Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cell Death Discov ; 8(1): 60, 2022 Feb 11.
Article in English | MEDLINE | ID: mdl-35149697

ABSTRACT

Epithelial ovarian cancer (EOC) is one of the most frequent and fatal gynecologic malignant tumors resulting in an unsatisfying prognosis. Long non-coding RNAs (lncRNAs) play pivotal roles in the tumorigenesis and progression of EOC. However, the profile of lncRNAs involved in EOC remains to be expanded to further improve clinical treatment strategy. In present study, we identified a novel tumor-suppressive lncRNA small nucleolar RNA host gene 10 (SNHG10) in EOC. Kaplan-Meier analysis and COX proportional hazard progression model showed that low expression of SNHG10 was correlated with a poor prognosis of EOC patients. Overexpressing SNHG10 suppressed the proliferation, colony formation, migration, and invasion of EOC cells. Furthermore, SNHG10 was predicted to sponge miR-200a-3p in EOC cells according to the LncBase v.2 experimental module. Then, the binding of SNHG10 and miR-200a-3p was confirmed by performing quantitative real-time PCR (qRT-PCR) and luciferase reporter assays. RNA immunoprecipitation (RIP) showed that SNHG10 and miR-200a-3p occupied the same Ago2 protein to form an RNA-induced silencing complex (RISC). By overlapping the results from the bioinformatics algorithms, tumor-suppressor bridging integrator-1 (BIN1) was found to be a main downstream target of the SNHG10/miR-200a-3p axis. Low expression of BIN1 in EOC tissues was detected by using immunohistochemistry (IHC). Besides, BIN1 and SNHG10 expression was positively correlated in EOC tissues. By performing miRNA rescue experiments, a SNHG10/miR-200a-3p/BIN1 axis and its promoting effects on malignant behaviors and epithelial-mesenchymal transition (EMT) process were verified in EOC cells. Moreover, SNHG10 overexpression significantly suppressed the tumorigenesis and EMT of EOC cells in vivo. Altogether, SNHG10 sponges miR-200a-3p to upregulate BIN1 and thereby exerting its tumor-suppressive effects in EOC. Therefore, the SNHG10/miR-200a-3p/BIN1 axis may act as a potential predictive biomarker and therapeutic target for treating EOC.

2.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-935049

ABSTRACT

@#[摘 要] 目的:探讨PD-1抗体联合化疗对比抗血管生成药物联合化疗在晚期驱动基因阴性肺腺癌一线治疗中的疗效和安全性。方法:收集2018年3月至2021年8月河北医科大学第四医院收治的141例不可手术切除的ⅢB/ⅢC和Ⅳ期驱动基因阴性肺腺癌患者,回顾性分析PD-1抗体联合化疗对比抗血管生成药物联合化疗在一线治疗中的疗效与安全性。主要研究终点为无进展生存期(PFS),次要终点为客观缓解率(ORR)、疾病控制率(DCR)和不良反应。结果:141例患者均纳入生存分析,中位随访时间为13.0个月(95% CI:12.0~14.0)。PD-1抗体联合化疗组(A组)和抗血管生成药物联合化疗组(B组)的ORR分别为33.33%和27.38%,DCR分别为98.25%和89.29%,差异均无统计学意义。A组和B组的中位PFS分别为8.4个月(95% CI: 7.3~9.9)和6.9个月(95% CI: 6.1~7.7),差异无统计学意义。亚组分析结果显示,ⅢB/ⅢC期、肝或脑转移患者中,A组中位PFS较B组均延长(均P<0.01)。A组和B组不良反应发生率分别为26.32%和14.29%,多数为1~2级。结论:PD-1抗体联合化疗对比抗血管生成药物联合化疗一线治疗晚期驱动基因阴性肺腺癌疗效相当,不良反应可耐受,可成为晚期驱动基因阴性肺腺癌标准一线治疗。

3.
Mol Cancer ; 20(1): 162, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34893064

ABSTRACT

BACKGROUND: Cis-diamminedichloro-platinum (CDDP)-based chemotherapy regimens are the most predominant treatment strategies for patients with esophageal squamous cell carcinoma (ESCC). Dysregulated long non-coding RNAs (lncRNAs) contribute to CDDP resistance, which results in treatment failure in ESCC patients. However, the majority of lncRNAs involved in CDDP resistance in ESCC remain to be elucidated. METHODS: The public Gene Expression Omnibus (GEO) dataset GSE45670 was analysed to reveal potential lncRNAs involved in CDDP resistance of ESCC. Candidate upregulated lncRNAs were detected in ESCC specimens by qRT-PCR to identify crucial lncRNAs. Non-coding RNA activated by DNA damage (NORAD) was selected for further study. Kaplan-Meier analysis and a COX proportional regression model were performed to analyse the potential of NORAD for predicting prognosis of ESCC patients. The role of NORAD in CDDP resistance were determined by conducting gain and loss-of-function experiments in vitro. Fluorescence in situ hybridization (FISH) was performed to determine the subcellular location of NORAD in ESCC cells. A public GEO dataset and bioinformatic algorithms were used to predict the microRNAs (miRNAs) that might be latently sponged by NORAD. qRT-PCR was conducted to verify the expression of candidate miRNAs. Luciferase reporter and Argonaute-2 (Ago2)-RNA immunoprecipitation (RIP) assays were conducted to evaluate the interaction between NORAD and candidate miRNAs. A miRNA rescue experiment was performed to authenticate the NORAD regulatory axis and its effects on CDDP resistance in ESCC cells. Western blotting was conducted to confirm the precise downstream signalling pathway of NORAD. A xenograft mouse model was established to reveal the effect of NORAD on CDDP resistance in vivo. RESULTS: The expression of NORAD was higher in CDDP-resistant ESCC tissues and cells than in CDDP-sensitive tissues and cells. NORAD expression was negatively correlated with the postoperative prognosis of ESCC patients who underwent CDDP-based chemotherapy. NORAD knockdown partially arrested CDDP resistance of ESCC cells. FISH showed that NORAD was located in the cytoplasm in ESCC cells. Furthermore, overlapping results from bioinformatic algorithms analyses and qRT-PCR showed that NORAD could sponge miR-224-3p in ESCC cells. Ago2-RIP demonstrated that NORAD and miR-224-3p occupied the same Ago2 to form an RNA-induced silencing complex (RISC) and subsequently regulated the expression of metadherin (MTDH) in ESCC cells. The NORAD/miR-224-3p/MTDH axis promoted CDDP resistance and progression in ESCC cells by promoting nuclear accumulation of ß-catenin in vitro and in vivo. CONCLUSIONS: NORAD upregulates MTDH to promote CDDP resistance and progression in ESCC by sponging miR-224-3p. Our results highlight the potential of NORAD as a therapeutic target in ESCC patients receiving CDDP-based chemotherapy.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Membrane Proteins/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA-Binding Proteins/genetics , Adult , Aged , Animals , Cell Line, Tumor , Computational Biology , Disease Models, Animal , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , RNA Interference , ROC Curve , Xenograft Model Antitumor Assays , beta Catenin/metabolism
4.
Cell Death Dis ; 10(8): 573, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31363080

ABSTRACT

Emerging evidence suggests that long noncoding RNA (lncRNA) plays pivotal roles in regulating various biological process in human cancers. Titin-antisense RNA1 (TTN-AS1) has been regarded as a tumor promoting lncRNA in numerous cancers. However, the clinical significance and biological function of TTN-AS1 in lung adenocarcinoma (LUAD) remain unclear. In the present study, we revealed that the expression of TTN-AS1 was upregulated in LUAD tissues and cell lines. High TTN-AS1 expression was associated with TNM stage and lymph node metastasis of LUAD patients. In addition, high expression of TTN-AS1 was correlated with poor postoperative prognosis of LUAD patients. Knockdown of TTN-AS1 significantly inhibited the growth, proliferation, migration, and invasion ability of LUAD cells in vitro. Then, by using bioinformation analysis and luciferase reporter experiment, we identified that TTN-AS1 could function as a competing endogenous RNA (ceRNA) by sponging miR-142-5p to regulate the expression of cyclin-dependent kinase 5 (CDK5) in LUAD. Since CDK5 is a key regulator in the process of epithelial mesenchymal transition (EMT), we detected the expression of EMT-related proteins, consequently, EMT was suppressed by knockdown of TTN-AS1 while this phenomenon was rescued by miR-142-5p inhibitor. Taken above, our study revealed that TTN-AS1 played an important role in LUAD progression. TTN-AS1/miR-142-5p/CDK5 regulatory axis may serve as a novel therapeutic target in the treatment of LUAD.


Subject(s)
Adenocarcinoma of Lung/genetics , Cyclin-Dependent Kinase 5/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , A549 Cells , Adenocarcinoma of Lung/pathology , Adult , Aged , Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
5.
Exp Ther Med ; 16(2): 649-656, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30112030

ABSTRACT

Diffuse large B cell lymphoma (DLBCL) is the most common type of non-Hodgkin's lymphoma in adults. Mda-7/IL-24 had been identified as a differentiation inducer of B phenotype lymphoma cells. Previous studies have revealed that knockdown of C-myb also leads to the terminal differentiation of B cell lymphoma. The aim of the present study was to investigate the association between the expression of Mda-7/IL-24 and C-myb, and their prognostic significance for DLBCL patients. The tumor tissues were collected from 72 cases of DLBCL patients and detected with reverse transcription-quantitative polymerase chain reaction, western blotting and immunohistochemistry assays. The results showed that, the expression of Mda-7/IL-24 mRNA and protein was lower while the expression of C-myb was higher in DLBCL tissues, compared with the specimens of normal lymph node tissues. Furthermore, C-myb expression was negatively correlated with Mda-7/IL-24 expression at mRNA and protein levels in DLBCL tissues. The expression of Mda-7/IL-24 and C-myb in DLBCL tissues was associated with some clinicopathological parameters such as clinical stage, infiltration in bone marrow, Ki67 expression level in the tumor tissues and overall survival rates. These results indicated that low expression of Mda-7/IL-24, along with high expression of C-myb, are predictor for poor prognosis of DLBCL patients, suggesting that Mda-7/IL-24 and C-myb may be potential targets for clinical treatment of DLBCL.

6.
Oncol Lett ; 15(4): 4649-4656, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29541237

ABSTRACT

Celecoxib is a newly-identified nonsteroidal anti-inflammatory drug, which has been used to treat fever in clinical practice. Celecoxib has been demonstrated to suppress the viability of various human tumor cells. However, the effect of celecoxib on response of T-cell lymphoma to chemotherapy agents remains unclear. The aim of the present study was to investigate the effect of celecoxib on chemosensitivity of human T-cell lymphoma, and to address the underlying mechanism of action. The cytotoxicity of CDDP, epirubicin and VCR on Jurkat and Hut-78 cells treated with celecoxib was assessed by MTT assay, and the half-maximal inhibitory concentration (IC50) value was calculated by Origin 75 software. The effect of celecoxib on apoptosis and intracellular concentration of Rhodamine-123 in Jurkat and Hut-78 cells was analyzed by flow cytometry. The expression of transcription factor p65 (p65), B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), multidrug resistance 1 (MDR1) and multidrug resistance-associated protein 1 (MRP1) at mRNA and protein levels were detected by reverse transcription quantitative polymerase chain reaction and western blotting, respectively. Proliferation suppression rates and apoptosis levels were significantly increased in Jurkat and Hut-78 cells combined with celecoxib compared with those without celecoxib, when treated with CDDP, epirubicin and VCR. The IC50 values of the chemotherapy agents were lower in Jurkat and Hut-78 cells treated with celecoxib compared with those that were not. The apoptosis level, expression of Bax and the intracellular concentration of Rhodamine-123 were increased, whereas the expression of p65, Bcl-2, MDR1 and MRP1 were decreased, in celecoxib-treated Jurkat and Hut-78 cells compared with those without celecoxib treatment. These results indicated that celecoxib may enhance the sensitivity of T-cell lymphoma to chemotherapy drugs by inhibiting the expression of multidrug resistance (MDR)-associated proteins via downregulating the activity of the nuclear factor-κB signaling pathway, suggesting that celecoxib may improve the curative effect of chemotherapy drugs in T-cell lymphoma.

7.
Hematology ; 23(8): 448-455, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29415639

ABSTRACT

Objectives Burkitt lymphoma is one of the most common types of haematopoietic malignancy in children and adolescents. Mda-7/IL-24 had been identified as a differentiation inducer of Burkitt lymphoma cells. Previous studies have revealed that knockdown of C-myb can also lead to the terminal differentiation of Burkitt lymphoma cells. The aim of the present study was to investigate the correlation between the expression of Mda-7/IL-24 and C-myb, as well as their prognostic significance, for Burkitt lymphoma patients. Methods The tumour tissues were collected from 59 cases of Burkitt lymphoma patients and detected with Western blotting and immunohistochemistry. Results The results showed that the expression of Mda-7/IL-24 was lower, whereas the expression of C-myb was higher in Burkitt lymphoma tissues compared to specimens of normal lymph node tissues. Furthermore, C-myb expression was negatively correlated with Mda-7/IL-24 expression at the protein level in Burkitt lymphoma tissues and cell lines. Both the expression of Mda-7/IL-24 and C-myb in Burkitt lymphoma tissues was associated with some clinicopathological parameters, such as clinical stage, infiltration in the bone marrow, Ki67 and overall survival rates. Conclusion These results indicated that low expression of Mda-7/IL-24 along with high expression of C-myb are predictors for poor prognosis of Burkitt lymphoma patients; this outcome suggests that Mda-7/IL-24 and C-myb might be potential targets for clinical treatment of Burkitt lymphoma. ABBREVIATIONS: Mda-7/IL-24: melanoma differentiation associated gene7/interleukin 24; FCM: flow cytometry; Ecog: Eastern Cooperative Oncology Group; IPI: International lymphoma prognosis index.


Subject(s)
Burkitt Lymphoma/metabolism , Gene Expression Regulation, Neoplastic , Interleukins/biosynthesis , Proto-Oncogene Proteins c-myb/biosynthesis , Adolescent , Burkitt Lymphoma/mortality , Burkitt Lymphoma/pathology , Burkitt Lymphoma/therapy , Cell Line, Tumor , Child , Child, Preschool , Female , Humans , Infant , Male
8.
Mol Med Rep ; 16(4): 5633-5642, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28849038

ABSTRACT

Interleukin 24 (IL­24) is a unique cytokine encoded by the melanoma differentiation associated gene­7 (Mda­7), and was first discovered inhuman melanoma cells. Exogenous Mda­7/IL­24 has been shown to inhibit the proliferation and invasion of a broad spectrum of human cancer cells, but its effect on the differentiation of B cell lymphoma is not yet clear. To the best of our knowledge, the present study demonstrated for the first time that overexpressing Mda­7/IL­24 can induce differentiation in human B cell lymphomacells, and the underlying mechanism was investigated. The proliferation of stable Mda­7/IL­24 overexpressing Raji and Daudi cells was assessed by the MTS method. The immunophenotype, apoptosis level and cell cycle distribution of Raji and Daudi cells were analyzed by flow cytometry. The expression of PR domain zinc finger protein 1 (Blimp1) and B­cell lymphoma 6 (Bcl6) were analyzed by western blotting. Additionally, western blotting assay was also performed to study the effect of Mda­7/IL­24 on the activity of the P38 mitogen activated protein kinase (MAPK) signaling pathway in Raji and Daudi cells. Proliferation of Raji and Daudi cells overexpressing Mda­7/IL­24 was inhibited significantly, compared with those of parent cells and cells transfected with the empty vector alone. Apoptosis was not involved in the proliferation inhibition, while the cell cycle was arrested in G1 phase in the Raji and Daudi cells overexpressing Mda­7/IL­24. Overexpressing Mda­7/IL­24 resulted in a significantly decreased expression of cluster of differentiation (CD)10, and increased expression of CD45 and CD138 in the cell surface of Raji and Daudi cells. The expression of Blimp1 was upregulated, while the levels of Bcl6 protein was downregulated, in Raji and Daudi cells overexpressing Mda­7/IL­24. Furthermore, the activities of the P38 MAPK signaling pathway in lymphoma cells were upregulated. These results indicated that Mda­7/IL­24 could induce terminal differentiation of B lymphoma cells by regulating the expression of Blimp1 and Bcl6 via altering the P38 MAPK signaling pathway, suggesting that Mda­7/IL­24 may therefore be a potential differentiation therapeutic agent to be applied in clinical treatment of B cell lymphoma.


Subject(s)
Interleukins/metabolism , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Flow Cytometry , Gene Expression , Humans , Immunophenotyping , Interleukins/genetics , Lymphoma, B-Cell/genetics
9.
Oncotarget ; 8(12): 19661-19673, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-28152502

ABSTRACT

Bridging integrator-1 (Bin1), as a tumor suppressor, is frequently attenuated or even abolished in multiple primary cancers. A reduced expression of Bin1 caused by DNA methylation, has been reported in breast and prostate cancers. However, the methylation status of Bin1 and potent biological functions in esophageal squamous cell carcinoma (ESCC) remain unclear. In a previous study, we showed that the Bin1 expression was low in ESCC tissues. Herein, we further characterized this mechanism, confirming that gene hypermethylation was significantly correlated with the aberrant attenuation of Bin1. In addition, the Bin1 hypermethylation was associated with the poorer clinical parameters and shorter survival times of ESCC patients. Methylation-specific reverse transcription-polymerase chain reaction (MS-RT-PCR) showed that Bin1 was hypermethylated in several ESCC cell lines, which might be the main cause of reduced Bin1 expression. In addition, treatment with the de-methylation agent Decitabine (DAC) could restore Bin1 expression and evidently restrained ESCC cell malignant behaviors, particularly the epithelial mesenchymal transition (EMT) via reactivating the PTEN/AKT signaling pathway to inhibit matrix metalloproteinase (MMP)-2 and MMP-9 expression in vitro and in vivo. In conclusion, these results demonstrated that Bin1 methylation could augment the malignant biological behaviors of ESCC and predict the poor prognosis for ESCC patients, thus indicating the potential clinical application value of DAC-based de-methylation therapy in ESCC.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Azacitidine/analogs & derivatives , Carcinoma, Squamous Cell/drug therapy , DNA Methylation , Epithelial-Mesenchymal Transition/drug effects , Esophageal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Azacitidine/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/secondary , Cell Cycle , Cell Movement , Cell Proliferation , Decitabine , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Follow-Up Studies , Genes, Tumor Suppressor , Humans , Lymphatic Metastasis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Nuclear Proteins/metabolism , Prognosis , Promoter Regions, Genetic , Signal Transduction , Survival Rate , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
10.
Int J Oncol ; 48(5): 1985-96, 2016 May.
Article in English | MEDLINE | ID: mdl-26936624

ABSTRACT

Radiotherapy has been widely used for the treatment of cancer patients, especially for esophageal cancer patients. Ring finger protein 2 (RNF2) plays an important role in promoting the growth of cancer cells after exposure to irradiation. The present study aims to characterize the proliferative effects of RNF2 on cancer cells, and its mechanisms on the growth of esophageal cancer cells. We demonstrate that expression of RNF2 was markedly upregulated in esophageal cancer cell lines and surgically resected cancer specimens. In addition, RNF2 expression level is positively correlated with the presence of tumor size, lymph node metastases and negatively correlated with patient survival rates, suggesting that it plays an important role in the progression of esophageal cancer. Furthermore, the expression of RNF2 at both mRNA and protein levels in esophageal cancer ECA109 and TE13 cells was detected by real-time PCR and western blot assay after shRNA targeting RNF2. Co-immunoprecipitation (Co-IP) assay and western blot analysis were employed to detect the interaction between RNF2 and r-H2AX, H2AK119ub, and the expression of proteins associated with cell cycle and apoptosis, respectively. We used flow cytometry assay to analyze cell cycle and apoptosis of transfected cells, and further examined cellular growth in vitro and in vivo. shRNA targeting RNF2 gene and protein downregulated RNF2 expression after transfection for 24 h. The proliferation of tumor cells in RNF2-shRNA group was suppressed after radiotherapy. In addition, the interaction of RNF2, H2AK119ub, r-H2AX was increased after exposure to IR, followed by increasing apoptosis rates and inducing the arrest at G0/G1 phase after irradiation and shRNA targeting RNF2. Expression of the short-hairpin RNA is also correlated with the upregulation of p16 and Bax, and the downregulation of cyclin D2, cyclin-dependent kinase (CDK)-4, H2AX and Bcl-2. RNF2 gene knockdown induces radiosensitivity of esophageal cancer cells in vitro and significantly inhibits the growth of tumor cells. The mechanisms include inducing the cell cycle arrest at G0/G1 phase and promoting apoptosis.


Subject(s)
Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Gene Knockdown Techniques/methods , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Apoptosis , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Esophageal Neoplasms/genetics , Esophageal Neoplasms/radiotherapy , Female , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Male , Prognosis , Survival Analysis , Up-Regulation
11.
J Insect Sci ; 142014.
Article in English | MEDLINE | ID: mdl-25527590

ABSTRACT

Midgut α-amylase is an important digestive enzyme involved in larval energy metabolism and carbohydrate assimilation. In this article, the properties of midgut α-amylase from the Oriental armyworm, Mythimna separata (Lepidoptera: Noctuidae), larvae were characterized, and its in vitro responses to chemical inhibitors were also determined. The kinetic parameters Km and Vmax of midgut α-amylase were 0.064 M, 4.81 U mg pro(-1) in phosphate buffer, and 0.128 M, 1.96 U mg pro(-1) in barbiturate-acetate buffer; α-amylase activity linearly increased as starch concentration increased. α-Amylase activity was not influenced by amino acids such as Pro, Met, Try, His, Ala, and Phe but was strongly activated by antioxidants. Reduced glutathione, 1,4-dithiothreitol, ß-mercaptoethanol, and ascorbic acid improved the activity of α-amylase about 2.06, 3.46, 3.37, and 6.38 times, respectively, relative to the control. Ethylenediaminetetraacetic acid, sodium dodecyl sulfonate, and N-bromosuccinimide (NBS) strongly inhibited α-amylase. α-, ß-, and γ-cyclodextrin were not the preferred substrates for α-amylase. Kinetic analysis showed that IC50 value of NBS against α-amylase was 1.52 (±0.26) µM, and the mode of action of NBS with Ki as 2.53 (0.35) µM was a mixed-type inhibition that indicated a combination of partial competitive and pure noncompetitive inhibition. The midgut α-amylase of armyworm larvae may be a potential target for novel insecticide development and pest control.


Subject(s)
Bromosuccinimide/pharmacology , Glycoside Hydrolase Inhibitors/pharmacology , Insect Proteins/metabolism , Moths/metabolism , alpha-Amylases/metabolism , Animals , Digestive System/drug effects , Digestive System/metabolism , Insecticides/pharmacology , Kinetics , Larva/drug effects , Larva/metabolism , Moths/drug effects , Moths/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...