Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 44(2): 338-347, 2022 Apr.
Article in Chinese | MEDLINE | ID: mdl-35538772

ABSTRACT

Programmed necrosis,a mode of cell death independent of Caspase,is mainly mediated by receptor-interacting protein kinase-1 (RIPK1),receptor-interacting protein kinase-3 (RIPK3),and mixed lineage kinase domain-like protein (MLKL).Studies have demonstrated that programmed necrosis has the dual role of promoting and inhibiting tumor growth and thus we can control the development of tumor by regulating programmed necrosis.The drugs capable of inducing programmed necrosis show potential anti-tumor activity.In addition,inducing programmed necrosis is an effective way to overcome tumor resistance to apoptosis.This paper summarized the mechanisms of programmed necrosis and its relationship with tumors.We focused on the antitumor activity of programmed necrosis inducers including natural products,chemotherapeutic drugs,death receptor ligands,kinase inhibitors,inorganic salts,metal complexes,and metal nanoparticles.These agents will provide new therapeutic candidates for the treatment of tumors,especially the tumors acquiring resistance to apoptosis.


Subject(s)
Neoplasms , Protein Kinases , Apoptosis , Cell Death , Humans , Necrosis/metabolism , Necrosis/pathology , Neoplasms/drug therapy , Protein Kinases/metabolism , Protein Kinases/pharmacology
2.
Front Psychiatry ; 13: 844578, 2022.
Article in English | MEDLINE | ID: mdl-35401285

ABSTRACT

Background: Previous research has suggested that children with autism spectrum disorder (ASD) display fewer prosocial behaviors, and the role of empathy or Theory of Mind (ToM) in prosocial behaviors of autistic children remains unclear. Methods: Data were obtained from an ongoing longitudinal study in Guangzhou, China. A total of 96 autistic children and 167 typically developing (TD) children were enrolled. Prosocial behaviors were assessed using a subscale of the Strength and Difficulties Questionnaire and Dictator Game (DG) paradigm with stickers as incentives. Empathic traits and ToM ability were measured using the children's Empathy Quotient and the Chinese version of ToM toolkit. Generalized linear models were used to assess the differences of prosocial behaviors and empathic traits, ToM ability between the two groups and the associations between empathic traits, ToM ability and prosocial behaviors in autistic children. Results: Compared with TD children, autistic children exhibited worse ToM ability and performed less pro-socially in the DG paradigm, while there were no differences regarding empathic traits. In autistic children, empathic traits especially affective empathy, were positively associated with parent-reported prosocial behaviors [ß = 0.17, 95% confidence interval (CI): 0.07-0.27; ß = 0.47, 95%CI: 0.33-0.60]. ToM ability was associated with DG paradigm (ß = 1.03, 95%CI: 0.16-1.89). Conclusion: Autistic children showed less pro-sociality and ToM ability than TD children. In autistic children, empathic trait was associated with parent-reported prosocial behaviors while their ToM ability was associated with prosocial behaviors in experimental condition. Our findings indicated that better ToM ability and empathic trait might promote prosocial behaviors in autistic children.

3.
Eur J Pharmacol ; 919: 174802, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35143830

ABSTRACT

IMB5046 is a nitrobenzoate microtubule inhibitor we reported previously. During screening of its structural analogues, we identified a novel compound IMB5476 with increased aqueous solubility. Here, its antitumor activity and the underlying mechanism were investigated. IMB5476 disrupted microtubule networks in cells and arrested cell cycle at G2/M phase. It inhibited purified tubulin polymerization in vitro. Competition assay indicated that it bound to tubulin at the colchicine pocket. Further experiments proved that it induced cell death by mitotic catastrophe and apoptosis. Notably, it was a poor substrate of P-glycoprotein and exhibited potent cytotoxicity against drug-resistant tumor cells. In addition, IMB5476 could inhibit angiogenesis in vitro. IMB5476 also inhibited the growth of drug-resistant KBV200 xenografts in mice. Conclusively, our data reveal a novel nitrobenzoate microtubule inhibitor with improved aqueous solubility and can overcome multidrug resistance.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Tubulin Modulators/pharmacology , Animals , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Microtubules/metabolism , Tubulin Modulators/therapeutic use , Xenograft Model Antitumor Assays
4.
Cancer Chemother Pharmacol ; 87(3): 425-436, 2021 03.
Article in English | MEDLINE | ID: mdl-33388950

ABSTRACT

PURPOSE: To investigate the antitumor efficacy of pingyangmycin (PYM) in combination with anti-PD-1 antibody and determine the capability of PYM to induce immunogenic cell death (ICD) in cancer cells. METHODS: The murine 4T1 breast cancer and B16 melanoma models were used for evaluation of therapeutic efficacy of the combination of PYM with anti-PD-1 antibody. The ELISA kits were used to quantify the ICD related ATP and HMGB1 levels. The Transwell assay was conducted to determine the chemotaxis ability of THP-1 cell in vitro. The flow cytometry was used to measure reactive oxygen species level and analyze the ratio of immune cell subsets. RESULTS: PYM induced ICD in murine 4T1 breast cancer and B16 melanoma cells and increased the release of nucleic acid fragments that may further promote the monocytic chemotaxis. In the 4T1 murine breast cancer model, PYM alone, anti-PD-1 antibody alone, and their combination suppressed tumor growth by 66.3%, 16.1% and 77.6%, respectively. PYM markedly enhanced the therapeutic efficacy of anti-PD-1 antibody against 4T1 breast cancer. The calculated CDI (coefficient of drug interaction) indicated synergistic effect. Evaluated by graphic analysis, the nucleated cells intensity in the femur bone marrow remained unchanged. Histopathological observations revealed no noticeable toxico-pathological changes in the lung and various organs, indicating that the PYM and anti-PD-1 antibody combination exerted enhanced efficacy at well-tolerated dosage level. By the combination treatment, a panel of immunological changes emerged. The ratio of CD3+ cells, NK cells and NKT cells increased and Tregs decreased in peripheral blood. The DCs increased in the spleen. Prominent changes occurred in tumor infiltrating lymphocytes. The ratio of CD8+ cells increased, while that of CD4+ cells decreased; however, the ratio of CD3+ cells remained unchanged, implying that certain immunological responses emerged in the tumor microenvironment. PYM alone could also increase CD8+ cells and reduce CD4+ cells in tumor infiltrating lymphocytes. CONCLUSIONS: The studies indicate that PYM, as an ICD inducer with mild myelosuppression effect, may enhance the therapeutic efficacy of anti-PD-1 antibody in association with tumor infiltrating CD8+ T cell augmentation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Mammary Neoplasms, Animal/drug therapy , Melanoma, Experimental/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antibodies/administration & dosage , Antibodies/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bleomycin/administration & dosage , Bleomycin/analogs & derivatives , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Lymphocytes, Tumor-Infiltrating/metabolism , Mammary Neoplasms, Animal/pathology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , Tumor Microenvironment/immunology
5.
J Med Virol ; 92(12): 3726-3735, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32557739

ABSTRACT

Mucins have long been regarded to play a role as a barrier to prevent mucosal infections; however, some studies report that overexpression of mucins induces obstruction and inflammation of airways. We investigated whether the secretion of overexpressed mucin, mucin5ac (MUC5AC), could improve protection against pathogens. To examine the possible roles of mucin hypersecretion in augmenting host defense against disease-promoting muco-obstructive lung disease, a mouse model that overexpressed MUC5AC was generated. We had previously proved that murine gammaherpesvirus-68 (MHV-68) infection could induce emphysema in mice, which later developed into combined pulmonary fibrosis and emphysema (CPFE). We further explored whether increased MUC5AC secretion could provide benefits against MHV-68 induced fibrosis. We initially developed a pcDNA3.1-MUC5AC mouse model. Next, the experimental mice were randomly divided into five groups: normal control, pcDNA3.1 control, pcDNA3.1-MUC5AC, CPFE, and pcDNA3.1- MUC5AC + CPFE. Morphometric analysis of each group was performed by hematoxylin and eosin staining and Masson trichrome staining. MUC5AC levels in lung tissues were analyzed by immunohistochemical staining, real-time polymerase chain reaction, and Western blot analysis. The airway inflammation was determined by differential cell counts of bronchoalveolar lavage fluid (BALF) and measurement of cytokines and chemokines in BALF by enzyme-linked immunosorbent assay. MUC5AC hypersecretion alone was not sufficient to drive goblet cell metaplasia to induce obvious mucus plugging and airway inflammation. However, MUC5AC overexpression served as a protective barrier against MHV-68 virus infection in vivo. Infectivity of MHV-68 was decreased in the pcDNA3.1-MUC5AC + CPFE group compared with that in CPFE group. Meanwhile, a reduction of MHV-68 virus attenuated the expressions of chemokine (C-C motif) ligand 2 (CCL2), chemokine (C-X-C motif) ligand 5 (CXCL5), interleukin-13 (IL-13), and transforming growth factor-ß1 (TGF-ß1), and weakened airway inflammation and fibrosis in the pcDNA3.1-MUC5AC + CPFE group. Overexpression of MUC5AC appears to exhibit a protective role against MHV-68 infection in mice with emphysema that subsequently developed into CPFE and to further decrease airway inflammation and fibrosis induced by MHV-68 by decreasing the expressions of CCL2, CXCL5, IL-13, and TGF-ß1.

6.
J Cell Mol Med ; 24(13): 7228-7238, 2020 07.
Article in English | MEDLINE | ID: mdl-32452639

ABSTRACT

Microtubule-depolymerizing agents can selectively disrupt tumor vessels via inducing endothelial membrane blebbing. However, the mechanism regulating blebbing is largely unknown. IMB5046 is a newly discovered microtubule-depolymerizing agent. Here, the functions of focal adhesion kinase (FAK) during IMB5046-induced blebbing and the relevant mechanism are studied. We found that IMB5046 induced membrane blebbing and reassembly of focal adhesions in human vascular endothelial cells. Both FAK inhibitor and knock-down expression of FAK inhibited IMB5046-induced blebbing. Mechanism study revealed that IMB5046 induced the activation of FAK via GEF-H1/ Rho/ ROCK/ MLC2 pathway. cRGD peptide, a ligand of integrin, also blocked IMB5046-induced blebbing. After activation, FAK further promoted the phosphorylation of MLC2. This positive feedback loop caused more intensive actomyosin contraction and continuous membrane blebbing. FAK inhibitor blocked membrane blebbing via inhibiting actomyosin contraction, and stimulated stress fibre formation via promoting the phosphorylation of HSP27. Conclusively, these results demonstrate that FAK is a molecular switch controlling endothelial blebbing and stress fibre formation. Our study provides a new molecular mechanism for microtubule-depolymerizing agents to be used as vascular disrupting agents.


Subject(s)
Benzoates/pharmacology , Cell Surface Extensions/metabolism , Endothelial Cells/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Microtubules/metabolism , Morpholines/pharmacology , Cardiac Myosins/metabolism , Cell Surface Extensions/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Heat-Shock Proteins/metabolism , Humans , Integrins/metabolism , Models, Biological , Molecular Chaperones/metabolism , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Quinolones/pharmacology , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/drug effects , Stress Fibers/drug effects , Stress Fibers/metabolism , Sulfones/pharmacology , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
7.
Cancer Biol Ther ; 21(4): 332-343, 2020 04 02.
Article in English | MEDLINE | ID: mdl-31906826

ABSTRACT

Gemcitabine (Gem) is currently used as the first-line therapy for liver and pancreatic cancer but has limited efficacy in most cases. Dexamethasone (Dex) have been applied as a chemoprotectant and chemosensitizer in cancer chemotherapy. This study further explored the potential of combination of Gem and Dex and tested the hypothesis that glucocorticoid receptor signaling is essential for the synergistic antitumor activity. In the HepG2 and AsPC-1 xenograft models, the combination treatment showed a significantly synergistic antitumor activity. Immunohistochemistry of post-treatment tumors showed a significant decrease in proliferation and angiogenesis as compared to either of the treatments alone. Dex alone and the combination with Gem inhibited the expression of glucocorticoid receptor. The combination of Dex and Gem showed synergistic cytotoxicity in cell lines in vitro. The antiproliferative synergism is prevented by used glucocorticoid receptor (GR) small interfering RNA, demonstrating that the glucocorticoid receptor is required for the antiproliferative synergism of Gem and Dex. The inhibition of glucocorticoid receptor signaling pathway and induction of apoptosis via activation of caspases 3, 8 and 9, PARP, contributed to the synergistic effect of this combination therapy. These results demonstrate that Dex could potentiate the antitumor efficacy of Gem. The synergistic antitumor activity of the combination of Dex and Gem was through glucocorticoid receptor signaling. Taken together, a combination of Dex and Gem shows a significant synergistic antitumor activity and lesser toxicity both in vitro and in vivo and could be a combination chemotherapy for the treatment of highly expression of glucocorticoid receptor patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Receptors, Glucocorticoid/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dexamethasone/administration & dosage , Female , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
8.
Oncol Rep ; 41(1): 475-482, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30542729

ABSTRACT

Apoptosis induction and differentiation of promyelocytic leukemic cells into mature cells are major strategies for the drug-based treatment of leukemia. Lidamycin (LDM) which is a member of the enediyne antibiotic family exhibits extreme cytotoxicity. In the present study, the induction of apoptosis and differentiation in human chronic myeloid leukemia K562 cells by low concentrations of lidamycin were investigated. K562 cells were treated with lidamycin at various concentrations for 48 h, and accumulated in the metaphase as determined in previous experiments. Cell viability was determined using a Cell Counting Kit-8 (CCK-8) assay and the IC50 value of lidamycin was 0.1±3.2 nM. Induction of apoptosis was investigated morphologically by acridine orange/ethidium bromide (AO/EtBr) staining. Growth inhibition and apoptosis induction were observed in cells treated with low concentrations of lidamycin. In addition, western blot analysis revealed that treatment of the K562 cells with lidamycin at low concentrations upregulated the expression of caspase-8 and caspase-3. The induction of differentiation in human chronic myeloid leukemia K562 cells by lidamycin at low concentrations was also investigated. The nitroblue tetrazolium reduction ability of K562 cells was increased following treatment with lidamycin. Low concentrations of lidamycin triggered erythroid differentiation among K562 cells, indicated by morphological changes, increased hemoglobin content, and the expression of cell surface antigens such as CD71. Additionally the expression of GATA-binding factor 1 (GATA-1) protein in low concentration lidamycin-treated K562 cells was increased. The results of the present study suggest that a low-concentration lidamycin exerts effects on apoptosis and erythroid differentiation induction by increasing the expression of caspases and GATA-1 protein. Lidamycin may serve a positive role in relevant targeted chemotherapy and may represent a potential candidate for chronic myelogenous leukemia differentiation-inducing treatment.


Subject(s)
Aminoglycosides/pharmacology , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Enediynes/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Aminoglycosides/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antigens, CD/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enediynes/therapeutic use , GATA1 Transcription Factor/metabolism , Hemoglobins/metabolism , Humans , K562 Cells , Nitroblue Tetrazolium/metabolism , Receptors, Transferrin/metabolism
9.
Mol Med Rep ; 18(1): 407-414, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29750303

ABSTRACT

Lung cancer is the leading cause of mortality due to tumor malignancy worldwide. In recent years, the treatment of lung cancer with chemotherapy has demonstrated notable resistance and insensitivity. Therefore, it is of great importance to investigate anti­lung cancer drugs with high efficiency and low toxicity. In the present study, the effects of isofraxidin on lung cancer cells and the associated mechanisms were investigated. The results revealed that, in vivo and in vitro, isofraxidin exhibited marked inhibitory effects on the A549 lung cancer cell line. The results of Cell Counting kit­8, Transwell migration and Matrigel invasion assays, and flow cytometry to determine apoptosis, revealed that isofraxidin significantly inhibited the proliferation, migration and invasion of A549 cells, and induced the cell apoptosis. Furthermore, western blot analysis demonstrated that isofraxidin treatment led to effects on the expression of apoptosis­associated proteins, including members of the Bcl­2 protein family, and invasion­associated proteins, including matrix metallopeptidase (MMP)­2 and MMP­9, which may occur via inhibition of the expression of phosphorylated (p)­epidermal growth factor receptor, p­AKT and p­extracellular signal­regulated kinase. This regulation of protein expression may contribute to the inhibition of proliferation, migration and invasion of A549lung cancer cells by isofraxidin. In addition, despite the inhibitory effects on the A549 lung cancer cell line, the present study revealed that isofraxidin exhibited low toxicity towards BEAS­2B normal lung epithelial cells within a certain dose range (0­160 µM), indicating that isofraxidin may be employed for lung cancer treatment with hypotoxicity and fewer side effects. In conclusion, isofraxidin may be a novel candidate for anti­lung cancer chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Coumarins/pharmacology , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Neoplasm Proteins/metabolism , Signal Transduction/drug effects , A549 Cells , ErbB Receptors/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasm Proteins/genetics , Signal Transduction/genetics
10.
Mol Carcinog ; 56(5): 1395-1404, 2017 05.
Article in English | MEDLINE | ID: mdl-27991698

ABSTRACT

CD13 is a marker of angiogenic endothelial cells, and recently it is proved to be a biomarker of human liver cancer stem cells (CSCs). Herein, the therapeutic effects of NGR-LDP-AE, a fusion protein composed of CD13-targeting peptide NGR and antitumor antibiotic lidamycin, on human liver cancer and its mechanism were studied. Western blot and immunofluorescence assay demonstrated that CD13 (WM15 epitope) was expressed in both human liver cancer cell lines and vascular endothelial cells, while absent in normal liver cells. MTT assay showed that NGR-LDP-AE displayed potent cytotoxicity to cultured tumor cell lines with IC50 values at low nanomolar level. NGR-LDP-AE inhibited tumorsphere formation of liver cancer cells, and the IC50 values were much lower than that in MTT assay, indicating selectively killing of CSCs. In endothelial tube formation assay, NGR-LDP-AE at low cytotoxic dose significantly inhibited the formation of intact tube networks. Animal experiment demonstrated that NGR-LDP-AE inhibited the growth of human liver cancer xenograft. Immunohistochemical analysis showed that NGR-LDP-AE induced the down-regulation of CD13. In vitro experiment using cultured tumor cells also confirmed this result. NGR-LDP-AE activated both apoptotic and autophagic pathways in cultured tumor cells, while the induced autophagy protected cells from death. Conclusively, NGR-LDP-AE exerts its antitumor activity via killing liver CSCs and inhibiting angiogenesis. With one targeting motif, NGR-LDP-AE acts on both liver CSCs and angiogenic endothelial cells. It is a promising dual targeting fusion protein for liver cancer therapy, especially for advanced or relapsed cancers.


Subject(s)
Antineoplastic Agents/pharmacology , CD13 Antigens/metabolism , Liver Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Neovascularization, Pathologic/drug therapy , Recombinant Proteins/pharmacology , Animals , Autophagy/drug effects , Cell Line, Tumor , Female , Humans , Liver/blood supply , Liver/drug effects , Liver/pathology , Liver Neoplasms/pathology , Mice, Nude , Molecular Targeted Therapy/methods , Oligopeptides , Xenograft Model Antitumor Assays
11.
Sci Rep ; 6: 31472, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27510727

ABSTRACT

Multidrug resistance is a major limitation for microtubule-binding agents in cancer treatment. Here we report a novel microtubule inhibitor (2-morpholin-4-yl-5-nitro-benzoic acid 4-methylsulfanyl-benzyl ester, IMB5046), its cytotoxicity against multidrug-resistant cell lines and its antitumor efficacy in animal models. IMB5046 disrupted microtubule structures in cells and inhibited purified tubulin polymerization in vitro. It bound to the colchicine pocket of tubulin. IMB5046 displayed potent cytotoxicity against multiple tumor cell lines with an IC50 range of 0.037-0.426 µM. Notably, several multidrug-resistant cell lines which were resistant to colchicine, vincristine and paclitaxel remained sensitive to IMB5046. IMB5046 was not a P-glycoprotein substrate. IMB5046 blocked cell cycle at G2/M phase and induced cell apoptosis. Microarray assay indicated that the differentially expressed genes after IMB5046 treatment were highly related to immune system, cell death and cancer. In a mouse xenograft model IMB5046 inhibited the growth of human lung tumor xenograft by 83% at a well-tolerated dose. It is concluded that IMB5046 is a tubulin polymerization inhibitor with novel chemical structure and can overcome multidrug resistance. It is a promising lead compound for cancer chemotherapy, especially for treatment of multidrug-resistant tumors.


Subject(s)
Benzoates/administration & dosage , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Morpholines/administration & dosage , Neoplasms/drug therapy , Nitrobenzoates/administration & dosage , Tubulin Modulators/administration & dosage , A549 Cells , Animals , Benzoates/chemistry , Benzoates/pharmacology , Cell Cycle/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , HT29 Cells , Humans , Mice , Morpholines/chemistry , Morpholines/pharmacology , NIH 3T3 Cells , Neoplasms/genetics , Nitrobenzoates/chemistry , Nitrobenzoates/pharmacology , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Xenograft Model Antitumor Assays
12.
J Huazhong Univ Sci Technolog Med Sci ; 36(2): 285-288, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27072977

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by high heritability. Recently, autism, the most profound form of ASD, has been increasingly attributed to synaptic abnormalities. Postsynaptic density 95 (PSD95), encoding PSD protein-95, was found essential for synaptic formation, maturation and plasticity at a PSD of excitatory synapse. It is possibly a crucial candidate gene for the pathogenesis of ASD. To identify the relationship between the rs13331 of PSD95 gene and ASD, we performed a case-control study in 212 patients and 636 controls in a Chinese population by using a polymerase chain reaction-restriction fragment length polymerase (PCR-RFLP) assay. The results showed that in genetic analysis of the heterozygous model, an association between the T allele of the rs13331 and ASD was found in the dominant model (OR=1.709, 95% CI 1.227-2.382, P=0.002) and the additive model (OR=1.409, 95% CI=1.104-1.800, P=0.006). Our data indicate that the genetic mutation C>T at the rs13331 in the PSD95 gene is strikingly associated with an increased risk of ASD.


Subject(s)
Autism Spectrum Disorder/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Polymorphism, Single Nucleotide , Aged , Case-Control Studies , Child , Child, Preschool , China , Disks Large Homolog 4 Protein , Female , Humans , Male , Middle Aged , Polymorphism, Restriction Fragment Length
13.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 36(7): 797-800, 2016 07.
Article in Chinese | MEDLINE | ID: mdl-30634204

ABSTRACT

Objective To study the effect of rhubarb combined medicinal activated carbon on levels of serum phosphorus and calcium-phosphorus product, as well as parathyroid hormone in hemodi- alysis patients with hyperphosphatemia. Methods Totally 126 hemodialysis patients with hyperphos- phatemia who had received treatment of calcium containing phosphorus binders were assigned to the treatment group and the control group according to random digit table, 63 in each group. Patients in the treatment group took rhubarb (80 mg/kg, twice per day, soaked in 150 mL boiled water, taken before dinner) combined with medicinal activated carbon (1. 5 g each time, thrice per day, taken during dinner). Those in the control group took medicinal activated carbon (1. 5 g each time, thrice per day, taken during dinner). The therapeutic course for all was 8 weeks. Levels of serum calcium , phosphorus, calcium- phosphorus product and parathyroid hormone were detected in the two groups before treatment, and at week 2, 4, 6, and 8 after treatment. Adverse reactions of gastrointestinal tract were observed as well. Results Twelve patients dropped out in the treatment group and the rest 51 completed the study. Nine patients dropped out in the treatment group and the rest 54 completed the study. Compared with before treatment in the same group, levels of serum phosphorus , calcium-phosphorus product, parathyroid hor- mone began to decline from week 2 to 8 in the treatment group (P <0. 05) ; and they began to decline from week 4 to 8 in the control group (P <0. 05). Compared with the control group, levels of serum phos- phorus, calcium-phosphorus product, parathyroid hormone obviously decreased in the treatment group (P <0.05). But there was no statistical difference in serum calcium level (P >0.05) between the two groups. Main adverse reactions of gastrointestinal tract mainly included abdominal distention, diarrhea, anal pedant expansion, and constipation. Compared with the control group, the incidence of abdominal distention and constipation, as well as the total incidence of gastrointestinal reactions all decreased in the treatment group (X2 =6. 815,7. 011 ,7.077, P <0. 05). Conclusion In addition to previous phosphorus binders therapy, rhubarb combined medicinal activated carbon could effectively reduce levels of serum phosphorus, calcium-phosphorus product, parathyroid hormone in hemodialysis patients with hyperphos- phatemia, and lessen complications of gastrointestinal tract.


Subject(s)
Hyperphosphatemia , Kidney Failure, Chronic , Parathyroid Hormone , Plant Extracts , Calcium/blood , Carbon/therapeutic use , Humans , Hyperphosphatemia/drug therapy , Hyperphosphatemia/etiology , Phosphorus/blood , Plant Extracts/therapeutic use , Renal Dialysis/adverse effects , Rheum/chemistry
14.
Cancer Chemother Pharmacol ; 69(5): 1323-32, 2012 May.
Article in English | MEDLINE | ID: mdl-22311160

ABSTRACT

PURPOSE: As reported, epidermal growth factor receptor (EGFR) is over expressed in a variety of cancers including esophageal squamous cell carcinoma. Therefore, it becomes one of the potential targets for treating esophageal cancer. Pingyangmycin (PYM), a single A5 component of bleomycin, is currently used for the treatment of different types of cancers of epidermal origin, especially for head and neck cancers. In this report, the effect of PYM on EGFR expression in human esophageal cancer cells and the therapeutic efficacy of the combination of PYM and cetuximab on esophageal cancer xenograft were investigated. METHODS: The effects of PYM, cetuximab and the combination on EGFR signaling, proliferation, cell cycle, apoptosis were evaluated by using MTT, Western blotting, RT-PCR assays and flow cytometry assays, respectively, in vitro and the therapeutic efficacy by a xenograft model in athymic mice. RESULTS: Cell volume and nucleus were enlarged after PYM treatment. PYM showed potent cytotoxicity in both cell lines of Kyse-150 and Eca-109 in time and dosage-depended manner in MTT assay. PYM treatment induced G(2)/M phase arrest and apoptosis. Notably, the expression of EGFR was down-regulated by PYM in EGFR highly expression esophageal cancer cells. PYM plus cetuximab resulted in a potentiation of antiproliferative activity. PYM combined with cetuximab displayed a much higher therapeutic effect than that of the single agent on esophageal cancer xenograft in athymic mice. CONCLUSIONS: PYM could down-regulate the expression of EGFR in esophageal cancer cells and potentiate the effects of cetuximab on esophageal cancer xenograft in nude mice. The combination of PYM and cetuximab, the EGFR-targeted combination of a chemotherapeutic agent and an antibody-based drug, might be useful in cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Down-Regulation/drug effects , ErbB Receptors/drug effects , Esophageal Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Bleomycin/administration & dosage , Bleomycin/analogs & derivatives , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cetuximab , Dose-Response Relationship, Drug , Drug Synergism , ErbB Receptors/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
15.
Yao Xue Xue Bao ; 47(12): 1593-8, 2012 Dec.
Article in Chinese | MEDLINE | ID: mdl-23460963

ABSTRACT

This study is to investigate the effects of ubenimex on tumor cell invasion and apoptosis, dose relationship and mechanism. Immunofluorescence staining was performed to detect the expression of CD13 in HT-1080 cells. MTT assay was used to analyze the effect of ubenimex on cell proliferation. Annexin V-EGFP/PI was used to detect apoptotic cells by flow cytometry. Cell cycle was analyzed using flow cytometry. Ala-pNA was used as substrate to evaluate the effect of ubenimex on the aminopeptidase activity. Transwell assay was used to analyze the effect of ubenimex on cell invasion and migration ability. Western blotting was used to detect the expression level of CD13. MMP activity was analyzed using gelatin zymography. The results showed that ubenimex at high concentration inhibited the proliferation of HT-1080 cells (IC50: 3.8 mg x mL(-1)), and induced cell apoptosis. Cell cycle was blocked at G1 phase. Ubenimex at low concentration inhibited the aminopeptidase activity of HT-1080 cells (IC50: 8.3 microg x mL(-1)) and inhibited cell invasion, but it had no effects on the cell migration and proliferation. Ubenimex had no effects on CD13 expression and MMP activity. In conclusion, ubenimex at low concentration can inhibit the invasion ability of tumor cells by directly inhibiting the aminopeptidase activity; ubenimex at high concentration can inhibit the proliferation of tumor cells and induce cell apoptosis by a CD13-independent pathway.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , CD13 Antigens/metabolism , Cell Movement/drug effects , Fibrosarcoma/pathology , Leucine/analogs & derivatives , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Fibrosarcoma/metabolism , Humans , Leucine/pharmacology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness
16.
Oncol Rep ; 23(6): 1747-56, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20428834

ABSTRACT

Potassium channels are essential for the regulation of cell proliferation. As reported, HERG protein is overexpressed in a wide range of human tumors, including colon carcinoma. The aim of this study was to investigate the effects of antibacterial agents sparfloxacin (SPFX), a blocker of HERG channel, on HERG K+ channel highly expressing colon cancer cells. Expression of HERG and apoptosis correlative proteins was examined by Western blotting. The MTT assay was used to detect the cytotoxicity of drugs and drug combination in vitro. Gene transfection was used to examine the changes in herg-related chemosensitivity. Cell apoptosis was analyzed by flow cytometry. The migration and invasion capacity of tumor cells by SPFX was determined by gelatin zymography assay and Boyden chamber. The in vivo efficacy of SPFX was assessed in murine colon carcinoma C26 in BALB/c mice and human colon carcinoma HCT116 xenografts in nude mice. High expression of HERG protein was detected in colon cancer C26, HCT116 and HT-29 cells. The cell viability of the colon cancer cells was inhibited by SPFX in a dose-dependent manner. SPFX induced apoptosis and inhibited migration and invasion of colon cancer HCT116 cells. The increase in apoptosis was associated with a decrease in procaspase-3 and Bcl-2 protein expression. Study with herg-transfected HEK293 cells and siRNA-knock down HCT116 cells confirmed that the cell viability inhibition by SPFX was correlated with HERG expression. When combined with 5-fluorouracil, SPFX showed synergistic anti-proliferation activity in HCT116 and HT-29 cells. Furthermore, SPFX inhibited the growth of human colon carcinoma HCT116 xenografts and showed synergistic effect with 5-fluorouracil in vivo. Our finding suggested that SPFX could be a biochemical modulator in treatment of colon cancer with chemotherapeutic drugs.


Subject(s)
Antitubercular Agents/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Ether-A-Go-Go Potassium Channels/metabolism , Fluoroquinolones/pharmacology , Animals , Blotting, Western , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , ERG1 Potassium Channel , Female , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Wound Healing/drug effects
17.
Guang Pu Xue Yu Guang Pu Fen Xi ; 29(3): 722-5, 2009 Mar.
Article in Chinese | MEDLINE | ID: mdl-19455808

ABSTRACT

The noise distribution of soil hyperspectra measured by ASD FieldSpec Pro FR was described, and then the quantitative evaluation of spectral denoising with six filters was compared. From the interpretation of soil hyperspectra, the continuum removed, first-order differential and high frequency curves, the UV/VNIR (350-1 050 nm) exhibit hardly noise except the coverage of 40 nm in the beginning 350 nm. However, the SWIR (1 000-2 500 nm) shows different noise distribution. Especially, the latter half of SWIR 2(1 800-2 500 nm) showed more noise, and the intersection spectrum of three spectrometers has more noise than the neighbor spectrum. Six filters were chosen for spectral denoising. The smoothing indexes (SI), horizontal feature reservation index (HFRI) and vertical feature reservation index (VFRI) were designed for evaluating the denoising performance of these filters. The comparison of their indexes shows that WD and MA filters are the optimal choice to filter the noise, in terms of balancing the contradiction between the smoothing and feature reservation ability. Furthermore the first-order differential data of 66 denoising soil spectra by 6 filters were respectively used as the input of the same PLSR model to predict the sand content. The different prediction accuracies caused by the different filters show that compared to the feature reservation ability, the filter's smoothing ability is the principal factor to influence the accuracy. The study can benefit the spectral preprocessing and analyzing, and also provide the scientific foundation for the related spectroscopy applications.

18.
Wei Sheng Wu Xue Bao ; 46(3): 373-8, 2006 Jun.
Article in Chinese | MEDLINE | ID: mdl-16933604

ABSTRACT

Isolate of Amanita spissa was obtained from basidiome stipe material collected from environment. It could utilize a broad range of carbon and nitrogen resources. Study on the influence of different conditions for solid culture was carried out. Optimal culture conditions were at 28 degrees C, pH6, in the dark. A. spissa was then fermentated in liquid culture for more mycelia. In flask and Airlift/ff bioreactor, maximum dry mycelia weight of A. spissa reached 0.893 g/L and 2.33 g/L, respectively. Mycelia obtained from solid culture and Airlift/ff bioreactor were then analyzed by HPLC. The results showed that mycelia from both cultures contained amatoxins but no phallotoxins. alpha-Amanitin in mycelia reached 26.02 microg/DWg under solid culture condition, and 15.25 microg/DWg under liquid culture condition. The amanitins were also confirmed by bud-inhibited assay. The results revealed that the effect of amanitin on mung bean cell was identical to that of authentic amanitins. This work suggests that it is possible to produce amatoxin by liquid culturing of A. spissa.


Subject(s)
Amanita/growth & development , Amanitins/analysis , Culture Techniques/methods , Amanita/chemistry , Amanita/drug effects , Amanitins/isolation & purification , Amanitins/toxicity , Bioreactors , Carbon/pharmacology , Chromatography, High Pressure Liquid , Darkness , Fabaceae/drug effects , Fabaceae/growth & development , Fermentation/drug effects , Hydrogen-Ion Concentration , Mycelium/chemistry , Mycelium/drug effects , Mycelium/growth & development , Nitrogen/pharmacology , Temperature
19.
Zhonghua Yi Xue Za Zhi ; 86(47): 3353-7, 2006 Dec 19.
Article in Chinese | MEDLINE | ID: mdl-17313833

ABSTRACT

OBJECTIVE: To investigate the effects of erythromycin on the proliferation of the human ether-a-go-go related gene (HERG) K(+) channel highly expressing cancer cells and its synergy with antitumor chemotherapeutic agents. METHODS: Human fibrosarcoma cell of the line HT-1080, murine colon carcinoma cells of the line C26, human colon carcinoma cells of the line HT-29, human pulmonary epithelial cells of the line A549, and human neuroblastoma cells of the line SH-SY5Y were cultured. Western blotting was used to detect the protein expression of the HERG K(+) channel protein. Erythromycin, taxol, and vincristine were added into the culture fluid. to observe the proliferation of the cancer cells. Collagen I was used to coat 96-well plate, HT-29 cells were put into, and Erythromycin, taxol, and vincristine were added, MTT method was used to examine the cell adhesion. SDS-PAGE was used to detect the secretion of gelatinase. Flow cytometry was used to detect the cell cycle and apoptosis. The coefficient of drug interaction (CDI) was calculated. RESULTS: HERG K(+) channel expression was found in both HT-29 human colon carcinoma cells and C26 murine colon carcinoma cells. The expression level in HT-29 cells was higher than that in the positive control SHSY5Y neuroblastoma cells. Erythromycin suppressed the proliferation of HT-29 and C26 cells in a dose-dependent manner. There existed a remarkable G(2)/M arrest after the cells were exposed to erythromycin. Induction of apoptosis in HT-29 cells and inhibition of cell adhesion to collagen I were found. Erythromycin inhibited the secretion of MMP-2 from HT-29 cells in a dose-dependent manner. At sub-cytotoxic concentration, erythromycin potentiated the cytotoxicity of vincristine, taxol, and hydroxyl-camptothecin to C26 cells. The IC(50) values for vincristine and vincristine plus erythromycin (50 micromol/L) were 62.65 nmol/Land and 4.68 nmol/L respectively. CONCLUSION: Erythromycin inhibits the proliferation and induces the apoptosis of cancer cells with high HERG K(+) channel expression. Synergy is found in the combination of erythromycin with other anticancer agents.


Subject(s)
Cell Proliferation/drug effects , Erythromycin/pharmacology , Ether-A-Go-Go Potassium Channels/metabolism , Vincristine/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , ERG1 Potassium Channel , Flow Cytometry , HT29 Cells , Humans , Neoplasms/metabolism , Neoplasms/pathology
20.
Wei Sheng Wu Xue Bao ; 45(5): 702-6, 2005 Oct.
Article in Chinese | MEDLINE | ID: mdl-16342759

ABSTRACT

The liquid culture of Laetiporus sulphureus var. sulphureus was lethal against fruit fly. It was found that extracellular metabolites were primary causation of the lethal effect against fruit fly, which was influenced by pH value. Isolation and analysis with ion-exchange resin column chromatography and HPLC demonstrated that oxalic acid was present in supernatant of Laetiporus sulphureus var. sulphureus, and it was one of the contributing factors to lethal effect against fruit fly and decrease of pH value of culture system. When cultured in airlift reactor, concentration of oxalic acid, quantity of mycelia and pH value was correlated with each other. Further analysis on elution revealed that a kind of oligidic pigment of amaranth in alkaline condition were also lethal against fruit fly.


Subject(s)
Polyporaceae/metabolism , Animals , Drosophila melanogaster/drug effects , Hydrogen-Ion Concentration , Oxalic Acid/analysis , Oxalic Acid/pharmacology , Polyporaceae/growth & development , Polyporaceae/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...