Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Life Sci ; 328: 121941, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37451400

ABSTRACT

AIMS: IMB5036 is a pyridazinone compound with antiproliferative and antitumour activity against hepatoma and pancreatic cancer. In this study, we attempted to identify the target protein of IMB5036 and test its potential for overcoming multidrug resistance and inducing pyroptosis. MATERIALS AND METHODS: We examined the effects of IMB5036 on cancer cells by in vitro assays, a molecular docking model and in vivo tumour models. We performed pull-down experiments using biotinylated IMB5036 and identified the binding proteins. Gene knockdown were used to investigate the oncogenic role of KH-type splicing regulatory protein (KSRP). Western blot was used to detect for mechanism-associated molecules. KEY FINDINGS: IMB5036 could overcome resistance to multiple chemotherapeutic drugs at the cellular level and in vivo. Furthermore, IMB5036 was not a P-glycoprotein (P-gp) substrate and downregulated the expression of P-gp. We identified KSRP as a binding protein of IMB5036. The knockdown of KSRP inhibited the proliferation of MCF7 and MCF7/adriamycin (MCF7/ADR) cells. In addition, IMB5036 induced pyroptosis in both MCF7 and MCF7/ADR cells via KSRP. SIGNIFICANCE: We found IMB5036 binds to KSRP and overcomes multidrug resistance via gasdermin E (GSDME)-dependent pyroptosis.


Subject(s)
Carcinoma, Hepatocellular , Pyroptosis , Humans , Molecular Docking Simulation , Drug Resistance, Multiple , Doxorubicin/pharmacology
2.
World Neurosurg ; 165: e628-e634, 2022 09.
Article in English | MEDLINE | ID: mdl-35772709

ABSTRACT

OBJECTIVE: To explore the application and effect of the "one disease, one product" project to the nursing care of patients who have undergone pituitary tumour surgery using the nasal sphenoid approach. METHODS: This is a prospective research study. In a standard treatment control study, 132 patients undergoing transnasal pituitary tumour surgery were divided into the control group (n = 71) and the observation group (n = 61). The control group was given routine pituitary tumor care, and the "one disease, one product" nursing model was used on the experimental group. The anxiety level of patients, the incidence of postoperative complications, postoperative hospitalization, and levels of satisfaction and capability of group members were measured between the control and experimental groups. RESULTS: There was no difference in the level of anxiety between the 2 groups before admission (P = 0.634). The anxiety level of the patients in the observation group decreased after the "one disease, one product" nursing intervention (P = 0.012), but in the control group, it did not decrease significantly (P = 0.149), and the anxiety level in the control group was significantly higher than in the observation group on day 1 preoperatively (P < 0.001). CONCLUSIONS: "One disease, one product" nursing can reduce the preoperative anxiety and postoperative satisfaction of pituitary adenoma surgery patients through the sphenoid sinus approach. It is worthy of popularization and application in pituitary adenoma resection through the sphenoid sinus approach.


Subject(s)
Adenoma , Pituitary Neoplasms , Adenoma/pathology , Adenoma/surgery , Humans , Pituitary Neoplasms/pathology , Pituitary Neoplasms/surgery , Prospective Studies , Retrospective Studies , Sphenoid Bone/pathology , Sphenoid Bone/surgery , Treatment Outcome
3.
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
4.
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
5.
Invest New Drugs ; 40(3): 487-496, 2022 06.
Article in English | MEDLINE | ID: mdl-35020067

ABSTRACT

Background Hepatocellular carcinoma (HCC) is one of the most common cancers with a high mortality rate due to metastasis and relapse. Purpose Here, we reported a small-molecule pyridazinone compound, designated as IMB5036. Its antitumor activity against HCC and underlying mechanism were studied. Methods In vitro cytotoxicity, apoptosis, DNA breaks, and cell motility assays were performed. Protein expression was analyzed by Western blot and microarray analysis. A xenograft tumor model in athymic mice was used to evaluate the antitumor activity. Results IMB5036 displayed potent cytotoxicity against various HCC cell lines. It caused double DNA breakages and induced cell death via apoptosis. It also significantly inhibited the motility of HCC cells. Western blot showed that IMB5036 induced the up-regulation of E-cadherin, while down-regulation of N-cadherin. The gene expression profile analysis and Western blot assay revealed that IMB5036 down-regulated the expression of Tau protein. Analysis of the TCGA dataset revealed that high expression of Tau decreased the survival rate of HCC patients. In vivo experiments proved that IMB5036 significantly inhibited the growth of HCC xenografts in athymic mice. Conclusions These results collectively demonstrate IMB5036 can be a promising therapeutic candidate for patients with HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Apoptosis , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/metabolism , Mice , Mice, Nude , Neoplasm Recurrence, Local/genetics , Xenograft Model Antitumor Assays
6.
Biochem Pharmacol ; 197: 114894, 2022 03.
Article in English | MEDLINE | ID: mdl-34968486

ABSTRACT

Mithramycin A (MIT) has reacquired extensive research attention due to its anti-solid tumor activity and improved pharmacological production. Mechanismly, MIT was broadly used as a c-Myc inhibitor, and c-Myc regulated CD47 and PD-L1 expression which has been demonstrated. However, how MIT affects immune check-point molecules remains unknown. In this study, we found CD47 expression was higher in melanoma of pan-tissue array. MIT inhibited CD47 expression both in mRNA and protein level in melanoma cells (SK-MEL-28 and B16). MIT inhibited c-Myc, Sp-1 and CD47 expression in a concentration-dependent way. MIT inhibited the surface CD47 expression and promoted the phagocytosis of SK-MEL-28 cells by THP-1 cells. We found MIT inhibited tumor growth in melanoma allograft mice and CD47 expression in tumor mass. We also found MIT upregulated PD-L1 expression in cancer cells possibly via inhibiting PD-L1 ubiquitination, increasing ROS and IFN-γ. Combination of MIT and anti-PD-1 antibody showed enhanced antitumor activity compared to MIT and anti-PD-1 antibody alone in MC38 allograft mice. Using immune checkpoint array we found MIT inhibited expression of FasL and Galectin3. These results suggest that MIT inhibits CD47 expression, while improves PD-L1 expression. Furthermore, the combination of MIT and anti-PD-1 antibody exerts potent antitumor effect.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , B7-H1 Antigen/biosynthesis , CD47 Antigen/biosynthesis , Melanoma, Experimental/metabolism , Plicamycin/therapeutic use , Animals , Antibiotics, Antineoplastic/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CD47 Antigen/antagonists & inhibitors , Dose-Response Relationship, Drug , Female , Gene Expression , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Plicamycin/pharmacology , THP-1 Cells , Xenograft Model Antitumor Assays/methods
7.
Basic Clin Pharmacol Toxicol ; 130(3): 375-384, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34841678

ABSTRACT

IMB5036 is a novel pyridazinone compound with potent cytotoxicity. In this study, we reported its antitumour activity against pancreatic cancer and the underlying mechanism. We found that IMB5036 induced rapid cell swelling and increased membrane permeability in pancreatic cancer cells. IMB5036 increased the ratio of PI+ cells, which could be rescued by necroptosis inhibitor. Furthermore, MLKL inhibitor NSA attenuated the killing effect of IMB5036 on pancreatic cancer cells. IMB5036 stimulated translocation of MLKL and p-MLKL from cytoplasm to cell membrane. IMB5036 upregulated the level of p-RIPK1, p-RIPK3, and p-MLKL. At the same time, IMB5036 also partially activated apoptosis and pyroptosis. IMB5036 inhibited tumour growth in pancreatic xenograft. IMB5036 induced larger necrosis area, increased p-MLKL level, and inhibited Ki67 expression in tumour mass. The study indicates that IMB5036 inhibits human pancreatic cancer growth primarily activating necroptosis.


Subject(s)
Necroptosis , Pancreatic Neoplasms , Apoptosis , Humans , Necrosis , Pancreatic Neoplasms/drug therapy , Protein Kinases/metabolism
8.
Cancer Biol Ther ; 21(8): 749-757, 2020 08 02.
Article in English | MEDLINE | ID: mdl-32644888

ABSTRACT

Previous studies have shown that DBDx, a combination consisting of dipyridamole, bestatin and dexamethasone is highly effective against several cancer xenografts in athymic mice. Here the therapeutic effects of DBDx and its combination with gemcitabine or capcitabine against human pancreatic cancer xenografts and the mechanism were studied. In vivo experiments performed in athymic mice showed that the antitumor efficacy of DBDx was much stronger than that of gemcitabine or capecitabine alone. Notably, the combination of DBDx and gemcitabine or capcitabine further enhanced the efficacy. In the case of DBDx (242 mg/kg) plus gemcitabine (100 mg/kg), tumor weight decreased about 97.7%, and tumor sizes were shrinking during the treatment. In the case of DBDx (242 mg/kg) plus capecitabine (718.7 mg/kg), tumor weight decreased about 94.9%. Moreover, DBDx and its combinations obviously prolonged theoverall survival of mice compared with gemcitabine or capcitabine alone. DBDx-based drug combination therapy showed no obvious systematic toxicity. The gene expression profile analysis showed that the genes changed by DBDx were related to immune system and tumor vasculature. The result of protein array showed that the changed proteins in the serum of treated mice were related to immune and inflammation system. These results show that DBDx-based drug combinations, a new strategy which integrates the use of low-cytotoxic drugs and cytotoxic chemotherapeutics, are highly effective regimens against human pancreatic cancer in athymic mice at well tolerated doses. DBDx-based drug combination therapy might provide new options for the treatment of pancreatic cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Pancreatic Neoplasms/drug therapy , Aged , Animals , Antineoplastic Agents/pharmacology , Disease Models, Animal , Drug Combinations , Humans , Mice , Mice, Nude , Middle Aged , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays
9.
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
10.
World J Gastroenterol ; 26(6): 657-669, 2020 Feb 14.
Article in English | MEDLINE | ID: mdl-32103874

ABSTRACT

BACKGROUND: The treatment outcome of transarterial chemoembolization (TACE) in unresectable hepatocellular carcinoma (HCC) varies greatly due to the clinical heterogeneity of the patients. Therefore, several prognostic systems have been proposed for risk stratification and candidate identification for first TACE and repeated TACE (re-TACE). AIM: To investigate the correlations between prognostic systems and radiological response, compare the predictive abilities, and integrate them in sequence for outcome prediction. METHODS: This nationwide multicenter retrospective cohort consisted of 1107 unresectable HCC patients in 15 Chinese tertiary hospitals from January 2010 to May 2016. The Hepatoma Arterial-embolization Prognostic (HAP) score system and its modified versions (mHAP, mHAP2 and mHAP3), as well as the six-and-twelve criteria were compared in terms of their correlations with radiological response and overall survival (OS) prediction for first TACE. The same analyses were conducted in 912 patients receiving re-TACE to evaluate the ART (assessment for re-treatment with TACE) and ABCR (alpha-fetoprotein, Barcelona Clinic Liver Cancer, Child-Pugh and Response) systems for post re-TACE survival (PRTS). RESULTS: All the prognostic systems were correlated with radiological response achieved by first TACE, and the six-and-twelve criteria exhibited the highest correlation (Spearman R = 0.39, P = 0.026) and consistency (Kappa = 0.14, P = 0.019), with optimal performance by area under the receiver operating characteristic curve of 0.71 [95% confidence interval (CI): 0.68-0.74]. With regard to the prediction of OS, the mHAP3 system identified patients with a favorable outcome with the highest concordance (C)-index of 0.60 (95%CI: 0.57-0.62) and the best area under the receiver operating characteristic curve at any time point during follow-up; whereas, PRTS was well-predicted by the ABCR system with a C-index of 0.61 (95%CI: 0.59-0.63), rather than ART. Finally, combining the mHAP3 and ABCR systems identified candidates suitable for TACE with an improved median PRTS of 36.6 mo, compared with non-candidates with a median PRTS of 20.0 mo (log-rank test P < 0.001). CONCLUSION: Radiological response to TACE is closely associated with tumor burden, but superior prognostic prediction could be achieved with the combination of mHAP3 and ABCR in patients with unresectable liver-confined HCC.


Subject(s)
Carcinoma, Hepatocellular/mortality , Chemoembolization, Therapeutic/mortality , Clinical Decision Rules , Liver Neoplasms/mortality , Severity of Illness Index , Aged , Area Under Curve , Carcinoma, Hepatocellular/therapy , Chemoembolization, Therapeutic/methods , Female , Humans , Liver Neoplasms/therapy , Male , Middle Aged , Prognosis , ROC Curve , Retrospective Studies , Risk Assessment , Risk Factors , Treatment Outcome , Tumor Burden , alpha-Fetoproteins/analysis
11.
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
12.
J Gene Med ; 22(3): e3134, 2020 03.
Article in English | MEDLINE | ID: mdl-31656043

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) has been recognized as a member of the most common human malignant tumors globally. According to multiple studies, long noncoding RNAs (lncRNAs) have been defined as vital regulators in tumor progression. Although previous studies have indicated that lncRNA long intergenic non-protein coding RNA 467 (LINC00467) exerts oncogenic effect in tumorigenesis and the development of cancers, the specific function that LINC00467 induces in HCC remains obscure. METHODS: LINC00467 expression was examined by a quantitative reverse transcriptase-polymerase chain reaction. CCK-8, EdU, transwell, western blotting and caspase-3 activity analyses were utilized to testify the role of LINC00467 in HCC. The interaction between IGF2BP3 and LINC00467 (or TRAF5) was investigated by luciferase reporter, RIP and RNA pull-down assays. RESULTS: LINC00467 upregulation in HCC tissues and cells was observed. LINC00467 silencing suppressed cell proliferation and metastasis, whereas it facilitated cell apoptosis in HCC. The gene for tumor necrosis factor receptor-associated factor 5 (TRAF5) was a neighboring gene of that for LINC00467 and its expression was positively modulated by LINC00467 in HCC. TRAF5 knockdown inhibited HCC progression. LINC00467 deficiency could decrease the mRNA stability of TRAF5 in HCC. Insulin-like growth factor-2 messenger RNA-binding protein 3 (IGF2BP3) could bind with LINC00467 and its depletion could lower TRAF5 mRNA stability in HCC. Final rescue assays further indicated that downregulation of IGF2BP3 or TRAF5 acted against LINC00467 upregulation-mediated function on HCC progression. CONCLUSIONS: LINC00467 promotes cell proliferation and metastasis by binding with IGF2BP3 to enhance the mRNA stability of TRAF5 in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , RNA, Long Noncoding/genetics , RNA-Binding Proteins/metabolism , TNF Receptor-Associated Factor 5/metabolism , Apoptosis/genetics , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Proliferation , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Liver Neoplasms/pathology , Neoplasm Metastasis , RNA Stability , TNF Receptor-Associated Factor 5/genetics , Up-Regulation
13.
Biochem Biophys Res Commun ; 518(1): 19-25, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31405563

ABSTRACT

INMAP was first identified as a spindle protein that plays important roles in cell-cycle progression, and previous studies have revealed that its abnormal expression leads to mitotic disorder and the growth inhibition of human tumor xenografts, but the underlying mechanism is still unclear. In this study, we knocked out INMAP in HEK293T cells, a strain of human embryonic renal cells, through CRISPR-Cas9 gene editing technology, resulting in obvious cell growth inhibition. In this system, the deletion of INMAP caused obviously apoptosis. And we also found that knockout of INMAP caused micronuclei formation, chromosome aberration, and γH2AX expression upregulation, suggesting DNA damage induction and genomic stability impairment. As a principal component of spindle, the expression of ß-tubulin, detected through Western blot, is obviously upregulated in HEK293T-INMAP-/-. Meanwhile, the level of Cyclin B is also upregulated, whereas, that of Cyclin E, downregulated, with the postponement of mitotic exit and the assembly anomaly of spindle. These results suggest that the deletion of INMAP block the formation of spindle, leading to arrest of cell cycle and DNA damage, finally blocking cell proliferation and inducing apoptosis. Therefore, INMAP is an indispensable factor for genomic integrity and normal mitotic exit.


Subject(s)
Apoptosis , Cell Cycle Proteins/metabolism , Gene Deletion , Mitosis , Nuclear Proteins/metabolism , Spindle Apparatus/metabolism , Cell Cycle Checkpoints , Cell Proliferation , DNA Damage , HEK293 Cells , Humans , Signal Transduction , Tubulin/metabolism
14.
J Hepatol ; 70(5): 893-903, 2019 05.
Article in English | MEDLINE | ID: mdl-30660709

ABSTRACT

BACKGROUND & AIMS: Previous prognostic scores for transarterial chemoembolization (TACE) were mainly derived from real-world settings, which are beyond guideline recommendations. A robust model for outcome prediction and risk stratification of recommended TACE candidates is lacking. We aimed to develop an easy-to-use tool specifically for these patients. METHODS: Between January 2010 and May 2016, 1,604 treatment-naïve patients with unresectable hepatocellular carcinoma (HCC), Child-Pugh A5-B7 and performance status 0 undergoing TACE were included from 24 tertiary centres. Patients were randomly divided into training (n = 807) and validation (n = 797) cohorts. A prognostic model was developed and subsequently validated. Predictive performance and discrimination were further evaluated and compared with other prognostic models. RESULTS: The final presentation of the model was "linear predictor = largest tumour diameter (cm) + tumour number", which consistently outperformed other currently available models in both training and validation datasets as well as in different subgroups. The thirtieth percentile and the third quartile of the linear predictor, namely 6 and 12, were further selected as cut-off values, leading to the "six-and-twelve" score which could divide patients into 3 strata with the sum of tumour size and number ≤6, >6 but ≤12, and >12 presenting significantly different median survival of 49.1 (95% CI 43.7-59.4) months, 32.0 (95% CI 29.9-37.5) months, and 15.8 (95% CI 14.1-17.7) months, respectively. CONCLUSIONS: The six-and-twelve score may prove an easy-to-use tool to stratify recommended TACE candidates (Barcelona Clinic Liver Cancer stage-A/B) and predict individual survival with favourable performance and discrimination. Moreover, the score could stratify these patients in clinical practice as well as help design clinical trials with comparable criteria involving these patients. Further external validation of the score is required. LAY SUMMARY: There is currently no prognostic model specifically developed for recommended or ideal transarterial chemoembolization (TACE) candidates with hepatocellular carcinoma, despite these patients being frequently identified as the best target population in pivotal randomized controlled trials. The six-and-twelve score provides patient survival prediction, especially in ideal candidates of TACE, outperforming other currently available models in both training and validation sets, as well as different subgroups. With cut-off values of 6 and 12, the score can stratify ideal TACE candidates into 3 strata with significantly different outcomes and may shed light on risk stratification of these patients in clinical practice as well as in clinical trials.


Subject(s)
Carcinoma, Hepatocellular/mortality , Chemoembolization, Therapeutic , Liver Neoplasms/mortality , Aged , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Female , Humans , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Middle Aged , Prognosis , Tumor Burden
15.
Exp Cell Res ; 373(1-2): 119-131, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30336116

ABSTRACT

When a cell migrates, the centrosome positions between the nucleus and the leading edge of migration via the microtubule system. The protein CrpF46 (centrosome-related protein F46) has a known role during mitosis and centrosome duplication. However, how CrpF46 efficiently regulates centrosome-related cell migration is unclear. Here, we report that knockdown of CrpF46 resulted in the disruption of microtubule arrangement, with impaired centrosomal reorientation, and slowed down cell migration. In cells that express low levels of CrpF46, stress fibers were weakened, which could be rescued by recovering Flag-CrpF46. We also found that CrpF46 interacted with non-muscle myosin high chain IIA (NMHC IIA) and that its three coiled-coil domains are pivotal for its binding to NMHC IIA. Additionally, analyses of phosphorylation of NMHC IIA and RLC (regulatory light chain) demonstrated that CrpF46 was associated with myosin IIA during filament formation. Indirect immunofluorescence images indicated that NM IIA filaments were inhibited when CrpF46 was under-expressed. Thus, CrpF46 regulates cell migration by centrosomal reorientation and altering the function of the actomyosin network by controlling specific phosphorylation of myosin.


Subject(s)
Actomyosin/metabolism , Autoantigens/physiology , Cell Movement , Molecular Motor Proteins/metabolism , Myosin Heavy Chains/metabolism , Actin Cytoskeleton/ultrastructure , Autoantigens/genetics , Autoantigens/metabolism , Cell Line, Tumor , Cell Polarity , Centrosome , HeLa Cells , Humans , Microtubules/ultrastructure
16.
PLoS One ; 13(2): e0191984, 2018.
Article in English | MEDLINE | ID: mdl-29394294

ABSTRACT

Through cell-based screening models, we have identified a new compound IMB5043, a thiophenylated pyridazinone, which exerted cytotoxicity against cancer cells. In the present study, we evaluated its antitumor efficacy and the possible mechanism. By MTT assay, IMB5043 inhibited the proliferation of various human cancer cells lines, especially hepatocarcinoma SMMC-7721 cells. IMB5043 blocked cell cycle with G2/M arrest, induced cell apoptosis, and inhibited the migration and invasion of SMMC-7721 cells. As verified by comet assay and γ-H2AX foci formation, IMB5043 caused DNA damage and activated ATM, Chk2 and p53 through phosphorylation. As shown by Gene microarray analysis, the differentially expressed genes in SMMC-7721 cells treated with IMB5043 were highly related to cell death and apoptosis. IMB5043 suppressed the growth of hepatocarcinoma SMMC-7721 xenograft in athymic mice. By histopathological examination, no lesions were found in bone marrow and various organs of the treated mice. Our findings reveal that IMB5043 as an active compound consisting of both pyridazinone and thiophene moieties exerts antitumor efficacy through activation of ATM-Chk2 pathway. IMB5043 may serve as a promising leading compound for the development of antitumor drugs.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Checkpoint Kinase 2/metabolism , Pyridazines/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , DNA Damage , Gene Expression Profiling , Humans , Mice , Mice, Nude , Xenograft Model Antitumor Assays
17.
Anticancer Res ; 37(8): 4635-4641, 2017 08.
Article in English | MEDLINE | ID: mdl-28739764

ABSTRACT

AIM: To prospectively estimate the safety, feasibility and accuracy of 1.0T open-magnetic resonance (MR)-guided percutaneous biopsy in free-hand of focal lesions located in the hepatic dome. MATERIALS AND METHODS: All percutaneous MR-guided biopsies of the lesions were performed at the Shandong Medical Imaging Research Institute. Thirty-six patients with focal liver lesions located in the hepatic dome measuring 6-36 mm in the maximum diameter were included in this study. Lesions were divided into two groups on the basis of maximum nodule diameters: ≤1.5 cm (n=14) and >1.5 cm (n=22). Surgical pathology of nodules or clinical and imaging follow-up for at least 12 months were used to establish final diagnosis. Diagnostic accuracy, specificity, sensitivity, average procedure time and complications were recorded and analyzed. RESULTS: All obtained samples were sufficient for diagnosis. Accuracy, sensitivity and specificity of MRI-guided percutaneous liver lesions biopsy in diagnosing malignant tumors were 97.2%, 96.7% and 100%, respectively. Accuracy, sensitivity and specificity were 92.9%, 90.9% and 100% for the lesions that were 1.5 cm or smaller in diameter and 100%, 100% and 100% for lesions larger than 1.5 cm in diameter, respectively. There was no significant difference between the two groups (p>0.05). The average procedure time for each pass of the needle from skin entry to the target lesion was 1.1 min and the total procedural time was 28.5 min. Biopsy-induced complications included peri-hepatic hemorrhage in 8.3% (3/36) of cases. No serious complications occurred. CONCLUSION: 1.0T open-MR-guided free-hand percutaneous biopsy is a safe, feasible and highly accurate diagnostic method for pathological diagnosis of focal liver lesions located in the hepatic dome.


Subject(s)
Fluoroscopy , Image-Guided Biopsy , Liver Diseases/diagnostic imaging , Liver Diseases/pathology , Liver/pathology , Magnetic Resonance Imaging , Adult , Aged , Female , Fluoroscopy/methods , Humans , Image-Guided Biopsy/instrumentation , Image-Guided Biopsy/methods , Magnetic Resonance Imaging/methods , Male , Middle Aged , Reproducibility of Results , Tomography, X-Ray Computed
18.
Anticancer Res ; 37(8): 4649-4653, 2017 08.
Article in English | MEDLINE | ID: mdl-28739766

ABSTRACT

Aim: To study the feasibility and curative effect of 125 I seeds articles combined with biliary stent implantation in the treatment of malignant obstructive jaundice. Patients and Methods: Fifty patients with malignant obstructive jaundice were included. Twenty-four were treated by biliary stent implantation combined with intraluminal brachytherapy by 125I seeds articles as the experimental group, while the remaining 26 were treated by biliary stent implantation only as the control group. The goal of this study was to evaluate total bilirubin, direct bilirubin and tumor markers (cancer antigen (CA)-199, CA-242 and carcinoembryonic antigen (CEA)), as well as biliary stent patency status and survival time before and after surgery. Results: Jaundice improved greatly in both groups. The decreases of CA-199 and CA-242 had statistical significance (p=0.003 and p=0.004) in the experimental group. The ratio of biliary stent patency was 83.3% (20/24) in the experimental group and 57.7% (15/26) in the control group (p=0.048). The biliary stent patency time in the experimental group was 1~15.5 (mean=9.84) months. The biliary stent patency time in the control group was 0.8~9 (mean=5.57) months, which was statistically significant (p=0.018). The median survival time was 10.2 months in the experimental group, while 5.4 months in control group (p<0.05). Conclusion: 125 I seeds articles combined with biliary stent implantation significantly prolongs biliary stent patency time and survival time for patients with malignant obstructive jaundice possibly by inhibiting the proliferation of vascular endothelial cells and the growth of tumor.


Subject(s)
Brachytherapy , Iodine Radioisotopes , Jaundice, Obstructive/etiology , Jaundice, Obstructive/therapy , Stents , Adult , Aged , Aged, 80 and over , Biliary Tract Surgical Procedures , Biomarkers, Tumor , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Jaundice, Obstructive/mortality , Kaplan-Meier Estimate , Male , Middle Aged , Postoperative Care , Treatment Outcome
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...