1
|
Torres-Llanos Y, Zabaleta J, Cruz-Rodriguez N, Quijano S, Guzmán PC, de los Reyes I, Poveda-Garavito N, Infante A, Lopez-Kleine L, Combita AL. MIR4435-2HG as a possible novel predictive biomarker of chemotherapy response and death in pediatric B-cell ALL. Front Mol Biosci 2024; 11:1385140. [PMID: 38745909 PMCID: PMC11091394 DOI: 10.3389/fmolb.2024.1385140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 02/28/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction: Although B-cell acute lymphoblastic leukemia (B-cell ALL) survival rates have improved in recent years, Hispanic children continue to have poorer survival rates. There are few tools available to identify at the time of diagnosis whether the patient will respond to induction therapy. Our goal was to identify predictive biomarkers of treatment response, which could also serve as prognostic biomarkers of death, by identifying methylated and differentially expressed genes between patients with positive minimal residual disease (MRD+) and negative minimal residual disease (MRD-). Methods: DNA and RNA were extracted from tumor blasts separated by immunomagnetic columns. Illumina MethlationEPIC and mRNA sequencing assays were performed on 13 bone marrows from Hispanic children with B-cell ALL. Partek Flow was used for transcript mapping and quantification, followed by differential expression analysis using DEseq2. DNA methylation analyses were performed with Partek Genomic Suite and Genome Studio. Gene expression and differential methylation were compared between patients with MRD-/- and MRD+/+ at the end of induction chemotherapy. Overexpressed and hypomethylated genes were selected and validated by RT-qPCR in samples of an independent validation cohort. The predictive ability of the genes was assessed by logistic regression. Survival and Cox regression analyses were performed to determine the association of genes with death. Results: DAPK1, BOC, CNKSR3, MIR4435-2HG, CTHRC1, NPDC1, SLC45A3, ITGA6, and ASCL2 were overexpressed and hypomethylated in MRD+/+ patients. Overexpression was also validated by RT-qPCR. DAPK1, BOC, ASCL2, and CNKSR3 can predict refractoriness, but MIR4435-2HG is the best predictor. Additionally, higher expression of MIR4435-2HG increases the probability of non-response, death, and the risk of death. Finally, MIR4435-2HG overexpression, together with MRD+, are associated with poorer survival, and together with overexpression of DAPK1 and ASCL2, it could improve the risk classification of patients with normal karyotype. Conclusion: MIR4435-2HG is a potential predictive biomarker of treatment response and death in children with B-cell ALL.
Collapse
Affiliation(s)
| | - Jovanny Zabaleta
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | | | - Sandra Quijano
- Department of Microbiology, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | | | | | - Ana Infante
- Department of Pediatrics, Hospital Universitario San Ignacio, Bogotá, Colombia
| | | | - Alba Lucía Combita
- Cancer Biology Group, Instituto Nacional de Cancerología, Bogotá, Colombia
- Department of Microbiology, School of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
2
|
Sun X, Kato H, Sato H, Han X, Hirofuji Y, Kato TA, Sakai Y, Ohga S, Fukumoto S, Masuda K. Dopamine-related oxidative stress and mitochondrial dysfunction in dopaminergic neurons differentiated from deciduous teeth-derived stem cells of children with Down syndrome. FASEB Bioadv 2022; 4:454-467. [PMID: 35812076 PMCID: PMC9254221 DOI: 10.1096/fba.2021-00086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 02/17/2022] [Accepted: 03/14/2022] [Indexed: 12/24/2022] Open
Abstract
Down syndrome (DS) is one of the common genetic disorders caused by the trisomy of human chromosome 21 (HSA21). Mitochondrial dysfunction and redox imbalance play important roles in DS pathology, and altered dopaminergic regulation has been demonstrated in the brain of individuals with DS. However, the pathological association of these elements is not yet fully understood. In this study, we analyzed dopaminergic neurons (DNs) differentiated from deciduous teeth-derived stem cells of children with DS or healthy control children. As previously observed in the analysis of a single case of DS, compared to controls, patient-derived DNs (DS-DNs) displayed shorter neurite outgrowth and fewer branches, as well as downregulated vesicular monoamine transporter 2 and upregulated dopamine transporter 1, both of which are key regulators of dopamine homeostasis in DNs. In agreement with these expression profiles, DS-DNs accumulated dopamine intracellularly and had increased levels of cellular and mitochondrial reactive oxygen species (ROS). DS-DNs showed downregulation of non-canonical Notch ligand, delta-like 1, which may contribute to dopamine accumulation and increased ROS levels through DAT1 upregulation. Furthermore, DS-DNs showed mitochondrial dysfunction in consistent with lower expression of peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) and upregulation of a HSA21-encoded negative regulator of PGC-1α, nuclear receptor-interacting protein 1. These results suggest that dysregulated dopamine homeostasis may participate in oxidative stress and mitochondrial dysfunction of the dopaminergic system in DS.
Collapse
Affiliation(s)
- Xiao Sun
- Section of Oral Medicine for Children, Division of Oral Health, Growth and DevelopmentFaculty of Dental Science, Kyushu UniversityFukuokaJapan
| | - Hiroki Kato
- Department of Molecular Cell Biology and Oral AnatomyKyushu University Graduate School of Dental ScienceFukuokaJapan
| | - Hiroshi Sato
- Section of Oral Medicine for Children, Division of Oral Health, Growth and DevelopmentFaculty of Dental Science, Kyushu UniversityFukuokaJapan
| | - Xu Han
- Section of Oral Medicine for Children, Division of Oral Health, Growth and DevelopmentFaculty of Dental Science, Kyushu UniversityFukuokaJapan
| | - Yuta Hirofuji
- Section of Oral Medicine for Children, Division of Oral Health, Growth and DevelopmentFaculty of Dental Science, Kyushu UniversityFukuokaJapan
| | - Takahiro A. Kato
- Department of NeuropsychiatryGraduate School of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Satoshi Fukumoto
- Section of Oral Medicine for Children, Division of Oral Health, Growth and DevelopmentFaculty of Dental Science, Kyushu UniversityFukuokaJapan
| | - Keiji Masuda
- Section of Oral Medicine for Children, Division of Oral Health, Growth and DevelopmentFaculty of Dental Science, Kyushu UniversityFukuokaJapan
| |
Collapse
|
3
|
Heterogeneous patterns of DNA methylation-based field effects in histologically normal prostate tissue from cancer patients. Sci Rep 2017; 7:40636. [PMID: 28084441 PMCID: PMC5233981 DOI: 10.1038/srep40636] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 12/09/2016] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer (PC) diagnosis is based on histological evaluation of prostate needle biopsies, which have high false negative rates. Here, we investigated if cancer-associated epigenetic field effects in histologically normal prostate tissue may be used to increase sensitivity for PC. We focused on nine genes (AOX1, CCDC181 (C1orf114), GABRE, GAS6, HAPLN3, KLF8, MOB3B, SLC18A2, and GSTP1) known to be hypermethylated in PC. Using quantitative methylation-specific PCR, we analysed 66 malignant and 134 non-malignant tissue samples from 107 patients, who underwent ultrasound-guided prostate biopsy (67 patients had at least one cancer-positive biopsy, 40 had exclusively cancer-negative biopsies). Hypermethylation was detectable for all genes in malignant needle biopsy samples (AUC: 0.80 to 0.98), confirming previous findings in prostatectomy specimens. Furthermore, we identified a four-gene methylation signature (AOX1xGSTP1xHAPLN3xSLC18A2) that distinguished histologically non-malignant biopsies from patients with vs. without PC in other biopsies (AUC = 0.65; sensitivity = 30.8%; specificity = 100%). This signature was validated in an independent patient set (59 PC, 36 adjacent non-malignant, and 9 normal prostate tissue samples) analysed on Illumina 450 K methylation arrays (AUC = 0.70; sensitivity = 40.6%; specificity = 100%). Our results suggest that a novel four-gene signature may be used to increase sensitivity for PC diagnosis through detection of epigenetic field effects in histologically non-malignant prostate tissue samples.
Collapse
|
4
|
Yao L, Ren S, Zhang M, Du F, Zhu Y, Yu H, Zhang C, Li X, Yang C, Liu H, Wang D, Meng H, Chang S, Han X, Sun Y, Sun Y. Identification of specific DNA methylation sites on the Y-chromosome as biomarker in prostate cancer. Oncotarget 2016; 6:40611-21. [PMID: 26485765 PMCID: PMC4747356 DOI: 10.18632/oncotarget.6141] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 09/26/2015] [Indexed: 12/31/2022] Open
Abstract
As a diagnostic biomarker, prostate special antigen (PSA) tests always generate false positive results and lead to unnecessary and/or repeat biopsies. Therefore, there is an urgent need for developing more sensitive, specific diagnostic biomarkers. We epigenotyped methylated sites in cancer tissues and adjacent normal tissues from 66 patients. In comparison with normal adjacent tissues, we observed that there were 6 aberrant methylation sites in prostate cancer tissues on the Y-chromosome. We further performed pyrosequencing using urine of PCa patients and we identified one methylated site (cg05163709) as a potential biomarker. We evaluated the predictive capacity of the aberrant methylated sites using the area under receiver operating characteristic (ROC) curve (AUC). The ROC analysis showed a higher AUC for cg05163709 (0.915) than prostate-specific antigen (PSA, 0.769). These results indicated that aberrant DNA methylation of cg05163709 on the Y-chromosome could serve as a potential diagnostic biomarker with high sensitivity and specificity.
Collapse
Affiliation(s)
- Lushuai Yao
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shancheng Ren
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Minjie Zhang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Fengxia Du
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Yasheng Zhu
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hui Yu
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chenyu Zhang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Xiaohua Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Caiyun Yang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Huixian Liu
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Dong Wang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hao Meng
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shuang Chang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiao Han
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yinghao Sun
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yingli Sun
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
5
|
Haldrup C, Lynnerup AS, Storebjerg TM, Vang S, Wild P, Visakorpi T, Arsov C, Schulz WA, Lindberg J, Grönberg H, Egevad L, Borre M, Ørntoft TF, Høyer S, Sørensen KD. Large-scale evaluation of SLC18A2 in prostate cancer reveals diagnostic and prognostic biomarker potential at three molecular levels. Mol Oncol 2016; 10:825-37. [PMID: 26905753 DOI: 10.1016/j.molonc.2016.02.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/23/2015] [Accepted: 02/01/2016] [Indexed: 12/18/2022] Open
Abstract
Limitations of current diagnostic and prognostic tools for prostate cancer (PC) have led to over-diagnosis and over-treatment. Here, we investigate the biomarker potential of the SLC18A2 (VMAT2) gene for PC at three molecular levels. Thus, SLC18A2 promoter methylation was analyzed in 767 malignant and 78 benign radical prostatectomy (RP) samples using methylation-specific qPCR and Illumina 450K methylation microarray data. SLC18A2 transcript levels were assessed in 412 malignant and 45 benign RP samples using RNAseq data. SLC18A2 protein was evaluated by immunohistochemistry in 502 malignant and 305 benign RP samples. Cancer-specificity of molecular changes was tested using Mann-Whitney U tests and/or receiver operating characteristic (ROC) analyses. Log rank, uni- and multivariate Cox regression tests were used for survival analyses. We found that SLC18A2 promoter hypermethylation was highly cancer-specific (area under the curve (AUC): 0.923-0.976) and associated with biochemical recurrence (BCR) after RP in univariate analyses. SLC18A2 transcript levels were reduced in PC and had independent prognostic value for BCR after RP (multivariate HR 0.13, P < 0.05). Likewise, SLC18A2 protein was down-regulated in PC (AUC 0.898) and had independent prognostic value for BCR (multivariate HR 0.51, P < 0.05). Reduced SLC18A2 protein expression was also associated with poor overall survival in univariate analysis (HR 0.29, P < 0.05). Our results highlight SLC18A2 as a new promising methylation marker candidate for PC diagnosis. Furthermore, SLC18A2 expression (RNA and protein) showed promising prognostic potential beyond routine clinicopathological variables. Thus, novel SLC18A2-based molecular tests could have useful future applications for PC detection and identification of high-risk patients.
Collapse
Affiliation(s)
- Christa Haldrup
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Anne-Sofie Lynnerup
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Tine Maj Storebjerg
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Vang
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Wild
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Tapio Visakorpi
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, and Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Christian Arsov
- Department of Urology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Wolfgang A Schulz
- Department of Urology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Lars Egevad
- Department of Oncolocy and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Michael Borre
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Søren Høyer
- Institute of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
6
|
Expression profiling of prostate cancer tissue delineates genes associated with recurrence after prostatectomy. Sci Rep 2015; 5:16018. [PMID: 26522007 PMCID: PMC4629186 DOI: 10.1038/srep16018] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/08/2015] [Indexed: 01/21/2023] Open
Abstract
Prostate cancer is a leading cause of cancer death amongst males. The main clinical dilemma in treating prostate cancer is the high number of indolent cases that confer a significant risk of overtreatment. In this study, we have performed gene expression profiling of tumor tissue specimens from 36 patients with prostate cancer to identify transcripts that delineate aggressive and indolent cancer. Key genes were validated using previously published data and by tissue microarray analysis. Two molecular subgroups were identified with a significant overrepresentation of tumors from patients with biochemical recurrence in one of the groups. We successfully validated key transcripts association with recurrence using two publically available datasets totaling 669 patients. Twelve genes were found to be independent predictors of recurrence in multivariate logistical regression analysis. SFRP4 gene expression was consistently up regulated in patients with recurrence in all three datasets. Using an independent cohort of 536 prostate cancer patients we showed SFRP4 expression to be an independent predictor of recurrence after prostatectomy (HR = 1.35; p = 0.009). We identified SFRP4 to be associated with disease recurrence. Prospective studies are needed in order to assess the clinical usefulness of the identified key markers in this study.
Collapse
|
7
|
Prognostic DNA methylation markers for prostate cancer. Int J Mol Sci 2014; 15:16544-76. [PMID: 25238417 PMCID: PMC4200823 DOI: 10.3390/ijms150916544] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/05/2014] [Accepted: 09/11/2014] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer (PC) is the most commonly diagnosed neoplasm and the third most common cause of cancer-related death amongst men in the Western world. PC is a clinically highly heterogeneous disease, and distinction between aggressive and indolent disease is a major challenge for the management of PC. Currently, no biomarkers or prognostic tools are able to accurately predict tumor progression at the time of diagnosis. Thus, improved biomarkers for PC prognosis are urgently needed. This review focuses on the prognostic potential of DNA methylation biomarkers for PC. Epigenetic changes are hallmarks of PC and associated with malignant initiation as well as tumor progression. Moreover, DNA methylation is the most frequently studied epigenetic alteration in PC, and the prognostic potential of DNA methylation markers for PC has been demonstrated in multiple studies. The most promising methylation marker candidates identified so far include PITX2, C1orf114 (CCDC181) and the GABRE~miR-452~miR-224 locus, in addition to the three-gene signature AOX1/C1orf114/HAPLN3. Several other biomarker candidates have also been investigated, but with less stringent clinical validation and/or conflicting evidence regarding their possible prognostic value available at this time. Here, we review the current evidence for the prognostic potential of DNA methylation markers in PC.
Collapse
|
8
|
Solovieff N, Roberts AL, Ratanatharathorn A, Haloosim M, De Vivo I, King AP, Liberzon I, Aiello A, Uddin M, Wildman DE, Galea S, Smoller JW, Purcell SM, Koenen KC. Genetic association analysis of 300 genes identifies a risk haplotype in SLC18A2 for post-traumatic stress disorder in two independent samples. Neuropsychopharmacology 2014; 39:1872-9. [PMID: 24525708 PMCID: PMC4059895 DOI: 10.1038/npp.2014.34] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 01/09/2014] [Accepted: 01/10/2014] [Indexed: 12/31/2022]
Abstract
The genetic architecture of post-traumatic stress disorder (PTSD) remains poorly understood with the vast majority of genetic association studies reporting on single candidate genes. We conducted a large genetic study in trauma-exposed European-American women (N=2538; 845 PTSD cases, 1693 controls) by testing 3742 SNPs across more than 300 genes and conducting polygenic analyses using results from the Psychiatric Genome-Wide Association Studies Consortium (PGC). We tested the association between each SNP and two measures of PTSD, a severity score and diagnosis. We found a significant association between PTSD (diagnosis) and SNPs (top SNP: rs363276, odds ratio (OR)=1.4, p=2.1E-05) in SLC18A2 (vesicular monoamine transporter 2). A haplotype analysis of 9 SNPs in SLC18A2, including rs363276, identified a risk haplotype (CGGCGGAAG, p=0.0046), and the same risk haplotype was associated with PTSD in an independent cohort of trauma-exposed African-Americans (p=0.049; N=748, men and women). SLC18A2 is involved in transporting monoamines to synaptic vesicles and has been implicated in a number of neuropsychiatric disorders including major depression. Eight genes previously associated with PTSD had SNPs with nominally significant associations (p<0.05). The polygenic analyses suggested that there are SNPs in common between PTSD severity and bipolar disorder. Our data are consistent with a genetic architecture for PTSD that is highly polygenic, influenced by numerous SNPs with weak effects, and may overlap with mood disorders. Genome-wide studies with very large samples sizes are needed to detect these types of effects.
Collapse
Affiliation(s)
- Nadia Solovieff
- Department of Center of Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Stanley Center for Psychiatric Research, the Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Andrea L Roberts
- Department of Society, Human Development and Health, Harvard School of Public Health, Boston, MA, USA
| | - Andrew Ratanatharathorn
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Michelle Haloosim
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Immaculata De Vivo
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, BWH, Harvard Medical School, Boston, MA, USA
| | - Anthony P King
- Mental Health Service, VA Ann Arbor Health Systems, MI, USA
| | - Israel Liberzon
- Mental Health Service, VA Ann Arbor Health Systems, MI, USA
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Allison Aiello
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Monica Uddin
- Center for Molecular Medicine and Genetics, Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Derek E Wildman
- Center for Molecular Medicine and Genetics, Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Sandro Galea
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Jordan W Smoller
- Department of Center of Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Stanley Center for Psychiatric Research, the Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Shaun M Purcell
- Department of Center of Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Stanley Center for Psychiatric Research, the Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Division of Psychiatric Genomics, Mount Sinai School of Medicine, New York, NY, USA
| | - Karestan C Koenen
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
9
|
Van Liefferinge J, Massie A, Portelli J, Di Giovanni G, Smolders I. Are vesicular neurotransmitter transporters potential treatment targets for temporal lobe epilepsy? Front Cell Neurosci 2013; 7:139. [PMID: 24009559 PMCID: PMC3757300 DOI: 10.3389/fncel.2013.00139] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 08/11/2013] [Indexed: 12/18/2022] Open
Abstract
The vesicular neurotransmitter transporters (VNTs) are small proteins responsible for packing synaptic vesicles with neurotransmitters thereby determining the amount of neurotransmitter released per vesicle through fusion in both neurons and glial cells. Each transporter subtype was classically seen as a specific neuronal marker of the respective nerve cells containing that particular neurotransmitter or structurally related neurotransmitters. More recently, however, it has become apparent that common neurotransmitters can also act as co-transmitters, adding complexity to neurotransmitter release and suggesting intriguing roles for VNTs therein. We will first describe the current knowledge on vesicular glutamate transporters (VGLUT1/2/3), the vesicular excitatory amino acid transporter (VEAT), the vesicular nucleotide transporter (VNUT), vesicular monoamine transporters (VMAT1/2), the vesicular acetylcholine transporter (VAChT) and the vesicular γ-aminobutyric acid (GABA) transporter (VGAT) in the brain. We will focus on evidence regarding transgenic mice with disruptions in VNTs in different models of seizures and epilepsy. We will also describe the known alterations and reorganizations in the expression levels of these VNTs in rodent models for temporal lobe epilepsy (TLE) and in human tissue resected for epilepsy surgery. Finally, we will discuss perspectives on opportunities and challenges for VNTs as targets for possible future epilepsy therapies.
Collapse
|
10
|
Haldrup C, Mundbjerg K, Vestergaard EM, Lamy P, Wild P, Schulz WA, Arsov C, Visakorpi T, Borre M, Høyer S, Orntoft TF, Sørensen KD. DNA methylation signatures for prediction of biochemical recurrence after radical prostatectomy of clinically localized prostate cancer. J Clin Oncol 2013; 31:3250-8. [PMID: 23918943 DOI: 10.1200/jco.2012.47.1847] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Diagnostic and prognostic tools for prostate cancer (PC) are suboptimal, causing overtreatment of indolent PC and risk of delayed treatment of aggressive PC. Here, we identify six novel candidate DNA methylation markers for PC with promising diagnostic and prognostic potential. METHODS Microarray-based screening and bisulfite sequencing of 20 nonmalignant and 29 PC tissue specimens were used to identify new candidate DNA hypermethylation markers for PC. Diagnostic and prognostic potential was evaluated in 35 nonmalignant prostate tissue samples, 293 radical prostatectomy (RP) samples (cohort 1, training), and 114 malignant RP samples (cohort 2, validation) collected in Denmark, Switzerland, Germany, and Finland. Sensitivity and specificity for PC were evaluated by receiver operating characteristic analyses. Correlations between DNA methylation levels and biochemical recurrence were assessed using log-rank tests and univariate and multivariate Cox regression analyses. RESULTS Hypermethylation of AOX1, C1orf114, GAS6, HAPLN3, KLF8, and MOB3B was highly cancer specific (area under the curve, 0.89 to 0.98). Furthermore, high C1orf114 methylation was significantly (P < .05) associated with biochemical recurrence in multivariate analysis in cohort 1 (hazard ratio [HR], 3.10; 95% CI, 1.89 to 5.09) and was successfully validated in cohort 2 (HR, 3.27; 95% CI, 1.17 to 9.12). Moreover, a significant (P < .05) three-gene prognostic methylation signature (AOX1/C1orf114/HAPLN3), classifying patients into low- and high-methylation subgroups, was trained in cohort 1 (HR, 1.91; 95% CI, 1.26 to 2.90) and validated in cohort 2 (HR, 2.33; 95% CI, 1.31 to 4.13). CONCLUSION We identified six novel candidate DNA methylation markers for PC. C1orf114 hypermethylation and a three-gene methylation signature were independent predictors of time to biochemical recurrence after RP in two PC patient cohorts.
Collapse
Affiliation(s)
- Christa Haldrup
- Christa Haldrup, Kamilla Mundbjerg, Else Marie Vestergaard, Philippe Lamy, Michael Borre, Søren Høyer, Torben F. Ørntoft, and Karina D. Sørensen, Aarhus University Hospital, Aarhus, Denmark; Peter Wild, University Hospital Zurich, Zurich, Switzerland; Wolfgang A. Schulz and Christian Arsov, Heinrich Heine University, Düsseldorf, Germany; and Tapio Visakorpi, University of Tampere and Tampere University Hospital, Tampere, Finland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Chen J, Zhang D, Yan W, Yang D, Shen B. Translational bioinformatics for diagnostic and prognostic prediction of prostate cancer in the next-generation sequencing era. BIOMED RESEARCH INTERNATIONAL 2013; 2013:901578. [PMID: 23957008 PMCID: PMC3727129 DOI: 10.1155/2013/901578] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 06/22/2013] [Indexed: 01/13/2023]
Abstract
The discovery of prostate cancer biomarkers has been boosted by the advent of next-generation sequencing (NGS) technologies. Nevertheless, many challenges still exist in exploiting the flood of sequence data and translating them into routine diagnostics and prognosis of prostate cancer. Here we review the recent developments in prostate cancer biomarkers by high throughput sequencing technologies. We highlight some fundamental issues of translational bioinformatics and the potential use of cloud computing in NGS data processing for the improvement of prostate cancer treatment.
Collapse
Affiliation(s)
- Jiajia Chen
- Center for Systems Biology, Soochow University, Suzhou 215006, China
- School of Chemistry, Biology and Material Engineering, Suzhou University of Science and Technology, Suzhou 215011, China
| | - Daqing Zhang
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| | - Wenying Yan
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| | - Dongrong Yang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Bairong Shen
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| |
Collapse
|
12
|
Abstract
Background: Novel biomarkers for prostate cancer (PC) are urgently needed. This study investigates the expression, epigenetic regulation, and prognostic potential of ANPEP in PC. Methods: Aminopeptidase N (APN; encoded by ANPEP) expression was analysed by immunohistochemistry using tissue microarrays representing 267 radical prostatectomy (RP) and 111 conservatively treated (CT) PC patients. Clinical end points were recurrence-free survival (RFS) and cancer-specific survival (CSS), respectively. The ANPEP promoter methylation levels were determined by bisulphite sequencing or MethyLight analysis in 278 nonmalignant and PC tissue samples, and in cell lines. Results: The APN expression was significantly downregulated in PC compared with nonmalignant prostate tissue samples. Aberrant promoter hypermethylation was frequently observed in PC tissue samples, and 5-aza-2′-deoxycytidine induced ANPEP expression in three hypermethylated prostate cell lines, suggesting epigenetic silencing. Negative APN immunoreactivity was significantly associated with short RFS and short CSS in the RP and CT cohort, respectively, independently of routine clinicopathological predictors. Combining APN with a known angiogenesis marker (vascular endothelial growth factor or microvessel density) improved risk prediction significantly in both cohorts. Conclusion: Our results suggest negative APN immunoreactivity as a new independent adverse prognostic factor for patients with clinically localised PC and, furthermore, that epigenetic mechanisms are involved in silencing of ANPEP in PC.
Collapse
|
13
|
Majumdar S, Buckles E, Estrada J, Koochekpour S. Aberrant DNA methylation and prostate cancer. Curr Genomics 2012; 12:486-505. [PMID: 22547956 PMCID: PMC3219844 DOI: 10.2174/138920211797904061] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2011] [Revised: 08/15/2011] [Accepted: 09/05/2011] [Indexed: 12/28/2022] Open
Abstract
Prostate cancer (PCa) is the most prevalent cancer, a significant contributor to morbidity and a leading cause of cancer-related death in men in Western industrialized countries. In contrast to genetic changes that vary among individual cases, somatic epigenetic alterations are early and highly consistent events. Epigenetics encompasses several different phenomena, such as DNA methylation, histone modifications, RNA interference, and genomic imprinting. Epigenetic processes regulate gene expression and can change malignancy-associated phenotypes such as growth, migration, invasion, or angiogenesis. Methylations of certain genes are associated with PCa progression. Compared to normal prostate tissues, several hypermethylated genes have also been identified in benign prostate hyperplasia, which suggests a role for aberrant methylation in this growth dysfunction. Global and gene-specific DNA methylation could be affected by environmental and dietary factors. Among other epigenetic changes, aberrant DNA methylation might have a great potential as diagnostic or prognostic marker for PCa and could be tested in tumor tissues and various body fluids (e.g., serum, urine). The DNA methylation markers are simple in nature, have high sensitivity, and could be detected either quantitatively or qualitatively. Availability of genome-wide screening methodologies also allows the identification of epigenetic signatures in high throughput population studies. Unlike irreversible genetic changes, epigenetic alterations are reversible and could be used for PCa targeted therapies.
Collapse
Affiliation(s)
- Sunipa Majumdar
- Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70122, USA
| | | | | | | |
Collapse
|
14
|
Lee C, Zhang Q, Zi X, Dash A, Soares MB, Rahmatpanah F, Jia Z, McClelland M, Mercola D. TGF-β mediated DNA methylation in prostate cancer. Transl Androl Urol 2012; 1:78-88. [PMID: 25133096 PMCID: PMC4131550 DOI: 10.3978/j.issn.2223-4683.2012.05.06] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 05/04/2012] [Indexed: 12/15/2022] Open
Abstract
Almost all tumors harbor a defective negative feedback loop of signaling by transforming growth factor-β (TGF-β). Epigenetic mechanisms of gene regulation, including DNA methylation, are fundamental to normal cellular function and also play a major role in carcinogenesis. Recent evidence demonstrated that TGF-β signaling mediates cancer development and progression. Many key events in TGF-β signaling in cancer included auto-induction of TGF-β1 and increased expression of DNA methyltransferases (DNMTs), suggesting that DNA methylation plays a significant role in cancer development and progression. In this review, we performed an extensive survey of the literature linking TGF-β signaling to DNA methylation in prostate cancer. It appeared that almost all DNA methylated genes detected in prostate cancer are directly or indirectly related to TGF-β signaling. This knowledge has provided a basis for our future directions of prostate cancer research and strategies for prevention and therapy for prostate cancer.
Collapse
|
15
|
Abstract
Epigenetic alterations contribute significantly to the development and progression of prostate cancer, the most prevalent malignant tumor in males of Western industrialized countries. Here, we review recent research on DNA methylation alterations in this cancer type. Hypermethylation of several genes including GSTP1 is well known to occur in a consistent and apparently coordinate fashion during the transition from intraepithelial neoplasia to frank carcinoma. These hypermethylation events have shown promise as biomarkers for detection of prostate carcinoma. Many other individual genes have been shown to undergo hypermethylation, which is typically associated with diminished expression. These investigations indicate additional candidates for biomarkers; in particular, hypermethylation events associated with progression can be employed to identify more aggressive cases. In addition, some of genes silenced by aberrant methylation in prostate have been shown to exhibit properties of tumor suppressors, revealing insights into mechanisms of carcinogenesis. Whereas most studies in the past have used candidate gene approaches, new techniques allowing genome-wide screening for altered methylation are increasingly employed in prostate cancer research and have already yielded encouraging results.
Collapse
Affiliation(s)
- Wolfgang Goering
- Department of Urology, Heinrich Heine University, Duesseldorf, Germany
| | | | | |
Collapse
|
16
|
Yang M, Park JY. DNA methylation in promoter region as biomarkers in prostate cancer. Methods Mol Biol 2012; 863:67-109. [PMID: 22359288 DOI: 10.1007/978-1-61779-612-8_5] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The prostate gland is the most common site of cancer and the second leading cause of cancer death in American men. Recent emerging molecular biological technologies help us to know that epigenetic alterations such as DNA methylation within the regulatory (promoter) regions of genes are associated with transcriptional silencing in cancer. Promoter hypermethylation of critical pathway genes could be potential biomarkers and therapeutic targets for prostate cancer. In this chapter, we updated current information on methylated genes associated with the development and progression of prostate cancer. Over 40 genes have been investigated for methylation in promoter region in prostate cancer. These methylated genes are involved in critical pathways, such as DNA repair, metabolism, and invasion/metastasis. The role of hypermethylated genes in regulation of critical pathways in prostate cancer is discussed. These findings may provide new information of the pathogenesis, the exciting potential to be predictive and to provide personalized treatment of prostate cancer. Indeed, some epigenetic alterations in prostate tumors are being translated into clinical practice for therapeutic use.
Collapse
Affiliation(s)
- Mihi Yang
- Division of Cancer Prevention and Controls, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | |
Collapse
|
17
|
Abildgaard MO, Borre M, Mortensen MM, Ulhøi BP, Tørring N, Wild P, Kristensen H, Mansilla F, Ottosen PD, Dyrskjøt L, Ørntoft TF, Sørensen KD. Downregulation of zinc finger protein 132 in prostate cancer is associated with aberrant promoter hypermethylation and poor prognosis. Int J Cancer 2011; 130:885-95. [PMID: 21445975 DOI: 10.1002/ijc.26097] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Accepted: 03/11/2011] [Indexed: 12/31/2022]
Abstract
This study investigates the expression and biomarker potential of zinc finger protein 132 (ZNF132) in prostate cancer (PC) by transcriptional profiling and immunohistochemical analysis of tissue microarrays, including tumor specimens from 615 radical prostatectomy (RP) patients and 199 conservatively treated patients. Primary clinical endpoints were time to PSA recurrence and cancer-specific death, respectively. Compared to normal prostate epithelial cells from men without PC, ZNF132 transcript levels were significantly reduced in PC cells from patients with localized PC and further downregulated in metastatic PC. Likewise, ZNF132 protein expression was significantly lower in primary tumors from patients with metastatic compared to localized PC and further reduced in castrate-refractory PC, indicating that ZNF132 downregulation correlates with disease progression. Reduced ZNF132 immunoreactivity was significantly associated with high Gleason score and advanced T stage in both PC patient cohorts. By univariate analysis, no/weak ZNF132 staining was a significant adverse predictor of PSA recurrence after RP (p = 0.024) and cancer-specific death following conservative treatment (p = 0.009). In multivariate models, however, ZNF132 did not add significant independent value to established prognostic factors. Finally, bisulfite sequencing revealed frequent promoter hypermethylation of ZNF132 in both PC cell lines and PC tissue samples, indicating that ZNF132 is epigenetically silenced in PC. In summary, our results show that downregulation of ZNF132 is associated with aggressive PC and furthermore identify ZNF132 as a new candidate methylation marker for PC.
Collapse
|
18
|
Bauerschlag DO, Ammerpohl O, Bräutigam K, Schem C, Lin Q, Weigel MT, Hilpert F, Arnold N, Maass N, Meinhold-Heerlein I, Wagner W. Progression-free survival in ovarian cancer is reflected in epigenetic DNA methylation profiles. Oncology 2011; 80:12-20. [PMID: 21577013 DOI: 10.1159/000327746] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Accepted: 02/18/2011] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Many patients with ovarian cancer disease relapse within 6 months after adjuvant chemotherapy, with a limited prognosis. Epigenetic modifications have been shown to play an important role in tumor development and formation. Therefore, global analysis of DNA methylation patterns might reveal specific CpG sites that correlate with progression-free interval (PFI) after therapy. METHODS Twenty samples of advanced ovarian cancer with a predominantly serous papillary histological subtype were subjected to DNA methylation profiling. Illumina HumanMethylation27 BeadChip technology was used for simultaneous analysis of 27,578 CpG sites in >14,000 genes. RESULTS Differential DNA methylation of various cytosines correlated with PFI. However, this becomes only significant by classification according to PFI with a cutoff of >28 months. Longer survival was associated with hypomethylation at specific CpG sites (e.g. GREB1, TGIF and TOB1) and hypermethylation in other genes (e.g. TMCO5, PTPRN and GUCY2C). Gene ontology analysis revealed that differentially methylated genes were significantly overrepresented in the categories telomere organization, mesoderm development and immune regulation. CONCLUSION Epigenetic modifications at specific CpG sites correlate with PFI in ovarian cancer. Therefore, such analysis might be of prognostic value.
Collapse
Affiliation(s)
- Dirk O Bauerschlag
- Department of Gynecology, University Medical Center Aachen, RWTH, Aachen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
BACKGROUND The prostate gland is the most common site of cancer and the second leading cause of cancer mortality in American men. It is well known that epigenetic alterations such as DNA methylation within the regulatory (promoter) regions of genes are associated with transcriptional silencing in cancer. Promoter hypermethylation of critical pathway genes could be potential biomarkers and therapeutic targets for prostate cancer. METHODS This review discusses current information on methylated genes associated with prostate cancer development and progression. RESULTS Over 30 genes have been investigated for promoter methylation in prostate cancer. These methylated genes are involved in critical pathways, such as DNA repair, metabolism, and invasion/metastasis. The role of hypermethylated genes in regulation of critical pathways in prostate cancer is reviewed. CONCLUSIONS These findings may provide new information of the pathogenesis of prostate cancer. Certain epigenetic alterations in prostate tumors are being translated into clinical practice for therapeutic use.
Collapse
Affiliation(s)
- Jong Y Park
- Division of Cancer Prevention and Control, Moffitt Cancer Center, Tampa, FL 33612, USA.
| |
Collapse
|
20
|
Yano K. Gene expression correlation analysis predicts involvement of high- and low-confidence risk genes in different stages of prostate carcinogenesis. Prostate 2010; 70:1746-59. [PMID: 20564324 DOI: 10.1002/pros.21210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Whole genome association studies have identified many loci associated with the risk of prostate cancer (PC). However, very few of the genes associated with these loci have been related to specific processes of prostate carcinogenesis. Therefore I inferred biological functions associated with these risk genes using gene expression correlation analysis. METHODS PC risk genes reported in the literature were classified as having high (P<10(-6)), medium (P<10(-4)), or low (P<10(-2)) statistical confidence. Correlation coefficients of the expression levels between the risk genes and other genes in cancerous prostates samples were compared against those in normal prostates using a microarray dataset from Gene Expression Omnibus. RESULTS Overall, significant decrease of correlations in PC was observed between the levels of expression of the high-confidence genes and other genes in the microarray dataset, whereas correlation between low-confidence genes and other genes in PC showed smaller decrease. Genes involved in developmental processes were significantly correlated with all risk gene categories. Ectoderm development genes, which may be related to squamous metaplasia, and genes enriched in fetal prostate stem cells (PSCs) showed strong association with the high-confidence genes. The association between the PSC genes and the low-confidence genes was weak, but genes related to neural system genes showed strong association with low-confidence genes. CONCLUSIONS The high-confidence risk genes may be associated with an early stage of prostate carcinogenesis, possibly involving PSCs and squamous metaplasia. The low-confidence genes may be involved in a later stage of carcinogenesis.
Collapse
Affiliation(s)
- Kojiro Yano
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
21
|
Vestergaard EM, Nexø E, Tørring N, Borre M, Ørntoft TF, Sørensen KD. Promoter hypomethylation and upregulation of trefoil factors in prostate cancer. Int J Cancer 2010; 127:1857-65. [PMID: 20112343 DOI: 10.1002/ijc.25209] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Trefoil factors, mucin-associated peptides, are overexpressed in prostate cancer (PC). We hypothesized that promoter methylation contributes to the regulation of trefoil factors (TFF1, TFF2 and TFF3) in human prostate cells. Here we show hypomethylation of promoter regions of TFF1 and TFF3 in PC cell lines with significant TFF expression as compared to benign immortalized prostate cell lines and PC cell lines not expressing trefoil factor. The most striking difference was observed for CpG sites located close to the AUG start codon overlapping several putative binding sites for cellular transcription factors. TFF2 was hypermethylated and had no or very low expression in all prostate cell lines investigated. Treatment of methylated cell lines with 5-aza-2'-deoxycytidine restored TFF expression in cell lines not expressing TFF and increased expression significantly in low-expressing cell lines. In clinical samples, methylation of the promoter/enhancer regions of TFF1 and TFF3 was significantly lower in PC compared to benign prostatic hyperplasia. The present study shows an inverse relation between promoter methylation and expression of trefoil factors. Preliminary analysis on clinical samples suggests that this regulatory mechanism is responsible for the increased levels of TFF1 and TFF3 observed in PC. The overexpression and promoter hypomethylation of trefoil factors may serve as biomarkers in PC.
Collapse
Affiliation(s)
- Else Marie Vestergaard
- Department of Clinical Biochemistry, Aarhus University Hospital Skejby, Brendstrupgaardsvej, Aarhus N, Denmark.
| | | | | | | | | | | |
Collapse
|
22
|
Sørensen KD, Ørntoft TF. Discovery of prostate cancer biomarkers by microarray gene expression profiling. Expert Rev Mol Diagn 2010; 10:49-64. [PMID: 20014922 DOI: 10.1586/erm.09.74] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Prostate cancer is the most commonly diagnosed malignancy in males in the Western world. This review focuses on advances in biomarker discovery for prostate cancer by microarray profiling of mRNA and microRNA expression. Novel biomarkers are strongly needed to enable more accurate detection of prostate cancer, improve prediction of tumor aggressiveness and facilitate discovery of new therapeutic targets for tailored medicine. Promising molecular markers identified from gene expression profiling studies include AMACR, EZH2, TMPRSS2-ERG, miR-221 and miR-141, which are described in more detail. In addition, a compilation of prognostic gene expression signatures for prediction of prostate cancer patient outcome is provided, and their possible clinical utility is discussed. Furthermore, limitations in the application of microarray-based expression profiling for identification of prostate cancer biomarkers are addressed.
Collapse
Affiliation(s)
- Karina Dalsgaard Sørensen
- Department of Molecular Medicine, Aarhus University Hospital, Skejby, Brendstrupgaardsvej 100, DK-8200 Aarhus N, Denmark.
| | | |
Collapse
|
23
|
Ahmed H. Promoter methylation in prostate cancer and its application for the early detection of prostate cancer using serum and urine samples. BIOMARKERS IN CANCER 2010; 2:17-33. [PMID: 24179382 PMCID: PMC2908742 DOI: 10.4137/bic.s3187] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Prostate cancer is the second most common cancer and the second leading cause of
cancer death in men. However, prostate cancer can be effectively treated and
cured, if it is diagnosed in its early stages when the tumor is still confined
to the prostate. Combined with the digital rectal examination, the PSA test has
been widely used to detect prostate cancer. But, the PSA screening method for
early detection of prostate cancer is not reliable due to the high prevalence of
false positive and false negative results. Epigenetic alterations including
hypermethylation of gene promoters are believed to be the early events in
neoplastic progression and thus these methylated genes can serve as biomarkers
for the detection of cancer from clinical specimens. This review discusses DNA
methylation of several gene promoters during prostate carcinogenesis and
evaluates the usefulness of monitoring methylated DNA sequences, such as
GSTP1, RASSF1A, RARβ2 and galectin-3, for early detection
of prostate cancer in tissue biopsies, serum and urine.
Collapse
Affiliation(s)
- Hafiz Ahmed
- Department of Biochemistry and Molecular Biology, Program in Oncology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
24
|
Deng D, Liu Z, Du Y. Epigenetic alterations as cancer diagnostic, prognostic, and predictive biomarkers. ADVANCES IN GENETICS 2010; 71:125-76. [PMID: 20933128 DOI: 10.1016/b978-0-12-380864-6.00005-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alterations of DNA methylation and transcription of microRNAs (miRNAs) are very stable phenomena in tissues and body fluids and suitable for sensitive detection. These advantages enable us to translate some important discoveries on epigenetic oncology into biomarkers for control of cancer. A few promising epigenetic biomarkers are emerging. Clinical trials using methylated CpG islands of p16, Septin9, and MGMT as biomarkers are carried out for predication of cancer development, diagnosis, and chemosensitivity. Circulating miRNAs are promising biomarkers, too. Breakthroughs in the past decade imply that epigenetic biomarkers may be useful in reducing the burden of cancer.
Collapse
Affiliation(s)
- Dajun Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry ofEducation), Peking University School of Oncology, Beijing Cancer Hospitaland Institute, Fu-Cheng-Lu, Haidian District, Beijing, 100142, PR China
| | | | | |
Collapse
|