Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Mar Pollut Bull ; 196: 115685, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37864862

ABSTRACT

Recently, dinoflagellate blooms have frequently occurred in the coastal waters of Fujian, East China Sea. In June 2022, a fish-killing bloom of Kareniaceae species occurred in this region. In this study, four species of Kareniaceae, namely, Karenia longicanalis, K. papilionacea, Karlodinium veneficum, and Karl. digitatum were identified from this bloom event based on the results of single-cell PCR and clone libraries, and intraspecies genetic diversity was found in the Karl. veneficum population. The results of acute toxicity assays of the bloom water to two zooplankton species (Brachionus plicatilis and Artemia salina) demonstrated this bloom event strongly inhibited their swimming capacities and survival. The results of this study suggested that the bloom events caused by multiple species of Kareniaceae in the Fujian coastal waters had adverse impacts on the local fishery resources and zooplankton community.


Subject(s)
Dinoflagellida , Rotifera , Animals , Harmful Algal Bloom , Artemia , Zooplankton
2.
Oncol Rep ; 50(4)2023 Oct.
Article in English | MEDLINE | ID: mdl-37681504

ABSTRACT

Subsequently to the publication of the above paper, an interested reader drew to the authors' attention that a pair of the wound­healing assay data panels featured in Fig. 2E on p. 1011 (namely, the PLZF / 0 h and 48 h data panels for the BGC823 cell line) had also appeared in another article containing a majority of the same authors that had already been published [Chen J­F, Wu P, Xia R, Yang J, Huo X­Y, Gu D­Y, Tang C­J, We D and Yang F: STAT3­induced lncRNA HAGLROS overexpression contributes to the malignant progression of gastric cancer cells via mTOR signal­mediated inhibition of autophagy. Mol Cancer 17: 6, 2018], where the same data had been been used to show the results from differently performed experiments. The authors were able to re­examine their original data files, and realized that this figure had been inadverently assembled incorrectly. The revised version of Fig. 2, containing the correct data for the PLZF / 0 h and 48 h data panels in Fig. 2E, is shown on the next page. Note that the revisions made to this figure do not affect the overall conclusions reported in the paper. The authors are grateful to the Editor of Oncology Reports for allowing them the opportunity to publish this Corrigendum, and apologize to the readership for any inconvenience caused. [Oncology Reports 41: 1007­1018, 2019; DOI: 10.3892/or.2018.6866].

3.
Transl Cancer Res ; 10(5): 2416-2427, 2021 May.
Article in English | MEDLINE | ID: mdl-35116557

ABSTRACT

BACKGROUND: Suppressors of cytokine signaling family member 4 (SOCS4) was shown to serve critical and multifaceted roles in the progression of numerous cancers, including hepatocellular carcinoma, thyroid cancer, breast cancer, and lung adenocarcinoma. While, the expression and the roles of SOCS4 in esophageal squamous cell carcinoma (ESCC) remain elusive. The current study is aimed to investigate the expression pattern and functions of SOCS4 in ESCC. METHODS: The relationship between SOCS4 and the clinicopathological features of ESCC was analyzed. SOCS4 expression in ESCC tissues was measured by western blot, quantitative real-time polymerase chain reaction (qRT-PCR), and immunohistochemical (IHC) staining. The roles of SOCS4 in modulating ESCC cell behaviors were examined using a series of assays, including cell proliferation assay, cell counting kit-8 (CCK-8) assay, cell cycle analysis, and wound-healing assay. RESULTS: In human ESCC tissues, SOCS4 expression was up-regulated and correlated with tumor size and lymph node metastasis, however was not correlated with the overall survival (OS) of patients. SOCS4 silencing in ESCC cells resulted in the suppression of cell growth, which was related to the cell cycle. SOCS4 knockdown also inhibited nuclear factor-kappa B (NF-κB) signaling and decreased the migratory potential of ESCC cells. CONCLUSIONS: These findings revealed that increased expression of SOCS4 in ESCC may promote the progression, proliferation, and migration by NF-κB signaling. Inhibition of SOCS4 may be a promising therapeutic strategy for ESCC.

4.
Zhonghua Nan Ke Xue ; 26(7): 588-594, 2020 Jul.
Article in Chinese | MEDLINE | ID: mdl-33377712

ABSTRACT

OBJECTIVE: To investigate the expression of Linc00662 in PCa and its influence on the biological function of PCa cells. METHODS: Using qRT-PCR, we detected the expression of Linc00662 in the PCa tissue and cell lines and that in the adjacent normal prostatic tissue and epithelial WPMY-1 cells and analyzed the correlation between the expression of Linc00662 and the clinicopathological features of the PCa tissue. We transfected PC-3 and DU145 cells with siRNA, and verified the interference efficiency by qRT-PCR. We examined the effects of interfering with the Linc00662 expression on the proliferation, apoptosis, migration and invasiveness of PC-3 and DU145 cells by CCK-8 assay, Caspase 3/9 activity assay, wound-healing assay and Transwell invasion assay. RESULTS: The expression of Linc00662 in the PCa tissue and cell lines was significantly up-regulated compared with that in the adjacent normal prostatic tissue and epithelial cells (P < 0.01), and the high expression of Linc00662 was positively correlated with the tumor stage (P = 0.002), primary tumor size (P = 0.006), lymph node metastasis (P = 0.001) and distant metastasis (P = 0.001). Transfection of si-Linc00662 into the PC-3 and DU145 cells significantly reduced the expression of Linc00662 (P < 0.01). Compared with the normal control, the PC-3 and DU145 cells in the Linc00662 interference group showed remarkably decreased proliferation, invasion and migration abilities (P < 0.01), but an increased rate of apoptosis (P < 0.01). CONCLUSIONS: Linc00662 is highly expressed in PCa tissues and cells relatively. Knockdown of the Linc00662 expression may inhibit the proliferation, migration and invasiveness and promote the apoptosis of PCa cells. Therefore, Linc00662 could be considered as a new marker of PCa.


Subject(s)
Carcinogenesis , Prostatic Neoplasms , RNA, Long Noncoding , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Male , Prostatic Neoplasms/genetics , RNA, Long Noncoding/genetics , RNA, Small Interfering
5.
Oncol Rep ; 41(2): 1007-1018, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30431129

ABSTRACT

Promyelocytic leukemia zinc finger (PLZF) plays important roles in tumorigenic and developmental processes of various types of cancers. However, the expression of PLZF in gastric cancer (GC) has not been reported. The aim of the present study was to investigate the expression level and potential status of PLZF in GC as well as its possible mechanism. In the present study, we found that PLZF was downregulated in the majority of GC cell lines and tumor tissues and that alteration of PLZF expression was closely correlated with a malignant phenotype, epithelial­mesenchymal transformation and overall survival. Evaluation of in vitro proliferation, colony information, migration and invasion indicated that PLZF gene transduction induced a less malignant phenotype, which was also confirmed through in vivo studies performed in athymic nude mice. Furthermore, we assessed the expression levels of the lncRNA ANRIL in GC and found that it was negatively associated with the level of PLZF and that ANRIL indirectly methylated PLZF to suppress its expression via binding with polycomb repressive complex 2. When GC cells were treated with the methylation inhibitor 5­Aza­2'­deoxycytidine, the expression of PLZF increased, which further confirmed that PLZF was methylated. These results indicated that constitutive ANRIL activation was a possible cause of the lack of PLZF expression in GC cells. Coupled deregulation of PLZF and ANRIL may account for most of the alterations described in GC, and PLZF may become a potential target of GC therapy.


Subject(s)
Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Promyelocytic Leukemia Zinc Finger Protein/genetics , RNA, Long Noncoding/metabolism , Stomach Neoplasms/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , DNA Methylation/genetics , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Histones/genetics , Histones/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic/genetics , Promyelocytic Leukemia Zinc Finger Protein/metabolism , RNA, Long Noncoding/genetics , Stomach/pathology , Stomach/surgery , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Survival Analysis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
6.
Hum Genomics ; 12(1): 40, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30134973

ABSTRACT

BACKGROUND: Massive occurrences of interstitial loss of heterozygosity (LOH) likely resulting from gene conversions were found by us in different cancers as a type of single-nucleotide variations (SNVs), comparable in abundance to the commonly investigated gain of heterozygosity (GOH) type of SNVs, raising the question of the relationships between these two opposing types of cancer mutations. METHODS: In the present study, SNVs in 12 tetra sample and 17 trio sample sets from four cancer types along with copy number variations (CNVs) were analyzed by AluScan sequencing, comparing tumor with white blood cells as well as tissues vicinal to the tumor. Four published "nontumor"-tumor metastasis trios and 246 pan-cancer pairs analyzed by whole-genome sequencing (WGS) and 67 trios by whole-exome sequencing (WES) were also examined. RESULTS: Widespread GOHs enriched with CG-to-TG changes and associated with nearby CNVs and LOHs enriched with TG-to-CG changes were observed. Occurrences of GOH were 1.9-fold higher than LOH in "nontumor" tissues more than 2 cm away from the tumors, and a majority of these GOHs and LOHs were reversed in "paratumor" tissues within 2 cm of the tumors, forming forward-reverse mutation cycles where the revertant LOHs displayed strong lineage effects that pointed to a sequential instead of parallel development from "nontumor" to "paratumor" and onto tumor cells, which was also supported by the relative frequencies of 26 distinct classes of CNVs between these three types of cell populations. CONCLUSIONS: These findings suggest that developing cancer cells undergo sequential changes that enable the "nontumor" cells to acquire a wide range of forward mutations including ones that are essential for oncogenicity, followed by revertant mutations in the "paratumor" cells to avoid growth retardation by excessive mutation load. Such utilization of forward-reverse mutation cycles as an adaptive mechanism was also observed in cultured HeLa cells upon successive replatings. An understanding of forward-reverse mutation cycles in cancer development could provide a genomic basis for improved early diagnosis, staging, and treatment of cancers.


Subject(s)
DNA Copy Number Variations/genetics , Genome, Human/genetics , Loss of Heterozygosity/genetics , Neoplasms/genetics , Genomics , HeLa Cells , High-Throughput Nucleotide Sequencing , Humans , Mutation , Neoplasms/pathology , Polymorphism, Single Nucleotide , Exome Sequencing
7.
Mol Cancer ; 17(1): 6, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29329543

ABSTRACT

BACKGROUND: Long noncoding RNAs (lncRNAs) are an important class of functional regulators involved in human cancers development, including gastric cancer (GC). Studying aberrantly expressed lncRNAs may provide us with new insights into the occurrence and development of gastric cancer by acting as oncogenes or tumor suppressors. In this study, we aim to examine the expression pattern of lncRNA HAGLROS in GC and its clinical significance as well as its biological role in tumor progression. METHODS: Bioinformatics analysis and qRT-PCR were performed to detect the relative expression of HAGLROS in GC tissues and cell lines. Gain or loss of function approaches were used to investigate the biological functions of HAGLROS. The effect of HAGLROS on proliferation was evaluated by MTT, colony formation assay and nude mouse xenograft model. Wound healing and Transwell assays were used to study the invasion and migration of GC cells. FISH, RIP, RNA-seq, Luciferase report assays, RNA pulldown and Western blot were fulfilled to measure molecular mechanisms. Results are shown as means ± S.D. and differences were tested for significance using Student's t-test (two-tailed). RESULTS: We screened out HAGLROS, whose expression was significantly increased and correlated with outcomes of GC patients by publicly available lncRNAs expression profiling and integrating analyses. Exogenous down-regulation of HAGLROS expression significantly suppressed the cell proliferation, invasion and migration. Mechanistic investigations showed that HAGLROS was a direct target of transcriptional factor STAT3. Moreover, HAGLROS knockdown decreased mTOR expression and increased autophagy-related genes ATG9A and ATG9B expression. Further investigation showed that HAGLROS regulated mTOR signals in two manners. In the one hand, HAGLROS competitively sponged miR-100-5p to increase mTOR expression by antagonizing miR-100-5p-mediated mTOR mRNA inhibition. On the other hand, HAGLROS interacted with mTORC1 components to activate mTORC1 signaling pathway which was known to be an important negative signal of autophagy. Here activation of mTORC1 signaling pathway by HAGLROS inhibited autophagy, thereby promoted excessive proliferation and maintained the malignant phenotype of GC cells. CONCLUSION: The present study demonstrates that HAGLROS overexpression contributes to GC development and poor prognosis and will be a target for GC therapy and further develop as a potential prognostic biomarker.


Subject(s)
Autophagy , Gene Expression Regulation, Neoplastic , RNA, Long Noncoding/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Binding Sites , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic , Databases, Genetic , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Heterografts , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Nucleotide Motifs , Prognosis , Protein Binding
8.
Pharmacol Ther ; 183: 137-151, 2018 03.
Article in English | MEDLINE | ID: mdl-29055715

ABSTRACT

Diet sources are closely involved in the pathogenesis of diverse neuropsychiatric disorders and cancers, in addition to inherited factors. Currently, natural products or nutraceuticals (commonly called medical foods) are increasingly employed for adjunctive therapy of these patients. However, the potential molecular mechanisms of the nutrient efficacy remain elusive. In this review, we summarized the neuroprotective and anti-cancer mechanisms of nutraceuticals. It was concluded that the nutraceuticals exerted neuroprotection and suppressed tumor growth possibly through the differential modulations of redox homeostasis. In addition, the balance between reactive oxygen species (ROS) production and ROS elimination was manipulated by multiple molecular mechanisms, including cell signaling pathways, inflammation, transcriptional regulation and epigenetic modulation, which were involved in the therapeutic potential of nutraceutical antioxidants against neurological diseases and cancers. We specifically proposed that ROS scavenging was integral in the neuroprotective potential of nutraceuticals, while alternation of ROS level (either increase or decrease) or disruption of redox homeostasis (ROS addiction) constituted the anti-cancer property of these compounds. We also hypothesized that ROS-associated ferroptosis, a novel type of lipid ROS-dependent regulatory cell death, was likely to be a critical mechanism for the nutraceutical antioxidants. Targeting ferroptosis is advantageous to develop new nutraceuticals with more effective and lower adverse reactions for curing patients with neuropsychiatric diseases or carcinomas.


Subject(s)
Antineoplastic Agents , Antioxidants , Dietary Supplements , Neuroprotective Agents , Animals , Dietary Supplements/classification , Humans , Neoplasms/therapy , Reactive Oxygen Species/metabolism
9.
Onco Targets Ther ; 8: 3255-63, 2015.
Article in English | MEDLINE | ID: mdl-26604792

ABSTRACT

PURPOSE: Taxane-containing induction chemotherapy (IC) regimens in combination with concurrent chemoradiotherapy (CCRT) have been compared with non-taxane-containing IC combined with CCRT in randomized controlled trials (RCTs) in Chinese patients with advanced nasopharyngeal carcinoma (NPC). This meta-analysis aimed to systematically evaluate their clinical efficacy and safety profiling in this ethnic population. METHODS: The electronic databases, PubMed, Embase, MEDLINE, and Chinese Biomedical Database, were searched for eligible studies. The outcomes included overall response rate (ORR), 1-year survival rate, and different types of adverse events. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to evaluate the strength of the associations. RESULTS: A total of 12 RCTs (representing 835 patients) were identified. The pooled analysis showed that taxane-containing regimens had a significant improvement in ORR for nasopharyngeal lesion (OR =4.57, 95% CI =1.14-18.30, P=0.032, z=2.15) but not in cervical lymph nodes (OR =1.23, 95% CI =0.65-2.36, P=0.532, z=0.64) and in 1-year survival rates (OR =1.19, 95% CI =0.10-14.82, P=0.893, z=0.13) compared with non-taxane-containing regimens. Regarding the adverse events and toxicities, grade 3-4 leukopenia and neutropenia were significantly different between the two groups (P<0.001) in favor of the non-taxane-containing regimens, but grade 3-4 vomiting was significantly different between the two groups (P<0.005) in favor of the taxane-containing regimens. CONCLUSION: When combined with CCRT, taxane-containing IC regimens may be more efficient for short-term local control in Chinese patients with locally advanced NPC than the non-taxane-containing IC regimens. Moreover, the major toxic effects, which were bone marrow suppression, could be tolerated by majority of patients. More long-term follow-up and high-quality trials of NPC are needed to validate our findings.

10.
BMC Med Genomics ; 8: 42, 2015 Jul 25.
Article in English | MEDLINE | ID: mdl-26208496

ABSTRACT

BACKGROUND: The presence of loss-of-heterozygosity (LOH) mutations in cancer cell genomes is commonly encountered. Moreover, the occurrences of LOHs in tumor suppressor genes play important roles in oncogenesis. However, because the causative mechanisms underlying LOH mutations in cancer cells yet remain to be elucidated, enquiry into the nature of these mechanisms based on a comprehensive examination of the characteristics of LOHs in multiple types of cancers has become a necessity. METHODS: We performed next-generation sequencing on inter-Alu sequences of five different types of solid tumors and acute myeloid leukemias, employing the AluScan platform which entailed amplification of such sequences using multiple PCR primers based on the consensus sequences of Alu elements; as well as the whole genome sequences of a lung-to-liver metastatic cancer and a primary liver cancer. Paired-end sequencing reads were aligned to the reference human genome to identify major and minor alleles so that the partition of LOH products between homozygous-major vs. homozygous-minor alleles could be determined at single-base resolution. Strict filtering conditions were employed to avoid false positives. Measurements of LOH occurrences in copy number variation (CNV)-neutral regions were obtained through removal of CNV-associated LOHs. RESULTS: We found: (a) average occurrence of copy-neutral LOHs amounting to 6.9% of heterologous loci in the various cancers; (b) the mainly interstitial nature of the LOHs; and (c) preference for formation of homozygous-major over homozygous-minor, and transitional over transversional, LOHs. CONCLUSIONS: The characteristics of the cancer LOHs, observed in both AluScan and whole genome sequencings, point to the formation of LOHs through repair of double-strand breaks by interhomolog recombination, or gene conversion, as the consequence of a defective DNA-damage response, leading to a unified mechanism for generating the mutations required for oncogenesis as well as the progression of cancer cells.


Subject(s)
DNA Damage/genetics , Gene Dosage/genetics , Genomics , Loss of Heterozygosity , Neoplasms/genetics , Alleles , Chromosomes, Human/genetics , Female , Genes, Neoplasm/genetics , High-Throughput Nucleotide Sequencing , Humans , Male , Sequence Analysis, DNA
11.
Oncotarget ; 5(8): 2276-92, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24810364

ABSTRACT

Long noncoding RNAs are involved in diseases including cancer. Here, we reported that ANRIL (CDKN2B-AS1), a 3.8-kb long noncoding RNA, recruiting and binding to PRC2, was generally upregulated in human gastric cancer (GC) tissues. In a cohort of 120 GC patients, the higher expression of ANRIL was significantly correlated with a higher TNM stage (P=0.041) and tumor size (P=0.001). Multivariate analyses revealed that ANRIL expression served as an independent predictor for overall survival (P=0.036). Further experiments revealed that ANRIL knockdown significantly repressed the proliferation both in vitro and in vivo. We also showed that E2F1 could induce ANRIL and ANRIL-mediated growth promotion is in part due to epigenetic repression of miR-99a/miR-449a in Trans (controlling the targets--mTOR and CDK6/E2F1 pathway) by binding to PRC2, thus forming a positive feedback loop, continuing to promote GC cell proliferation. To our knowledge, this is the first report showed that the role of ANRIL in the progression of GC and ANRIL could crosstalk with microRNAs in epigenetic level. Our results suggest that ANRIL, as a growth regulator, may serve as a candidate prognostic biomarker and target for new therapies in human gastric cancer.


Subject(s)
Gene Silencing , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Animals , Blotting, Western , Chromatin Immunoprecipitation , Disease-Free Survival , Epigenesis, Genetic , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred BALB C , Middle Aged , Prognosis , Proportional Hazards Models , Real-Time Polymerase Chain Reaction , Stomach Neoplasms/mortality , Transfection
12.
Mol Cancer ; 13: 68, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24655544

ABSTRACT

BACKGROUND: Recent evidence indicates that long noncoding RNAs (lncRNAs) play a critical role in the regulation of cellular processes, such as differentiation, proliferation and metastasis. These lncRNAs are found to be dysregulated in a variety of cancers. BRAF activated non-coding RNA (BANCR) is a 693-bp transcript on chromosome 9 with a potential functional role in melanoma cell migration. The clinical significance of BANCR, and its' molecular mechanisms controlling cancer cell migration and metastasis are unclear. METHODS: Expression of BANCR was analyzed in 113 non-small cell lung cancer (NSCLC) tissues and seven NSCLC cell lines using quantitative polymerase chain reaction (qPCR) assays. Gain and loss of function approaches were used to investigate the biological role of BANCR in NSCLC cells. The effects of BANCR on cell viability were evaluated by MTT and colony formation assays. Apoptosis was evaluated by Hoechst staining and flow cytometry. Nude mice were used to examine the effects of BANCR on tumor cell metastasis in vivo. Protein levels of BANCR targets were determined by western blotting and fluorescent immunohistochemistry. RESULTS: BANCR expression was significantly decreased in 113 NSCLC tumor tissues compared with normal tissues. Additionally, reduced BANCR expression was associated with larger tumor size, advanced pathological stage, metastasis distance, and shorter overall survival of NSCLC patients. Reduced BANCR expression was found to be an independent prognostic factor for NSCLC. Histone deacetylation was involved in the downregulation of BANCR in NSCLC cells. Ectopic expression of BANCR impaired cell viability and invasion, leading to the inhibition of metastasis in vitro and in vivo. However, knockdown of BANCR expression promoted cell migration and invasion in vitro. Overexpression of BANCR was found to play a key role in epithelial-mesenchymal transition (EMT) through the regulation of E-cadherin, N-cadherin and Vimentin expression. CONCLUSION: We determined that BANCR actively functions as a regulator of EMT during NSCLC metastasis, suggesting that BANCR could be a biomarker for poor prognosis of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Epithelial-Mesenchymal Transition/physiology , Lung Neoplasms/metabolism , Proto-Oncogene Proteins B-raf/biosynthesis , RNA, Long Noncoding/biosynthesis , Animals , Apoptosis/genetics , Blotting, Western , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Cell Movement/physiology , Down-Regulation , Flow Cytometry , Heterografts , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Prognosis , Proto-Oncogene Proteins B-raf/genetics , RNA, Long Noncoding/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
J Clin Bioinforma ; 4(1): 15, 2014.
Article in English | MEDLINE | ID: mdl-25558350

ABSTRACT

BACKGROUND: AluScan combines inter-Alu PCR using multiple Alu-based primers with opposite orientations and next-generation sequencing to capture a huge number of Alu-proximal genomic sequences for investigation. Its requirement of only sub-microgram quantities of DNA facilitates the examination of large numbers of samples. However, the special features of AluScan data rendered difficult the calling of copy number variation (CNV) directly using the calling algorithms designed for whole genome sequencing (WGS) or exome sequencing. RESULTS: In this study, an AluScanCNV package has been assembled for efficient CNV calling from AluScan sequencing data employing a Geary-Hinkley transformation (GHT) of read-depth ratios between either paired test-control samples, or between test samples and a reference template constructed from reference samples, to call the localized CNVs, followed by use of a GISTIC-like algorithm to identify recurrent CNVs and circular binary segmentation (CBS) to reveal large extended CNVs. To evaluate the utility of CNVs called from AluScan data, the AluScans from 23 non-cancer and 38 cancer genomes were analyzed in this study. The glioma samples analyzed yielded the familiar extended copy-number losses on chromosomes 1p and 9. Also, the recurrent somatic CNVs identified from liver cancer samples were similar to those reported for liver cancer WGS with respect to a striking enrichment of copy-number gains in chromosomes 1q and 8q. When localized or recurrent CNV-features capable of distinguishing between liver and non-liver cancer samples were selected by correlation-based machine learning, a highly accurate separation of the liver and non-liver cancer classes was attained. CONCLUSIONS: The results obtained from non-cancer and cancerous tissues indicated that the AluScanCNV package can be employed to call localized, recurrent and extended CNVs from AluScan sequences. Moreover, both the localized and recurrent CNVs identified by this method could be subjected to machine-learning selection to yield distinguishing CNV-features that were capable of separating between liver cancers and other types of cancers. Since the method is applicable to any human DNA sample with or without the availability of a paired control, it can also be employed to analyze the constitutional CNVs of individuals.

14.
Oncol Lett ; 6(6): 1595-1603, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24273600

ABSTRACT

The periostin protein, encoded by the POSTN gene, is a component of the extracellular matrix, which is expressed by fibroblasts and has been observed in a variety of human malignancies. The present study aimed to detect the expression of periostin in the tissues of non-small cell lung cancer (NSCLC) patients and benign lung tumors, and to correlate the results with the clinicopathological data of the subjects, in order to evaluate periostin as a potential prognostic marker. In total, 49 NSCLC patients and 6 benign lung tumors were included in this study. The protein level of periostin was detected in paired normal/paratumor/cancer tissues by a western blot analysis and the mRNA level in paired normal/cancer tissues was detected by quantitative polymerase chain reaction (qPCR). The results were then correlated with established biological and prognostic factors. Immunohistochemistry was used to confirm the location of periostin in the NSCLC tissues. Uni- and multivariate analyses were performed using Cox's proportional hazards regression model. The protein level of periostin was elevated in the cancer tissue of the NSCLC patients compared with the normal (P=0.017) and paratumor (P=0.000) tissues. The expression level in the male patients was much higher than in the female patients at the protein (P=0.001) and mRNA (P=0.010) levels. The mRNA level in the non-adenocarcinoma (non-ADC) patients was much higher than in the adenocarcinoma (ADC) patients (P=0.029). Periostin was demonstrated higher expression at the protein level in the pseudotumors and tuberculosis patients than in the adjacent (P=0.016) and surrounding tissues (P=0.001). Immunostaining indicated that high levels of periostin were present in the mesenchymal areas, but not in the cancer cells themselves. The patients with tumors exhibiting high-level periostin expression showed a significantly shorter survival time (P=0.036, log-rank test). The 3-year survival rate was 81.5% for patients with low-level periostin expression (periostin-L; n=27) and 45.4% for patients with high-level periostin expression (periostin-H; n=22). Similarly, pathological node (pN) status was a significant prognostic marker in the univariate Cox survival analysis. Notably, periostin-H expression was also identified as an independent prognostic factor by the multivariate analysis (P=0.011). These results showed that the overexpression of periostin predicts a poor prognosis, therefore it may be regarded as a novel molecule in the progression and development of NSCLC. The results provide an additional target for the adjuvant treatment of NSCLC.

15.
Asian Pac J Cancer Prev ; 13(10): 4943-6, 2012.
Article in English | MEDLINE | ID: mdl-23244087

ABSTRACT

Studies have reported an association between the TERT rs2736098 single nucleotide polymorphism (SNP) and cancer susceptibility, but the results remain inconclusive. Toprovide a more precise estimation of the relationship, a meta-analysis of 8 published studies including 8,070 cases and 10,239 controls was performed. Stratification by sample size, genotyping method, source of controls and ethnicity were used to explore the source of heterogeneity. In the overall analysis, no significant association was found between the TERT rs2736098 polymorphism and cancer risk. However, the result showed the rs2736098 was significantly associated with an increased cancer risk and the heterogeneity was effectively decreased for homozygote comparison by removal of two studies: OR = 1.337 (95% CI = 1.183-1.511; Pheterogeneity = 0.087). In the subgroup analysis by ethnicity, a significantly increased risk of cancers was found among Asians (OR = 1.413, 95% CI = 1.187-1.683 for AA versus GG). Our meta-analysis did not show that the TERT rs2736098 plays an important role in cancer risk. More studies with larger sample size and well-matched controls are needed to confirm the findings.


Subject(s)
Genetic Predisposition to Disease , Neoplasms/etiology , Polymorphism, Genetic/genetics , Telomerase/genetics , Case-Control Studies , Humans , Prognosis , Risk Factors
16.
World J Gastroenterol ; 18(21): 2712-8, 2012 Jun 07.
Article in English | MEDLINE | ID: mdl-22690082

ABSTRACT

AIM: To investigate the correlation between expression of phosphatase and tensin homolog (PTEN) and cetuximab effects in colorectal cancer. METHODS: We searched PubMed, EMBASE and ASCO to identify eligible studies. Finally, 8 randomized control studies were included in the meta-analysis. STATA 10.0 Software was used to investigate heterogeneity among individual studies and to summarize all the studies. Risk ratios (RRs) and hazard ratios (HRs) with 95% confidence intervals (CIs) were used to assess the strength of the association. RESULTS: Compared with 20 of 266 patients with loss of PTEN, 206 of 496 patients with intact PTEN protein expression had a better objective response rate to cetuximab-based therapy (RR, 4.75; 95% CI, 2.59-8.72; P < 0.001). PTEN positivity was associated with better progression-free survival (PFS) (HR, 0.675; 95% CI, 0.473-0.964; P = 0.031) but not with better overall survival (OS) (HR, 0.608; 95% CI, 0.411-0.899; P = 0.013). In patients with KRAS wild-type status, PTEN positivity did not predict a longer PFS or OS (PFS: HR, 0.707; 95% CI, 0.440-1.138; P = 0.154; OS: HR, 0.943; 95% CI, 0.646-1.377; P = 0.761). CONCLUSION: Expression of PTEN is related to the effect of cetuximab in colorectal cancer patients and should be considered in treatment with cetuximab.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , PTEN Phosphohydrolase/biosynthesis , Antibodies, Monoclonal, Humanized , Cetuximab , Disease-Free Survival , Humans , Medical Oncology/methods , Models, Statistical , Neoplasm Metastasis , Prognosis , Randomized Controlled Trials as Topic , Treatment Outcome , ras Proteins/biosynthesis
17.
World J Gastroenterol ; 16(46): 5889-94, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21155012

ABSTRACT

AIM: To assess the efficiency and toxicities of irinotecan (CPT-11)-involved regimens in patients with advanced gastric cancer. METHODS: Randomized phases II and III clinical trials on chemotherapy for advanced gastric cancer were searched from MEDLINE, EMbase, Cochrane Controlled Trials Register, and EBSCO. Relevant abstracts were manually searched. A total of 657 patients were analyzed for their overall response rate (ORR), time to treatment failure (TTF), overall survival (OS) rate, and toxicities. Overall survival rate, reported as hazard ratio (HR) with 95% CI, was used as the primary outcome measure. RESULTS: Four randomized controlled trials on chemotherapy for advanced gastric cancer were detected. The CPT-11-containing combination chemotherapy was not significantly advantageous over the non CPT-11-containing combination chemotherapy for OS rate (HR = 1.12, 95% CI: 0.92-1.36, P = 0.266) and ORR [risk ratio (RR) = 1.23, 95% CI: 0.71-2.14, P = 0.458]. However, the CPT-11-containing combination chemotherapy was significantly advantageous over the non CPT-11-containing combination chemotherapy for TTF (HR = 1.35, 95% CI: 1.12-1.64, P = 0.002). Grade 3/4 haematological toxicity (thrombocytopenia: RR = 0.20, 95% CI: 0.09-0.48; P < 0.001) and gastrointestinal toxicity (diarrhea: RR = 4.09, 95% CI: 2.42-6.93, P < 0.001) were lower in patients with advanced gastric cancer after CPT-11-containing combination chemotherapy than after non CPT-11 -containing combination chemotherapy. CONCLUSION: CPT-11-containing combination chemotherapy is advantageous over non CPT-11 -containing combination chemotherapy for TTF with no significant toxicity. CPT-11-containing combination chemotherapy can be used in treatment of advanced gastric cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/analogs & derivatives , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Randomized Controlled Trials as Topic , Stomach Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols , Camptothecin/therapeutic use , Databases, Factual , Drug Therapy, Combination , Humans , Irinotecan , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...