1
|
Li X, Wang X, Yu F, Li Z, Chen D, Qi Y, Lu Z, Liu Y, Chen D, Wu Y. Development and validation of a prognostic and drug sensitivity model for gastric cancer utilizing telomere-related genes. Transl Oncol 2025; 52:102232. [PMID: 39647324 PMCID: PMC11667168 DOI: 10.1016/j.tranon.2024.102232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/10/2024] [Accepted: 11/28/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) poses a major global health challenge because of its unfavorable prognosis. Elevated telomerase activity has been linked to the rapid growth and invasiveness of GC tumors. Investigating the expression profiles of telomerase could improve our understanding of the mechanisms underlying telomere-related GC advancement and its applicability as potential targets for diverse therapeutic strategies for GC. METHODS The TCGA and GEO databases were utilized to access transcriptome and clinical data related to GC. After assessing differentially expressed genes (DEGs), a prognostic risk model was developed through Cox univariate regression, LASSO-Cox regression. The prognostic risk model was validated using data from the GSE62254 cohort. The significant influence of the risk model on the tumor immune microenvironment (TIME) and its sensitivity to various drugs was assessed. RESULTS Differential expression analysis identified 328 significantly telomere-related DEGs in GC, with 35 of them showing a significant association with GC prognosis. A predictive risk model composed of four telomere-related genes (TRGs) was established, enabling the accurate stratification of GC patients into two distinct prognostic groups. The LASSO risk model demonstrated notable variations in immune-cell infiltration and drug sensitivity patterns between high- and low-risk groups. CONCLUSIONS The study establishes suggestive relationships between four TRGs (LRRN1, SNCG, GAMT, and PDE1B) and the prognosis of GC. The comprehensive characterization of the TRG model reveals their possible roles in the prognosis, TIME, and drug sensitivity in GC.
Collapse
Affiliation(s)
- Xiaoxiao Li
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaoxuan Wang
- The state Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co.,Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Fuxiang Yu
- Department of General Surgery, Dandong First Hospital, Dandong, Liaoning, China
| | - Zhongguo Li
- Department of General Surgery, Dandong First Hospital, Dandong, Liaoning, China
| | - Daxin Chen
- Department of General Surgery, Dandong First Hospital, Dandong, Liaoning, China
| | - Yingxue Qi
- The state Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co.,Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Zhongyu Lu
- The state Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co.,Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Yaqin Liu
- The state Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co.,Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Dongsheng Chen
- The state Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co.,Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China; Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China.
| | - Yaoqiang Wu
- Department of General Surgery, Dandong First Hospital, Dandong, Liaoning, China.
| |
Collapse
|
2
|
Lili M, Yuxiang F, Zhongcheng H, Ying S, Ru C, Rong X, Jiang L. Genetic variations associated with telomere length affect the risk of gastric carcinoma. Medicine (Baltimore) 2020; 99:e20551. [PMID: 32502020 PMCID: PMC7306382 DOI: 10.1097/md.0000000000020551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 03/13/2020] [Accepted: 05/03/2020] [Indexed: 11/26/2022] Open
Abstract
This study aimed to further understand the role of relative telomere length (RTL) in susceptibility to gastric carcinoma (GC) and investigate the association between genetic polymorphisms in the telomere length related genes and GC risk.RTL was measured using the real-time quantitative polymerase chain reaction from 1000 patients and 1100 healthy controls. Genotyping was performed using the Agena MassARRAY platform. The statistical analysis was performed using the chi-square/ Welch T tests, Mann-Whitney U test, and logistic regression analysis.The association analysis of telomere length and GC showed that the RTL in the case group was shorter than in the controls, and the shorter RTL was associated with an increased risk of GC. The association analysis between telomere length related genes polymorphisms and genetic susceptibility to GC indicated that: In the allele models and genetic models, TERT (rs10069690, rs2242652 and rs2853676) and TN1F1 (rs7708392 and rs10036748) were significantly associated with an increased risk of GC. In addition, the haplotype 'Grs10069690Crs2242652" of TERT and the haplotype 'Grs7708392Trs10036748" of TNIP1 were associated with an increased risk of GCOur results suggested that shorter RTL was associated with an increased risk of GC; The association analysis have identified that the TERT (rs10069690, rs2242652 and rs2853676) and TN1P1 (rs7708392 and rs10036748) were associated with GC risk.
Collapse
Affiliation(s)
- Ma Lili
- Department of Oncology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Fan Yuxiang
- The Second Department of Oncology, Traditional Chinese Medical Hospital of Xinjiang Uygur Autonomous Region (The Fourth Affiliated Hospital of Xinjiang Medical University), Urumqi, Xinjiang, China
| | - Han Zhongcheng
- Department of Oncology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Su Ying
- Department of Oncology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Chen Ru
- Department of Oncology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Xu Rong
- Department of Oncology, People's Hospital of Xinjiang Uygur Autonomous Region
| | - Liu Jiang
- Department of Oncology, People's Hospital of Xinjiang Uygur Autonomous Region
| |
Collapse
|
3
|
Trybek T, Kowalik A, Góźdź S, Kowalska A. Telomeres and telomerase in oncogenesis. Oncol Lett 2020; 20:1015-1027. [PMID: 32724340 PMCID: PMC7377093 DOI: 10.3892/ol.2020.11659] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/25/2020] [Indexed: 12/24/2022] Open
Abstract
Telomeres are located at the ends of chromosomes and protect them from degradation. Suppressing the activity of telomerase, a telomere-synthesizing enzyme, and maintaining short telomeres is a protective mechanism against cancer in humans. In most human somatic cells, the expression of telomerase reverse transcriptase (TERT) is repressed and telomerase activity is inhibited. This leads to the progressive shortening of telomeres and inhibition of cell growth in a process called replicative senescence. Most types of primary cancer exhibit telomerase activation, which allows uncontrolled cell proliferation. Previous research indicates that TERT activation also affects cancer development through activities other than the canonical function of mediating telomere elongation. Recent studies have improved the understanding of the structure and function of telomeres and telomerase as well as key mechanisms underlying the activation of TERT and its role in oncogenesis. These advances led to a search for drugs that inhibit telomerase as a target for cancer therapy. The present review article summarizes the organization and function of telomeres, their role in carcinogenesis, and advances in telomerase-targeted therapy.
Collapse
Affiliation(s)
- Tomasz Trybek
- Endocrinology Clinic, Holycross Cancer Center, 25-734 Kielce, Poland
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holycross Cancer Center, 25-734 Kielce, Poland
| | - Stanisław Góźdź
- The Faculty of Health Sciences, Jan Kochanowski University, 25-319 Kielce, Poland.,Oncology Clinic, Holycross Cancer Center, 25-734 Kielce, Poland
| | - Aldona Kowalska
- Endocrinology Clinic, Holycross Cancer Center, 25-734 Kielce, Poland.,The Faculty of Health Sciences, Jan Kochanowski University, 25-319 Kielce, Poland
| |
Collapse
|
4
|
Ko E, Kim JS, Bae JW, Kim J, Park SG, Jung G. SERPINA3 is a key modulator of HNRNP-K transcriptional activity against oxidative stress in HCC. Redox Biol 2019; 24:101217. [PMID: 31121493 PMCID: PMC6529774 DOI: 10.1016/j.redox.2019.101217] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/26/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023] Open
Abstract
Most studies about serpin peptidase inhibitor, clade A member 3 (SERPINA3) has been limited to its inhibitory functions and mechanisms. Herein, we report a novel role of SERPINA3 in transcriptional regulation of HCC progression-related genes. Among 19 selected genes through HCC cell isolation system based on telomere length, microarray analyses, and cell-based studies, SERPINA3 was the strongest determinant of increases in telomere length, HCC cell proliferation, survival, migration, and invasion. We also found that SERPINA3 strongly interacted with heterogeneous nuclear ribonucleoprotein K (HNRNP-K) under H2O2 exposure, and the oxidation-elicited SERPINA3-HNRNP-K complex enhanced the promoter activities and transcript levels of a telomere-relating gene (POT1) and HCC-promoting genes (UHRF1 and HIST2H2BE). Intriguingly, the inhibition of SERPINA3 oxidation rendered the transcriptional activity of the SERPINA3-HNRNP-K complex suppressed. Moreover, the co-immunoprecipitated HNRNP-K with SERPINA3 quantitatively correlated with not only the level of SERPINA3 oxidation but also the level of POT1, UHRF1, and HIST2H2BE transcripts and telomere length in HCC tissues. Therefore, the upregulated transcriptional activity of HNRNP-K mediated by SERPINA3 promotes HCC cell survival and proliferation and could be an indicator of poor prognosis for HCC patients. SERPINA3-HNRNP-K complex promotes HCC survival and proliferation. Oxidation of SERPINA3 accentuated the role of complex on target regulatory DNA. Blockade of the SERPINA3-HNRNP-K complex could be valuable in HCC therapy.
Collapse
Affiliation(s)
- Eunkyong Ko
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Jong-Seo Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea; Center for RNA Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Jong Woo Bae
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea; Center for RNA Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Jeesoo Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea; Center for RNA Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Sung-Gyoo Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Guhung Jung
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
5
|
Hanif F, Perveen K, Malhi SM, Jawed H, Simjee SU. Verapamil potentiates anti-glioblastoma efficacy of temozolomide by modulating apoptotic signaling. Toxicol In Vitro 2018; 52:306-313. [PMID: 30003979 DOI: 10.1016/j.tiv.2018.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 11/27/2022]
Abstract
Glioblastoma Multiforme (GBM) is the most malignant and invasive tumor of the CNS. Although temozolomide (TMZ) has improved the survival, long-lasting responses have not been reported. Therefore, there is a need to develop improved treatments, one of which might be newly identified drugs which can be used in combination therapy with low doses of standard drugs. Verapamil (VP) a known antihypertensive drug has been shown to enhance the activity of bis-chloroethylnitrosourea (BCNU), a drug used to treat GBM. Since, TMZ has replaced BCNU as the standard GBM chemotherapy; therefore, we aimed to study in vitro interaction of VP and TMZ against GBM. Anti-proliferative and apoptotic activities were studied using MTT, TUNEL assay and DAPI staining. Synergy was assessed using combination index method. Apoptotic markers were evaluated by RT-PCR, and immunocytochemistry. Both VP and TMZ significantly inhibited the growth of U87 cells in dose dependent manner. The combine effect of TMZ with VP was synergistic with a CDI value of <1. Combination of TMZ and VP increased the ratio of Bax to Bcl-2 expression and thus shifted the equilibrium of cells towards apoptosis. Our findings suggest that the synergistic growth inhibition that was observed in combination treatment group may in part relate to increase in apoptosis. The combine administration of VP and TMZ may be therapeutically exploited for the management of GBM.
Collapse
Affiliation(s)
- Farina Hanif
- Institute of Biomedical Sciences, Dow University of Health Sciences, Ojha Campus, SUPARCO Road, Karachi, Pakistan; Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| | - Kahkashan Perveen
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan; Department of Biochemistry, Baqai Medical University, Karachi, Pakistan
| | - Saima M Malhi
- Institute of Biomedical Sciences, Dow University of Health Sciences, Ojha Campus, SUPARCO Road, Karachi, Pakistan; H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| | - Huma Jawed
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan; Mohammad Ali Jinnah University, Karachi, Pakistan
| | - Shabana U Simjee
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan; H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
6
|
Gomez DLM, Armando RG, Cerrudo CS, Ghiringhelli PD, Gomez DE. Telomerase as a Cancer Target. Development of New Molecules. Curr Top Med Chem 2017; 16:2432-40. [PMID: 26873194 PMCID: PMC4997958 DOI: 10.2174/1568026616666160212122425] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 09/15/2015] [Accepted: 10/25/2015] [Indexed: 12/26/2022]
Abstract
Telomeres are the terminal part of the chromosome containing a long repetitive and non-codifying sequence that has as function protecting the chromosomes. In normal cells, telomeres lost part of such repetitive sequence in each mitosis, until telomeres reach a critical point, triggering at that time senescence and cell death. However, in most of tumor cells in each cell division a part of the telomere is lost, however the appearance of an enzyme called telomerase synthetize the segment that just has been lost, therefore conferring to tumor cells the immortality hallmark. Telomerase is significantly overexpressed in 80–95% of all malignant tumors, being present at low levels in few normal cells, mostly stem cells. Due to these characteristics, telomerase has become an attractive target for new and more effective anticancer agents. The capability of inhibiting telomerase in tumor cells should lead to telomere shortening, senescence and apoptosis. In this work, we analyze the different strategies for telomerase inhibition, either in development, preclinical or clinical stages taking into account their strong points and their caveats. We covered strategies such as nucleosides analogs, oligonucleotides, small molecule inhibitors, G-quadruplex stabilizers, immunotherapy, gene therapy, molecules that affect the telomere/telomerase associated proteins, agents from microbial sources, among others, providing a balanced evaluation of the status of the inhibitors of this powerful target together with an analysis of the challenges ahead.
Collapse
Affiliation(s)
| | | | | | | | - D E Gomez
- Laboratory of Molecular Oncology, Department of Science and Technology. Quilmes National University, Bernal, Buenos Aires, Argentina. R. Saenz Peña 352, (1876) Buenos Aires, Argentina.
| |
Collapse
|
7
|
Wincewicz A, Kowalik A, Zięba S, Lewitowicz P, Góźdź S, Sulkowski S. α-Fetoprotein-Producing Hepatoid Gastric Adenocarcinoma With Osteoclast-Like Giant Cells and Neuroendocrine Differentiation: A Case Study With Molecular Profiling. Int J Surg Pathol 2015; 23:537-41. [PMID: 26009570 DOI: 10.1177/1066896915586807] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Here we present the case of a 73-year-old woman with an ulcerated, advanced, hepatoid, and α-fetoprotein-producing poorly differentiated (G3) primary gastric adenocarcinoma pT3 N3a M1 with multinucleated cells and evident neuroendocrine component. This tumor was consistent with giant cell tumor type gastric carcinoma with osteoclast-like giant cells (OGCs). The cancer was HER2 and E-cadherin negative, chromogranin A dispersedly and moderately positive, and strongly α-fetoprotein-positive with evident CK AE1/AE3 immunoreactivity, while OGCs expressed CD68. To provide an insight into the molecular background of this peculiar neoplasm, next-generation sequencing (NGS) was performed to analyze the 50 most frequently mutated oncogenes and tumor suppressors. We detected mutations in the primary tumor in the following genes: KIT, EGFR, PTEN, ATM, and RB1. In the liver metastasis, we revealed mutations in 3 genes: PIK3CA, KIT, and CDKN2A.
Collapse
Affiliation(s)
- Andrzej Wincewicz
- Department of Anatomy, Faculty of Health Sciences, Jan Kochanowski University, Kielce, Poland Non Public Health Care Unit - Department of Pathology, Kielce, Poland, Specialist Medical Practice-Pathologist Kielce
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holy Cross Cancer Center, Kielce, Poland
| | - Sebastian Zięba
- Department of Molecular Diagnostics, Holy Cross Cancer Center, Kielce, Poland
| | - Piotr Lewitowicz
- Non Public Health Care Unit - Department of Pathology, Kielce, Poland, Specialist Medical Practice-Pathologist Kielce Department of Pathology, Faculty of Health Sciences, Jan Kochanowski University of Kielce
| | - Stanisław Góźdź
- Department of Clinical Oncology Holy Cross Cancer Centre, Kielce, Poland Department of Prevention and Epidemiology of Neoplasms, Institute of Public Health, Faculty of Health Sciences, Jan Kochanowski University, Poland
| | - Stanisław Sulkowski
- Department of General Pathomorphology, Collegium Pathologicum, Medical University of Bialystok, Poland
| |
Collapse
|
8
|
Hanif F, Perveen K, Jawed H, Ahmed A, Malhi SM, Jamall S, Simjee SU. N-(2-hydroxyphenyl)acetamide (NA-2) and Temozolomide synergistically induce apoptosis in human glioblastoma cell line U87. Cancer Cell Int 2014; 14:133. [PMID: 25663820 PMCID: PMC4319240 DOI: 10.1186/s12935-014-0133-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/19/2014] [Indexed: 01/24/2023] Open
Abstract
Background Despite the modern therapies available for treating glioblastoma multiforme (GBM), it is still a deadly disease. The development of new therapeutic strategies for the management of gliomas is therefore crucial. The present study is designed to analyze the therapeutic potentials of synthetic compound N-(2-hydroxyphenyl)acetamide (NA-2) in the treatment of GBM as a single agent or in combination with Temozolomide (TMZ) on glioblastoma cells. Methods MTT and TUNEL assays were used to detect the growth inhibitory effect and apoptotic activity of NA-2 alone and in combination with TMZ. Synergy was assessed using combination Index method. The expression of apoptosis related markers Bax, Bcl-2 and caspase-3 were assessed by RT-PCR, whereas, the active caspase-3 protein expression was determined using imunocytochemistry. Results Both NA-2 and TMZ inhibited the growth of U87 in a dose dependent manner. The combine administration of NA-2 (0.33 mM) and temozolomide (0.1 mM) significantly enhanced the cell growth inhibition and apoptosis. Furthermore RT-PCR and imunocytochemistry data revealed that cooperative apoptosis induction was associated with increased ratio of Bax to Bcl-2 and active Caspase-3 expression. Conclusion Our findings support that NA-2 possesses strong apoptotic activity and the combined administration of NA-2 and TMZ may be therapeutically exploited for the management of GBM. Electronic supplementary material The online version of this article (doi:10.1186/s12935-014-0133-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Farina Hanif
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270 Pakistan
| | - Kahkashan Perveen
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270 Pakistan
| | - Huma Jawed
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270 Pakistan
| | - Aqeel Ahmed
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270 Pakistan
| | - Saima M Malhi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270 Pakistan
| | - Siddiqua Jamall
- Department of Biochemistry, University of Karachi, Karachi, 75270 Pakistan
| | - Shabana U Simjee
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270 Pakistan ; H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270 Pakistan
| |
Collapse
|
9
|
Hohman TJ, Koran MEI, Thornton-Wells TA. Genetic modification of the relationship between phosphorylated tau and neurodegeneration. Alzheimers Dement 2014; 10:637-645.e1. [PMID: 24656848 PMCID: PMC4169762 DOI: 10.1016/j.jalz.2013.12.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 12/04/2013] [Accepted: 12/09/2013] [Indexed: 01/18/2023]
Abstract
BACKGROUND A subset of individuals present at autopsy with the pathologic features of Alzheimer's disease having never manifest the clinical symptoms. We sought to identify genetic factors that modify the relationship between phosphorylated tau (PTau) and dilation of the lateral inferior ventricles. METHODS We used data from 700 subjects enrolled in the Alzheimer's Disease Neuroimaging Initiative (ADNI). A genome-wide association study approach was used to identify PTau × single nucleotide polymorphism (SNP) interactions. Variance explained by these interactions was quantified using hierarchical linear regression. RESULTS Five SNP × PTau interactions passed a Bonferroni correction, one of which (rs4728029, POT1, 2.6% of variance) was consistent across ADNI-1 and ADNI-2/GO subjects. This interaction also showed a trend-level association with memory performance and levels of interleukin-6 receptor. CONCLUSIONS Our results suggest that rs4728029 modifies the relationship between PTau and both ventricular dilation and cognition, perhaps through an altered neuroinflammatory response.
Collapse
Affiliation(s)
- Timothy J Hohman
- The Center for Human Genetics Research, Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Mary Ellen I Koran
- The Center for Human Genetics Research, Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Tricia A Thornton-Wells
- The Center for Human Genetics Research, Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
10
|
Telomere 1 (POT1) gene expression and its association with telomerase activity in colorectal tumor samples with different pathological features. Biomed Pharmacother 2014; 68:841-6. [DOI: 10.1016/j.biopha.2014.08.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 08/07/2014] [Indexed: 01/18/2023] Open
|
11
|
Fridley BL, Armasu SM, Cicek MS, Larson MC, Wang C, Winham SJ, Kalli KR, Koestler DC, Rider DN, Shridhar V, Olson JE, Cunningham JM, Goode EL. Methylation of leukocyte DNA and ovarian cancer: relationships with disease status and outcome. BMC Med Genomics 2014; 7:21. [PMID: 24774302 PMCID: PMC4102255 DOI: 10.1186/1755-8794-7-21] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 04/16/2014] [Indexed: 12/22/2022] Open
Abstract
Background Genome-wide interrogation of DNA methylation (DNAm) in blood-derived leukocytes has become feasible with the advent of CpG genotyping arrays. In epithelial ovarian cancer (EOC), one report found substantial DNAm differences between cases and controls; however, many of these disease-associated CpGs were attributed to differences in white blood cell type distributions. Methods We examined blood-based DNAm in 336 EOC cases and 398 controls; we included only high-quality CpG loci that did not show evidence of association with white blood cell type distributions to evaluate association with case status and overall survival. Results Of 13,816 CpGs, no significant associations were observed with survival, although eight CpGs associated with survival at p < 10-3, including methylation within a CpG island located in the promoter region of GABRE (p = 5.38 x 10-5, HR = 0.95). In contrast, 53 CpG methylation sites were significantly associated with EOC risk (p <5 x10-6). The top association was observed for the methylation probe cg04834572 located approximately 315 kb upstream of DUSP13 (p = 1.6 x10-14). Other disease-associated CpGs included those near or within HHIP (cg14580567; p =5.6x10-11), HDAC3 (cg10414058; p = 6.3x10-12), and SCR (cg05498681; p = 4.8x10-7). Conclusions We have identified several CpGs in leukocytes that are differentially methylated by case-control status. Since a retrospective study design was used, we cannot differentiate whether DNAm was etiologic or resulting from EOC; thus, prospective studies of EOC-associated loci are the critical next step.
Collapse
Affiliation(s)
- Brooke L Fridley
- Department of Biostatistics, University of Kansas, Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hao SY, Yu JC. Shelterin complex and digestive system tumor. Shijie Huaren Xiaohua Zazhi 2012; 20:3124-3129. [DOI: 10.11569/wcjd.v20.i32.3124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Shelterin complex is the crucial components of telomere binding proteins. The regulation of this complex, together with telomerase and the alterative lengthening of telomeres (ALT mechanism), plays a critical role in maintaining telomere functions. Telomeres are DNA-protein complexes that contain short repeat sequences added on to the ends of chromosome by the telomerase for protecting the ends of chromosome and preventing chromosome fusion. The loss of protective function of telomeres is closely related to genome instability, and this is the molecular basis for tumor development. Thus, telomeres play key roles in the process of malignant tumor development. Many studies have shown that telomere binding proteins are associated with gastric, colorectal and liver cancers, and other digestive system tumors. This review will focus on the role of the shelterin complex in digestive system neoplasms to provide an insight into prevention and targeted therapy of these malignancies.
Collapse
|
13
|
Wan SM, Tie J, Zhang YF, Guo J, Yang LQ, Wang J, Xia SH, Yang SM, Wang RQ, Fang DC. Silencing of the hPOT1 gene by RNA inference promotes apoptosis and inhibits proliferation and aggressive phenotype of gastric cancer cells, likely through up-regulating PinX1 expression. J Clin Pathol 2011; 64:1051-7. [PMID: 21778296 DOI: 10.1136/jclinpath-2011-200211] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND The human protection of telomeres 1 (hPOT1) protein, a single-strand telomeric DNA binding protein, plays an important role in telomere protection and telomere length regulation. However, its effect on invasion of gastric cancer remains unclear. AIMS To explore the role of hPOT1 in the proliferation and invasion of gastric cancer cells. METHODS The gastric expression of hPOT1 was examined in normal gastric mucosa (n=25), intestinal metaplasia (n=20), gastric dysplasia (n=20) and gastric cancer (n=150) by immunohistochemistry. The mean optical density (MOD) of the immunostaining was determined by semi-quantitative image analysis. The role of hPOT1 in the cell proliferation, apoptosis and invasion of gastric cancer 7901 cells was determined by means of the RNA interference (RNAi) of hPOT1 mRNA. The effects of hPOT1 RNAi on the expression of hPinX1 and hTERT were detected with western blotting. RESULTS The hPOT1 MOD was progressively increased from the normal mucosa to intestinal metaplasia, dysplasia, and gastric cancer. An increased hPOT1 expression significantly correlated with tumour serosal invasion, node metastasis and advanced stage. Transfection of hPOT1 siRNA into SGC-7901 cells led to a decrease in cell proliferation, colony formation and invasion, and also an increase of apoptosis. An up-regulation of hPinX1 and down-regulation of hTERT were found in gastric cancer cells with hPOT1 siRNA. CONCLUSIONS Increased hPOT1 expression is associated with an advanced tumour stage. hPOT1 RNAi inhibits proliferation and invasion, and induces apoptosis of gastric cancer cells. The effects of hPOT1 RNAi seem to be functionally linked to up-regulation of PinX1 and down-regulation of hTERT.
Collapse
Affiliation(s)
- Shun-Mei Wan
- Department of Gastroenterology, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Xu DZ, Geng QR, Tian Y, Cai MY, Fang XJ, Zhan YQ, Zhou ZW, Li W, Chen YB, Sun XW, Guan YX, Li YF, Lin TY. Activated mammalian target of rapamycin is a potential therapeutic target in gastric cancer. BMC Cancer 2010; 10:536. [PMID: 20929525 PMCID: PMC2958947 DOI: 10.1186/1471-2407-10-536] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2009] [Accepted: 10/07/2010] [Indexed: 12/12/2022] Open
Abstract
Background The mammalian target of rapamycin (mTOR) plays a key role in cellular growth and homeostasis. The purpose of our present study is to investigate the expression of activated mTOR (p-mTOR) in gastric cancer patients, their prognostic significance and the inhibition effect of RAD001 on tumor growth and to determine whether targeted inhibition of mTOR could be a potential therapeutic strategy for gastric cancer. Methods The expression of p-mTOR was detected in specimens of 181 gastric cancers who underwent radical resection (R0) by immunohistochemistry. The correlation of p-mTOR expression to clinicopathologic features and survival of gastric cancer was studied. We also determined the inhibition effect of RAD001 on tumor growth using BGC823 and AGS human gastric cancer cell lines. Results Immunostaining for p-mTOR was positive in 93 of 181 (51.4%) gastric cancers, closely correlated with lymph node status and pTNM stage. Patients with p-mTOR positive showed significantly shorter disease-free survival (DFS) and overall survival (OS) rates than those with p-mTOR-negative tumors in univariable analyses, and there was a trend toward a correlation between p-mTOR expression and survival in multivariable analyses. RAD001 markedly inhibited dose-dependently proliferation of human gastric carcinoma cells by down-regulating expression of p70s6k, p-p70s6k, C-myc, CyclinD1 and Bcl-2, up-regulating expression of P53. Conclusions In gastric cancer, p-mTOR is a potential therapeutic target and RAD001 was a promising treatment agent with inducing cell cycle arrest and apoptosis by down-regulating expression of C-myc, CyclinD1 and Bcl-2, up-regulating expression of P53.
Collapse
Affiliation(s)
- Da-zhi Xu
- State Key Laboratory of Oncology in South China, Guangzhou 510060, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|