Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Toxicology ; 506: 153838, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38797228

RESUMO

Aristolochic acid (AA)-IIIa is an AA analog present in Aristolochiaceae plants. To evaluate the chronic toxicity of AA-IIIa, mice were intragastrically administered with media control, 1 mg/kg AA-IIIa, and 10 mg/kg AA-IIIa, and designated as the control (CTL), AA-IIIa low dose (AA-IIIa-L), and AA-IIIa high dose (AA-IIIa-H) groups, respectively. AA-IIIa was administered three times a week, every other day, for 24 weeks (24-week time point). Thereafter, some mice were sacrificed immediately, while others were sacrificed 29 or 50 weeks after AA-IIIa withdrawal (53- or 74-week time point). Serum and organs were collected for biochemical and pathological analyses, respectively. Whole-genome sequencing was performed on the kidney, liver, and stomach tissues of AA-IIIa-treated mice for single-nucleotide polymorphism (SNP) detection. AA-IIIa-H mice died at 66 weeks, and the remaining mice showed moribund conditions at the 69 weeks. AA-IIIa induced minor kidney tubule injury, fibroblast hyperplasia, and forestomach carcinoma in mice. Bladder, intestine, liver, heart, spleen, lung, and testis tissues were not pathologically altered by AA-IIIa. In addition, AA-IIIa increased the C:G > A:T mutation in the kidney; however, no SNP mutation changes were observed in the liver and forestomach tissues of AA-IIIa-H mice at the 24-week time point compared with control mice. Therefore, we suspect that AA-IIIa is potentially mutagenic for mice after overdose and long-term administration. On the other hand, the forestomach is a unique organ in mice, but it does not exist in humans; thus, we hypothesize that the stomach toxicity induced by AA-IIIa is not a suitable reference for toxicological evaluation in humans. We recommend that Aristolochiaceae plants containing AA-IIIa should be properly supervised, and overdosing and long-term administration of drugs containing AA-IIIa should be avoided.


Assuntos
Ácidos Aristolóquicos , Animais , Ácidos Aristolóquicos/toxicidade , Camundongos , Masculino , Rim/efeitos dos fármacos , Rim/patologia , Polimorfismo de Nucleotídeo Único , Feminino , Fígado/efeitos dos fármacos , Fígado/patologia , Estômago/efeitos dos fármacos , Estômago/patologia , Testes de Toxicidade Crônica/métodos , Relação Dose-Resposta a Droga
3.
Neoplasma ; 70(4): 500-513, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37789776

RESUMO

Long noncoding RNAs (lncRNAs) play important roles in the progression of human cancer. It is reported that lncRNA plasmacytoma variant translocation 1 (PVT1) is involved in colorectal cancer (CRC), however, the underlying mechanism remains to be explored deeply, especially by in vivo models. In the present study, bioinformatics analysis showed that the expression level of PVT1 was upregulated in CRC tissues and highly associated with poor prognosis of CRC patients. In cultured CRC cells, knockdown of PVT1 inhibited cell proliferation and migration of CRC cells, while overexpression of PVT1 promoted the progression of CRC cells. In zebrafish xenografts, the silencing of PVT1 also suppressed the growth and metastasis of CRC cells. For mechanism studies, the binding relationships among PVT1, miR-24-3p, and Neuropilin 1 (NRP1) were predicted by starBase firstly. The luciferase reporter assays verified that PVT1 and NRP1 could bind with miR-24-3p directly. Further studies showed miR-24-3p negatively regulated the progression of CRC cells, the inhibition of miR-24-3p counteracted the repression effects of CRC progression when knocking down PVT1. In addition, the expression of NRP1 was regulated by PVT1, and NRP1 overexpression could partially rescue the inhibition effects of CRC progression when knocking down PVT1 in vitro and in vivo. Our study reveals that PVT1 promotes the proliferation and metastasis of CRC via regulating the miR-24-3p/NRP1 axis, which provides a prognosis biomarker and a potential therapeutic target for CRC patients.


Assuntos
Neoplasias Colorretais , MicroRNAs , RNA Longo não Codificante , Animais , Humanos , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , MicroRNAs/genética , MicroRNAs/metabolismo , Neuropilina-1/genética , Prognóstico , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
4.
Pharm Biol ; 58(1): 98-106, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31957525

RESUMO

Context: Aristolochia manshuriensis Kom (Aristolochiaceae) (AMK) is known for toxicity and mutagenicity.Objective: The tumorigenic role of AMK has yet to be understood.Materials and methods: AMK extracts were extracted from root crude drug. SD (Sprague Dawley) rats underwent gavage with AMK (0.92 g/kg) every other day for 10 (AMK-10) or 20 (AMK-20) weeks. Stomach samples were gathered for histopathological evaluation, microarray and mRNA analysis.Results: The gastric weight to body weight ratio (GW/BW) is 1.7 in the AMK-10 cohort, and 1.8 in AMK-20 cohort compared to control (CTL) cohort. Liver function was damaged in AMK-10 and AMK-20 rats compared to CTL rats. There were no significant changes of CRE (creatinine) in AMK-10 and AMK-20 rats. Histopathological analysis revealed that rats developed dysplasia in the forestomach in AMK-10 rats, and became gastric carcinoma in AMK-20 rats. Genes including Mapk13, Nme1, Gsta4, Gstm1, Jun, Mgst2, Ggt6, Gpx2, Gpx8, Calml3, Rasgrp2, Cd44, Gsr, Dgkb, Rras, and Amt were found to be critical in AMK-10 and AMK-20 rats. Pik3cb, Plcb3, Tp53, Hras, Myc, Src, Akt1, Gnai3, and Fgfr3 worked in AMK-10 rats, and PDE2a and PDE3a played a pivotal role in AMK-20 rats.Discussion and conclusions: AMK induced benign or malignant gastric tumours depends on the period of AMK administration. Genes including Mapk13, Nme1, Gsta4, Gstm1, Jun, Mgst2, Ggt6, Gpx2, Gpx8, Calml3, Rasgrp2, Cd44, Gsr, Dgkb, Rras, Amt, Pik3cb, Plcb3, Tp53, Hras, Myc, Src, Akt1, Gnai3, Fgfr3, PDE2a, and PDE3a were found to be critical in aristolochic acid-induced gastric tumour process.


Assuntos
Aristolochia/química , Extratos Vegetais/toxicidade , Neoplasias Gástricas/induzido quimicamente , Animais , Ácidos Aristolóquicos/isolamento & purificação , Ácidos Aristolóquicos/toxicidade , Análise em Microsséries , Extratos Vegetais/administração & dosagem , Ratos , Ratos Sprague-Dawley , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Fatores de Tempo
5.
Int J Mol Sci ; 20(20)2019 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-31600977

RESUMO

A high incidence of hypersensitivity reactions (HSRs) largely limits the use of paclitaxel injection. Currently, these reactions are considered to be mediated by histamine release and complement activation. However, the evidence is insufficient and the molecular mechanism involved in paclitaxel injection-induced HSRs is still incompletely understood. In this study, a mice model mimicking vascular hyperpermeability was applied. The vascular leakage induced merely by excipients (polyoxyl 35 castor oil) was equivalent to the reactions evoked by paclitaxel injection under the same conditions. Treatment with paclitaxel injection could cause rapid histamine release. The vascular exudation was dramatically inhibited by pretreatment with a histamine antagonist. No significant change in paclitaxel injection-induced HSRs was observed in complement-deficient and complement-depleted mice. The RhoA/ROCK signaling pathway was activated by paclitaxel injection. Moreover, the ROCK inhibitor showed a protective effect on vascular leakage in the ears and on inflammation in the lungs. In conclusion, this study provided a suitable mice model for investigating the HSRs characterized by vascular hyperpermeability and confirmed the main sensitization of excipients in paclitaxel injection. Histamine release and RhoA/ROCK pathway activation, rather than complement activation, played an important role in paclitaxel injection-induced HSRs. Furthermore, the ROCK inhibitor may provide a potential preventive approach for paclitaxel injection side effects.


Assuntos
Antineoplásicos Fitogênicos/efeitos adversos , Hipersensibilidade a Drogas/etiologia , Hipersensibilidade a Drogas/metabolismo , Paclitaxel/efeitos adversos , Transdução de Sinais , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Biópsia , Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Modelos Animais de Doenças , Hipersensibilidade a Drogas/patologia , Feminino , Liberação de Histamina , Masculino , Camundongos , Paclitaxel/administração & dosagem
6.
Oncol Lett ; 18(1): 599-608, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31289532

RESUMO

SRY-box 9 (SOX9) is overexpressed in a number of human tumors, including gastric cancer (GC). However, the function of SOX9 in the development of GC remains unknown. In the present study, SOX9 activated the Hippo-yes-associated protein (YAP) signaling pathway to enhance the epithelial-mesenchymal transition in GC cell lines. The results suggested that SOX9 knockdown inhibited invasion, proliferation and migration of GC cells. Furthermore, SOX9 silencing upregulated the expression of E-cadherin, an epithelial marker, and downregulated the expression of mesenchymal markers, including snail family transcriptional repressor 1, vimentin and N-cadherin. SOX9 overexpression increased the expression of the aforementioned markers. SOX9 significantly affected YAP phosphorylation and total YAP protein levels, suggesting that SOX9 is involved in the Hippo-YAP signaling pathway. The current study revealed that SOX9 may be involved in the pathogenesis of GC, and further elucidation of the pathways involved may support the development of novel therapeutic options for the treatment of GC.

7.
Biomed Res Int ; 2019: 1427871, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30937307

RESUMO

Butyrate is a short-chain fatty acid decomposed from dietary fiber and has been shown to have effects on inhibition of proliferation but induction of apoptosis in colorectal cancer cells. However, clinical trials have yielded ambiguous outcomes with regard to its antitumor activities. In this study, we aimed to explore the molecular mechanisms underlying the sensitivity of colorectal cancer cells to sodium butyrate (NaB). RNA sequencing was used to establish the whole-transcriptome profile in NaB-treated versus untreated colorectal cancer cells. Differentially expressed genes were bioinformatically analyzed to predict their possible involvement in NaB-triggered cell death, and the expression of eight dysregulated genes was validated by quantitative real-time PCR. We found that there were a total of 7192 genes (5720 upregulated and 1472 downregulated, fold-change ≥ 2 or ≤ 0.5 for upregulation or downregulation, q-value < 0.05) differentially expressed in NaB-treated cells as compared with the untreated controls. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis demonstrated that the differentially expressed genes were enriched in DNA replication, cell cycle, homologous recombination, pyrimidine metabolism, mismatch repair, and other signaling pathways and may take part in NaB-induced cell death. Among the identified factors, the MCM2-7 complex might be a target of NaB. Our findings provide an important basis for further studies of the complicate network that might regulate sensitivity of colorectal cancer cells to NaB.


Assuntos
Ácido Butírico/farmacologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Análise de Sequência de RNA , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Ontologia Genética , Humanos , Proteínas de Manutenção de Minicromossomo/metabolismo
8.
J Ethnopharmacol ; 225: 81-89, 2018 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-30008395

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Aristolochia manshuriensis Kom (AMK), belonging to the Aristolochia family, is traditionally used in China to remove heart fire, promote dieresis, restore menstruation, and enhance milk secretion. The active constitutes of AMK are aristolochic acids (AAs, I and II) that are reported to cause serious side effects including nephrotoxicity and carcinogenicity. AIM OF THE STUDY: The tumorigenic role of AMK is far to be understood. We analyzed the toxicity reactions after long-term exposure of AMK in rats. MATERIALS AND METHODS: Sprague-Dawley rats underwent gavage with AMK doses of 51 mg/kg (AMK-1), 253 mg/kg (AMK-2), 508 mg/kg (AMK-3), 1029 mg/kg (AMK-4) or AAs of 15 mg/kg (AAs), and then sacrificed at the 6th, 10th, 14th, 18th, 22th, 26th and 30th weeks. Endpoint measurements included clinical observations, body weights, blood biochemistry, haematology and histomorphological observations. RESULTS: Body weight decreased after AMK or AAs treatment in rats. AMK destroyed renal function, and induced anemia in rats. AMK caused kidney, stomach, bladder and subcutaneous tumors in rats. In addition, primary hepatic carcinoma was not observed in rats. CONCLUSIONS: AMK had significant toxic effects in rats with regard to decreased body weight, diminished renal function, increased anemia and tumor incidence. Kidney, stomach, bladder and subcutaneous tissue are carcinogenic target organs of AMK or AAs, however liver is no- carcinogenic target organ of AMK or AAs in rats. AMK is carcinogenic in rats, and not be safe for humans.


Assuntos
Aristolochia , Carcinógenos/toxicidade , Neoplasias/induzido quimicamente , Extratos Vegetais/toxicidade , Administração Oral , Anemia/induzido quimicamente , Animais , Peso Corporal/efeitos dos fármacos , Feminino , Rim/efeitos dos fármacos , Rim/patologia , Rim/fisiologia , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Neoplasias/patologia , Tamanho do Órgão/efeitos dos fármacos , Ratos Sprague-Dawley , Estômago/efeitos dos fármacos , Estômago/patologia , Testes de Toxicidade Crônica , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/patologia
9.
Oncotarget ; 8(60): 101899-101910, 2017 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-29254212

RESUMO

Lung cancer is one of the leading causes of cancer-related mortality, and responds badly to existing treatment. Thus, it is of urgent need to identify novel diagnostic markers and therapeutic targets. Increasing evidences have indicated that long non-coding RNAs (lncRNAs) play an important role in initiation and progression of lung cancer. However, the role of lncRNA Taurine upregulated 1 (TUG1) in lung adenocarcinoma (LAD) progression is not well known. In this study, we determined the diagnostic value of TUG1 in LAD patients, and further uncovered the underlying functional mechanism. Our results showed that TUG1 was significantly upregulated in LAD cells and serum samples. Receiver operator characteristic (ROC) analysis suggested a relatively higher area under the curve (AUC) of TUG1 (0.756) contrast to cyfra21-1 (0.619). In addition, high TUG1 level was associated with enhanced tumor size, degree of differentiation, lymph node metastases, distant metastasis and TNM stage. Cell functional assays showed that knockdown of TUG1 suppressed LAD cell viability and promoted cell apoptosis. We then sought to reveal the underlying regulatory mechanism, and the pro-apoptotic protein BAX was then identified as the downstream target of TUG1. Gain and loss functional assays showed that inhibition of BAX reversed the induced apoptosis by TUG1 knockdown. Finally, RNA immunoprecipitation and Chromatin immunoprecipitation revealed that TUG1 suppressed BAX expression through physically interacting with EZH2. In conclusion, lncRNA TUG1 is a promising diagnostic marker for LAD patients and suppression of TUG1 levels could be a future direction to promote the prognosis of LAD patients.

10.
PLoS One ; 11(9): e0163447, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27668427

RESUMO

BACKGROUND: Ovarian cancer is the most lethal type of malignant tumor in gynecological cancers and is associated with a high percentage of late diagnosis and chemotherapy resistance. Thus, it is urgent to identify a tumor marker or a molecular target that allows early detection and effective treatment. RNA-binding proteins (RBPs) are crucial in various cellular processes at the post-transcriptional level. The eukaryotic translation initiation factor 4 gamma, 1(eIF4G1), an RNA-binding protein, facilitates the recruitment of mRNA to the ribosome, which is a rate-limiting step during the initiation phase of protein synthesis. However, little is known regarding the characteristics of eIF4G1 expression and its clinical significance in ovarian cancer. Therefore, we propose to investigate the expression and clinicopathological significance of eIF4G1 in ovarian cancer patients. METHODS: We performed Real-time PCR in 40 fresh serous ovarian cancer tissues and 27 normal ovarian surface epithelial cell specimens to assess eIF4G1mRNA expression. Immunohistochemistry (IHC) was used to examine the expression of eIF4G1 at the protein level in 134 patients with serous ovarian cancer and 18 normal ovarian tissues. Statistical analysis was conducted to determine the correlation of the eIF4G1 protein levels with the clinicopathological characteristics and prognosis in ovarian cancer. RESULTS: The expression of eIF4G1 was upregulated in serous ovarian cancer tissues at both the mRNA (P = 0.0375) and the protein (P = 0.0007) levels. The eIF4G1 expression was significantly correlated with the clinical tumor stage (P = 0.0004) and omentum metastasis (P = 0.024). Moreover, patients with low eIF4G1 protein expression had a longer overall survival time (P = 0.026). CONCLUSIONS: These data revealed that eIF4G1 is markedly expressed in serous ovarian cancer and that upregulation of the eIF4G1 protein expression is significantly associated with an advanced tumor stage. Besides, the patients with lower expression of eIF4G1 tend to have a longer overall survival time. Thus, eIF4G1 may contribute to the occurrence and metastasis of ovarian cancer and can serve as a potential therapeutic target for the treatment of ovarian cancer.

11.
J Clin Lab Anal ; 30(6): 1220-1225, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27265845

RESUMO

AIMS: The aim of this study was to investigate the association between CYP1A1 gene polymorphism and cervical cancer risk, and the impact of SNP-SNP interaction on cervical cancer risk in Chinese women. METHODS: A total of 728 females with a mean age of 60.1 ± 14.5 years old were selected, including 360 cervical cancer patients and 368 normal controls. Logistic regression was performed to investigate association between single-nucleotide polymorphisms (SNP) and cervical cancer risk. Generalized multifactor dimensionality reduction (GMDR) was used to analyze the SNP-SNP interaction. RESULTS: Logistic analysis showed a significant association between rs4646903 and increased cervical cancer risk. The carriers of homozygous mutant of rs4646903 polymorphism revealed increased cervical cancer risk than those with wild-type homozygotes, OR (95%CI) were 1.45 (1.20-1.95). There was a significant two-locus model (P = 0.0107) involving rs4646903 and rs1048943, indicating a potential SNP-SNP interaction between rs4646903 and rs1048943. Overall, the two-locus models had a cross-validation consistency of 10 of 10, and had the testing accuracy of 60.72%. Subjects with TC or CC of rs4646903 and AG or GG of rs1048943 genotype have the highest cervical cancer risk, compared to subjects with TT of rs4646903 and AA of rs1048943 genotype, OR (95%CI) was 2.03 (1.42-2.89). CONCLUSIONS: rs4646903 minor alleles and interaction between rs4646903 and rs1048943 were associated with increased cervical cancer risk.


Assuntos
Citocromo P-450 CYP1A1/genética , Predisposição Genética para Doença/genética , Polimorfismo de Nucleotídeo Único/genética , Neoplasias do Colo do Útero/genética , Adulto , Idoso , Estudos de Casos e Controles , China , Feminino , Genótipo , Humanos , Modelos Logísticos , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Risco
12.
Am J Transl Res ; 8(2): 698-704, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27158362

RESUMO

To explore the subcellular localization of Polyphenol oxidase (PPO) from Pyrus bretschneideri, the 1779 bp cDNA of PPO gene excluding the termination codon TAA was cloned and fused with GFP to construct a binary vector pBI121-PPO-GFP. Then, the binary vector was transformed into Nicotiana tabacum by the tumefanciens-mediated method. Using confocal laser scanning microscopy, green fluorescent signals were localized in chloroplasts of the transformed Nicotiana tabacum cell, suggesting that the Polyphenol oxidase from Pyrus bretschneideri was a chloroplast protein.

13.
Zhongguo Zhong Yao Za Zhi ; 40(14): 2696-700, 2015 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-26666011

RESUMO

Genotoxicity research takes an important place in traditional Chinese medicine safety evaluation. Genotoxicity test on traditional Chinese medicine has been paid great attention since 1970s. Currently, the most developed genotoxicity test methods included: bacterial reverse mutation test and mouse lymphoma assay which are used to detect relevant genetic changes, micronucleus test and chromosomal analysis which are used to measure chromosomal aberration, and single cell electrophoresis assay which is used to test DNA damage. This article reviews research progress on genotoxicity of traditional Chinese medicine, evaluation methods of genotoxicity, the problems and solutions on genotoxicity evaluation of traditional Chinese medicine, and new technique used in genotoxicity test.


Assuntos
Medicina Tradicional Chinesa/efeitos adversos , Testes de Mutagenicidade/métodos , Animais , Pesquisa Biomédica , Humanos
14.
Mol Med Rep ; 10(5): 2595-600, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25175940

RESUMO

Histone deacetylases (HDACs) are important in chromatin remodeling and epigenetic regulation of gene expression. Histone deacetylase inhibitors (HDACi) have highly effective anti-metastatic and anti-angiogenic activity in various types of cancer, while the molecular mechanisms involved in this process are not fully understood. In the present study, trichostatin A (TSA), a HDACi, was found to suppress MCF-7 breast carcinoma cell invasion and upregulate TET1 expression in a dose-dependent manner. TET1, a dioxygenase involved in cytosine demethylation, is downregulated during breast cancer progression. TET1 knockdown in MCF-7 cells facilitates cell invasion, inhibits the expression of tissue inhibitors of metalloproteinase 2/3 (TIMP2/3) and promotes matrix metalloproteinases (MMP) 2/9 transcriptional activity. Importantly, TET1 depletion impaired the inhibitory effect of TSA on breast cancer cell invasion. Together, these results illustrated a mechanism by which TET1 partially mediates HDACi elicited suppression of breast cancer invasion.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Movimento Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/fisiologia , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Proteínas Proto-Oncogênicas/fisiologia , Adulto , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Oxigenases de Função Mista , Invasividade Neoplásica , Inibidor Tecidual de Metaloproteinase-2/genética , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Inibidor Tecidual de Metaloproteinase-3/genética , Inibidor Tecidual de Metaloproteinase-3/metabolismo
15.
Zhongguo Zhong Yao Za Zhi ; 39(3): 511-4, 2014 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-24946558

RESUMO

Pseudoallergic reactions of Qingkailing injection (QKLI) was assessed by vascular hyperpermeability which were indicated by ear blue staining in ICR mice after single intravenous injection of QKLI mixed with Evans blue (EB) and skin blue spot formation in SD rats after intradermal injection of QKLI and intravenous injection of EB. In addition, QKLI-induced histamine, VEGF, TNF-alpha release was measured after ICR mice received the single dosing of QKLI iv. The mild vascular hyperpermeability characterized by ear blue staining could be observed in mice after intravenous injection of QKLI and EB. Intracutaneous injection of 50 micro L of test solution containing QKLI (25,50 microL) in rat back skin caused obvious local vascular hyperpermeability at the injection sites so as to result the larger diameters of blue spots than that in negative control group (P <0. 01). QKLI induced a significant increase of VEGF and a slight elevation of histamine in mice after intravenous administration, while TNF-alpha showed no change after QKLI iv. The results in this study indicated that both intravenous injection and intracutanous injection of QKLI could induce vascular hyperpemeability so as to cause pseudoallergic reaction in mice and rats. QKLI-induced pseudoallergic reaction may be associated with the release of histamine and VEGF.


Assuntos
Hipersensibilidade a Drogas/etiologia , Medicamentos de Ervas Chinesas/administração & dosagem , Medicamentos de Ervas Chinesas/efeitos adversos , Animais , Hipersensibilidade a Drogas/sangue , Hipersensibilidade a Drogas/imunologia , Histamina/sangue , Injeções , Masculino , Camundongos , Ratos , Pele/efeitos dos fármacos , Pele/imunologia , Fator de Necrose Tumoral alfa/sangue , Fator A de Crescimento do Endotélio Vascular/sangue
16.
Tumour Biol ; 33(6): 1915-25, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22782504

RESUMO

The differentially expressed in adenocarcinoma of the lung-1 (DAL-1) protein is a member of the membrane-associated cytoskeleton protein 4.1 family. This protein was previously found to be downregulated or lost in more than half of primary non-small cell lung cancers (NSCLC). In this study, the relationship between DAL-1 expression and NSCLC metastasis was examined. DAL-1 mRNA and protein levels were measured in NSCLC cell lines and in tumor cells isolated from the pleural fluid of NSCLC patients clinically diagnosed with distant metastases to the bone or brain. The results revealed that DAL-1 expression was observed in two (GLC-82 and NCI-H460) out of seven metastatic NSCLC cell lines examined. DAL-1 expression was not observed in the cells isolated from the pleural fluid in nine out of ten patients. Overexpression of DAL-1 in A549 cells, a cell line lacking endogenous DAL-1, inhibited cell migration and invasion by approximately 38 and 48 %, respectively. In contrast, DAL-1 knockdown in NCI-H460 cells enhanced the migration and invasion potential of this cell line 4.6- and 3-fold, respectively. Furthermore, DAL-1 promoter methylation was observed in six of nine pleural fluid NSCLC cell isolates and in two cell lines (A549 and H1299), as evidenced by a lack of endogenous DAL-1. Demethylation in A549 cells successfully restored DAL-1 mRNA and protein expression levels, resulting in a parallel remarkable inhibition of migration and invasion. These results indicated that DAL-1 was pivotal in triggering NSCLC migration and invasion and that loss of DAL-1 expression was due to the epigenetic methylation.


Assuntos
Neoplasias Ósseas/metabolismo , Neoplasias Encefálicas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , Proteínas dos Microfilamentos/metabolismo , Derrame Pleural/metabolismo , Western Blotting , Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/secundário , Adesão Celular , Movimento Celular , Proliferação de Células , Metilação de DNA , Humanos , Neoplasias Pulmonares/patologia , Proteínas dos Microfilamentos/genética , Derrame Pleural/genética , Derrame Pleural/patologia , Prognóstico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA