1
|
Zhao R, Ding Y, Han R, Ding R, Liu J, Zhu C, Ding D, Bao M. Prognostic correlation between specialized capillary endothelial cells and lung adenocarcinoma. Heliyon 2024; 10:e28236. [PMID: 38533005 PMCID: PMC10963648 DOI: 10.1016/j.heliyon.2024.e28236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Background In-depth analysis of the functional changes occurring in endothelial cells (ECs) involved in capillary formation can help to elucidate the mechanism of tumour vascular growth. Methods Appropriate datasets were retrieved from the GEO database to obtain single-cell data on LUAD samples and adjacent normal tissue samples. ECs were selected by an automatic annotation program in R and further subdivided based on reported EC marker genes. Functional changes in different types of capillary ECs were then visualized, and the concrete expression was classified by genetic data in the TCGA. Finally, a prognostic model was constructed to predict immunoinfiltration status, survival and drug therapy effects. Results The LUAD data contained in the GSE183219 dataset were suitable for our analysis. After dimensionality reduction analysis and cell annotation, EC general capillary and EC aerocyte subsets as capillary specialized phenotypes showed a series of functional changes in tumour samples, with a total of 108 genes found to undergo functional changes. Use of CellPhoneDB revealed a close interaction of activity between ECs. After integration of TCGA, GSE68465 and GSE11969 datasets, the genes obtained were analysed by cluster analysis and risk model construction, identifying 8 genes. Drug sensitivity, immune cell and molecular differences can be accurately predicted. Conclusions EC general capillary and EC aerocyte subsets are recognized capillary ECs in the tumour microenvironment, and the functional changes between them are relevant to the prognosis and treatment of LUAD patients and have the potential to be used in target therapy.
Collapse
Affiliation(s)
- Rongchang Zhao
- Department of Oncology, Taixing People's Hospital, Taixing, China
| | - Yan Ding
- Department of Oncology, Taixing People's Hospital, Taixing, China
| | - Rongbo Han
- Department of Oncology, The Fourth Affiliated Hospital Of Nanjing Medical University, Nanjing, China
| | - Rongjie Ding
- Department of Oncology, Taixing People's Hospital, Taixing, China
| | - Jun Liu
- Department of Oncology, Taixing People's Hospital, Taixing, China
| | - Chunrong Zhu
- Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dan Ding
- Department of Oncology, Taixing People's Hospital, Taixing, China
| | - Minhui Bao
- Department of Oncology, Taixing People's Hospital, Taixing, China
| |
Collapse
|
2
|
Wang X, Li L, Yang Y, Fan L, Ma Y, Mao F. Reveal the Heterogeneity in the Tumor Microenvironment of Pancreatic Cancer and Analyze the Differences in Prognosis and Immunotherapy Responses of Distinct Immune Subtypes. Front Oncol 2022; 12:832715. [PMID: 35252003 PMCID: PMC8891159 DOI: 10.3389/fonc.2022.832715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose The current clinical classification of pancreatic ductal adenocarcinoma (PDAC) cannot well predict the patient’s possible response to the treatment plan, nor can it predict the patient’s prognosis. We use the gene expression patterns of PDAC patients to reveal the heterogeneity of the tumor microenvironment of pancreatic cancer and analyze the differences in the prognosis and immunotherapy response of different immune subtypes. Methods Firstly, use ICGC’s PACA-AU PDAC expression profile data, combined with the ssGSEA algorithm, to analyze the immune enrichment of the patient’s tumor microenvironment. Subsequently, the spectral clustering algorithm was used to extract different classifications, the PDAC cohort was divided into four subtypes, and the correlation between immune subtypes and clinical characteristics and survival prognosis was established. The patient’s risk index is obtained through the prognostic prediction model, and the correlation between the risk index and immune cells is prompted. Results We can divide the PDAC cohort into four subtypes: immune cell and stromal cell enrichment (Immune-enrich-Stroma), non-immune enrichment but stromal cell enrichment (Non-immune-Stroma), immune-enriched Collective but non-matrix enrichment (Immune-enrich-non-Stroma) and non-immune enrichment and non-stromal cell enrichment (Non-immune-non-Stroma). The five-year survival rate of immune-enrich-Stroma and non-immune-Stroma of PACA-CA is quite different. TCGA-PAAD’s immune-enrich-Stroma and immune-enrich-non-Stroma groups have a large difference in productivity in one year. The results of the correlation analysis between the risk index and immune cells show that the patient’s disease risk is significantly related to epithelial cells, megakaryocyte-erythroid progenitor (MEP), and Th2 cells. Conclusion The tumor gene expression characteristics of pancreatic cancer patients are related to immune response, leading to morphologically recognizable PDAC subtypes with prognostic/predictive significance.
Collapse
Affiliation(s)
- Xiaoqin Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Feifei Mao, ; Xiaoqin Wang,
| | - Lifang Li
- Emergency Department, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yang Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First People’s Hospital of Changzhou, Changzhou, China
| | - Linlin Fan
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ying Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Feifei Mao
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Feifei Mao, ; Xiaoqin Wang,
| |
Collapse
|
3
|
Jiang HK, Liang Y. Penalized logistic regression based on L1/2 penalty for high-dimensional DNA methylation data. Technol Health Care 2021; 28:161-171. [PMID: 32364148 PMCID: PMC7369078 DOI: 10.3233/thc-209016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND: DNA methylation is a molecular modification of DNA that is vital and occurs in gene expression. In cancer tissues, the 5’–C–phosphate–G–3’(CpG) rich regions are abnormally hypermethylated or hypomethylated. Therefore, it is useful to find out the diseased CpG sites by employing specific methods. CpG sites are highly correlated with each other within the same gene or the same CpG island. OBJECTIVE: Based on this group effect, we proposed an efficient and accurate method for selecting pathogenic CpG sites. METHODS: Our method aimed to combine a L1/2 regularized solver and a central node fully connected network to penalize group constrained logistic regression model. Consequently, both sparsity and group effect were brought in with respect to the correlated regression coefficients. RESULTS: Extensive simulation studies were used to compare our proposed approach with existing mainstream regularization in respect of classification accuracy and stability. The simulation results show that a greater predictive accuracy was attained in comparison to previous methods. Furthermore, our method was applied to over 20000 CpG sites and verified using the ovarian cancer data generated from Illumina Infinium HumanMethylation 27K Beadchip. In the result of the real dataset, not only the indicators of predictive accuracy are higher than the previous methods, but also more CpG sites containing genes are confirmed pathogenic. Additionally, the total number of CpG sites chosen is less than other methods and the results show higher accuracy rates in comparison to other methods in simulation and DNA methylation data. CONCLUSION: The proposed method offers an advanced tool to researchers in DNA methylation and can be a powerful tool for recognizing pathogenic CpG sites.
Collapse
Affiliation(s)
- Hong-Kun Jiang
- Corresponding author: Hong-Kun Jiang, Faculty of Information Technology, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, China. E-mail:
| | | |
Collapse
|
4
|
Gong Y, Zhao W, Jia Q, Dai J, Chen N, Chen Y, Gu D, Huo X, Chen J. IKBKB rs2272736 is Associated with Gastric Cancer Survival. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2020; 13:345-352. [PMID: 32884329 PMCID: PMC7443400 DOI: 10.2147/pgpm.s258761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/20/2020] [Indexed: 12/18/2022]
Abstract
Background IKBKB/IKKβ, as the core catalytic subunit of IκB kinase complex, participates in mediation of the classical NF-κB pathway, which has been linked to inflammation and tumorigenesis. Previous studies have shown that single nucleotide polymorphisms in IKBKB have been related to gastric cancer, but how they associate to the clinical outcome is not yet clear. In this study, we retrospectively investigated the associations between single nucleotide polymorphisms located in IKBKB and gastric cancer survival. Materials and Methods IKBKB rs2272736 was genotyped in 1210 patients with primary gastric cancer in a Han Chinese population, and the relationships between rs2272736 and overall survival were evaluated. We conducted Cox proportional hazards regression, which was performed to estimate the effects of single nucleotide polymorphisms on the overall survival of patients, adjusted for potential confounding variables. Results We found that patients with rs2272736 A allele in IKBKB had significantly prolonged overall survival time compared to those with the G allele (HR = 0.83, 95% CI = 0.68–1.00, P = 0.050). In addition, AA genotype was demonstrated to have reduced risk of death for gastric cancer compared with that associated with the GG/GA genotypes, which was more common in patients with cardiac carcinoma, well-differentiated and moderately differentiated tumors, TNM Ⅰ/Ⅱ stages and intestinal type. Conclusion Our findings have shown that single nucleotide polymorphism rs2272736 in IKBKB may be a promising prognostic biomarker which should promote personalized treatment.
Collapse
Affiliation(s)
- Yang Gong
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Wenjing Zhao
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Qiong Jia
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Jiali Dai
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Nan Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Yuetong Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Dongying Gu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Xinying Huo
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Jinfei Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.,Cancer Center, Taikang Xianlin Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
5
|
Tsoy TD, Kruglova NA, Filatov AV. Lymphocyte Phosphatase-Associated Phosphoprotein Is a Substrate of Protein Kinase CK2. BIOCHEMISTRY (MOSCOW) 2018; 83:1380-1387. [PMID: 30482149 DOI: 10.1134/s0006297918110081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Lymphocyte phosphatase-associated phosphoprotein (LPAP) is a molecular partner of CD45 phosphatase that plays a key role in the regulation of antigen-specific activation of lymphocytes. The functions of LPAP still remain unknown. We believe that studying LPAP phosphorylation pathways could shed light on its functions. In this work, we studied the phosphorylation of LPAP ectopically expressed in non-lymphoid cells in order to determine the effect of LPAP interaction partners on its phosphorylation. We found that phosphorylation at Ser153 and Ser163 in non-hematopoietic HEK293 cells was conserved, while phosphorylation at Ser99 and Ser172 was almost absent. The pattern of LPAP phosphorylation in K562 erythroid and U937 myeloid cells expressing endogenous CD45 protein was similar to that observed in T and B lymphocytes. We demonstrated for the first time that LPAP is a substrate for protein kinase CK2 that phosphorylates it at Ser153, presumably ensuring LPAP resistance to degradation.
Collapse
Affiliation(s)
- T D Tsoy
- Institute of Immunology National Research Center, Federal Medical-Biological Agency, Moscow, 115522, Russia. .,Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234, Russia
| | - N A Kruglova
- Institute of Immunology National Research Center, Federal Medical-Biological Agency, Moscow, 115522, Russia.,Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234, Russia
| | - A V Filatov
- Institute of Immunology National Research Center, Federal Medical-Biological Agency, Moscow, 115522, Russia. .,Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234, Russia
| |
Collapse
|
6
|
Lee WR, Na H, Lee SW, Lim WJ, Kim N, Lee JE, Kang C. Transcriptomic analysis of mitochondrial TFAM depletion changing cell morphology and proliferation. Sci Rep 2017; 7:17841. [PMID: 29259235 PMCID: PMC5736646 DOI: 10.1038/s41598-017-18064-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/05/2017] [Indexed: 12/27/2022] Open
Abstract
Human mitochondrial transcription factor A (TFAM) has been implicated in promoting tumor growth and invasion. TFAM activates mitochondrial DNA (mtDNA) transcription, and affects nuclear gene expression through mitochondrial retrograde signaling. In this study, we investigated the effects of TFAM depletion on the morphology and transcriptome of MKN45 gastric cancer cells. Morphology alteration became visible at 12 h after TFAM knockdown: the proportion of growth-arrested polygonal cells versus oval-shaped cells increased, reaching a half-maximum at 24 h and a near-maximum at 36 h. TFAM knockdown upregulated four genes and downregulated six genes by more than threefold at 24 h and similarly at 48 h. Among them, the knockdown of CFAP65 (cilia and flagella associated protein 65) or PCK1 (cytoplasmic phosphoenolpyruvate carboxykinase) rescued the effects of TFAM depletion on cell morphology and proliferation. PCK1 was found to act downstream of CFAP65 in calcium-mediated retrograde signaling. Furthermore, mtDNA depletion by 2',3'-dideoxycytidine was sufficient for induction of CFAP65 and PCK1 expression and inhibition of cell proliferation, but oxidative phosphorylation blockade or mitochondrial membrane potential depolarization was not. Thus, the TFAM-mtDNA-calcium-CFAP65-PCK1 axis participates in mitochondrial retrograde signaling, affecting tumor cell differentiation and proliferation.
Collapse
Affiliation(s)
- Woo Rin Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Korea
- Center for Bioanalysis, Korea Research Institute of Standards and Science, Daejeon, 34113, Korea
| | - Heeju Na
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Korea
| | - Seon Woo Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Korea
| | - Won-Jun Lim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Korea
- Department of Bioinformatics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34141, Korea
| | - Namshin Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Korea
- Department of Bioinformatics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34141, Korea
| | - J Eugene Lee
- Center for Bioanalysis, Korea Research Institute of Standards and Science, Daejeon, 34113, Korea.
| | - Changwon Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Korea.
| |
Collapse
|
7
|
Landskron J, Kraggerud SM, Wik E, Dørum A, Bjørnslett M, Melum E, Helland Ø, Bjørge L, Lothe RA, Salvesen HB, Taskén K. C77G in PTPRC (CD45) is no risk allele for ovarian cancer, but associated with less aggressive disease. PLoS One 2017; 12:e0182030. [PMID: 28759630 PMCID: PMC5536273 DOI: 10.1371/journal.pone.0182030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/11/2017] [Indexed: 12/12/2022] Open
Abstract
The pan lymphocyte marker CD45 exists in various isoforms arising from alternative splicing of the exons 4, 5 and 6. While naïve T cells express CD45RA translated from an mRNA containing exon 4, exons 4–6 are spliced out to encode the shorter CD45R0 in antigen-experienced effector/memory T cells. The SNP C77G (rs17612648) is located in exon 4 and blocks the exon’s differential splicing from the pre-mRNA, enforcing expression of CD45RA. Several studies have linked C77G to autoimmune diseases but lack of validation in other cohorts has left its role elusive. An incidental finding in an ovarian cancer patient cohort from West Norway (Bergen region, n = 312), suggested that the frequency of C77G was higher among ovarian cancer patients than in healthy Norwegians (n = 1,357) (3.0% vs. 1.8% allele frequency). However, this finding could not be validated in a larger patient cohort from South-East Norway (Oslo region, n = 1,198) with 1.2% allele frequency. Hence, C77G is not associated with ovarian cancer in the Norwegian population. However, its frequency was increased in patients with FIGO stage II, endometrioid histology or an age at diagnosis of 60 years or older indicating a possible association with a less aggressive cancer type.
Collapse
Affiliation(s)
- Johannes Landskron
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Centre for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| | - Sigrid M. Kraggerud
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Elisabeth Wik
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Section for Pathology, University of Bergen, Bergen, Norway
| | - Anne Dørum
- Department of Gynaecologic Oncology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Merete Bjørnslett
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Espen Melum
- Norwegian PSC Research Centre, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital, The National Hospital, Oslo, Norway
- K.G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | - Øystein Helland
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Line Bjørge
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Ragnhild A. Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Helga B. Salvesen
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Kjetil Taskén
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Centre for Cancer Immunotherapy, University of Oslo, Oslo, Norway
- K.G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
- * E-mail:
| |
Collapse
|
8
|
Wang J, Wang T, Xu J, Chen W, Shi W, Cheng J, Liu P, Zhou X. Prognostic significance of X-ray cross-complementing gene 1 expression in gastric cancer. Chin J Cancer Res 2016; 28:355-61. [PMID: 27478321 PMCID: PMC4949281 DOI: 10.21147/j.issn.1000-9604.2016.03.10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Objective The aim of this study is to identify the prognostic significance of X-ray cross-complementing gene 1 (XRCC1) in patients with gastric cancer undergoing surgery and platinum-based adjuvant chemotherapy.
Methods Immunohistochemistry (IHC) was used to evaluate XRCC1 protein expression profiles on surgical specimens of 612 gastric cancer patients. The relationship between XRCC1 expression and existing prognostic factors, platinum-based adjuvant chemotherapy, disease-free survival (DFS) and overall survival (OS) were analyzed. Results Among 612 patients staged Ⅱ/Ⅲ in our study, 182 (29.74%) were evaluated as XRCC1 IHC positive. XRCC1 expression was not significantly related to OS (P = 0.347) or DFS (P = 0.297). Compared with surgery only, platinum-based adjuvant chemotherapy significantly improved the OS (P = 0.031). And the patients with negative XRCC1 expression benefited more from platinum-based adjuvant chemotherapy (P = 0.049). Multivariate analysis demonstrated that tumor size, T category, N category, vascular or nerve invasion and platinum-based chemotherapy were good prognostic factors for OS (P < 0.05). Though XRCC1 plays an important role in DNA repair pathways, no significant relationship is found in XRCC1 expression and OS among gastric cancer in our study.
Conclusions XRCC1 might be an alternative prognostic marker for the patients of gastric cancer after radical resection. The patients with negative XRCC1 expression can benefit more from platinum-based adjuvant chemotherapy.
Collapse
Affiliation(s)
- Jian Wang
- Department of Medical Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention & Treatment, Cancer Center, Nanjing Medical University, Nanjing 210029, China
| | - Tongshan Wang
- Department of Medical Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention & Treatment, Cancer Center, Nanjing Medical University, Nanjing 210029, China
| | - Jun Xu
- Department of Medical Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - WenJiao Chen
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wei Shi
- Department of Medical Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jianfeng Cheng
- Carolinas Medical Center, University of North Carolina Charlotte Campus, Charlotte, NC 28203, USA
| | - Ping Liu
- Carolinas Medical Center, University of North Carolina Charlotte Campus, Charlotte, NC 28203, USA
| | - Xiqiao Zhou
- Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
9
|
Yu CY, Chen HY. Genetic Variations and Gastric Cancer. Gastrointest Tumors 2015. [DOI: 10.1159/000431265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
<b><i>Background:</i></b> Gastric cancer (GC) has an apparent hereditary component. However, in a large fraction of gastric cases, no known genetic syndrome or family history can be identified, suggesting the presence of ‘missing heritability' in GC etiology. Genome-wide association studies (GWAS) and traditional candidate gene studies have both led to the identification of multiple replicable common genetic variants associated with GC risk. <b><i>Summary:</i></b> We summarize the genetic variants associated with GC risk identified up to date. Achievements derived from translational cancer research including the following aspects: (a) contribution to the our understanding of gastric tumorigenesis, (b) guidance to individualized treatment and (c) prediction of patient prognosis. We also prospect future research direction such as post-GWAS analyses and rare variants studies. <b><i>Key Message:</i></b> Many genetic variants were found through GWAS or candidate gene studies, and interpreting their underlying mechanisms will help us translate risk profiles generated from these variations into use in the clinical setting for targeted screening and treatment. <b><i>Practical Implications:</i></b> Investigation of the potential use of genetic variations as prognostic and predictive markers is a developing field. Many people could benefit from a better understanding of genetic polymorphisms to potentially identify a priori individuals who might have the best chance of survival and therefore derive most clinical benefit from treatment. Outcomes of particular scientific interest for molecular epidemiologic studies should include overall survival, recurrence- and progression-free survival, response to treatment, and early and late toxicities stemming from chemotherapy and radiation.
Collapse
|
10
|
Feigelson HS, Goddard KAB, Hollombe C, Tingle SR, Gillanders EM, Mechanic LE, Nelson SA. Approaches to integrating germline and tumor genomic data in cancer research. Carcinogenesis 2014; 35:2157-63. [PMID: 25115441 DOI: 10.1093/carcin/bgu165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cancer is characterized by a diversity of genetic and epigenetic alterations occurring in both the germline and somatic (tumor) genomes. Hundreds of germline variants associated with cancer risk have been identified, and large amounts of data identifying mutations in the tumor genome that participate in tumorigenesis have been generated. Increasingly, these two genomes are being explored jointly to better understand how cancer risk alleles contribute to carcinogenesis and whether they influence development of specific tumor types or mutation profiles. To understand how data from germline risk studies and tumor genome profiling is being integrated, we reviewed 160 articles describing research that incorporated data from both genomes, published between January 2009 and December 2012, and summarized the current state of the field. We identified three principle types of research questions being addressed using these data: (i) use of tumor data to determine the putative function of germline risk variants; (ii) identification and analysis of relationships between host genetic background and particular tumor mutations or types; and (iii) use of tumor molecular profiling data to reduce genetic heterogeneity or refine phenotypes for germline association studies. We also found descriptive studies that compared germline and tumor genomic variation in a gene or gene family, and papers describing research methods, data sources, or analytical tools. We identified a large set of tools and data resources that can be used to analyze and integrate data from both genomes. Finally, we discuss opportunities and challenges for cancer research that integrates germline and tumor genomics data.
Collapse
Affiliation(s)
- Heather Spencer Feigelson
- Institute for Health Research, Kaiser Permanente Colorado, Denver, CO, USA, Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA and Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Katrina A B Goddard
- Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA and
| | - Celine Hollombe
- Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA and
| | - Sharna R Tingle
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Elizabeth M Gillanders
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Leah E Mechanic
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Stefanie A Nelson
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| |
Collapse
|
11
|
Wang HM, Chang TH, Lin FM, Chao TH, Huang WC, Liang C, Chu CF, Chiu CM, Wu WY, Chen MC, Weng CT, Weng SL, Chiang FF, Huang HD. A new method for post Genome-Wide Association Study (GWAS) analysis of colorectal cancer in Taiwan. Gene 2012; 518:107-13. [PMID: 23262349 DOI: 10.1016/j.gene.2012.11.067] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 11/27/2012] [Indexed: 01/09/2023]
Abstract
Recently, single nucleotide polymorphisms (SNPs) located in specific loci or genes have been identified associated with susceptibility to colorectal cancer (CRC) in Genome-Wide Association Studies (GWAS). However, in different ethnicities and regions, the genetic variations and the environmental factors can widely vary. Therefore, here we propose a post-GWAS analysis method to investigate the CRC susceptibility SNPs in Taiwan by conducting a replication analysis and bioinformatics analysis. One hundred and forty-four significant SNPs from published GWAS results were collected by a literature survey, and two hundred and eighteen CRC samples and 385 normal samples were collected for post-GWAS analysis. Finally, twenty-six significant SNPs were identified and reported as associated with susceptibility to colorectal cancer, other cancers, obesity, and celiac disease in a previous GWAS study. Functional analysis results of 26 SNPs indicate that most biological processes identified are involved in regulating immune responses and apoptosis. In addition, an efficient prediction model was constructed by applying Jackknife feature selection and ANOVA testing. As compared to another risk prediction model of CRC for European Caucasians population, which performs 0.616 of AUC by using 54 SNPs, the proposed model shows good performance in predicting CRC risk within the Taiwanese population, i.e., 0.724 AUC by using 16 SNPs. We believe that the proposed risk prediction model is highly promising for predicting CRC risk within the Taiwanese population. In addition, the functional analysis results could be helpful to explore the potential associated regulatory mechanisms that may be involved in CRC development.
Collapse
Affiliation(s)
- Hwei-Ming Wang
- Division of Colorectal Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Kim M, Ju H, Lim B, Kang C. Maspin genetically and functionally associates with gastric cancer by regulating cell cycle progression. Carcinogenesis 2012; 33:2344-50. [DOI: 10.1093/carcin/bgs280] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
13
|
Lu R, Gao X, Chen Y, Ni J, Yu Y, Li S, Guo L. Association of an NFKB1 intron SNP (rs4648068) with gastric cancer patients in the Han Chinese population. BMC Gastroenterol 2012; 12:87. [PMID: 22776619 PMCID: PMC3407756 DOI: 10.1186/1471-230x-12-87] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 07/10/2012] [Indexed: 12/13/2022] Open
Abstract
Background Hyperactivation of nuclear factor-κB (NF-κB) is associated with various types of tumors. This study investigated the susceptibility of the rs4648068 A/G genotype in the intron region of NFKB1 to gastric cancer and the association of this polymorphism with clinicopathologic variables in gastric cancer patients. Methods A hospital-based case–control study of 248 gastric cancer patients and 192 control individuals was conducted in Fudan University Shanghai Cancer Center (Shanghai, China). Single nucleotide polymorphism (SNP) rs4648068 genotype in NFKB1 from blood samples of a total of 440 people was analyzed by polymerase chain reaction-based genotyping. Results The frequencies of the AA, AG, and GG genotypes of the rs4648068 polymorphism were 31.5%, 47.2%, and 21.3% in the gastric cancer patients and 29.7%, 59.9%, and 10.4% in the control individuals, respectively. We found that the GG genotype was associated with a significantly increased risk of gastric cancer (P = 0.042). Furthermore, among the gastric cancer cases, the rs4648068 GG genotype was associated with high clinical stage (AOR = 2.27, 95% CI: 1.11- 4.66), lymph node involvement (AOR = 2.90, 95% CI = 1.40- 6.03) and serosa invasion (AOR = 2.78, 95% CI = 1.34- 5.75). However, rs4648068 genotypes were not associated with tumor differentiation in gastric cancer patients. Conclusions Homozygous rs4648068 GG was associated with an increased risk of gastric cancer, especially for the lymph node status and serosa invasion in Han Chinese population.
Collapse
Affiliation(s)
- Renquan Lu
- Department of Clinical Laboratory, Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | | | | | | | | | | | | |
Collapse
|
14
|
The interconnectedness of cancer cell signaling. Neoplasia 2012; 13:1183-93. [PMID: 22241964 DOI: 10.1593/neo.111746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 12/14/2011] [Accepted: 12/14/2011] [Indexed: 11/18/2022] Open
Abstract
The elegance of fundamental and applied research activities have begun to reveal a myriad of spatial and temporal alterations in downstream signaling networks affected by cell surface receptor stimulation including G protein-coupled receptors and receptor tyrosine kinases. Interconnected biochemical pathways serve to integrate and distribute the signaling information throughout the cell by orchestration of complex biochemical circuits consisting of protein interactions and covalent modification processes. It is clear that scientific literature summarizing results from both fundamental and applied scientific research activities has served to provide a broad foundational biologic database that has been instrumental in advancing our continued understanding of underlying cancer biology. This article reflects on historical advances and the role of innovation in the competitive world of grant-sponsored research.
Collapse
|
15
|
Lim B, Ju H, Kim M, Kang C. Increased genetic susceptibility to intestinal-type gastric cancer is associated with increased activity of the RUNX3 distal promoter. Cancer 2011; 117:5161-71. [PMID: 21523770 DOI: 10.1002/cncr.26161] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 02/24/2011] [Accepted: 03/10/2011] [Indexed: 01/17/2023]
Abstract
BACKGROUND The runt-related transcription factor RUNX3 plays essential roles in various types of tumors, including gastric cancer. Epigenetic changes in the methylation of the RUNX3 proximal promoter, but not common genetic changes in RUNX3, have been associated with both changes in the gene expression and development of the cancer. METHODS A case-control association study was conducted by genotyping 865 unrelated Korean subjects. Subsequent functional studies were performed to reveal functional implication of genetic association. RESULTS Several single-nucleotide polymorphisms (SNPs) in RUNX3 were significantly associated with susceptibility to intestinal-type gastric cancer (.0028 ≤ P ≤ .022) but not diffuse-type gastric cancer (.70 ≤ P ≤ .96). The risk-associated, minor variant of an intestinal-type gastric cancer-associated SNP in the RUNX3 distal promoter (rs7528484) significantly increased promoter activity in a CREB1-dependent manner. The distal promoter-derived, 33 kDa isoform of RUNX3 increased the activity of transcription factor nuclear factor kappa B (NF-κB), which had been activated by Helicobacter pylori infection, a risk factor for intestinal-type gastric cancer, and the expression of the interleukin-1β gene (IL1B), an NF-κB target genetically and functionally associated with gastric cancer. In contrast, the proximal promoter-derived, 44 kDa isoform of RUNX3 decreased both NF-κB activity and IL1B expression. CONCLUSIONS In addition to epigenetic changes in the RUNX3 proximal promoter, genetic changes in the distal promoter may be associated with susceptibility to intestinal-type gastric cancer by increasing promoter activity. Functionally, 2 RUNX3 isoforms may contribute differentially to intestinal-type gastric cancer susceptibility, at least in part through regulating NF-κB activity and IL1B expression.
Collapse
Affiliation(s)
- Byungho Lim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | | | | | | |
Collapse
|
16
|
Dinosaurs and ancient civilizations: reflections on the treatment of cancer. Neoplasia 2011; 12:957-68. [PMID: 21170260 DOI: 10.1593/neo.101588] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 11/15/2010] [Accepted: 11/15/2010] [Indexed: 12/14/2022] Open
Abstract
Research efforts in the area of palaeopathology have been seen as an avenue to improve our understanding of the pathogenesis of cancer. Answers to questions of whether dinosaurs had cancer, or if cancer plagued ancient civilizations, have captured the imagination as well as the popular media. Evidence for dinosaurian cancer may indicate that cancer may have been with us from the dawn of time. Ancient recorded history suggests that past civilizations attempted to fight cancer with a variety of interventions. When contemplating the issue why a generalized cure for cancer has not been found, it might prove useful to reflect on the relatively limited time that this issue has been an agenda item of governmental attention as well as continued introduction of an every evolving myriad of manmade carcinogens relative to the total time cancer has been present on planet Earth. This article reflects on the history of cancer and the progress made following the initiation of the "era of cancer chemotherapy."
Collapse
|
17
|
Abstract
Gastric cancer (GC) is an important cause of morbidity and mortality worldwide. In addition to environmental factors, genetic factors also play an important role in GC etiology, as demonstrated by the fact that only a small proportion of individuals exposed to the known environmental risk factors develop GC. Molecular studies have provided evidence that GC arises not only from the combined effects of environmental factors and susceptible genetic variants but also from the accumulation of genetic and epigenetic alterations that play crucial roles in the process of cellular immortalization and tumorigenesis. This review is intended to focus on the recently described basic aspects that play key roles in the process of gastric carcinogenesis. Genetic variation in the genes DNMT3A, PSCA, VEGF, and XRCC1 has been reported to modify the risk of developing gastric carcinoma. Several genes have been newly associated with gastric carcinogenesis, both through oncogenic activation (MYC, SEMA5A, BCL2L12, RBP2 and BUBR1) and tumor suppressor gene inactivation mechanisms (KLF6, RELN, PTCH1A, CLDN11, and SFRP5). At the level of gastric carcinoma treatment, the HER-2 tyrosine kinase receptor has been demonstrated to be a molecular target of therapy.
Collapse
Affiliation(s)
- Carlos Resende
- Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s ⁄ n, Porto, Portugal
| | | | | |
Collapse
|
18
|
The War on Cancer rages on. Neoplasia 2010; 11:1252-63. [PMID: 20019833 DOI: 10.1593/neo.91866] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 11/03/2009] [Accepted: 11/03/2009] [Indexed: 02/08/2023] Open
Abstract
In 1971, the "War on Cancer" was launched by the US government to cure cancer by the 200-year anniversary of the founding of the United States of America, 1976. This article briefly looks back at the progress that has been made in cancer research and compares progress made in other areas of human affliction. While progress has indeed been made, the battle continues to rage on.
Collapse
|