismo , Feminino , Humanos , beta-Ciclodextrinas/farmacologia
13.
Commun Biol ; 7(1): 585, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38755288

RESUMO

Exposure to pollutants is a potentially crucial but overlooked driver of population declines in shorebirds along the East Asian-Australasian Flyway. We combined knowledge of moult strategy and life history with a standardised sampling protocol to assess mercury (Hg) contamination in 984 individuals across 33 migratory shorebird species on an intercontinental scale. Over one-third of the samples exceeded toxicity benchmarks. Feather Hg was best explained by moulting region, while habitat preference (coastal obligate vs. non-coastal obligate), the proportion of invertebrates in the diet and foraging stratum (foraging mostly on the surface vs. at depth) also contributed, but were less pronounced. Feather Hg was substantially higher in South China (Mai Po and Leizhou), Australia and the Yellow Sea than in temperate and Arctic breeding ranges. Non-coastal obligate species (Tringa genus) frequently encountered in freshwater habitats were at the highest risk. It is important to continue and expand biomonitoring research to assess how other pollutants might impact shorebirds.


Assuntos
Migração Animal , Mercúrio , Animais , Austrália , Aves , Charadriiformes , China , Ecossistema , Monitoramento Ambiental , Poluentes Ambientais/análise , Plumas/química , Mercúrio/análise , Mercúrio/toxicidade , Poluentes Químicos da Água/análise , Poluentes Químicos da Água/efeitos adversos , Ásia Oriental , Australásia
14.
Biomed Pharmacother ; 176: 116847, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38823277

RESUMO

Luteolin, a monomeric substance, is a natural product of the Brucea javanica (BJ) plant. Brucea javanica oil emulsion injection (BJOEI) is a proprietary Chinese medicine purified from BJ that is widely used clinically as an anti-tumor treatment. Although a growing body of research suggests that luteolin and BJOEI have anti-tumor effects, the molecular mechanism of action has not been fully elucidated. In this study, through molecular docking technology, we found that luteolin can interact directly with GPSM2 and regulate the FoxO signaling pathway through GPSM2. In addition, the inhibitory effect of luteolin on colon adenocarcinoma (COAD) cells was found to be offset by knockdown of GPSM2. In contrast, the anti-proliferative effects of luteolin could be notably reversed by overexpression of GPSM2. The results reveal that GPSM2 is crucial in luteolin-mediated anti-proliferative effects. The mediation of anti-proliferative effects by GPSM2 has also been indirectly demonstrated in RKO and SW480 xenograft mice models. In addition, we verified that BJOEI inhibits the progression of COAD by mediating GPSM2 and regulating the FoxO signaling pathway. We also found that BJOEI achieved a better anti-tumor effect when combined with fluorouracil injection. Collectively, our data show that the anti-tumor effects of BJOEI and luteolin on COAD are GPSM2-dependent and downregulating the expression of GPSM2 to regulate the FoxO signaling pathway may be an effective way to treat COAD.


Assuntos
Adenocarcinoma , Proliferação de Células , Neoplasias do Colo , Fluoruracila , Luteolina , Camundongos Nus , Luteolina/farmacologia , Humanos , Animais , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Neoplasias do Colo/metabolismo , Fluoruracila/farmacologia , Linhagem Celular Tumoral , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Adenocarcinoma/metabolismo , Proliferação de Células/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Camundongos Endogâmicos BALB C , Transdução de Sinais/efeitos dos fármacos , Camundongos , Produtos Biológicos/farmacologia , Produtos Biológicos/isolamento & purificação , Produtos Biológicos/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Simulação de Acoplamento Molecular
15.
Int J Mol Sci ; 14(12): 24399-411, 2013 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-24351824

RESUMO

5-Fluorouracil (5-FU) is an essential component of anticancer chemotherapy against gastric cancer. However, the response rate of single drug is still limited. The ubiquitin ligase Cbl-b is a negative regulator of growth factor receptor signaling and is involved in the suppression of cancer cell proliferation. However, whether Cbl-b could affect 5-FU sensitivity remains unclear. The present study showed that Cbl-b knockdown caused higher proliferation concomitant with the decrease of apoptosis induced by 5-FU treatment in gastric cancer cell. Further mechanism investigation demonstrated that Cbl-b knockdown caused significant increase of phosphorylation of EGFR, ERK and Akt, decrease of mitochondrial membrane potential, and increase of expression ratio of Bcl-2/Bax. These results suggest that Cbl-b enhances sensitivity to 5-FU via EGFR- and mitochondria-mediated pathways in gastric cancer cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose/efeitos dos fármacos , Receptores ErbB/metabolismo , Fluoruracila/toxicidade , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-cbl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-cbl/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Proteína X Associada a bcl-2/metabolismo
16.
Sci Rep ; 12(1): 11470, 2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35794224

RESUMO

Methylmercury (MeHg) is a global pollutant that can cause metabolic disruptions in animals and thereby potentially compromise the energetic capacity of birds for long-distance migration, but its effects on avian lipid metabolism pathways that support endurance flight and stopover refueling have never been studied. We tested the effects of short-term (14-d), environmentally relevant (0.5 ppm) dietary MeHg exposure on lipid metabolism markers in the pectoralis and livers of yellow-rumped warblers (Setophaga coronata) that were found in a previous study to have poorer flight endurance in a wind tunnel than untreated conspecifics. Compared to controls, MeHg-exposed birds displayed lower muscle aerobic and fatty acid oxidation capacity, but similar muscle glycolytic capacity, fatty acid transporter expression, and PPAR expression. Livers of exposed birds indicated elevated energy costs, lower fatty acid uptake capacity, and lower PPAR-γ expression. The lower muscle oxidative enzyme capacity of exposed birds likely contributed to their weaker endurance in the prior study, while the metabolic changes observed in the liver have potential to inhibit lipogenesis and stopover refueling. Our findings provide concerning evidence that fatty acid catabolism, synthesis, and storage pathways in birds can be dysregulated by only brief exposure to MeHg, with potentially significant consequences for migratory performance.


Assuntos
Mercúrio , Compostos de Metilmercúrio , Aves Canoras , Animais , Ácidos Graxos , Metabolismo dos Lipídeos , Fígado , PPAR gama , Músculos Peitorais
17.
Sci Total Environ ; 762: 143109, 2021 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-33162143

RESUMO

Migratory songbirds breeding in the Canadian Boreal forest are exposed to mercury (Hg), a potent neurotoxin that impairs avian health, however, the degree of exposure depends on many factors. As breeding grounds are geographically remote and vast, the measurement of Hg in individual birds is impractical particularly at large spatial scales. Here, we present a Canada-wide dataset of nearly 2000 migratory songbirds that were used to assess summer Hg exposure of 15 songbird species sampled during fall migration. We measured Hg concentrations in tail feathers and related those to dietary guild, geographic capture location, age, sex and probable breeding ground locations using feather δ2H. Overall mean (±SE) feather Hg concentration was 1.49 ± 0.03 µg/g (N = 1946): however, a clear geographic gradient in feather Hg concentrations emerged being highest in East and lowest in West. Dietary guild was the next strongest predictor of feather Hg with insectivorous songbirds in Eastern Canada at particular risk due to Hg exposure on summer breeding grounds. This broad-scale assessment of Hg exposure in migratory songbirds in Canada can be used to guide future studies on finer-scale determinants of Hg exposure in birds.


Assuntos
Poluentes Ambientais , Mercúrio , Aves Canoras , Animais , Canadá , Monitoramento Ambiental , Poluentes Ambientais/análise , Plumas/química , Isótopos , Mercúrio/análise
18.
Environ Pollut ; 288: 117752, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34284209

RESUMO

Mercury (Hg), as a global pollutant, its contamination has been documented in environmental compartments of the Himalayan region. However, little research exists regarding to Hg accumulation in terrestrial wildlife, as well as its driving factors. In this study, surface soil and small mammals were collected in the Lebu Valley, East Himalayas of China, in order to measure the uptake of the long-distance transported Hg along an elevational gradient approximately from 2300 to 5000 m a.s.l. The soil Hg concentrations were measured and predicted mostly by vegetation type as well as soil organic matter, while the Hg in hair of small mammals (Muridae and Cricetidae) showed deeply influenced by soil Hg. Notably, combined with the field survey data, soil and hair Hg were both enhanced in low and mid-elevations, which overlapped the distribution ranges of a majority of mammals. Overall, this indicates that Hg contamination in low- and mid-elevations poses a potential threat to the top predators that consuming small mammals directly or indirectly. Furthermore, our data advances the understanding of Hg dynamics in remote, high mountain ecosystems and provides baseline data for biomonitoring for reduction of Hg emission globally.


Assuntos
Mercúrio , Poluentes do Solo , Animais , Ecossistema , Monitoramento Ambiental , Mamíferos , Mercúrio/análise , Solo , Poluentes do Solo/análise
20.
Front Oncol ; 11: 592761, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33747912

RESUMO

Background: NOS3 (endothelial NOS, eNOS) is a member of the nitric oxide synthase (NOS) enzyme family, mainly participating in nitric oxide (NO) generation. NOS3 has been reported to inhibit apoptosis and promote angiogenesis, proliferation, and invasiveness. However, the expression pattern of NOS3 and its diagnostic and prognostic potential has not been investigated in a pan-cancer perspective. Methods: Data from the Genotype-Tissue Expression (GTEx), the Cancer Genome Atlas (TCGA), the Cancer Cell Line Encyclopedia (CCLE), and the Cancer Therapeutics Response Portal (CTRP) were employed and NOS3 expression was comprehensively analyzed in normal tissues, cancer tissues, and cell lines. Immunohistochemical staining of tissue sections were used to validate the prognostic role of NOS3 in gastric cancer patients. GSVA and GSEA analyses were performed to investigate signaling pathways related to NOS3 expression. Results: In normal tissues, NOS3 was expressed highest in the spleen and lowest in the blood. NOS3 expression was increased in stomach adenocarcinoma (STAD) and significantly associated with poor prognosis of patients. Immunohistochemical staining validated that NOS3 was an independent prognostic factor of gastric cancer. Several canonical cancer-related pathways were found to be correlated with NOS3 expression in STAD. The expression of NOS3 was related to the response to QS-11 and brivinib in STAD. Conclusions: NOS3 was an independent prognostic factor for patients with STAD. Increased expression of NOS3 influenced occurrence and development of STAD through several canonical cancer-related pathways. Drug response analysis reported drugs to suppress NOS3. NOS3 might be a novel target for gastric cancer treatment.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA