1
|
Khalid S, Kearney M, McReynolds DE. Can social adversity alter the epigenome, trigger oral disease, and affect future generations? Ir J Med Sci 2024; 193:2597-2606. [PMID: 38740675 PMCID: PMC11450135 DOI: 10.1007/s11845-024-03697-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/27/2024] [Indexed: 05/16/2024]
Abstract
The nature versus nurture debate has intrigued scientific circles for decades. Although extensive research has established a clear relationship between genetics and disease development, recent evidence has highlighted the insufficiency of attributing adverse health outcomes to genetic factors alone. In fact, it has been suggested that environmental influences, such as socioeconomic position (SEP), may play a much larger role in the development of disease than previously thought, with extensive research suggesting that low SEP is associated with adverse health conditions. In relation to oral health, a higher prevalence of caries (tooth decay) exists among those of low SEP. Although little is known about the biological mechanisms underlying this relationship, epigenetic modifications resulting from environmental influences have been suggested to play an important role. This review explores the intersection of health inequalities and epigenetics, the role of early-life social adversity and its long-term epigenetic impacts, and how those living within the lower hierarchies of the socioeconomic pyramid are indeed at higher risk of developing diseases, particularly in relation to oral health. A deeper understanding of these mechanisms could lead to the development of targeted interventions for individuals of low SEP to improve oral health or identify those who are at higher risk of developing oral disease.
Collapse
Affiliation(s)
- Sakr Khalid
- Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland
| | - Michaela Kearney
- Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland
| | - David E McReynolds
- Dublin Dental University Hospital, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
2
|
Gretarsson KH, Abini-Agbomson S, Gloor SL, Weinberg DN, McCuiston JL, Kumary VUS, Hickman AR, Sahu V, Lee R, Xu X, Lipieta N, Flashner S, Adeleke OA, Popova IK, Taylor HF, Noll K, Windham CL, Maryanski DN, Venters BJ, Nakagawa H, Keogh MC, Armache KJ, Lu C. Cancer-associated DNA hypermethylation of Polycomb targets requires DNMT3A dual recognition of histone H2AK119 ubiquitination and the nucleosome acidic patch. SCIENCE ADVANCES 2024; 10:eadp0975. [PMID: 39196936 PMCID: PMC11352909 DOI: 10.1126/sciadv.adp0975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/24/2024] [Indexed: 08/30/2024]
Abstract
During tumor development, promoter CpG islands that are normally silenced by Polycomb repressive complexes (PRCs) become DNA-hypermethylated. The molecular mechanism by which de novo DNA methyltransferase(s) [DNMT(s)] catalyze CpG methylation at PRC-regulated regions remains unclear. Here, we report a cryo-electron microscopy structure of the DNMT3A long isoform (DNMT3A1) amino-terminal region in complex with a nucleosome carrying PRC1-mediated histone H2A lysine-119 monoubiquitination (H2AK119Ub). We identify regions within the DNMT3A1 amino terminus that bind H2AK119Ub and the nucleosome acidic patch. This bidentate interaction is required for effective DNMT3A1 engagement with H2AK119Ub-modified chromatin in cells. Further, aberrant redistribution of DNMT3A1 to Polycomb target genes recapitulates the cancer-associated DNA hypermethylation signature and inhibits their transcriptional activation during cell differentiation. This effect is rescued by disruption of the DNMT3A1-acidic patch interaction. Together, our analyses reveal a binding interface critical for mediating promoter CpG island DNA hypermethylation, a major molecular hallmark of cancer.
Collapse
Affiliation(s)
- Kristjan H. Gretarsson
- Department of Genetics and Development and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Stephen Abini-Agbomson
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | | | - Daniel N. Weinberg
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | - Varun Sahu
- Department of Genetics and Development and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rachel Lee
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Xinjing Xu
- Department of Genetics and Development and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Natalie Lipieta
- Department of Genetics and Development and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Samuel Flashner
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | - Hiroshi Nakagawa
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY 10032, USA
| | | | - Karim-Jean Armache
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Chao Lu
- Department of Genetics and Development and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
3
|
Gretarsson KH, Abini-Agbomson S, Gloor SL, Weinberg DN, McCuiston JL, Kumary VUS, Hickman AR, Sahu V, Lee R, Xu X, Lipieta N, Flashner S, Adeleke OA, Popova IK, Taylor HF, Noll K, Windham CL, Maryanski DN, Venters BJ, Nakagawa H, Keogh MC, Armache KJ, Lu C. Cancer-associated DNA Hypermethylation of Polycomb Targets Requires DNMT3A Dual Recognition of Histone H2AK119 Ubiquitination and the Nucleosome Acidic Patch. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585588. [PMID: 38562823 PMCID: PMC10983913 DOI: 10.1101/2024.03.18.585588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
During tumor development, promoter CpG islands (CGIs) that are normally silenced by Polycomb repressive complexes (PRCs) become DNA hypermethylated. The molecular mechanism by which de novo DNA methyltransferase(s) catalyze CpG methylation at PRC-regulated regions remains unclear. Here we report a cryo-EM structure of the DNMT3A long isoform (DNMT3A1) N-terminal region in complex with a nucleosome carrying PRC1-mediated histone H2A lysine 119 monoubiquitination (H2AK119Ub). We identify regions within the DNMT3A1 N-terminus that bind H2AK119Ub and the nucleosome acidic patch. This bidentate interaction is required for effective DNMT3A1 engagement with H2AK119Ub-modified chromatin in cells. Furthermore, aberrant redistribution of DNMT3A1 to Polycomb target genes inhibits their transcriptional activation during cell differentiation and recapitulates the cancer-associated DNA hypermethylation signature. This effect is rescued by disruption of the DNMT3A1-acidic patch interaction. Together, our analyses reveal a binding interface critical for countering promoter CGI DNA hypermethylation, a major molecular hallmark of cancer.
Collapse
|
4
|
Qiu M, Zhang N, Yao S, Zhou H, Chen X, Jia Y, Zhang H, Li X, Jiang Y. DNMT3A-mediated high expression of circ_0057504 promotes benzo[a]pyrene-induced DNA damage via the NONO-SFPQ complex in human bronchial epithelial cells. ENVIRONMENT INTERNATIONAL 2022; 170:107627. [PMID: 36399942 DOI: 10.1016/j.envint.2022.107627] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 06/16/2023]
Abstract
Benzo[a]pyrene (B[a]P) is a class I carcinogen and hazardous environmental pollutant with genetic toxicity. Understanding the molecular mechanisms underlying genetic deterioration and epigenetic alterations induced by environmental contaminants may contribute to the early detection and prevention of cancer. However, the role and regulatory mechanisms of circular RNAs (circRNAs) in the B[a]P-induced DNA damage response (DDR) have not been elucidated. In this study, human bronchial epithelial cell lines (16HBE and BEAS-2B) were exposed to various concentrations of B[a]P, and BALB/c mice were treated with B[a]P intranasally. B[a]P exposure was found to induce DNA damage and upregulate circular RNA hsa_circ_0057504 (circ_0057504) expression in vitro and in vivo. In addition, B[a]P upregulated TMEM194B mRNA and circ_0057504 expression through inhibition of DNA methyltransferase 3 alpha (DNMT3A) expression in vitro. Modulation (overexpression or knockdown) of circ_0057504 expression levels using a lentiviral system in human bronchial epithelial cells revealed that circ_0057504 promoted B[a]P-induced DNA damage. RNA pull-down and western blot assays showed that circ_0057504 interacted with non-POU domain-containing octamer-binding (NONO) and splicing factor proline and glutamine rich (SFPQ) proteins and regulated formation of the NONO-SFPQ protein complex. Thus, our findings indicate that circ_0057504 acts as a novel regulator of DNA damage in human bronchial epithelial cells exposed to B[a]P. The current study reveals novel insights into the role of circRNAs in the regulation of genetic damage, and describes the effect and regulatory mechanisms of circ_0057504 on B[a]P genotoxicity.
Collapse
Affiliation(s)
- Miaoyun Qiu
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Nan Zhang
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Shuwei Yao
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Hanyu Zhou
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Xintong Chen
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Yangyang Jia
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Han Zhang
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Xin Li
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China
| | - Yiguo Jiang
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
5
|
Pietrasz D, Sereni E, Lancelotti F, Pea A, Luchini C, Innamorati G, Salvia R, Bassi C. Circulating tumour DNA: a challenging innovation to develop "precision onco-surgery" in pancreatic adenocarcinoma. Br J Cancer 2022; 126:1676-1683. [PMID: 35197581 PMCID: PMC9174156 DOI: 10.1038/s41416-022-01745-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 12/13/2021] [Accepted: 02/04/2022] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is predicted to become the third leading cause of cancer-related mortality within the next decade. Management of PDAC remains challenging with limited effective treatment options and a dismal long-term prognosis. Liquid biopsy and circulating biomarkers seem to be promising to improve the multidisciplinary approach in PDAC treatment. Circulating tumour DNA (ctDNA) is the most studied blood liquid biopsy analyte and can provide insight into the molecular profile and individual characteristics of the tumour in real-time and in advance of standard imaging modalities. This could pave the way for identifying new therapeutic targets and markers of tumour response to supplement diagnostic and provide enhanced stratified treatment. Although its specificity seems excellent, the current sensitivity of ctDNA remains a limitation for clinical use, especially in patients with a low tumour burden. Increasing evidence suggests that ctDNA is a pertinent candidate biomarker to assess minimal residual disease after surgery but also a strong independent prognostic biomarker. This review explores the current knowledge and recent developments in ctDNA as a screening, diagnostic, prognostic and predictive biomarker in the management of resectable PDAC but also technical and analytical challenges that must be overcome to move toward "precision onco-surgery."
Collapse
Affiliation(s)
- Daniel Pietrasz
- APHP Hôpital Paul-Brousse, Centre Hépato-Biliaire, Université Paris-Saclay, 94800, Villejuif, France.
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy.
| | - Elisabetta Sereni
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Francesco Lancelotti
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Antonio Pea
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Giulio Innamorati
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Roberto Salvia
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Claudio Bassi
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy
| |
Collapse
|
6
|
Fu J, Zhang L, Li D, Tian T, Wang X, Sun H, Ge A, Liu Y, Zhang X, Huang H, Meng S, Zhang D, Zhao L, Sun S, Zheng T, Jia C, Zhao Y, Pang D. DNA Methylation of Imprinted Genes KCNQ1, KCNQ1OT1, and PHLDA2 in Peripheral Blood Is Associated with the Risk of Breast Cancer. Cancers (Basel) 2022; 14:cancers14112652. [PMID: 35681632 PMCID: PMC9179312 DOI: 10.3390/cancers14112652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 02/01/2023] Open
Abstract
Methylation alterations of imprinted genes lead to loss of imprinting (LOI). Although studies have explored the mechanism of LOI in breast cancer (BC) development, the association between imprinted gene methylation in peripheral blood and BC risk is largely unknown. We utilized HumanMethylation450 data from TCGA and GEO (n = 1461) to identify the CpG sites of imprinted genes associated with BC risk. Furthermore, we conducted an independent case-control study (n = 1048) to validate DNA methylation of these CpG sites in peripheral blood and BC susceptibility. cg26709929, cg08446215, cg25306939, and cg16057921, which are located at KCNQ1, KCNQ1OT1, and PHLDA2, were discovered to be associated with BC risk. Subsequently, the association between cg26709929, cg26057921, and cg25306939 methylation and BC risk was validated in our inhouse dataset. All 22 CpG sites in the KCNQ1OT1 region were associated with BC risk. Individuals with a hypermethylated KCNQ1OT1 region (>0.474) had a lower BC risk (OR: 0.553, 95% CI: 0.397−0.769). Additionally, the methylation of the KCNQ1OT1 region was not significantly different among B cells, monocytes, and T cells, which was also observed at CpG sites in PHLDA2. In summary, the methylation of KCNQ1, KCNQ1OT1, and PHLDA2 was associated with BC risk, and KCNQ1OT1 methylation could be a potential biomarker for BC risk assessment.
Collapse
Affiliation(s)
- Jinming Fu
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Lei Zhang
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Dapeng Li
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Tian Tian
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Xuan Wang
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Hongru Sun
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Anqi Ge
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Yupeng Liu
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Xianyu Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China;
| | - Hao Huang
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Shuhan Meng
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Ding Zhang
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Liyuan Zhao
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Simin Sun
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Ting Zheng
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Chenyang Jia
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
| | - Yashuang Zhao
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin 150081, China; (J.F.); (L.Z.); (D.L.); (T.T.); (X.W.); (H.S.); (A.G.); (Y.L.); (H.H.); (S.M.); (D.Z.); (L.Z.); (S.S.); (T.Z.); (C.J.)
- Correspondence: (Y.Z.); (D.P.); Tel.: +86-451-8750-2823 (Y.Z.); +86-451-8750-2885 (D.P.)
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China;
- Correspondence: (Y.Z.); (D.P.); Tel.: +86-451-8750-2823 (Y.Z.); +86-451-8750-2885 (D.P.)
| |
Collapse
|
7
|
Skaar DA, Dietze EC, Alva-Ornelas JA, Ann D, Schones DE, Hyslop T, Sistrunk C, Zalles C, Ambrose A, Kennedy K, Idassi O, Miranda Carboni G, Gould MN, Jirtle RL, Seewaldt VL. Epigenetic Dysregulation of KCNK9 Imprinting and Triple-Negative Breast Cancer. Cancers (Basel) 2021; 13:6031. [PMID: 34885139 PMCID: PMC8656495 DOI: 10.3390/cancers13236031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 11/26/2021] [Indexed: 12/02/2022] Open
Abstract
Genomic imprinting is an inherited form of parent-of-origin specific epigenetic gene regulation that is dysregulated by poor prenatal nutrition and environmental toxins. KCNK9 encodes for TASK3, a pH-regulated potassium channel membrane protein that is overexpressed in 40% of breast cancer. However, KCNK9 gene amplification accounts for increased expression in <10% of these breast cancers. Here, we showed that KCNK9 is imprinted in breast tissue and identified a differentially methylated region (DMR) controlling its imprint status. Hypomethylation at the DMR, coupled with biallelic expression of KCNK9, occurred in 63% of triple-negative breast cancers (TNBC). The association between hypomethylation and TNBC status was highly significant in African-Americans (p = 0.006), but not in Caucasians (p = 0.70). KCNK9 hypomethylation was also found in non-cancerous tissue from 77% of women at high-risk of developing breast cancer. Functional studies demonstrated that the KCNK9 gene product, TASK3, regulates mitochondrial membrane potential and apoptosis-sensitivity. In TNBC cells and non-cancerous mammary epithelial cells from high-risk women, hypomethylation of the KCNK9 DMR predicts for increased TASK3 expression and mitochondrial membrane potential (p < 0.001). This is the first identification of the KCNK9 DMR in mammary epithelial cells and demonstration that its hypomethylation in breast cancer is associated with increases in both mitochondrial membrane potential and apoptosis resistance. The high frequency of hypomethylation of the KCNK9 DMR in TNBC and non-cancerous breast tissue from high-risk women provides evidence that hypomethylation of the KNCK9 DMR/TASK3 overexpression may serve as a marker of risk and a target for prevention of TNBC, particularly in African American women.
Collapse
Affiliation(s)
- David A. Skaar
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA;
| | - Eric C. Dietze
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Jackelyn A. Alva-Ornelas
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - David Ann
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Dustin E. Schones
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Terry Hyslop
- Department of Biostatistics, School of Medicine, Duke University, Durham, NC 27710, USA;
| | - Christopher Sistrunk
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Carola Zalles
- Department of Pathology, Mercy Hospital, Miami, FL 33133, USA;
| | - Adrian Ambrose
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Kendall Kennedy
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Ombeni Idassi
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| | - Gustavo Miranda Carboni
- Laboratory of Oncology, Department of Oncology, School of Medicine, University of Tennessee Health Science, Memphis, TN 38163, USA;
| | - Michael N. Gould
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Randy L. Jirtle
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA;
| | - Victoria L. Seewaldt
- Beckman Research Institute, Department of Population Sciences, City of Hope, Duarte, CA 91010, USA; (E.C.D.); (J.A.A.-O.); (D.A.); (D.E.S.); (C.S.); (A.A.); (K.K.); (O.I.)
| |
Collapse
|
8
|
Genetic and Non-Genetic Mechanisms Underlying Cancer Evolution. Cancers (Basel) 2021; 13:cancers13061380. [PMID: 33803675 PMCID: PMC8002988 DOI: 10.3390/cancers13061380] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Our manuscript summarizes the up-to-date data on the complex and dynamic nature of adaptation mechanisms and evolutionary processes taking place during cancer initiation, development and progression. Although for decades cancer has been viewed as a process governed by genetic mechanisms, it is becoming more and more clear that non-genetic mechanisms may play an equally important role in cancer evolution. In this review, we bring together these fundamental concepts and discuss how those tightly interconnected mechanisms lead to the establishment of highly adaptive quickly evolving cancers. Furthermore, we argue that in depth understanding of cancer progression from the evolutionary perspective may allow the prediction and direction of the evolutionary path of cancer populations towards drug sensitive phenotypes and thus facilitate the development of more effective anti-cancer approaches. Abstract Cancer development can be defined as a process of cellular and tissular microevolution ultimately leading to malignancy. Strikingly, though this concept has prevailed in the field for more than a century, the precise mechanisms underlying evolutionary processes occurring within tumours remain largely uncharacterized and rather cryptic. Nevertheless, although our current knowledge is fragmentary, data collected to date suggest that most tumours display features compatible with a diverse array of evolutionary paths, suggesting that most of the existing macro-evolutionary models find their avatar in cancer biology. Herein, we discuss an up-to-date view of the fundamental genetic and non-genetic mechanisms underlying tumour evolution with the aim of concurring into an integrated view of the evolutionary forces at play throughout the emergence and progression of the disease and into the acquisition of resistance to diverse therapeutic paradigms. Our ultimate goal is to delve into the intricacies of genetic and non-genetic networks underlying tumour evolution to build a framework where both core concepts are considered non-negligible and equally fundamental.
Collapse
|
9
|
Gropman AL. Epigenetics and pervasive developmental disorders. EPIGENETICS IN PSYCHIATRY 2021:519-552. [DOI: 10.1016/b978-0-12-823577-5.00011-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
10
|
Radhakrishnan VK, Ravichandran K, Eke C, Ortiz-Vicil A, Tan Q, León MD, León DDD. Methylation of a newly identified region of the INS-IGF2 gene determines IGF2 expression in breast cancer tumors and in breast cancer cells. Oncotarget 2020; 11:3904-3920. [PMID: 33216823 PMCID: PMC7646830 DOI: 10.18632/oncotarget.27655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 06/01/2020] [Indexed: 11/25/2022] Open
Abstract
IGF2 is essential in breast differentiation, lactation, tumor growth, and in breast cancer (BC) development and progression. This growth factor also inhibits apoptosis and promotes metastasis and chemoresistance, contributing to more aggressive tumors. We previously demonstrated that IGF2 protein levels are higher in BC tissues from African American women than in Caucasian women. We also showed that high IGF2 protein levels are expressed in normal breast tissues of African American women while little or no IGF2 was detected in tissues from Caucasian women. Others showed that decreased DNA methylation of the IGF2 gene leads to different BC clinical features. Thus, we designed this study to determine if differentially methylated regions of the IGF2 gene correspond to IGF2 protein expression in paired (Normal/Tumor) breast tissues and in BC cell lines. Methylation analysis was performed using Sodium Bisulphite Analysis and Methylation Sensitive Restriction Enzyme digestion methods. Our results show that a unique site in the INS-IGF2 region is hypermethylated in normal breast and hypomethylated in breast cancer. We designated this region the DVDMR. Furthermore, the methylation levels in the DVDMR significantly correlated with IGF2 protein levels. This novel DMR consists of 257bp localized in the INS-IGF2 gene. We propose that methylation of DVDMR represents a novel epigenetic biomarker that determines the levels of IGF2 protein expression in breast cancer. Since IGF2 promotes metastasis and chemoresistance, we propose that IGF2 levels contribute to BC aggressiveness. Validation of IGF2 as a biomarker will improve diagnosis and treatment of BC patients.
Collapse
Affiliation(s)
- Vinodh Kumar Radhakrishnan
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Kameswaran Ravichandran
- Division of Renal Diseases and Hypertension, University of Colorado at Denver, Aurora, CO 80045, USA
| | - Chibuzo Eke
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Amanda Ortiz-Vicil
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Qianwei Tan
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Marino De León
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Daisy D De León
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
11
|
Kumaresan A, Das Gupta M, Datta TK, Morrell JM. Sperm DNA Integrity and Male Fertility in Farm Animals: A Review. Front Vet Sci 2020; 7:321. [PMID: 32637425 PMCID: PMC7317013 DOI: 10.3389/fvets.2020.00321] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022] Open
Abstract
The accurate prediction of male fertility is of major economic importance in the animal breeding industry. However, the results of conventional semen analysis do not always correlate with field fertility outcomes. There is evidence to indicate that mammalian fertilization and subsequent embryo development depend, in part, on the inherent integrity of the sperm DNA. Understanding the complex packaging of mammalian sperm chromatin and assessment of DNA integrity could potentially provide a benchmark in clinical infertility. In the era of assisted reproduction, especially when in-vitro fertilization or gamete intrafallopian transfer or intracytoplasmic sperm injection is used, assessment of sperm DNA integrity is important because spermatozoa are not subjected to the selection process occurring naturally in the female reproductive tract. Although sperm DNA integrity testing measures a significant biological parameter, its precise role in the infertility evaluation in farm animals remains unclear. In this review, the earlier findings on sperm DNA integrity in relation to male fertility are compiled and analyzed. Furthermore, the causes and consequences of sperm DNA damage are described, together with a review of advances in methods for detection of sperm DNA damage, and the prognostic value of sperm DNA quality on male fertility.
Collapse
Affiliation(s)
- Arumugam Kumaresan
- Theriogenology Laboratory, Southern Regional Station of National Dairy Research Institute (ICAR), Bengaluru, India
| | - Mohua Das Gupta
- Theriogenology Laboratory, Southern Regional Station of National Dairy Research Institute (ICAR), Bengaluru, India
| | - Tirtha Kumar Datta
- Animal Genomics Laboratory, National Dairy Research Institute (ICAR), Karnal, India
| | - Jane M. Morrell
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
12
|
Khosravizadeh Z, Hassanzadeh G, Tavakkoly Bazzaz J, Alizadeh F, Totonchi M, Salehi E, Khodamoradi K, Khanehzad M, Hosseini SR, Abolhassani F. The effect of cryopreservation on DNA methylation patterns of the chromosome 15q11-q13 region in human spermatozoa. Cell Tissue Bank 2020; 21:433-445. [PMID: 32253620 DOI: 10.1007/s10561-020-09828-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 03/11/2020] [Indexed: 01/23/2023]
Abstract
Human sperm cryopreservation is a common technique which is used in assisted reproductive technologies. Despite the existence of evidence supporting the production of ROS and DNA fragmentation during sperm cryopreservation, there is little and equivocal information about the cryopreservation effects on methylation of imprinted genes and imprinting control regions. In this study, we have investigated the effects of cryopreservation on DNA methylation in promoter regions of SNURF-SNRPN and UBE3A imprinted genes, PWS-ICR and AS-ICR in the chromosome 15q11-q13 region. Semen samples from 10 healthy normozoospermic men were collected and each sample was divided into four equal aliquots: fresh, cryoprotectant, cryopreservation, and H2O2. We measured the ROS levels and DNA fragmentation using DCFH-DA and TUNEL assay respectively by flow cytometry. DNA methylation in promoter regions of SNURF-SNRPN and UBE3A imprinted genes, PWS-ICR and AS-ICR in the chromosome 15q11-q13 region were evaluated by quantitative methylation-specific PCR technique. Intracellular levels of ROS and percentage of TUNEL-positive spermatozoa significantly increased in cryopreservation group compared to fresh group. Exposure to cryoprotectant had no significant effect on ROS levels and DNA fragmentation. Neither cryopreservation nor exposure to cryoprotectant significantly affected DNA methylation of the selected gene regions. However, DNA fragmentation had positive correlation with DNA methylation of AS-ICR. In conclusion, based on our study, clinical use of sperm cryopreservation for fertility treatments appear to be safe in regard to DNA methylation in the chromosome 15q11-q13 region.
Collapse
Affiliation(s)
- Zahra Khosravizadeh
- Faculty of Anatomy, School of Medicine, Tehran University of Medical Science, 16 Azar Avenue, Poor Sina Street, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Faculty of Anatomy, School of Medicine, Tehran University of Medical Science, 16 Azar Avenue, Poor Sina Street, Tehran, Iran
| | - Javad Tavakkoly Bazzaz
- Faculty of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, 16 Azar Avenue, Poor Sina Street, Tehran, Iran
| | - Fatemeh Alizadeh
- Faculty of Genomic Psychiatry and Behavioral Genomics (DGPBG), Roozbeh Hospital, School of Medicine, Tehran University of Medical Science, South Kargar Avenue, Tehran, Iran
| | - Mehdi Totonchi
- Faculty of Stem Cells and Developmental Biology, Royan Institute, Tehran, Iran
| | - Ensieh Salehi
- Faculty of Gynecology, School of Medicine, Fertility and Infertility Research Center, Bandar Abbas, Iran.,Dr. Ali Shariati Hospital, Shahid Mohammadi Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Kajal Khodamoradi
- Faculty of Anatomy, School of Medicine, Tehran University of Medical Science, 16 Azar Avenue, Poor Sina Street, Tehran, Iran
| | - Maryam Khanehzad
- Faculty of Anatomy, School of Medicine, Tehran University of Medical Science, 16 Azar Avenue, Poor Sina Street, Tehran, Iran
| | - Seyed Reza Hosseini
- Faculty of Urology, Sina Hospital, Tehran University of Medical Science, Imam Khomeini Street, Tehran, Iran.
| | - Farid Abolhassani
- Faculty of Anatomy, School of Medicine, Tehran University of Medical Science, 16 Azar Avenue, Poor Sina Street, Tehran, Iran.
| |
Collapse
|
13
|
Abstract
Stroke is the first cause of disability in the population and post-stroke patients admitted to rehabilitation units often present a malnutrition status which can influence nutritional indices and then vitamin levels. Vitamin D deficiency seems implicated beyond stroke severity and stroke risk, and also affects post-stroke recovery. Some studies on vitamin D levels and outcome in stroke patients are available but very few data on vitamin D levels and outcome after rehabilitation treatment are reported. This literature review shows the possible relationship between vitamin D deficiency and recovery in post-stroke patients undergoing rehabilitation treatment. Moreover, because several studies have reported that single nucleotide polymorphisms and promoter methylation in genes are involved in vitamin D metabolism and might affect circulating vitamin D levels, these aspects are evaluated in the current paper. From the studies evaluated in this review, it emerges that vitamin D deficiency could not only have an important role in the recovery of patients undergoing rehabilitation after a stroke, but that genetic and epigenetic factors related to vitamin D levels could have a crucial role on the rehabilitation outcome of patients after stroke. Therefore, further studies are necessary on stroke patients undergoing rehabilitation treatment, including: (a) the measurement of the 25(OH) vitamin D serum concentrations at admission and post rehabilitation treatment; (b) the identification of the presence/absence of CYP2R1, CYP27B1, CYP24A1 and VDR polymorphisms, and (c) analysis of the methylation levels of these genes pre- and post-rehabilitation treatment.
Collapse
|
14
|
Barati E, Nikzad H, Karimian M. Oxidative stress and male infertility: current knowledge of pathophysiology and role of antioxidant therapy in disease management. Cell Mol Life Sci 2020; 77:93-113. [PMID: 31377843 PMCID: PMC11105059 DOI: 10.1007/s00018-019-03253-8] [Citation(s) in RCA: 290] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/11/2019] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
Abstract
Infertility is a global health problem involving about 15% of couples. Approximately half of the infertility cases are related to male factors. The oxidative stress, which refers to an imbalance in levels of reactive oxygen species (ROS) and antioxidants, is one of the main causes of infertility in men. A small amount of ROS is necessary for the physiological function of sperm including the capacitation, hyperactivation and acrosomal reaction. However, high levels of ROS can cause infertility through not only by lipid peroxidation or DNA damage but inactivation of enzymes and oxidation of proteins in spermatozoa. Oxidative stress (OS) is mainly caused by factors associated with lifestyle. Besides, immature spermatozoa, inflammatory factors, genetic mutations and altering levels of sex hormones are other main source of ROS. Since OS occurs due to the lack of antioxidants and its side effects in semen, lifestyle changes and antioxidant regimens can be helpful therapeutic approaches to overcome this problem. The present study aimed to describe physiological ROS production, roles of genetic and epigenetic factors on the OS and male infertility with various mechanisms such as lipid peroxidation, DNA damage, and disorder of male hormone profile, inflammation, and varicocele. Finally, the roles of oral antioxidants and herbs were explained in coping with OS in male infertility.
Collapse
Affiliation(s)
- Erfaneh Barati
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Hossein Nikzad
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Karimian
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran.
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
15
|
Jiang F, Yang X, He X, Yang M. Circulating DNA, a Potentially Sensitive and Specific Diagnostic Tool for Future Medicine. Dose Response 2019; 17:1559325819891010. [PMID: 31827416 PMCID: PMC6886285 DOI: 10.1177/1559325819891010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/21/2019] [Accepted: 10/24/2019] [Indexed: 11/28/2022] Open
Abstract
Liquid biopsy has the great potential of detecting early diseases before deterioration and is valued for screening abnormalities at early stage. In oncology, circulating DNA derived from shed cancer cells reflects the tissue of origin, so it could be used to locate tissue sites during early screening. However, the heterogenous parameters of different types limit the clinical application, making it inaccessible to encompass all the cancer types. Instead, for reproducible scenario as pregnancy, fetal cell-free DNA has been well utilized for screening aneuploidies. Noninvasive and convenient as is, it would be of great value in the next decades far more than early diagnosis. This review recapitulates the discovery and development of tumor and fetal cell-free DNA. The common factors are also present that could be taken into consideration when collecting, transporting, and preserving samples. Meanwhile, several protocols used for purifying cell-free DNA, either classic ones or through commercial kits, are compared carefully. In addition, the development of technologies for analyzing cell-free DNA have been summarized and discussed in detail, especially some up-to-date approaches. At the end, the potential prospect of circulating DNA is bravely depicted. In summary, although there would be a lot of efforts before it’s prevalent, cell-free DNA remains a promising tool in point-of-care diagnostic medicine.
Collapse
Affiliation(s)
- Fan Jiang
- Department of Rehabilitation Medicine, The First People's Hospital of Wenling, Wenzhou Medical University, Wenling, Zhejiang, China
| | - Xiaoxiao Yang
- Department of Rehabilitation Medicine, The First People's Hospital of Wenling, Wenzhou Medical University, Wenling, Zhejiang, China
| | - Xiping He
- Department of Rehabilitation Medicine, The First People's Hospital of Wenling, Wenzhou Medical University, Wenling, Zhejiang, China
| | - Mingming Yang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
16
|
Gall TMH, Belete S, Khanderia E, Frampton AE, Jiao LR. Circulating Tumor Cells and Cell-Free DNA in Pancreatic Ductal Adenocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:71-81. [PMID: 30558725 DOI: 10.1016/j.ajpath.2018.03.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/06/2018] [Accepted: 03/26/2018] [Indexed: 12/21/2022]
Abstract
Pancreatic cancer is detected late in the disease process and has an extremely poor prognosis. A blood-based biomarker that can enable early detection of disease, monitor response to treatment, and potentially allow for personalized treatment would be of great benefit. This review analyzes the literature regarding two potential biomarkers, circulating tumor cells (CTCs) and cell-free DNA (cfDNA), with regard to pancreatic ductal adenocarcinoma. The origin of CTCs and the methods of detection are discussed and a decade of research examining CTCs in pancreatic cancer is summarized, including both levels of CTCs and analyzing their molecular characteristics and how they may affect survival in both advanced and early disease and allow for treatment monitoring. The origin of cfDNA is discussed, and the literature over the past 15 years is summarized. This includes analyzing cfDNA for genetic mutations and methylation abnormalities, which have the potential to be used for the detection and prognosis of pancreatic ductal adenocarcinoma. However, the research certainly remains in the experimental stage, warranting future large trials in these areas.
Collapse
Affiliation(s)
- Tamara M H Gall
- Hepato-Pancreato-Biliary Surgical Unit, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital Campus, London, United Kingdom.
| | - Samuel Belete
- Hepato-Pancreato-Biliary Surgical Unit, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital Campus, London, United Kingdom
| | - Esha Khanderia
- Hepato-Pancreato-Biliary Surgical Unit, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital Campus, London, United Kingdom
| | - Adam E Frampton
- Hepato-Pancreato-Biliary Surgical Unit, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital Campus, London, United Kingdom
| | - Long R Jiao
- Hepato-Pancreato-Biliary Surgical Unit, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital Campus, London, United Kingdom
| |
Collapse
|
17
|
Klawitter J, Klawitter J, Pennington A, Kirkpatrick B, Roda G, Kotecha NC, Thurman JM, Christians U. Cyclophilin D knockout protects the mouse kidney against cyclosporin A-induced oxidative stress. Am J Physiol Renal Physiol 2019; 317:F683-F694. [PMID: 31188033 DOI: 10.1152/ajprenal.00417.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Mitochondrial dysfunction and oxidative stress have been implicated in cyclosporin A (CsA)-induced nephrotoxicity. CsA interacts with cyclophilin D (CypD), an essential component of the mitochondrial permeability transition pore and regulator of cell death processes. Controversial reports have suggested that CypD deletion may or may not protect cells against oxidative stress-induced cell death. In the present study, we treated wild-type (WT) mice and mice lacking CypD [peptidylprolyl isomerase F knockout (Ppif-/-) mice] with CsA to test the role and contribution of CypD to the widely described CsA-induced renal toxicity and oxidative stress. Our results showed an increase in the levels of several known uremic toxins as well as the oxidative stress markers PGF2α and 8-isoprostane in CsA-treated WT animals but not in Ppif-/- animals. Similarly, a decline in S-adenosylmethionine and the resulting methylation potential indicative of DNA hypomethylation were observed only in CsA-treated WT mice. This confirms previous reports of the protective effects of CypD deletion on the mouse kidney mediated through a stronger resistance of these animals to oxidative stress and DNA methylation damage. However, a negative effect of CsA on the glycolysis and overall energy metabolism in Ppif-/- mice also indicated that additional, CypD-parallel pathways are involved in the toxic effects of CsA on the kidney. In summary, CsA-mediated induction of oxidative stress is associated with CypD, with CypD deletion providing a protective effect, whereas the reduction of energy production observed upon CsA exposure did not depend on the animals' CypD status.
Collapse
Affiliation(s)
- Jelena Klawitter
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
- Division of Nephrology and Hypertension, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Jost Klawitter
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | - Alexander Pennington
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | - Bruce Kirkpatrick
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | - Galen Roda
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | - Nidhi C Kotecha
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | - Joshua M Thurman
- Division of Nephrology and Hypertension, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Uwe Christians
- Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
18
|
Lu S, Xu F, Hu W, Niu Z, Cai H, Chen Y, Tu Q, Zhang Y, Chen W, Liu W, Tang S, Zhang Z. SCD1 methylation in subcutaneous adipose tissue associated with menopausal age. Climacteric 2019; 22:395-402. [PMID: 30777456 DOI: 10.1080/13697137.2019.1571028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- S. Lu
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - F. Xu
- Department of Gastroenterology, Hangzhou Third People's Hospital, Hangzhou, China
| | - W. Hu
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - Z. Niu
- Department of Obstetrics and Gynecology, The Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou, China
| | - H. Cai
- Department of Obstetrics and Gynecology, The Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou, China
| | - Y. Chen
- Laboratory of Gene Diagnosis, The Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou, China
| | - Q. Tu
- Laboratory of Gene Diagnosis, The Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou, China
| | - Y. Zhang
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - W. Chen
- Department of Obstetrics and Gynecology, The Second People's Hospital of Tonglu, Hangzhou, China
| | - W. Liu
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - S. Tang
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - Z. Zhang
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| |
Collapse
|
19
|
Santana VP, Miranda-Furtado CL, Pedroso DCC, Eiras MC, Vasconcelos MAC, Ramos ES, Calado RT, Ferriani RA, Esteves SC, dos Reis RM. The relationship among sperm global DNA methylation, telomere length, and DNA fragmentation in varicocele: a cross-sectional study of 20 cases. Syst Biol Reprod Med 2019; 65:95-104. [DOI: 10.1080/19396368.2018.1557762] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Viviane Paiva Santana
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Daiana Cristina Chielli Pedroso
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Matheus Credendio Eiras
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Ester Silveira Ramos
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rodrigo Tocantins Calado
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rui Alberto Ferriani
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Rosana Maria dos Reis
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
20
|
Wood PJ, Strong R, McArthur GA, Michael M, Algar E, Muscat A, Rigby L, Ferguson M, Ashley DM. A phase I study of panobinostat in pediatric patients with refractory solid tumors, including CNS tumors. Cancer Chemother Pharmacol 2018; 82:493-503. [PMID: 29987369 DOI: 10.1007/s00280-018-3634-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/25/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE This was an open label, phase I (3 + 3 design), multi-centre study evaluating panobinostat in pediatric patients with refractory solid tumors. METHODS Primary endpoints were to establish MTD, define and describe associated toxicities, including dose limiting toxicities (DLT) and to characterize its pharmacokinetics (PK). Secondary endpoints included assessing the anti-tumour activity of panobinostat, and its biologic activity, by measuring acetylation of histones in peripheral blood mononuclear cells. RESULTS Nine patients were enrolled and treated with intravenous panobinostat at a dosing level of 15 mg/m2 which was tolerated. Six were evaluable for adverse events. Two (33%) patients experienced Grade 3-4 thrombocytopenia, 1 (17%) experienced Grade 3 anemia, and 2 (33%) experienced Grade 3 neutropenia. Grade 4 drug related pain occurred in 2 (33%) of the patients studied. Two (33%) patients experienced a Grade 2 QTcF change (0.478 ± 0.006 ms). One cardiac DLT (T wave changes) was reported. PK values for 15 mg/m2 (n = 9) dosing were: Tmax 0.8 h, Cmax 235.2 ng/mL, AUC0-t 346.8 h ng/mL and t1/2 7.3 h. Panobinostat significantly induced acetylation of histone H3 and H4 at all time points measured when compared to pre-treatment samples (p < 0.05). Pooled quantitative Western blot data confirmed that panobinostat significantly induced acetylation of histone H4 at 6 h (p < 0.01), 24 h (p < 0.01) and 28-70 h (p < 0.01) post dose. CONCLUSION A significant biological effect of panobinostat, measured by acetylation status of histone H3 and H4, was achieved at a dose of 15 mg/m2. PK data and drug tolerability at 15 mg/m2 was similar to that previously published.
Collapse
Affiliation(s)
- Paul J Wood
- Department of Paediatrics, Monash University, Melbourne, Australia. .,Children's Cancer Centre, Monash Children's Hospital, Melbourne, Australia. .,Molecular Oncology and Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, Australia.
| | - Robyn Strong
- Australian & New Zealand Children's Haematology/Oncology Group (ANZCHOG), Melbourne, Australia
| | - Grant A McArthur
- Molecular Oncology and Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, Australia.,Department of Medicine, St. Vincent's Hospital, Melbourne, Australia
| | - Michael Michael
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Elizabeth Algar
- Monash University, Melbourne, Australia.,Hudson Institute of Medical Research, Melbourne, Australia
| | - Andrea Muscat
- Deakin University, School of Medicine, Geelong, Australia
| | - Lin Rigby
- Murdoch Children's Research Institute, Melbourne, Australia
| | | | - David M Ashley
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
21
|
Tejedor JR, Bueno C, Cobo I, Bayón GF, Prieto C, Mangas C, Pérez RF, Santamarina P, Urdinguio RG, Menéndez P, Fraga MF, Fernández AF. Epigenome-wide analysis reveals specific DNA hypermethylation of T cells during human hematopoietic differentiation. Epigenomics 2018; 10:903-923. [DOI: 10.2217/epi-2017-0163] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Aim: Epigenetic regulation plays an important role in cellular development and differentiation. A detailed map of the DNA methylation dynamics that occur during cell differentiation would contribute to decipher the molecular networks governing cell fate commitment. Methods: Illumina MethylationEPIC BeadChip platform was used to describe the genome-wide DNA methylation changes observed throughout hematopoietic maturation by analyzing multiple myeloid and lymphoid hematopoietic cell types. Results: We identified a plethora of DNA methylation changes that occur during human hematopoietic differentiation. We observed that T lymphocytes display substantial enhancement of de novo CpG hypermethylation as compared with other hematopoietic cell populations. T-cell-specific hypermethylated regions were strongly associated with open chromatin marks and enhancer elements, as well as binding sites of specific key transcription factors involved in hematopoietic differentiation, such as PU.1 and TAL1. Conclusion: These results provide novel insights into the role of DNA methylation at enhancer elements in T-cell development.
Collapse
Affiliation(s)
- J Ramón Tejedor
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Principado de Asturias, Spain
- Fundación para la Investigación Biosanitaria de Asturias (FINBA), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Principado de Asturias, Spain
| | - Clara Bueno
- Department of Biomedicine, School of Medicine, Josep Carreras Leukemia Research Institute, University of Barcelona, Barcelona, Spain
| | - Isabel Cobo
- Department of Biomedicine, School of Medicine, Josep Carreras Leukemia Research Institute, University of Barcelona, Barcelona, Spain
| | - Gustavo F Bayón
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Principado de Asturias, Spain
| | - Cristina Prieto
- Department of Biomedicine, School of Medicine, Josep Carreras Leukemia Research Institute, University of Barcelona, Barcelona, Spain
| | - Cristina Mangas
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Principado de Asturias, Spain
| | - Raúl F Pérez
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Principado de Asturias, Spain
- Nanomaterials & Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo, Principado de Asturias, Spain
| | - Pablo Santamarina
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Principado de Asturias, Spain
- Fundación para la Investigación Biosanitaria de Asturias (FINBA), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Principado de Asturias, Spain
| | - Rocío G Urdinguio
- Nanomaterials & Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo, Principado de Asturias, Spain
| | - Pablo Menéndez
- Department of Biomedicine, School of Medicine, Josep Carreras Leukemia Research Institute, University of Barcelona, Barcelona, Spain
- Instituciò Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Josep Carreras Leukemia Research Institute-Campus ICO, Research Institut Germans Trias i Pujol (IGTP), Badalona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCIII, Barcelona, Spain
| | - Mario F Fraga
- Nanomaterials & Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo, Principado de Asturias, Spain
| | - Agustín F Fernández
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Principado de Asturias, Spain
- Fundación para la Investigación Biosanitaria de Asturias (FINBA), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Principado de Asturias, Spain
| |
Collapse
|
22
|
Ou Y, Zhang Q, Tang Y, Lu Z, Lu X, Zhou X, Liu C. DNA methylation enzyme inhibitor RG108 suppresses the radioresistance of esophageal cancer. Oncol Rep 2018; 39:993-1002. [PMID: 29328411 PMCID: PMC5802040 DOI: 10.3892/or.2018.6210] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/15/2017] [Indexed: 01/28/2023] Open
Abstract
Esophageal cancer (EC) is the eighth most common highly aggressive cancer worldwide. The purpose of this study was to investigate the effect of the DNA methyltransferase inhibitor RG108 on the radiosensitivity of EC cells. MTT and clonogenic assays were performed to assess the effect of RG108 on the proliferation and radiosensitivity of Eca-109 and TE-1 human EC cells. The cell cycle progression and alterations in apoptosis were analyzed by flow cytometry. For the in vivo analysis, the Eca-109 cells were inoculated into nude mice to establish tumors. Tissues from xenografts were obtained to detect changes to microvessels and tumor growth by immunohistochemistry (IHC). RNA-seq was used to identify differentially expressed genes. We found that RG108 increased the radiosensitivity of EC cells. Apoptosis and G2/M-phase arrest were induced by X-ray irradiation and were significantly enhanced by RG108. In addition, growth of tumor xenografts from the Eca-109 cells was significantly inhibited by irradiation in combination with RG108. The RNA-seq analysis revealed that, compared with radiation alone, X-ray irradiation in combination with RG108 altered the expression of 121 genes in multiple pathways, including the TGF-β signaling pathway and the Epstein-Barr virus infection pathway. In conclusion, RG108 induced radiosensitivity in EC cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Yao Ou
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Quan Zhang
- Department of Radiotherapy, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Yiting Tang
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Zhonghua Lu
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Xujing Lu
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Xifa Zhou
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Changmin Liu
- Department of Oncology, The Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| |
Collapse
|
23
|
Fernandez AF, Valledor L, Vallejo F, Cañal MJ, Fraga MF. Quantification of Global DNA Methylation Levels by Mass Spectrometry. Methods Mol Biol 2018; 1708:49-58. [PMID: 29224138 DOI: 10.1007/978-1-4939-7481-8_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Global DNA methylation was classically considered the relative percentage of 5-methylcysine (5mC) with respect to total cytosine (C). Early approaches were based on the use of high-performance separation technologies and UV detection. However, the recent development of protocols using mass spectrometry for the detection has increased sensibility and permitted the precise identification of peak compounds based on their molecular masses. This allows work to be conducted with much less genomic DNA starting material and also to quantify 5-hydroxymethyl-cytosine (5hmC), a recently identified form of methylated cytosine that could play an important role in active DNA demethylation. Here, we describe the protocol that we currently use in our laboratory to analyze 5mC and 5hmC by mass spectrometry. The protocol, which is based on the method originally developed by Le and colleagues using Ultra Performance Liquid Chromatography (UPLC) and mass spectrometry (triple Quadrupole (QqQ)) detection, allows for the rapid and accurate quantification of relative global 5mC and 5hmC levels starting from just 1 μg of genomic DNA, which allows for the rapid and accurate quantification of relative global 5mC and 5hmC levels.
Collapse
Affiliation(s)
- Agustin F Fernandez
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (IISPA), Hospital Universitario Central de Asturias (HUCA), Fundación para la Investigación Biosanitaria de Asturias (FINBA), Grupo de Epigenética del Cáncer y Nanomedicina, Avda. de Roma, s/n., 33011, Oviedo, Spain
| | - Luis Valledor
- Area de Fisiología Vegetal, Dpto. Biología de Organismos y Sistemas, Universidad de Oviedo, Oviedo, Spain
| | - Fernando Vallejo
- Centro de Edafología y Biología Aplicada del Segura (CEBAS-CSIC), Campus Universitario de Espinardo, Murcia, Spain
| | - Maria Jesús Cañal
- Area de Fisiología Vegetal, Dpto. Biología de Organismos y Sistemas, Universidad de Oviedo, Oviedo, Spain
| | - Mario F Fraga
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Oviedo, Spain. .,Department of Immunology and Oncology, National Center for Biotechnology, CNB-CSIC, Cantoblanco, 28049, Madrid, Spain. .,Instituto de Investigación Sanitaria del Principado de Asturias (IISPA), Hospital Universitario Central de Asturias (HUCA), Fundación para la Investigación Biosanitaria de Asturias (FINBA), Grupo de Epigenética del Cáncer y Nanomedicina, Avda. de Roma, s/n., 33011, Oviedo, Spain.
| |
Collapse
|
24
|
Santana VP, Miranda-Furtado CL, de Oliveira-Gennaro FG, Dos Reis RM. Genetics and epigenetics of varicocele pathophysiology: an overview. J Assist Reprod Genet 2017; 34:839-847. [PMID: 28523408 DOI: 10.1007/s10815-017-0931-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/19/2017] [Indexed: 11/26/2022] Open
Abstract
Varicocele is found in approximately 20% of adults and adolescents and in 19-41% of men seeking treatment for infertility. It is associated with a decrease in sperm count as well as sperm motility and morphology. The currently accepted description of the pathophysiology of varicocele does not explain all its clinical manifestations; therefore, other factors such as genetic and epigenetic changes, associated with the environment, might be involved in causing infertility and decrease in sperm quality. It has been reported that the varicocele-induced deterioration of testicular function is progressive and interferes with fertility; hence, early and efficient assessment of the genetic manifestations in patients would be important for developing future medical interventions. Chromosomal disorders, mutations, polymorphisms, changes in gene expression, and epigenetic changes have all been reported to be associated with varicocele. Several studies are underway to unravel the genetic basis of this disease, as it is important to understand the origin and the aggravating factors to ensure appropriate guidance and intervention. Here, we review the available literature regarding the genetic and epigenetic changes associated with varicocele, and how these alterations are related to the different clinical manifestations of the disease.
Collapse
Affiliation(s)
- Viviane Paiva Santana
- Department of Gynecology and Obstetrics, Ribeirao Preto Medical School, University of São Paulo (USP), 3900 Bandeirantes Ave, São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Cristiana Libardi Miranda-Furtado
- Department of Gynecology and Obstetrics, Ribeirao Preto Medical School, University of São Paulo (USP), 3900 Bandeirantes Ave, São Paulo, Ribeirão Preto, 14049-900, Brazil
- Oswaldo Cruz Foundation (FIOCRUZ), Ceará, Fortaleza, Brazil
| | - Flavia Gaona de Oliveira-Gennaro
- Department of Gynecology and Obstetrics, Ribeirao Preto Medical School, University of São Paulo (USP), 3900 Bandeirantes Ave, São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Rosana Maria Dos Reis
- Department of Gynecology and Obstetrics, Ribeirao Preto Medical School, University of São Paulo (USP), 3900 Bandeirantes Ave, São Paulo, Ribeirão Preto, 14049-900, Brazil.
| |
Collapse
|
25
|
Abstract
Epigenetic mechanisms play important roles in properly occurring mammalian oogenesis. One of these mechanisms is DNA methylation adding a methyl group to the fifth carbon atom of the cytosine residues using S-adenosyl-L-methionine as a methyl donor. DNA methylation generally takes place at cytosine-phosphate-guanine (CpG) dinucleotide sites and rarely occurs at cytosine-phosphate-thymine (CpT), cytosine-phosphate-adenine (CpA), or cytosine-phosphate-cytosine sites, known as non-CpG sites. Basically, two different DNA methylation processes are identified: de novo methylation and maintenance methylation. While the de novo methylation functions in methylation of unmethylated DNA strands, maintenance methylation is capable of methylating hemi-methylated DNA strands following DNA replication. Both DNA methylation processes are catalyzed by special DNA methyltransferase (DNMT) enzymes. To date, five different DNMTs have been identified: DNMT1, DNMT3A, DNMT3B, DNMT3L, and DNMT2. In this chapter, we focus particularly on temporal and spatial expression of DNMTs in mammalian oocytes and granulosa cells.
Collapse
Affiliation(s)
- Fatma Uysal
- Department of Histology and Embryology, School of Medicine, Akdeniz University, Campus, 07070, Antalya, Turkey
| | - Saffet Ozturk
- Department of Histology and Embryology, School of Medicine, Akdeniz University, Campus, 07070, Antalya, Turkey.
| |
Collapse
|
26
|
Villota-Salazar NA, Mendoza-Mendoza A, González-Prieto JM. Epigenetics: from the past to the present. FRONTIERS IN LIFE SCIENCE 2016. [DOI: 10.1080/21553769.2016.1249033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
27
|
Chisholm RH, Lorenzi T, Clairambault J. Cell population heterogeneity and evolution towards drug resistance in cancer: Biological and mathematical assessment, theoretical treatment optimisation. Biochim Biophys Acta Gen Subj 2016; 1860:2627-45. [PMID: 27339473 DOI: 10.1016/j.bbagen.2016.06.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/25/2016] [Accepted: 06/05/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Drug-induced drug resistance in cancer has been attributed to diverse biological mechanisms at the individual cell or cell population scale, relying on stochastically or epigenetically varying expression of phenotypes at the single cell level, and on the adaptability of tumours at the cell population level. SCOPE OF REVIEW We focus on intra-tumour heterogeneity, namely between-cell variability within cancer cell populations, to account for drug resistance. To shed light on such heterogeneity, we review evolutionary mechanisms that encompass the great evolution that has designed multicellular organisms, as well as smaller windows of evolution on the time scale of human disease. We also present mathematical models used to predict drug resistance in cancer and optimal control methods that can circumvent it in combined therapeutic strategies. MAJOR CONCLUSIONS Plasticity in cancer cells, i.e., partial reversal to a stem-like status in individual cells and resulting adaptability of cancer cell populations, may be viewed as backward evolution making cancer cell populations resistant to drug insult. This reversible plasticity is captured by mathematical models that incorporate between-cell heterogeneity through continuous phenotypic variables. Such models have the benefit of being compatible with optimal control methods for the design of optimised therapeutic protocols involving combinations of cytotoxic and cytostatic treatments with epigenetic drugs and immunotherapies. GENERAL SIGNIFICANCE Gathering knowledge from cancer and evolutionary biology with physiologically based mathematical models of cell population dynamics should provide oncologists with a rationale to design optimised therapeutic strategies to circumvent drug resistance, that still remains a major pitfall of cancer therapeutics. This article is part of a Special Issue entitled "System Genetics" Guest Editor: Dr. Yudong Cai and Dr. Tao Huang.
Collapse
Affiliation(s)
- Rebecca H Chisholm
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Tommaso Lorenzi
- School of Mathematics and Statistics, University of St Andrews, North Haugh, KY16 9SS, St Andrews, Scotland, United Kingdom. http://www.tommasolorenzi.com
| | - Jean Clairambault
- INRIA Paris, MAMBA team, 2, rue Simone Iff, CS 42112, 75589 Paris Cedex 12, France; Sorbonne Universités, UPMC Univ. Paris 6, UMR 7598, Laboratoire Jacques-Louis Lions, Boîte courrier 187, 4 Place Jussieu, 75252 Paris Cedex 05, France.
| |
Collapse
|
28
|
Wang F, Zhang N, Wang J, Wu H, Zheng X. Tumor purity and differential methylation in cancer epigenomics. Brief Funct Genomics 2016; 15:408-419. [PMID: 27199459 DOI: 10.1093/bfgp/elw016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
DNA methylation is an epigenetic modification of DNA molecule that plays a vital role in gene expression regulation. It is not only involved in many basic biological processes, but also considered an important factor for tumorigenesis and other human diseases. Study of DNA methylation has been an active field in cancer epigenomics research. With the advances of high-throughput technologies and the accumulation of enormous amount of data, method development for analyzing these data has gained tremendous interests in the fields of computational biology and bioinformatics. In this review, we systematically summarize the recent developments of computational methods and software tools in high-throughput methylation data analysis with focus on two aspects: differential methylation analysis and tumor purity estimation in cancer studies.
Collapse
|
29
|
Li Z, Li J, Liu Y, Wang Z. DNA demethylation during Chrysanthemum floral transition following short-day treatment. ELECTRON J BIOTECHN 2016. [DOI: 10.1016/j.ejbt.2016.02.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
30
|
Negative regulation of DNMT3A de novo DNA methylation by frequently overexpressed UHRF family proteins as a mechanism for widespread DNA hypomethylation in cancer. Cell Discov 2016; 2:16007. [PMID: 27462454 PMCID: PMC4849474 DOI: 10.1038/celldisc.2016.7] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/29/2016] [Indexed: 12/19/2022] Open
Abstract
Global DNA hypomethylation is a most common epigenetic alteration in cancer, but the mechanism remains elusive. Previous studies demonstrate that UHRF1 but not UHRF2 is required for mediating DNA maintenance methylation by DNMT1. Here we report unexpectedly a conserved function for UHRF1 and UHRF2: inhibiting de novo DNA methylation by functioning as E3 ligases promoting DNMT3A degradation. UHRF1/2 are frequently overexpressed in cancers and we present evidence that UHRF1/2 overexpression downregulates DNMT3A proteins and consequently leads to DNA hypomethylation. Abrogating this negative regulation on DNMT3A or overexpression of DNMT3A leads to increased DNA methylation and impaired tumor growth. We propose a working model that UHRF1/2 safeguards the fidelity of DNA methylation and suggests that UHRF1/2 overexpression is likely a causal factor for widespread DNA hypomethylation in cancer via suppressing DNMT3A.
Collapse
|
31
|
Tabano S, Bonaparte E, Miozzo M. Detection of Loss of Imprinting by Pyrosequencing®. Methods Mol Biol 2016; 1315:241-58. [PMID: 26103904 DOI: 10.1007/978-1-4939-2715-9_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Genomic imprinting is an epigenetically regulated process determining allele-specific expression in a parent-of-origin dependent manner. Altered expression of imprinted genes characterizes numerous congenital diseases including Beckwith-Wiedemann, Silver-Russell, Angelman, and Prader-Willi syndromes as well as acquired disorders such as cancer. The detection of imprinting alterations has important translational implications in clinics and the application of the Pyrosequencing(®) technology offers the possibility to identify accurately also subtle modifications in allele-specific expression and in DNA methylation levels.Here, we describe two methods to investigate genomic imprinting defects (loss of imprinting, LOI) using Pyrosequencing: (1) Allele-specific expression analysis based on single nucleotide polymorphism (SNP), and (2) quantification of DNA methylation.The protocol for the quantification of the allele-specific expression is carried out by analyzing an informative SNP located within the transcribed portion of an imprinted gene. The method includes the cDNA amplification of the region containing the SNP and the Pyrosequencing-based analysis for the quantitative allelic discrimination comparing the ratio of the two alleles.The second protocol allows the accurate quantification of the DNA methylation levels at the Imprinting Control Regions (ICRs). Imprinted genes are clustered in chromosomal regions and their expression is mainly regulated by DNA methylation at CpG sites located within the ICRs. After bisulfite modification of the genomic DNA, the region of interest is amplified by PCR and analyzed by Pyrosequencing. The methylation value at each CpG site is calculated by the CpG software, which determines the ratio of the incorporation of "C" and "T" and converts the value in methylation percentage.
Collapse
Affiliation(s)
- Silvia Tabano
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via Festa Del Perdono 7, Milan, 20122, Italy
| | | | | |
Collapse
|
32
|
Lue JK, Amengual JE, O'Connor OA. Epigenetics and Lymphoma: Can We Use Epigenetics to Prime or Reset Chemoresistant Lymphoma Programs? Curr Oncol Rep 2016; 17:40. [PMID: 26141799 DOI: 10.1007/s11912-015-0464-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Non-Hodgkin lymphoma is a diverse group of lymphocyte-derived neoplasms. Although a heterogeneous group of malignancies, it has become apparent that epigenetic alterations, such as disturbances of DNA methylation and histone modification, are a common occurrence in both B cell and T cell lymphomas, contributing to lymphomagenesis. As a result, the use of epigenetic targeted therapy has been incorporated into various pre-clinical and clinical studies, demonstrating significant efficacy in lymphoma, with vorinostat becoming the first epigenetic therapy to receive FDA approval in any malignancy. The role of epigenetic drugs is evolving, with its potential use in combination therapy as well as a means of overcoming chemotherapy resistance. In this review, we discuss the epigenetic alterations in non-Hodgkin lymphomas as well as provide an overview of current epigenetic drugs and their role in clinical practice, and on-going clinical trials.
Collapse
Affiliation(s)
- Jennifer K Lue
- Center for Lymphoid Malignancies, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, Columbia University, New York, NY, USA
| | | | | |
Collapse
|
33
|
Schultz B, Yao X, Deng Y, Waner M, Spock C, Tom L, Persing J, Narayan D. A Common Polymorphism within the IGF2 Imprinting Control Region Is Associated with Parent of Origin Specific Effects in Infantile Hemangiomas. PLoS One 2015; 10:e0113168. [PMID: 26496499 PMCID: PMC4619854 DOI: 10.1371/journal.pone.0113168] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 10/24/2014] [Indexed: 11/30/2022] Open
Abstract
Infantile hemangioma (IH) is the most common tumor of the pediatric age group, affecting up to 4% of newborns ranging from inconsequential blemishes, to highly aggressive tumors. Following well defined growth phases (proliferative, plateau involutional) IH usually regress into a fibro-fatty residuum. Despite the high prevalence of IH, little is known regarding the pathogenesis of disease. A reported six fold decrease in IGF2 expression (correlating with transformation of proliferative to involuted lesions) prompted us to study the IGF-2 axis further. We demonstrate that IGF2 expression in IH is strongly related to the expression of a cancer testes and suspected oncogene BORIS (paralog of CTCF), placing IH in the unique category of being the first known benign BORIS positive tumor. IGF2 expression was strongly and positively related to BORIS transcript expression. Furthermore, a stronger association was made when comparing BORIS levels against the expression of CTCF via either a percentage or difference between the two. A common C/T polymorphism at CTCF BS6 appeared to modify the correlation between CTCF/BORIS and IGF2 expression in a parent of origin specific manner. Moreover, these effects may have phenotypic consequences as tumor growth also correlates with the genotype at CTCF BS6. This may provide a framework for explaining the clinical variability seen in IH and suggests new insights regarding CTCF and BORIS related functionality in both normal and malignant states.
Collapse
Affiliation(s)
- Brent Schultz
- University of Washington, Division of Plastic Surgery, Seattle, WA, United States of America
| | - Xiaopan Yao
- Yale Center for Analytic Sciences at YSPH, New Haven, CT, United States of America
| | - Yanhong Deng
- Yale Center for Analytic Sciences at YSPH, New Haven, CT, United States of America
| | - Milton Waner
- Vascular Birthmark Institute, New York, NY, United States of America
| | | | - Laura Tom
- Division of Plastic Surgery, University of Washington, Seattle, WA, United States of America
| | - John Persing
- Yale Plastic and Reconstructive Surgery, New Haven, United States of America
| | - Deepak Narayan
- Yale Plastic and Reconstructive Surgery, New Haven, United States of America
- * E-mail:
| |
Collapse
|
34
|
Uysal F, Akkoyunlu G, Ozturk S. Dynamic expression of DNA methyltransferases (DNMTs) in oocytes and early embryos. Biochimie 2015; 116:103-13. [PMID: 26143007 DOI: 10.1016/j.biochi.2015.06.019] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/26/2015] [Indexed: 11/26/2022]
Abstract
Epigenetic mechanisms play critical roles in oogenesis and early embryo development in mammals. One of these epigenetic mechanisms, DNA methylation is accomplished through the activities of DNA methyltransferases (DNMTs), which are responsible for adding a methyl group to the fifth carbon atom of the cytosine residues within cytosine-phosphate-guanine (CpG) and non-CpG dinuclotide sites. Five DNMT enzymes have been identified in mammals including DNMT1, DNMT2, DNMT3A, DNMT3B, and DNMT3L. They function in two different methylation processes: maintenance and de novo. For maintenance methylation, DNMT1 preferentially transfers methyl groups to the hemi-methylated DNA strands following DNA replication. However, for de novo methylation activities both DNMT3A and DNMT3B function in the methylation of the unmodified cytosine residues. Although DNMT3L indirectly contributes to de novo methylation process, DNMT2 enables the methylation of the cytosine 38 in the anticodon loop of aspartic acid transfer RNA and does not methylate DNA. In this review article, we have evaluated and discussed the existing published studies to characterize the spatial and temporal expression patterns of the DNMTs in mouse, bovine and human oocytes and early embryos. We have also reviewed the effects of in vitro culture conditions (serum abundance and glucose concentration), aging, superovulation, vitrification, and somatic cell nuclear transfer technology on the dynamics of DNMTs.
Collapse
Affiliation(s)
- Fatma Uysal
- Department of Histology and Embryology, Akdeniz University, School of Medicine, Antalya, Turkey
| | - Gokhan Akkoyunlu
- Department of Histology and Embryology, Akdeniz University, School of Medicine, Antalya, Turkey
| | - Saffet Ozturk
- Department of Histology and Embryology, Akdeniz University, School of Medicine, Antalya, Turkey.
| |
Collapse
|
35
|
Sun S, Li P. HMPL: A Pipeline for Identifying Hemimethylation Patterns by Comparing Two Samples. Cancer Inform 2015; 14:235-45. [PMID: 26308520 PMCID: PMC4530977 DOI: 10.4137/cin.s17286] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 06/18/2015] [Accepted: 06/28/2015] [Indexed: 01/03/2023] Open
Abstract
DNA methylation (the addition of a methyl group to a cytosine) is an important epigenetic event in mammalian cells because it plays a key role in regulating gene expression. Most previous methylation studies assume that DNA methylation occurs on both positive and negative strands. However, a few studies have reported that in some genes, methylation occurs only on one strand (ie, hemimethylation) and has clustering patterns. These studies report that hemimethylation occurs on individual genes. It is unclear whether hemimethylation occurs genome-wide and whether there are hemimethylation differences between cancerous and noncancerous cells. To address these questions, we have developed the first-ever pipeline, named hemimethylation pipeline (HMPL), to identify hemimethylation patterns. Utilizing the available software and the newly developed Perl and R scripts, HMPL can identify hemimethylation patterns for a single sample and can also compare two different samples.
Collapse
Affiliation(s)
- Shuying Sun
- Department of Mathematics, Texas State University, San Marcos, TX, USA
| | - Peng Li
- Department of Electrical Engineering and Computer Sciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
36
|
Abstract
Oral squamous cell carcinoma (OSCC) is a multistep process which is modulated by several endogenous and environmental factors. Epigenetic changes have been found to be equally responsible for OSCC as genetic changes. A plethora of genes showing hypermethylation have been discovered in OSCC. Since these changes are reversible, a lot of emphasis is on using the natural compounds for their ability to cause demethylation which could lead to reactivation of the inactivated tumor suppressor genes. This review encompasses the promoter hypermethylation of tumor suppressor genes in OSCC and its possible reversal using natural compounds. In addition, new compounds which could be screened for their demethylating ability have also been proposed.
Collapse
|
37
|
Urdinguio RG, Bayón GF, Dmitrijeva M, Toraño EG, Bravo C, Fraga MF, Bassas L, Larriba S, Fernández AF. Aberrant DNA methylation patterns of spermatozoa in men with unexplained infertility. Hum Reprod 2015; 30:1014-28. [PMID: 25753583 DOI: 10.1093/humrep/dev053] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 02/16/2015] [Indexed: 12/17/2022] Open
Abstract
STUDY QUESTION Are there DNA methylation alterations in sperm that could explain the reduced biological fertility of male partners from couples with unexplained infertility? SUMMARY ANSWER DNA methylation patterns, not only at specific loci but also at Alu Yb8 repetitive sequences, are altered in infertile individuals compared with fertile controls. WHAT IS KNOWN ALREADY Aberrant DNA methylation of sperm has been associated with human male infertility in patients demonstrating either deficiencies in the process of spermatogenesis or low semen quality. STUDY DESIGN, SIZE, DURATION Case and control prospective study. This study compares 46 sperm samples obtained from 17 normospermic fertile men and 29 normospermic infertile patients. PARTICIPANTS/MATERIALS, SETTING, METHODS Illumina Infinium HD Human Methylation 450K arrays were used to identify genomic regions showing differences in sperm DNA methylation patterns between five fertile and seven infertile individuals. Additionally, global DNA methylation of sperm was measured using the Methylamp Global DNA Methylation Quantification Ultra kit (Epigentek) in 14 samples, and DNA methylation at several repetitive sequences (LINE-1, Alu Yb8, NBL2, D4Z4) measured by bisulfite pyrosequencing in 44 sperm samples. A sperm-specific DNA methylation pattern was obtained by comparing the sperm methylomes with the DNA methylomes of differentiated somatic cells using data obtained from methylation arrays (Illumina 450 K) of blood, neural and glial cells deposited in public databases. MAIN RESULTS AND THE ROLE OF CHANCE In this study we conduct, for the first time, a genome-wide study to identify alterations of sperm DNA methylation in individuals with unexplained infertility that may account for the differences in their biological fertility compared with fertile individuals. We have identified 2752 CpGs showing aberrant DNA methylation patterns, and more importantly, these differentially methylated CpGs were significantly associated with CpG sites which are specifically methylated in sperm when compared with somatic cells. We also found statistically significant (P < 0.001) associations between DNA hypomethylation and regions corresponding to those which, in somatic cells, are enriched in the repressive histone mark H3K9me3, and between DNA hypermethylation and regions enriched in H3K4me1 and CTCF, suggesting that the relationship between chromatin context and aberrant DNA methylation of sperm in infertile men could be locus-dependent. Finally, we also show that DNA methylation patterns, not only at specific loci but also at several repetitive sequences (LINE-1, Alu Yb8, NBL2, D4Z4), were lower in sperm than in somatic cells. Interestingly, sperm samples at Alu Yb8 repetitive sequences of infertile patients showed significantly lower DNA methylation levels than controls. LIMITATIONS, REASONS FOR CAUTION Our results are descriptive and further studies would be needed to elucidate the functional effects of aberrant DNA methylation on male fertility. WIDER IMPLICATIONS OF THE FINDINGS Overall, our data suggest that aberrant sperm DNA methylation might contribute to fertility impairment in couples with unexplained infertility and they provide a promising basis for future research. STUDY FUNDING/COMPETING INTERESTS This work has been financially supported by Fundación Cientifica de la AECC (to R.G.U.); IUOPA (to G.F.B.); FICYT (to E.G.T.); the Spanish National Research Council (CSIC; 200820I172 to M.F.F.); Fundación Ramón Areces (to M.F.F); the Plan Nacional de I+D+I 2008-2011/2013-2016/FEDER (PI11/01728 to AF.F., PI12/01080 to M.F.F. and PI12/00361 to S.L.); the PN de I+D+I 2008-20011 and the Generalitat de Catalunya (2009SGR01490). A.F.F. is sponsored by ISCIII-Subdirección General de Evaluación y Fomento de la Investigación (CP11/00131). S.L. is sponsored by the Researchers Stabilization Program from the Spanish National Health System (CES09/020). The IUOPA is supported by the Obra Social Cajastur, Spain.
Collapse
Affiliation(s)
- Rocío G Urdinguio
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Oviedo 33006, Spain
| | - Gustavo F Bayón
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Oviedo 33006, Spain
| | - Marija Dmitrijeva
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Oviedo 33006, Spain
| | - Estela G Toraño
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Oviedo 33006, Spain
| | - Cristina Bravo
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Oviedo 33006, Spain
| | - Mario F Fraga
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Oviedo 33006, Spain Department of Immunology and Oncology, National Center for Biotechnology, CNB-CSIC, Cantoblanco, Madrid 28049, Spain
| | - Lluís Bassas
- Laboratory of Seminology and Embryology, Andrology Service-Fundació Puigvert, Barcelona 08025, Spain
| | - Sara Larriba
- Human Molecular Genetics Group-IDIBELL, L'Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Agustín F Fernández
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), HUCA, Universidad de Oviedo, Oviedo 33006, Spain
| |
Collapse
|
38
|
Maroni L, Pierantonelli I, Banales JM, Benedetti A, Marzioni M. The significance of genetics for cholangiocarcinoma development. ANNALS OF TRANSLATIONAL MEDICINE 2014; 1:28. [PMID: 25332972 DOI: 10.3978/j.issn.2305-5839.2012.10.04] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 10/15/2012] [Indexed: 12/15/2022]
Abstract
Cholangiocarcinoma (CCA) is a rare malignancy of the liver, arising from bile ducts. The incidence is increasing worldwide, but the prognosis has remained dismal and virtually unchanged in the past 30 years. Although several risk factors have been associated with the development of this cancer, none of them are normally identified in most patients. Diagnosis in advanced stages of the disease and limited therapeutic options contribute to poor survival rates. The recent analysis of genetic and epigenetic alterations occurring in CCA has shed new light in the understanding of the molecular mechanisms leading to the malignant transformation of biliary cells. Further studies in this direction may foster new diagnostic, prognostic and therapeutic approaches. This review provides a global overview of recent advances in CCA and describes the most important genetic mutations and epigenetic alterations so far reported in CCA.
Collapse
Affiliation(s)
- Luca Maroni
- 1 Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy ; 2 Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands ; 3 Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria ; 4 Division of Hepatology and Gastroenterology, Biodonostia Research Institute (Donostia University Hospital), CIBERehd, University of Basque Country, San Sebastián, Spain - IKERBASQUE (Basque Foundation of Science), and "Asociación Española Contra el Cáncer, (AECC)"
| | - Irene Pierantonelli
- 1 Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy ; 2 Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands ; 3 Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria ; 4 Division of Hepatology and Gastroenterology, Biodonostia Research Institute (Donostia University Hospital), CIBERehd, University of Basque Country, San Sebastián, Spain - IKERBASQUE (Basque Foundation of Science), and "Asociación Española Contra el Cáncer, (AECC)"
| | - Jesus M Banales
- 1 Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy ; 2 Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands ; 3 Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria ; 4 Division of Hepatology and Gastroenterology, Biodonostia Research Institute (Donostia University Hospital), CIBERehd, University of Basque Country, San Sebastián, Spain - IKERBASQUE (Basque Foundation of Science), and "Asociación Española Contra el Cáncer, (AECC)"
| | - Antonio Benedetti
- 1 Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy ; 2 Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands ; 3 Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria ; 4 Division of Hepatology and Gastroenterology, Biodonostia Research Institute (Donostia University Hospital), CIBERehd, University of Basque Country, San Sebastián, Spain - IKERBASQUE (Basque Foundation of Science), and "Asociación Española Contra el Cáncer, (AECC)"
| | - Marco Marzioni
- 1 Department of Gastroenterology, Università Politecnica delle Marche, Ancona, Italy ; 2 Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands ; 3 Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria ; 4 Division of Hepatology and Gastroenterology, Biodonostia Research Institute (Donostia University Hospital), CIBERehd, University of Basque Country, San Sebastián, Spain - IKERBASQUE (Basque Foundation of Science), and "Asociación Española Contra el Cáncer, (AECC)"
| |
Collapse
|
39
|
Zheng X, Zhao Q, Wu HJ, Li W, Wang H, Meyer CA, Qin QA, Xu H, Zang C, Jiang P, Li F, Hou Y, He J, Wang J, Wang J, Zhang P, Zhang Y, Liu XS. MethylPurify: tumor purity deconvolution and differential methylation detection from single tumor DNA methylomes. Genome Biol 2014; 15:419. [PMID: 25103624 PMCID: PMC4165374 DOI: 10.1186/s13059-014-0419-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 08/07/2014] [Indexed: 12/14/2022] Open
Abstract
We propose a statistical algorithm MethylPurify that uses regions with bisulfite reads showing discordant methylation levels to infer tumor purity from tumor samples alone. MethylPurify can identify differentially methylated regions (DMRs) from individual tumor methylome samples, without genomic variation information or prior knowledge from other datasets. In simulations with mixed bisulfite reads from cancer and normal cell lines, MethylPurify correctly inferred tumor purity and identified over 96% of the DMRs. From patient data, MethylPurify gave satisfactory DMR calls from tumor methylome samples alone, and revealed potential missed DMRs by tumor to normal comparison due to tumor heterogeneity.
Collapse
|
40
|
Gall TMH, Frampton AE, Krell J, Habib NA, Castellano L, Stebbing J, Jiao LR. Cell-free DNA for the detection of pancreatic, liver and upper gastrointestinal cancers: has progress been made? Future Oncol 2014; 9:1861-9. [PMID: 24295416 DOI: 10.2217/fon.13.152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Detecting alterations in blood cell-free DNA (cfDNA) is hoped to be a novel, noninvasive method for diagnosing, prognosing and monitoring cancer patients. Several studies have assessed the usefulness of measuring tumor-specific genetic and epigenetic changes of cfDNA, such as loss of heterozygosity, frequency of mutations, alterations of microsatellites and the methylation of genes in patient blood samples. However, few well-designed trials have been carried out to translate these findings effectively. In this review, we have assessed the clinical utility of cfDNA in pancreatic, liver and upper gastrointestinal malignancies.
Collapse
Affiliation(s)
- Tamara M H Gall
- HPB Surgical Unit, Department of Surgery & Cancer, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London, W12 0HS, UK
| | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Epigenetic mechanisms play a crucial role in regulating gene expression. The main mechanisms involve methylation of DNA and covalent modifications of histones by methylation, acetylation, phosphorylation, or ubiquitination. The complex interplay of different epigenetic mechanisms is mediated by enzymes acting in the nucleus. Modifications in DNA methylation are performed mainly by DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) proteins, while a plethora of enzymes, such as histone acetyltransferases (HATs), histone deacetylases (HDACs), histone methyltransferases (HMTs), and histone demethylases (HDMs) regulate covalent histone modifications. In many diseases, such as cancer, the epigenetic regulatory system is often disturbed. Vitamin D interacts with the epigenome on multiple levels. Firstly, critical genes in the vitamin D signaling system, such as those coding for vitamin D receptor (VDR) and the enzymes 25-hydroxylase (CYP2R1), 1α-hydroxylase (CYP27B1), and 24-hydroxylase (CYP24A1) have large CpG islands in their promoter regions and therefore can be silenced by DNA methylation. Secondly, VDR protein physically interacts with coactivator and corepressor proteins, which in turn are in contact with chromatin modifiers, such as HATs, HDACs, HMTs, and with chromatin remodelers. Thirdly, a number of genes encoding for chromatin modifiers and remodelers, such as HDMs of the Jumonji C (JmjC)-domain containing proteins and lysine-specific demethylase (LSD) families are primary targets of VDR and its ligands. Finally, there is evidence that certain VDR ligands have DNA demethylating effects. In this review we will discuss regulation of the vitamin D system by epigenetic modifications and how vitamin D contributes to the maintenance of the epigenome, and evaluate its impact in health and disease.
Collapse
Affiliation(s)
- Irfete S Fetahu
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Comprehensive Cancer Center, Medical University of Vienna Vienna, Austria
| | - Julia Höbaus
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Comprehensive Cancer Center, Medical University of Vienna Vienna, Austria
| | - Enikő Kállay
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Comprehensive Cancer Center, Medical University of Vienna Vienna, Austria
| |
Collapse
|
42
|
Gropman AL. Epigenetics and Pervasive Developmental Disorders. EPIGENETICS IN PSYCHIATRY 2014:395-424. [DOI: 10.1016/b978-0-12-417114-5.00019-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
43
|
Fernández AF, Toraño EG, Urdinguio RG, Lana AG, Fernández IA, Fraga MF. The Epigenetic Basis of Adaptation and Responses to Environmental Change: Perspective on Human Reproduction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 753:97-117. [DOI: 10.1007/978-1-4939-0820-2_6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
44
|
Jacobs DI, Mao Y, Fu A, Kelly WK, Zhu Y. Dysregulated methylation at imprinted genes in prostate tumor tissue detected by methylation microarray. BMC Urol 2013; 13:37. [PMID: 23890537 PMCID: PMC3751920 DOI: 10.1186/1471-2490-13-37] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 07/15/2013] [Indexed: 11/18/2022] Open
Abstract
Background Imprinting is an important epigenetic regulator of gene expression that is often disrupted in cancer. While loss of imprinting (LOI) has been reported for two genes in prostate cancer (IGF2 and TFPI2), disease-related changes in methylation across all imprinted gene regions has not been investigated. Methods Using an Illumina Infinium Methylation Assay, we analyzed methylation of 396 CpG sites in the promoter regions of 56 genes in a pooled sample of 12 pairs of prostate tumor and adjacent normal tissue. Selected LOI identified from the array was validated using the Sequenom EpiTYPER assay for individual samples and further confirmed by expression data from publicly available datasets. Results Methylation significantly increased in 52 sites and significantly decreased in 17 sites across 28 unique genes (P < 0.05), and the strongest evidence for loss of imprinting was demonstrated in tumor suppressor genes DLK1, PLAGL1, SLC22A18, TP73, and WT1. Differential expression of these five genes in prostate tumor versus normal tissue using array data from a publicly available database were consistent with the observed LOI patterns, and WT1 hypermethylation was confirmed using quantitative DNA methylation analysis. Conclusions Together, these findings suggest a more widespread dysregulation of genetic imprinting in prostate cancer than previously reported and warrant further investigation.
Collapse
Affiliation(s)
- Daniel I Jacobs
- Yale School of Public Health, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | |
Collapse
|
45
|
Jacobs DI, Hansen J, Fu A, Stevens RG, Tjonneland A, Vogel UB, Zheng T, Zhu Y. Methylation alterations at imprinted genes detected among long-term shiftworkers. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2013; 54:141-146. [PMID: 23193016 PMCID: PMC4993623 DOI: 10.1002/em.21752] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 10/19/2012] [Indexed: 06/02/2023]
Abstract
Exposure to light at night through shiftwork has been linked to alterations in DNA methylation and increased risk of cancer development. Using an Illumina Infinium Methylation Assay, we analyzed methylation levels of 397 CpG sites in the promoter regions of 56 normally imprinted genes to investigate whether shiftwork is associated with alteration of methylation patterns. Methylation was significantly higher at 20 CpG sites and significantly lower at 30 CpG sites (P < 0.05) in 10 female long-term shiftworkers as compared to 10 female age- and folate intake-matched day workers. The strongest evidence for altered methylation patterns in shiftworkers was observed for DLX5, IGF2AS, and TP73 based on the magnitude of methylation change and consistency in the direction of change across multiple CpG sites, and consistent results were observed using quantitative DNA methylation analysis. We conclude that long-term shiftwork may alter methylation patterns at imprinted genes, which may be an important mechanism by which shiftwork has carcinogenic potential and warrants further investigation.
Collapse
Affiliation(s)
- Daniel I. Jacobs
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Johnni Hansen
- Institute of Cancer Epidemiology, Danish Cancer Society, Copenhagen, Denmark
| | - Alan Fu
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | | | - Anne Tjonneland
- Institute of Cancer Epidemiology, Danish Cancer Society, Copenhagen, Denmark
| | - Ulla B. Vogel
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Tongzhang Zheng
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Yong Zhu
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| |
Collapse
|
46
|
Varley KE, Gertz J, Bowling KM, Parker SL, Reddy TE, Pauli-Behn F, Cross MK, Williams BA, Stamatoyannopoulos JA, Crawford GE, Absher DM, Wold BJ, Myers RM. Dynamic DNA methylation across diverse human cell lines and tissues. Genome Res 2013; 23:555-67. [PMID: 23325432 PMCID: PMC3589544 DOI: 10.1101/gr.147942.112] [Citation(s) in RCA: 531] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As studies of DNA methylation increase in scope, it has become evident that methylation has a complex relationship with gene expression, plays an important role in defining cell types, and is disrupted in many diseases. We describe large-scale single-base resolution DNA methylation profiling on a diverse collection of 82 human cell lines and tissues using reduced representation bisulfite sequencing (RRBS). Analysis integrating RNA-seq and ChIP-seq data illuminates the functional role of this dynamic mark. Loci that are hypermethylated across cancer types are enriched for sites bound by NANOG in embryonic stem cells, which supports and expands the model of a stem/progenitor cell signature in cancer. CpGs that are hypomethylated across cancer types are concentrated in megabase-scale domains that occur near the telomeres and centromeres of chromosomes, are depleted of genes, and are enriched for cancer-specific EZH2 binding and H3K27me3 (repressive chromatin). In noncancer samples, there are cell-type specific methylation signatures preserved in primary cell lines and tissues as well as methylation differences induced by cell culture. The relationship between methylation and expression is context-dependent, and we find that CpG-rich enhancers bound by EP300 in the bodies of expressed genes are unmethylated despite the dense gene-body methylation surrounding them. Non-CpG cytosine methylation occurs in human somatic tissue, is particularly prevalent in brain tissue, and is reproducible across many individuals. This study provides an atlas of DNA methylation across diverse and well-characterized samples and enables new discoveries about DNA methylation and its role in gene regulation and disease.
Collapse
|
47
|
Pogribny IP, Rusyn I. Environmental toxicants, epigenetics, and cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 754:215-32. [PMID: 22956504 PMCID: PMC4281087 DOI: 10.1007/978-1-4419-9967-2_11] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumorigenesis, a complex and multifactorial progressive process of transformation of normal cells into malignant cells, is characterized by the accumulation of multiple cancer-specific heritable phenotypes triggered by the mutational and/or non-mutational (i.e., epigenetic) events. Accumulating evidence suggests that environmental and occupational exposures to natural substances, as well as man-made chemical and physical agents, play a causative role in human cancer. In a broad sense, carcinogenesis may be induced through either genotoxic or non-genotoxic mechanisms; however, both genotoxic and non-genotoxic carcinogens also cause prominent epigenetic changes. This review presents current evidence of the epigenetic alterations induced by various chemical carcinogens, including arsenic, 1,3-butadine, and pharmaceutical and biological agents, and highlights the potential for epigenetic changes to serve as markers for carcinogen exposure and cancer risk assessment.
Collapse
Affiliation(s)
- Igor P. Pogribny
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA
| | - Ivan Rusyn
- Department of Environmental Sciences & Engineering, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
48
|
Bogen KT. Efficient tumorigenesis by mutation-induced failure to terminate microRNA-mediated adaptive hyperplasia. Med Hypotheses 2012. [PMID: 23183421 DOI: 10.1016/j.mehy.2012.10.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Seven current contending cancer theories consider different sets of critical events as sufficient for tumorigenesis. These theories, most recently the microRNA dysregulation (MRD) theory, have overlapping attributes and extensive empirical support, but also some discrepancies, and some do not address both benign and malignant tumorigenesis. By definition, the most efficient tumorigenic pathways will dominate under conditions that selectively activate those pathways. The MRD theory provides a mechanistic basis to combine elements of the current theories into a new hypothesis that: (i) tumors arise most efficiently under stress that induces and sustains either protective or regenerative states of adaptive hyperplasia (AH) that normally are epigenetically maintained unless terminated; and (ii) if dysregulated by a somatic mutation that prevents normal termination, these two AH states can generate benign and malignant tumors, respectively. This hypothesis, but not multistage cancer theory, predicts that key participating AH-stem-cell populations expand markedly when triggered by stress, particularly chronic metabolic or oxidative stress, mechanical irritation, toxic exposure, wounding, inflammation, and/or infection. This hypothesis predicts that microRNA expression patterns in benign vs. malignant tumor tissue will correlate best with those governing protective vs. regenerative AH in that tissue, and that tumors arise most efficiently inmutagen-exposed stem cells that either happen to be in, or incidentally later become recruited into, an AH state.
Collapse
Affiliation(s)
- Kenneth T Bogen
- DrPH DABT, Exponent Inc., Health Sciences, 475, 14th Street, Ste 400, Oakland, CA 94612, USA.
| |
Collapse
|
49
|
Akhtar M, Holmgren C, Göndör A, Vesterlund M, Kanduri C, Larsson C, Ekström TJ. Cell type and context-specific function of PLAG1 for IGF2 P3 promoter activity. Int J Oncol 2012; 41:1959-66. [PMID: 23023303 PMCID: PMC3583874 DOI: 10.3892/ijo.2012.1641] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/14/2012] [Indexed: 12/23/2022] Open
Abstract
The fetal transcription factor PLAG1 is found to be overexpressed in cancers, and has been suggested to bind the insulin like growth factor 2 (IGF2) P3 promoter, and to activate the IGF2 gene. The expression of IGF2 has partly been linked to loss of CTCF-dependent chromatin insulator function at the H19 imprinting control region (ICR). We investigated the role of PLAG1 for IGF2 regulation in Hep3B and JEG-3 cell lines. Chromatin immunoprecipitation revealed cell type-specific binding of PLAG1 to the IGF2 P3 promoter, which was substantially insensitive to recombinant PLAG1 overexpression in the endogenous context. We hypothesized that the H19 chromatin insulator may be involved in the cell type-specific PLAG1 response. By using a GFP reporter gene/insulator assay plasmid construct with and without the H19 ICR and/or an SV40 enhancer, we confirm that the effect of the insulator is specifically associated with the activity of the IGF2 P3 promoter in the GFP reporter system, and furthermore, that the reporter insulator is functional in JEG-3 but not in Hep3B cells. FACS analysis was used to assess the function of PLAG1 in low endogenously expressing, but Zn-inducible stable PLAG1 expressing JEG-3 cell clones. Considerable increase in IGF2 expression upon PLAG1 induction with a partial insulator overriding activity was found using the reporter constructs. This is in contrast to the effect of the endogenous IGF2 gene which was insensitive to PLAG1 expression in JEG-3, while modestly induced the already highly expressed IGF2 gene in Hep3B cells. We suggest that the PLAG1 binding to the IGF2 P3 promoter and IGF2 expression is cell type-specific, and that the PLAG1 transcription factor acts as a transcriptional facilitator that partially overrides the insulation by the H19 ICR.
Collapse
Affiliation(s)
- Monira Akhtar
- Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
50
|
Pogribny IP, Beland FA. DNA methylome alterations in chemical carcinogenesis. Cancer Lett 2012; 334:39-45. [PMID: 23010082 DOI: 10.1016/j.canlet.2012.09.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 08/28/2012] [Accepted: 09/14/2012] [Indexed: 01/30/2023]
Abstract
Carcinogenesis, a complex multifactorial process of the transformation of normal cells into malignant cells, is characterized by many biologically significant and interdependent alterations triggered by the mutational and/or non-mutational (i.e., epigenetic) events. One of these events, specific to all types of cancer, is alterations in DNA methylation. This review summarizes the current knowledge of the role of DNA methylation changes induced by various genotoxic chemicals (carcinogenic agents that interact with DNA) and non-genotoxic carcinogens (chemicals causing tumor by mechanisms other than directly damaging DNA) in the lung, colorectal, liver, and hematologic carcinogenesis. It also emphasizes the potential role for epigenetic changes to serve as markers for carcinogen exposure and carcinogen risk assessment.
Collapse
Affiliation(s)
- Igor P Pogribny
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA.
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA.
| |
Collapse
|