Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
bioRxiv ; 2024 May 18.
Article in English | MEDLINE | ID: mdl-38405779

ABSTRACT

Extrachromosomal DNA (ecDNA) is a central mechanism for focal oncogene amplification in cancer, occurring in approximately 15% of early stage cancers and 30% of late-stage cancers. EcDNAs drive tumor formation, evolution, and drug resistance by dynamically modulating oncogene copy-number and rewiring gene-regulatory networks. Elucidating the genomic architecture of ecDNA amplifications is critical for understanding tumor pathology and developing more effective therapies. Paired-end short-read (Illumina) sequencing and mapping have been utilized to represent ecDNA amplifications using a breakpoint graph, where the inferred architecture of ecDNA is encoded as a cycle in the graph. Traversals of breakpoint graph have been used to successfully predict ecDNA presence in cancer samples. However, short-read technologies are intrinsically limited in the identification of breakpoints, phasing together of complex rearrangements and internal duplications, and deconvolution of cell-to-cell heterogeneity of ecDNA structures. Long-read technologies, such as from Oxford Nanopore Technologies, have the potential to improve inference as the longer reads are better at mapping structural variants and are more likely to span rearranged or duplicated regions. Here, we propose CoRAL (Complete Reconstruction of Amplifications with Long reads), for reconstructing ecDNA architectures using long-read data. CoRAL reconstructs likely cyclic architectures using quadratic programming that simultaneously optimizes parsimony of reconstruction, explained copy number, and consistency of long-read mapping. CoRAL substantially improves reconstructions in extensive simulations and 9 datasets from previously-characterized cell-lines as compared to previous short-read-based tools. As long-read usage becomes wide-spread, we anticipate that CoRAL will be a valuable tool for profiling the landscape and evolution of focal amplifications in tumors.

2.
Open Med (Wars) ; 18(1): 20230849, 2023.
Article in English | MEDLINE | ID: mdl-38045857

ABSTRACT

The gut microbiota and microbial metabolites influence the enteric nervous system and the central nervous system via the microbial-gut-brain axis. Increasing body of evidence suggests that disturbances in the metabolism of peripheral branched-chain amino acids (BCAAs) can contribute to the development of neurodegenerative diseases through neuroinflammatory signaling. Preliminary research has shown that longitudinal changes in serum amino acid levels in mouse models of Parkinson's disease (PD) are negatively correlated with disease progression. Therefore, the aim of the present study was to determine the changes in serum levels of short-chain fatty acids (SCFAs) in a mouse model of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD after dietary BCAA supplementation. In our research, gas chromatography-mass spectrometry was used to detect serum SCFA concentrations. The data were then analyzed with principal component analysis and orthogonal partial least squares discriminant analysis. Finally, the correlations of serum SCFA levels with gut and motor function in MPTP-induced PD mice were explored. Propionic acid, acetic acid, butyric acid, and isobutyric acid concentrations were elevated in MPTP + H-BCAA mice compared with MPTP mice. Propionic acid concentration was increased the most, while the isovaleric acid concentration was decreased. Propionic acid concentration was positively correlated with fecal weight and water content and negatively correlated with the pole-climbing duration. In conclusion, these results not only suggest that propionic acid may be a potential biomarker for PD, but also indicate the possibility that PD may be treated by altering circulating levels of SCFA.

3.
Nutr Metab Cardiovasc Dis ; 33(5): 934-946, 2023 05.
Article in English | MEDLINE | ID: mdl-36958976

ABSTRACT

BACKGROUND AND AIMS: Prospective cohorts are inconsistent regarding the association between dietary calcium intake and the risk of stroke. The aim was to perform a meta-analysis to determine whether an association exists between them in cohort studies. METHODS AND RESULTS: Relevant studies were identified by searching PubMed, EMBASE and Web of Science databases that published before December 2022. Prospective cohort studies that provided relative risk (RR) estimates with 95% confidence intervals (CIs) for the association were included. Study-specific risk estimates were combined by using a random effects model. Eighteen prospective studies, including 19,557 stroke cases among 882,181 participants, were pooled in the meta-analysis. We observed a nonlinear association between calcium intake and risk of stroke (Pnonlinearity < 0.003). Compared with the lowest value of zero assumed as the reference, the RRs (95% CI) of stroke across levels of calcium intake were 0.95 (0.92, 0.98) for 200 mg/day, 0.94 (0.90, 0.98) for 300 mg/day, 0.95 (0.90, 0.99) for 500 mg/day, 0.98 (0.93, 1.03) for 700 mg/day, and 1.04 (0.97, 1.11) for 1000 mg/day. The stratified analyses by geographic region showed nonlinear associations and indicated that the protective effect was observed in Asian countries (Pnonlinearity = 0.001) but not in non-Asian regions (Pnonlinearity = 0.047). CONCLUSION: This meta-analysis suggests that dietary calcium intake might play an effective role in the prevention of stroke, especially in Asian countries. Future research among Asia population should attempt to establish whether this association is causal. SYSTEMATIC REVIEW REGISTRATION: PROSPERO CRD42022357710.


Subject(s)
Calcium, Dietary , Stroke , Humans , Prospective Studies , Risk Factors , Calcium, Dietary/adverse effects , Calcium , Cohort Studies , Stroke/diagnosis , Stroke/epidemiology , Stroke/prevention & control
4.
Front Endocrinol (Lausanne) ; 13: 1072419, 2022.
Article in English | MEDLINE | ID: mdl-36686472

ABSTRACT

Background: Children and adolescents increasingly commonly suffer from obesity and headache. It has been confirmed that there is an association between obesity and headache in adults; however, evidence of such an association in paediatric populations is still controversial. Therefore, this study examined the relationship between obesity and headache among children and adolescents in the US. Methods: The cross-sectional data of 3948 participants were obtained from the National Health and Nutrition Examination Survey 1999-2004. Weighted logistic regression models were applied to investigate the association between obesity and headache. Subgroup analysis stratified by sex and age was performed to explore the potential difference in the association of paediatric obesity with headache. The performance of paediatric obesity on headache was assessed by receiver operating characteristic (ROC) curve. Results: The present study involved 3948 participants, of whom 713 (18.1%) had headache. Compared to those without headache, participants with headache tended to be girls and adolescents, have less calcium intake, and have higher levels of body mass index (BMI), C-reactive protein (CRP), serum ferritin and triglycerides (TGs) (all P < 0.05). After fully adjusting for potential confounders, the ORs with 95% CIs for headache were 1.03 (0.58-1.54) and 1.25 (0.68-2.30) for overweight and obese participants in comparison with normal-weight controls, respectively, implying no association of paediatric obesity with headache independent of other potential confounding factors. In addition, although higher odds of headache were noted in girls and adolescents (aged 10-17 years), no statistically significant difference was found across any subgroups. The area under the ROC (AUC) of paediatric obesity on headache was 0.634. Conclusions: In summary, our study indicated that obesity is not associated with headache among US children and adolescents. Further prospective studies with larger sample size are needed to validate our findings.


Subject(s)
Pediatric Obesity , Adult , Female , Humans , Child , Adolescent , Pediatric Obesity/complications , Pediatric Obesity/epidemiology , Nutrition Surveys , Cross-Sectional Studies , Prospective Studies , Headache/epidemiology , Headache/etiology
5.
J Oncol ; 2021: 2182761, 2021.
Article in English | MEDLINE | ID: mdl-34804157

ABSTRACT

BACKGROUND: Osteosarcoma is the most prevalent bone cancer that affects young adults and adolescents. It is the most frequent malignancy of the bone. In spite of the fact that complete surgical resection and chemotherapy have increased the overall survival of osteosarcoma patients considerably, the prognosis remains dismal in patients with recurring and/or metastasized osteosarcoma. Thus, finding predictive biomarkers representing osteosarcoma's biological variability may result in more effective treatment for osteosarcoma patients. METHODS: In this research, RNA data and clinical information were obtained from TARGET database. The risk score was calculated using a technique that incorporated both univariate and multivariate Cox regression. A variety of statistical methods were employed to assess the risk score's accuracy. These included ROC curves, nomograms, and Kaplan-Meier curves. Following that, bioinformatics studies were carried out in order to investigate the possible biological processes that influence the prognosis of osteosarcoma patients. GSEA was used to investigate the variations in pathway enrichment among the different groups of genes. To examine the disparities in the immune microenvironment, the analytical methods CIBERSORT and ssGSEA were employed. RESULTS: We discovered three differentially expressed lncRNAs (RPARP-AS1, AC009159.3, and AC124312.3) that are linked to osteosarcoma prognosis. Kaplan-Meier analysis showed the presence of a signature of high-risk lncRNAs linked with a poor prognosis for osteosarcoma. Furthermore, the AUC of the lncRNAs signature was 0.773, indicating that they are useful in predicting osteosarcoma prognosis in certain cases. In predicting osteosarcoma prognosis, our risk assessment approach outperformed conventional clinicopathological characteristics. In the high-risk group of people, GSEA showed the presence of tumor-related pathways as well as immune-related pathways. Furthermore, TARGET revealed that immune-related functions such as checkpoint, T-cell coinhibition, and costimulation were significantly different between the high-risk and low-risk groups. LAIR1, LAG3, CD44, and CD22, as well as other immune checkpoints, were shown to be expressed differentially across the two risk groups. CONCLUSION: This study established that pyroptosis-derived lncRNAs had a significant predictive value for osteosarcoma patients' survival, indicating that they may be a viable target for future therapy.

6.
Arch Virol ; 166(8): 2217-2224, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34091783

ABSTRACT

Swine influenza is an economically important respiratory disease in swine, but it also constantly poses a threat to human health. Therefore, developing rapid, sensitive, and efficient detection methods for swine influenza virus (SIV) is important. By aligning the haemagglutinin (HA) gene sequences of SIVs circulating in China over a 10-year period, an H1 primer-probe set targeting both Eurasian avian-like H1N1 (EA H1N1) and pandemic 2009 H1N1 ((H1N1)pdm09) lineages plus a H3 primer-probe set targeting the prevalent human-like H3N2 (HL H3N2) subtype were designed. Subsequently, a TaqMan-MGB-based duplex one-step real-time RT-PCR (RT-qPCR) assay was established and evaluated. The duplex RT-qPCR has a detection limit of 5 copies/µL of HA plasmid for EA H1N1, (H1N1)pdm09, and HL H3N2 subtype SIVs, and its overall detection sensitivity of 100% and specificity of 91.67% matches that of traditional virus isolation through chicken embryo inoculation using experimentally infected mouse lung samples. The method showed high repeatability both within run and between runs, and there was no cross-reactivity against several other porcine viruses that are commonly circulating in China. Furthermore, the duplex RT-qPCR method revealed a higher prevalence of subtype H1 than subtype H3 in 166 nasal swabs from pigs collected from one slaughterhouse between October and December 2019. This assay could be very helpful in the rapid differential detection and routine surveillance of EA H1N1, (H1N1)pdm09, and HL H3N2 SIVs in China.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/isolation & purification , Orthomyxoviridae Infections/diagnosis , Animals , China , Disease Models, Animal , Early Diagnosis , Female , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/classification , Influenza A Virus, H3N2 Subtype/genetics , Mice , Multiplex Polymerase Chain Reaction , Nose/virology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Swine
7.
BMC Vet Res ; 17(1): 80, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33588843

ABSTRACT

BACKGROUND: Genotype S H9N2 viruses have become predominant in poultry in China since 2010. These viruses frequently donate their whole internal gene segments to other emerging influenza A subtypes such as the novel H7N9, H5N6, and H10N8 viruses. We recently reported that the PB2 and M genes of the genotype S H9N2 virus, which are derived from the G1-like virus, enhance the fitness of H5Nx and H7N9 avian influenza viruses in chickens and mice. However, whether the G1-like PB2 and M genes are preferentially incorporated into progeny virions during virus reassortment remains unclear; whether the G1-like PB2 and M genes from different subtypes are differentially incorporated into new virion progeny remains unknown. RESULTS: We conducted a reassortment experiment with the use of a H7N9 virus as the backbone and found that G1-like M/PB2 genes were preferentially incorporated in progeny virions over F/98-like M/PB2 genes. Importantly, the preference varied among G1-like M/PB2 genes of different subtypes. When competing with F/98-like M/PB2 genes during reassortment, both the M and PB2 genes from the H7N9 virus GD15 showed an advantage, whereas only the PB2 gene from the H9N2 virus CZ73 and the M gene from the H9N2 virus AH320 displayed the advantage. CONCLUSION: Our findings highlight the preferential and variable advantages of H9N2-derived G1-like M and PB2 genes in incorporating them into H7N9 progeny virions over SH14-derived F/98-like M/PB2 genes.


Subject(s)
Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H9N2 Subtype/genetics , Reassortant Viruses/genetics , Animals , Chick Embryo , Coinfection , Dogs , Genotype , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Reassortant Viruses/growth & development , Viral Matrix Proteins/genetics , Virion
8.
J Res Med Sci ; 25: 49, 2020.
Article in English | MEDLINE | ID: mdl-32765619

ABSTRACT

BACKGROUND: The reasonable use of amino acids (AAs) in parenteral nutrition (PN) is very critical to the growth and development of premature infants. However, the appropriate dose of AAs has not been determined. Our study was designed to investigate the clinical effect of two different doses of AAs in PN for low birth weight premature infants. MATERIALS AND METHODS: This randomized controlled study included 191 preterm infants who admitted to the neonatal intensive care unit of the First Affiliated Hospital of Nanjing Medical University from June 2015 to December 2016 and they were randomly divided into Group 1 (n = 81) and Group 2 (n = 110). In Group 1, the starting dose of AAs dose was 1.0-1.5 g/kg/day, which was increased by 0.5 g/kg with the maximum dose at 3.5 g/kg/day. In Group 2, the starting dose of AAs was 1.8-2.5 g/kg/day and was increased by 1.0 g/kg with the maximum dose at 4.0-4.5 g/kg/day. We analyzed the clinical characteristics, body weight, body length, total calorie intake, nonprotein calorie intake, total protein intake, liver and kidney function, and complications of the two groups of preterm infants. RESULTS: The start of enteral feeding and the recovery of birth weight in Group 2 were earlier than those in Group 1 (3.83 ± 3.15 day vs. 5.53 ± 5.63 day, P = 0.016 and 6.36 ± 4.88 day vs. 8.48 ± 9.27 day, P = 0.043, respectively). The duration of PN and the time before total enteral nutrition were shorter in Group 2 than in Group 1 (16.46 ± 10.33 day vs. 21.41 ± 18.00 day, P = 0.029 and 15.47 ± 10.54 day vs. 19.47 ± 14.57 day, P = 0.038; respectively). The duration of mechanical ventilation (1.12 ± 2.62 day vs. 3.31 ± 8.13 day, P = 0.028) in Group 2 was shorter than that in Group 1. CONCLUSION: High doses of AAs in the early PN for preterm infants facilitate the promotion of early growth and development, advance recovery of birth weight, reduce the duration of PN, and reduce respiratory support without increasing the incidence of complications.

9.
Cancer Invest ; 29(7): 485-93, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21740086

ABSTRACT

Tumor necrosis factor (TNF)-α has been proved as an adjuvant therapy for tumor by FDA. However, the effect of chronic TNF-α expression for tumor is still controversial. In this study, we investigated the effect of low-dose TNF-α on tumor growth. We confirmed that low-dose TNF-α promoted angiogenesis of tumor in vivo, vascular endothelial growth factor (VEGF) and hypoxia-inducible factor (HIF)-1α, the transcription factor of VEGF, were both upregulated. Our results suggested that low-dose TNF-α was a powerful activator of angiogenesis in tumor and HIF-1α-VEGF pathway seemed to be the most important molecular mechanism.


Subject(s)
Melanoma, Experimental/pathology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Allantois/blood supply , Animals , Antigens, CD34/biosynthesis , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chick Embryo , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Melanoma, Experimental/blood supply , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/etiology , Neovascularization, Physiologic
10.
J Biol Chem ; 286(28): 25007-15, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21592963

ABSTRACT

Mesenchymal stem cells (MSCs), which are modulated by cytokines present in the tumor microenvironment, play an important role in tumor progression. It is well documented that inflammation is an important part of the tumor microenvironment, so we investigated whether stimulation of MSCs by inflammatory cytokines would contribute to their ability to promote tumor growth. We first showed that MSCs could increase C26 colon cancer growth in mice. This growth-promoting effect was further accelerated when the MSCs were pre-stimulated by inflammatory factors IFN-γ and TNF-α. At the same time, we demonstrated that MSCs pre-stimulated by both inflammatory factors could promote tumor angiogenesis in vivo to a greater degree than untreated MSCs or MSCs pre-stimulated by either IFN-γ or TNF-α alone. A hen egg test-chorioallantoic membrane (HET-CAM) assay showed that treatment of MSC-conditioned medium can promote chorioallantoic membrane angiogenesis in vitro, especially treatment with conditioned medium of MSCs pretreated with IFN-γ and TNF-α together. This mechanism of promoting angiogenesis appears to take place via an increase in the expression of vascular endothelial growth factor (VEGF), which itself takes place through an increase in signaling in the hypoxia-inducible factor 1α (HIF-1α)-dependent pathway. Inhibition of HIF-1α in MSCs by siRNA was found to effectively reduce the ability of MSC to affect the growth of colon cancer in vivo in the inflammatory microenviroment. These results indicate that MSCs stimulated by inflammatory cytokines such as IFN-γ and TNF-α in the tumor microenvironment express higher levels of VEGF via the HIF-1α signaling pathway and that these MSCs then enhance tumor angiogenesis, finally leading to colon cancer growth in mice.


Subject(s)
Colonic Neoplasms/metabolism , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Mesenchymal Stem Cells/metabolism , Neovascularization, Pathologic/metabolism , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/metabolism , Chorioallantoic Membrane/pathology , Colonic Neoplasms/pathology , Culture Media, Conditioned/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation Mediators/pharmacology , Interferon-gamma/pharmacology , Male , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/metabolism
11.
J Cell Mol Med ; 15(11): 2343-52, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21091630

ABSTRACT

Mesenchymal stem cells (MSCs) are studied for their potential clinical use in regenerative medicine, tissue engineering and tumour therapy. However, the therapeutic application of MSCs in tumour therapy still remains limited unless the immunosuppressive role of MSCs for tumour growth in vivo is better understood. In this study, we investigated the mechanism of MSCs favouring tumour escape from immunologic surveillance in inflammatory microenvironment. We first compared the promotive capacity of bone marrow-derived MSCs on B16 melanoma cells growth in vivo, pre-incubated or not with the inflammatory cytokines interferon (IFN)-γ and tumour necrosis factor (TNF)-α. We showed that the development of B16 melanoma cells is faster when co-injected with MSCs pre-incubated with IFN-γ and TNF-α compared with control groups. Moreover, tumour incidence increases obviously in allogeneic recipients when B16 melanoma cells were co-injected with MSCs pre-incubated with IFN-γ and TNF-α. We then demonstrated that the immunosuppressive function of MSCs was elicited by IFN-γ and TNF-α. These cytokine combinations provoke the expression of inducible nitric oxide synthase (iNOS) by MSCs. The impulsive effect of MSCs treated with inflammatory cytokines on B16 melanoma cells in vivo can be reversed by inhibitor or short interfering RNA of iNOS. Our results suggest that the MSCs in tumour inflammatory microenvironment may be elicited of immunosuppressive function, which will help tumour to escape from the immunity surveillance.


Subject(s)
Immune Tolerance , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mesenchymal Stem Cells , Nitric Oxide Synthase Type II/metabolism , Tumor Escape , Animals , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cell Proliferation , Immunosuppression Therapy , Interferon-gamma/immunology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , RNA Interference , RNA, Small Interfering , Tumor Necrosis Factor-alpha/immunology
12.
Cancer Lett ; 288(1): 68-74, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19660860

ABSTRACT

Autophagy is a membrane process that results in the transporting of cellular contents to lysosomes for degradation. Autophagic cell death is another way of programed cell death called type II PCD, which has complicated connection with apoptosis, both of these two types of cell death play an important role in tumor development. In this study, we investigated chemotherapeutic agent induced cell death pathway in wild type (WT), Bax(-/-) and PUMA(-/-) HCT116 cells. Bax or PUMA deficient cells had similar chemosensitivity to WT cells but were defective in undergoing apoptosis. The results of electron microscopy and GFP-LC3 localization assay showed that autophagy was induced in Bax or PUMA deficient cells but not in WT cells. mTOR activity was decreased in Bax or PUMA deficient cells which further indicated the up-regulation of autophagy. Inhibition of autophagy by 3-Methyladenine (3-MA) decreased the cell death in Bax or PUMA deficient cells. Taken together, these results suggest that autophagic cell death can be used as an alternative cell death pathway in apoptosis defective cells and may bring a new target for cancer therapy.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis Regulatory Proteins/deficiency , Autophagy/drug effects , Colonic Neoplasms/pathology , Fluorouracil/pharmacology , Proto-Oncogene Proteins/deficiency , bcl-2-Associated X Protein/deficiency , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis Regulatory Proteins/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Dose-Response Relationship, Drug , HCT116 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , bcl-2-Associated X Protein/genetics
13.
Exp Mol Pathol ; 88(1): 112-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19879258

ABSTRACT

CpG island methylator phenotype (CIMP), in which multiple genes are concurrently methylated, is an important mechanism in hepatocellular carcinoma development. We determined a hypermethylation profile in hepatocellular carcinoma (HCC). We examined the promoter methylation status of 10 genes in 60 cases of hepatocellular carcinoma (HCC), 60 cases of paired non-tumor tissues, and 6 cases of normal tissues by methylation-specific PCR. The average methylated gene numbers were significantly different between HCC and nontumor tissues (p<0.001). We found metastasis, gamma-glutamyl transpeptidase (GGT) and tumor node metastasis (TNM) stage were significantly different among patients with different CIMP status. Patients with high frequency CIMP tumors had significantly worse survival than patients with intermediate frequency or no CIMP tumors (p<0.01 and p<0.05, respectively). Our results suggested that CIMP could serve as a molecular marker of late stage and poorly prognostic HCC development.


Subject(s)
Carcinoma, Hepatocellular/genetics , CpG Islands/genetics , Liver Neoplasms/genetics , Promoter Regions, Genetic/genetics , Adult , Aged , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/secondary , DNA Methylation/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Phenotype , Prognosis , Survival Rate , gamma-Glutamyltransferase/metabolism
14.
Oncol Rep ; 22(3): 549-56, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19639202

ABSTRACT

The telomerase is specifically activated in most malignant tumors but is usually inactive in normal somatic cells. It has been reported that telomerase has an anti-apoptotic role and up-regulation of telomerase helps cancer cells to be resistant to chemotherapeutic agent-induced cell death. The effect of cisplatin on telomerase activity is complex, and the exact mechanism remains largely unknown. In this study, we found that cisplatin activated telomerase activity and human telomerase reverse transcriptase (hTERT) expression in SMMC7721 human hepatocellular carcinoma cell line. Low-dose cisplatin up-regulated hTERT and NF-kappaB p65 expression and increased telomerase and NF-kappaB activity. Inhibition of NF-kappaB attenuated the hTERT expression and telomerase activity exposed to cisplatin, suggesting that NF-kappaB is responsible for the cisplatin-induced activation of the hTERT. Furthermore, preincubation of low-dose cisplatin which induced high expression of hTERT help hepatocellular carcinoma SMMC7721 cells survive under the high concentration of anti-cancer drugs. Inhibition of hTERT increased sensitivity of SMMC7721 cells to chemotherapy. Taken together, these results suggested that up-regulation of hTERT expression by low-dose cisplatin is NF-kappaB-dependent and contributes to chemotherapy resistance in human hepatocellular cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cisplatin/pharmacology , Liver Neoplasms/drug therapy , Telomerase/genetics , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , DNA Damage , Drug Resistance, Neoplasm , Gene Expression Regulation, Enzymologic , Humans , Liver Neoplasms/genetics , NF-kappa B/physiology , Telomerase/analysis , Up-Regulation
15.
BMC Cancer ; 8: 375, 2008 Dec 18.
Article in English | MEDLINE | ID: mdl-19091131

ABSTRACT

BACKGROUND: Chemoresistance is one of the main obstacles to successful cancer therapy and is frequently associated with Multidrug resistance (MDR). Many different mechanisms have been suggested to explain the development of an MDR phenotype in cancer cells. One of the most studied mechanisms is the overexpression of P-glycoprotein (P-gp), which is a product of the MDR1 gene. Tumor cells often acquire the drug-resistance phenotype due to upregulation of the MDR1 gene. Overexpression of MDR1 gene has often been reported in primary gastric adenocarcinoma. METHODS: This study investigated the role of p38-MAPK signal pathway in vincristine-resistant SGC7901/VCR cells. P-gp and MDR1 RNA were detected by Western blot analysis and RT-PCR amplification. Mitgen-activated protein kinases and function of P-gp were demonstrated by Western blot and FACS Aria cytometer analysis. Ap-1 activity and cell apoptosis were detected by Dual-Luciferase Reporter Assay and annexin V-PI dual staining. RESULTS: The vincristine-resistant SGC7901/VCR cells with increased expression of the multidrug-resistance 1 (MDR1) gene were resistant to P-gp-related drug and P-gp-unrelated drugs. Constitutive increases of phosphorylated p38-MAPK and AP-1 activities were also found in the drug-resistant cells. Inhibition of p38-MAPK by SB202190 reduced activator protein-1 (AP-1) activity and MDR1 expression levels and increased the sensitivity of SGC7901/VCR cells to chemotherapy. CONCLUSION: Activation of the p38-MAPK pathway might be responsible for the modulation of P-glycoprotein-mediated and P-glycoprotein-unmediated multidrug resistance in the SGC7901/VCR cell line.


Subject(s)
Drug Resistance, Neoplasm/physiology , Stomach Neoplasms/drug therapy , Vincristine/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Blotting, Western , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Flow Cytometry , Humans , Imidazoles/pharmacology , Inhibitory Concentration 50 , MAP Kinase Signaling System/physiology , Phosphorylation , Pyridines/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcription Factor AP-1/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
16.
Cancer Biol Ther ; 7(10): 1555-60, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18708758

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide. However, there is no effective treatment for HCC. It has been shown that sustained activation of telomerase is essential for the growth and progression of HCC, suggesting that telomerase is a rational target for HCC therapy. Here, we investigated the effects of siRNA-mediated knockdown of hTERT, the catalytic and rate-limiting subunit of telomerase, on the sensitivity of HCC cells to cisplatin. While silencing of hTERT and the resultant inhibition of telomerase activity by infection with the recombinant adenovirus expressing a hTERT siRNA (Ad-si/hTERT) alone did not affect the proliferation and viability of SMMC7721 and HepG2 HCC cells within five days, co-administration of Ad-si/hTERT, but not the empty adenovirus vector, with cisplatin caused much greater extent of apoptosis in vitro under the same conditions and induced significantly more robust inhibition of SMMC7721 and HepG2 tumors growth in a mouse tumor xenograft model than cisplatin monotherapy. Our results demonstrated the synergistic effect between hTERT siRNA and cisplatin in the suppression of HCC progression and indicated that the combination of hTERT-specific siRNA and cisplatin could be an effective therapy for HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , RNA, Small Interfering/metabolism , Telomerase/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cisplatin/pharmacology , Disease Progression , Gene Silencing , Humans , Inhibitory Concentration 50 , Mice , Neoplasm Transplantation
17.
Cancer Biol Ther ; 7(3): 392-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18075305

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide with an annual occurrence of one million new cases. At present there is no effective treatment for HCC individuals that not amenable to curative therapies. Recent studies show the PI3K/Akt/mTOR signal pathway is involved in multiple cellular functions including proliferation, differentiation, tumorigenesis, and apoptosis. Rapamycin (a specific Mtor inhibitor) could lead to G(1) arrest of many malignant cell lines, and currently analogs of rapamycin are being investigated as a cancer chemotherapeutic adjuvant. This study investigated rapamycin and chemotherapeutic agent 5-fluorouracil (5-Fu) in combination treatment induced apoptosis and cell senescence in hepatocarcinoma cell line SMMC-7721 cells. Treating SMMC-7721 cells with rapamycin plus 5-Fu led to not only apoptosis but also cell senescence, and the senescent cells exhibited significantly less clonogenic potential than 5-Fu individually treated cells. Further study showed rapamycin plus 5-Fu-induced senescence-like growth arrest was accompanied by down-regulation of AP-1 and NF kappa B transcription activity. These results suggest that inhibitors of mTOR may have anticancer potential when used together with some other chemotherapeutic agents, and that down-regulation of AP-1 and NF kappa B transcription activity might take part in a senescence-like growth arrest program induced by rapamycin plus 5-Fu.


Subject(s)
Apoptosis/physiology , Carcinoma, Hepatocellular/pathology , Cellular Senescence/physiology , Fluorouracil/pharmacology , Liver Neoplasms/pathology , Sirolimus/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cellular Senescence/drug effects , Humans , Protein Kinases , TOR Serine-Threonine Kinases
18.
BMC Cancer ; 7: 208, 2007 Nov 12.
Article in English | MEDLINE | ID: mdl-17996122

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is the most invasive and frequently diagnosed malignancy and the second leading cause of cancer death in many regions of Asia. The PI3K/Akt/mTOR signal pathway is involved in multiple cellular functions including proliferation, differentiation, tumorigenesis, and apoptosis. Up-regulation of telomerase activity is thought to be a critical step leading to cell transformation. METHODS: This study investigated changes in mTOR pathway and telomerase activity in hepatocarcinoma cell line SMMC-7721 treated with chemotherapeutic agent 5-fluorouracil (5-Fu). We detected apoptosis of hepatocarcinoma cells by TUNEL assay. Telomerase activity, hTERT transcription level and p- p70 S6k was demonstrated by the telomeric repeat amplification protocol and silver staining assay, Dual-Luciferase Reporter Assay and Western blot analysis respectively. RESULTS: Treating SMMC-7721 cells with 5-Fu leads to apoptosis of the cells, and reduction in telomerase activity, as well as a dramatic reduction in the activated form of p70 S6 kinase, a mTOR substrate. The 5-Fu treatment nearly abolishes transcription of hTERT (the major component of telomerase) mRNA. Treating SMMC-7721 cells with Rapamycin, a specific mTOR inhibitor, significantly reduce hTERT protein level but did not affect hTERT transcription. 5-Fu and rapamycin were synergistic in regards to down-regulation of telomerase activity in hepatocarcinoma cells. CONCLUSION: These results suggest that chemotherapeutic agent 5-Fu may down-regulate telomerase activity at both transcriptional level and PI3K/Akt/mTOR pathway-dependent post-transcriptional level to facilitate hepatocellular carcinoma cell apoptosis.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Protein Kinases/biosynthesis , Telomerase/biosynthesis , Cell Line, Tumor , Down-Regulation , Humans , In Situ Nick-End Labeling , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Telomerase/physiology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...