501
|
Saini RK, Nile SH, Keum YS. Folates: Chemistry, analysis, occurrence, biofortification and bioavailability. Food Res Int 2016; 89:1-13. [PMID: 28460896 DOI: 10.1016/j.foodres.2016.07.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/18/2016] [Accepted: 07/22/2016] [Indexed: 01/27/2023]
Abstract
Folates (Vitamin B9) include both naturally occurring folates and synthetic folic acid used in fortified foods and dietary supplements. Folate deficiency causes severe abnormalities in one-carbon metabolism can result chronic diseases and developmental disorders, including neural tube defects. Mammalian cells cannot synthesize folates de novo; therefore, diet and dietary supplements are the only way to attain daily folate requirements. In the last decade, significant advancements have been made to enhance the folate content of rice, tomato, common bean and lettuce by using genetic engineering approaches. Strategies have been developed to improve the stability of folate pool in plants. Folate deglutamylation through food processing and thermal treatment has the potential to enhance the bioavailability of folate. This review highlights the recent developments in biosynthesis, composition, bioavailability, enhanced production by elicitation and metabolic engineering, and methods of analysis of folate in food. Additionally, future perspectives in this context are identified. Detailed knowledge of folate biosynthesis, degradation and salvage are the prime requirements to efficiently engineer the plants for the enhancement of overall folate content. Similarly, consumption of a folate-rich diet with enhanced bioavailability is the best way to maintain optimum folate levels in the body.
Collapse
Affiliation(s)
- Ramesh Kumar Saini
- Department of Bioresources and Food Science, College of Life and Environmental Sciences, Konkuk University, Seoul 143-701, Republic of Korea.
| | - Shivraj Hariram Nile
- Department of Bioresources and Food Science, College of Life and Environmental Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - Young-Soo Keum
- Department of Bioresources and Food Science, College of Life and Environmental Sciences, Konkuk University, Seoul 143-701, Republic of Korea.
| |
Collapse
|
502
|
Naz N, Jimenez AR, Sanjuan-Vilaplana A, Gurney M, Miyan J. Neonatal hydrocephalus is a result of a block in folate handling and metabolism involving 10-formyltetrahydrofolate dehydrogenase. J Neurochem 2016; 138:610-23. [DOI: 10.1111/jnc.13686] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 04/29/2016] [Accepted: 05/23/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Naila Naz
- Faculty of Life Sciences; The University of Manchester; Manchester UK
| | | | | | - Megan Gurney
- Faculty of Life Sciences; The University of Manchester; Manchester UK
| | - Jaleel Miyan
- Faculty of Life Sciences; The University of Manchester; Manchester UK
| |
Collapse
|
503
|
Liu Y, Zhi L, Shen J, Li S, Yao J, Yang X. Effect of in ovo folic acid injection on hepatic IGF2 expression and embryo growth of broilers. J Anim Sci Biotechnol 2016; 7:40. [PMID: 27453780 PMCID: PMC4957392 DOI: 10.1186/s40104-016-0099-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 06/28/2016] [Indexed: 11/12/2022] Open
Abstract
Background Insulin-like factor 2 (IGF2) plays an important role in embryonic growth process by modulating intermediary metabolism and cell proliferation. Folic acid is involved in one carbon metabolism and contributes to DNA methylation which is related to gene expression. The purpose of this study was to explore whether folic acid could regulate IGF2 expression via epigenetic mechanism and further promote embryonic growth of new-hatched broilers. Methods In the present study, 360 fertile eggs were selected and randomly assigned to four treatments. On 11 embryonic day of incubation (E11), 0, 50, 100 and 150 μg folic acid were injected into eggs respectively. After hatched, growth performance of broilers were calculated. Hepatic IGF2 expression, methylation level and chromatin structure of promoter region were analyzed. Results Results have showed that IGF2 expression was up-regulated in 150 μg folic acid group (P < 0.05) and other two dose of folic acid did not affect gene expression (P > 0.05). Meanwhile, methylation level of IGF2 promoter were lower in 100 and 150 μg groups, which was consistent with lower expression of DNA methyltransferase 1 (DNMT1) (P < 0.05). What’s more, chromatin looseness of IGF2 promoter was higher in 150 μg group than control group (P < 0.05). Further, birth weight (BW), liver and bursa index of new-hatched chickens in 150 μg folic acid group were higher than the other groups (P < 0.05). There were positive correlations between hepatic IGF2 expression and BW and organs index (P < 0.05). Conclusion In conclusion, our data have demonstrated that 150 μg folic acid injection on E11 could up-regulate IGF2 expression by modulating DNA hypomethylation and improving chromatin accessibility in the gene promoter region, and ulteriorly facilitate embryonic growth and organ development of broilers. Electronic supplementary material The online version of this article (doi:10.1186/s40104-016-0099-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 China
| | - Lihui Zhi
- School of Mathematics and Computer Science, ShanXi Normal University, Linfen, 041000 China
| | - Jing Shen
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 China
| | - Shizhao Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 China
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 China
| |
Collapse
|
504
|
Characterization of nutritionally important phytoconstituents in bitter melon (Momordica charantia L.) fruits by HPLC–DAD and GC–MS. JOURNAL OF FOOD MEASUREMENT AND CHARACTERIZATION 2016. [DOI: 10.1007/s11694-016-9378-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
505
|
Shojaei Saadi HA, Gagné D, Fournier É, Baldoceda Baldeon LM, Sirard MA, Robert C. Responses of bovine early embryos to S-adenosyl methionine supplementation in culture. Epigenomics 2016; 8:1039-60. [PMID: 27419740 DOI: 10.2217/epi-2016-0022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM There is a growing concern about the potential adverse effects of high dose folic acid (FA) supplementation before and during pregnancy. FA metabolism generates S-adenosyl methionine (SAM) which is an important cofactor of epigenetic programming. We sought to assess the impact of a large dose of SAM on early embryo development. MATERIALS & METHODS In vitro cultured bovine embryos were treated with SAM from the eight-cell stage to the blastocyst stage. In addition to the phenotype, the genome-wide epigenetic and transcription profiles were analyzed. RESULTS Treatment significantly improved embryo hatching and caused a shift in sex ratio in favor of males. SAM caused genome-wide hypermethylation mainly in exonic regions and in CpG islands. Although differentially expressed genes were associated with response to nutrients and developmental processes, no correspondence was found with the differentially methylated regions, suggesting that cellular responses to SAM treatment during early embryo development may not require DNA methylation-driven changes. CONCLUSION Since bovine embryos were not indifferent to SAM, effects of large-dose FA supplements on early embryonic development in humans cannot be ruled out.
Collapse
Affiliation(s)
- Habib A Shojaei Saadi
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Département des sciences animales, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Dominic Gagné
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Département des sciences animales, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Éric Fournier
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Département des sciences animales, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Luis Manuel Baldoceda Baldeon
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Département des sciences animales, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Marc-André Sirard
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Département des sciences animales, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Claude Robert
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Département des sciences animales, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, QC, G1V 0A6, Canada
| |
Collapse
|
506
|
Saa PA, Nielsen LK. Construction of feasible and accurate kinetic models of metabolism: A Bayesian approach. Sci Rep 2016; 6:29635. [PMID: 27417285 PMCID: PMC4945864 DOI: 10.1038/srep29635] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/20/2016] [Indexed: 12/24/2022] Open
Abstract
Kinetic models are essential to quantitatively understand and predict the behaviour of metabolic networks. Detailed and thermodynamically feasible kinetic models of metabolism are inherently difficult to formulate and fit. They have a large number of heterogeneous parameters, are non-linear and have complex interactions. Many powerful fitting strategies are ruled out by the intractability of the likelihood function. Here, we have developed a computational framework capable of fitting feasible and accurate kinetic models using Approximate Bayesian Computation. This framework readily supports advanced modelling features such as model selection and model-based experimental design. We illustrate this approach on the tightly-regulated mammalian methionine cycle. Sampling from the posterior distribution, the proposed framework generated thermodynamically feasible parameter samples that converged on the true values, and displayed remarkable prediction accuracy in several validation tests. Furthermore, a posteriori analysis of the parameter distributions enabled appraisal of the systems properties of the network (e.g., control structure) and key metabolic regulations. Finally, the framework was used to predict missing allosteric interactions.
Collapse
Affiliation(s)
- Pedro A. Saa
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Lars K. Nielsen
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
507
|
Walker CL. Minireview: Epigenomic Plasticity and Vulnerability to EDC Exposures. Mol Endocrinol 2016; 30:848-55. [PMID: 27355193 DOI: 10.1210/me.2016-1086] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The epigenome undergoes significant remodeling during tissue and organ development, which coincides with a period of exquisite sensitivity to environmental exposures. In the case of endocrine-disrupting compounds (EDCs), exposures can reprogram the epigenome of developing tissues to increase susceptibility to diseases later in life, a process termed "developmental reprogramming." Both DNA methylation and histone modifications have been shown to be vulnerable to disruption by EDC exposures, and several mechanisms have been identified by which EDCs can reprogram the epigenome. These include altered methyl donor availability, loss of imprinting control, changes in dioxygenase activity, altered expression of noncoding RNAs, and activation of cell signaling pathways that can phosphorylate, and alter the activity of, histone methyltransferases. This altered epigenomic programming can persist across the life course, and in some instances generations, to alter gene expression in ways that correlate with increased disease susceptibility. Together, these studies on developmental reprogramming of the epigenome by EDCs are providing new insights into epigenomic plasticity that is vulnerable to disruption by environmental exposures.
Collapse
Affiliation(s)
- Cheryl Lyn Walker
- Institute of Biosciences and Technology, Texas A&M University System Health Science Center, Houston, Texas 77030
| |
Collapse
|
508
|
Bhattacharyya S, Varshney U. Evolution of initiator tRNAs and selection of methionine as the initiating amino acid. RNA Biol 2016; 13:810-9. [PMID: 27322343 DOI: 10.1080/15476286.2016.1195943] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Transfer RNAs (tRNAs) have been important in shaping biomolecular evolution. Initiator tRNAs (tRNAi), a special class of tRNAs, carry methionine (or its derivative, formyl-methionine) to ribosomes to start an enormously energy consuming but a highly regulated process of protein synthesis. The processes of tRNAi evolution, and selection of methionine as the universal initiating amino acid remain an enigmatic problem. We constructed phylogenetic trees using the whole sequence, the acceptor-TψC arm ('minihelix'), and the anticodon-dihydrouridine arm regions of tRNAi from 158 species belonging to all 3 domains of life. All the trees distinctly assembled into 3 domains of life. Large trees, generated using data for all the tRNAs of a vast number of species, fail to reveal the major evolutionary events and identity of the probable elongator tRNA sequences that could be ancestor of tRNAi. Therefore, we constructed trees using the minihelix or the whole sequence of species specific tRNAs, and iterated our analysis on 50 eubacterial species. We identified tRNA(Pro), tRNA(Glu), or tRNA(Thr) (but surprisingly not elongator tRNA(Met)) as probable ancestors of tRNAi. We then determined the factors imposing selection of methionine as the initiating amino acid. Overall frequency of occurrence of methionine, whose metabolic cost of synthesis is the highest among all amino acids, remains almost unchanged across the 3 domains of life. Our correlation analysis shows that its high metabolic cost is independent of many physicochemical properties of the side chain. Our results indicate that selection of methionine, as the initiating amino acid was possibly a consequence of the evolution of one-carbon metabolism, which plays an important role in regulating translation initiation.
Collapse
Affiliation(s)
- Souvik Bhattacharyya
- a Department of Microbiology and Cell Biology , Indian Institute of Science , Bangalore , India
| | - Umesh Varshney
- a Department of Microbiology and Cell Biology , Indian Institute of Science , Bangalore , India.,b Jawaharlal Nehru Center for Advanced Scientific Research, Jakkur , Bangalore , India
| |
Collapse
|
509
|
Andlauer TFM, Buck D, Antony G, Bayas A, Bechmann L, Berthele A, Chan A, Gasperi C, Gold R, Graetz C, Haas J, Hecker M, Infante-Duarte C, Knop M, Kümpfel T, Limmroth V, Linker RA, Loleit V, Luessi F, Meuth SG, Mühlau M, Nischwitz S, Paul F, Pütz M, Ruck T, Salmen A, Stangel M, Stellmann JP, Stürner KH, Tackenberg B, Then Bergh F, Tumani H, Warnke C, Weber F, Wiendl H, Wildemann B, Zettl UK, Ziemann U, Zipp F, Arloth J, Weber P, Radivojkov-Blagojevic M, Scheinhardt MO, Dankowski T, Bettecken T, Lichtner P, Czamara D, Carrillo-Roa T, Binder EB, Berger K, Bertram L, Franke A, Gieger C, Herms S, Homuth G, Ising M, Jöckel KH, Kacprowski T, Kloiber S, Laudes M, Lieb W, Lill CM, Lucae S, Meitinger T, Moebus S, Müller-Nurasyid M, Nöthen MM, Petersmann A, Rawal R, Schminke U, Strauch K, Völzke H, Waldenberger M, Wellmann J, Porcu E, Mulas A, Pitzalis M, Sidore C, Zara I, Cucca F, Zoledziewska M, Ziegler A, Hemmer B, Müller-Myhsok B. Novel multiple sclerosis susceptibility loci implicated in epigenetic regulation. SCIENCE ADVANCES 2016; 2:e1501678. [PMID: 27386562 PMCID: PMC4928990 DOI: 10.1126/sciadv.1501678] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/27/2016] [Indexed: 05/11/2023]
Abstract
We conducted a genome-wide association study (GWAS) on multiple sclerosis (MS) susceptibility in German cohorts with 4888 cases and 10,395 controls. In addition to associations within the major histocompatibility complex (MHC) region, 15 non-MHC loci reached genome-wide significance. Four of these loci are novel MS susceptibility loci. They map to the genes L3MBTL3, MAZ, ERG, and SHMT1. The lead variant at SHMT1 was replicated in an independent Sardinian cohort. Products of the genes L3MBTL3, MAZ, and ERG play important roles in immune cell regulation. SHMT1 encodes a serine hydroxymethyltransferase catalyzing the transfer of a carbon unit to the folate cycle. This reaction is required for regulation of methylation homeostasis, which is important for establishment and maintenance of epigenetic signatures. Our GWAS approach in a defined population with limited genetic substructure detected associations not found in larger, more heterogeneous cohorts, thus providing new clues regarding MS pathogenesis.
Collapse
Affiliation(s)
- Till F. M. Andlauer
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Dorothea Buck
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Gisela Antony
- Central Information Office KKNMS, Philipps University Marburg, 35043 Marburg, Germany
| | - Antonios Bayas
- Department of Neurology, Klinikum Augsburg, 86156 Augsburg, Germany
| | - Lukas Bechmann
- Department of Neurology, University of Leipzig, 04103 Leipzig, Germany
- Institute of Medical Microbiology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Achim Berthele
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Andrew Chan
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Neurology, University Hospital Bern and University of Bern, 3010 Bern, Switzerland
| | - Christiane Gasperi
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Christiane Graetz
- Department of Neurology, Focus Program Translational Neurosciences (FTN) and Research Center for Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Jürgen Haas
- Department of Neurology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Michael Hecker
- Department of Neurology, University of Rostock, 18147 Rostock, Germany
| | - Carmen Infante-Duarte
- NeuroCure Clinical Research Center, Department of Neurology, and Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine, and Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Matthias Knop
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, Ludwigs-Maximilians-Universität, 81377 Munich, Germany
| | - Volker Limmroth
- Department of Neurology, Hospital Köln-Merheim, 51109 Köln, Germany
| | - Ralf A. Linker
- Department of Neurology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Verena Loleit
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Felix Luessi
- Department of Neurology, Focus Program Translational Neurosciences (FTN) and Research Center for Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Sven G. Meuth
- Department of Neurology, Klinik für Allgemeine Neurologie, University of Münster, 48149 Münster, Germany
| | - Mark Mühlau
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | | | - Friedemann Paul
- NeuroCure Clinical Research Center, Department of Neurology, and Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine, and Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Michael Pütz
- Clinical Neuroimmunology Group, Department of Neurology, Philipps-University of Marburg, 35043 Marburg, Germany
| | - Tobias Ruck
- Department of Neurology, Klinik für Allgemeine Neurologie, University of Münster, 48149 Münster, Germany
| | - Anke Salmen
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Neurology, University Hospital Bern and University of Bern, 3010 Bern, Switzerland
| | - Martin Stangel
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Jan-Patrick Stellmann
- Institute of Neuroimmunology and Multiple Sclerosis and Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Klarissa H. Stürner
- Institute of Neuroimmunology and Multiple Sclerosis and Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Björn Tackenberg
- Clinical Neuroimmunology Group, Department of Neurology, Philipps-University of Marburg, 35043 Marburg, Germany
| | - Florian Then Bergh
- Department of Neurology and Translational Center for Regenerative Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Hayrettin Tumani
- Department of Neurology, University of Ulm, 89081 Ulm, Germany
- Neurological Clinic Dietenbronn, 88477 Schwendi, Germany
| | - Clemens Warnke
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Frank Weber
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Neurological Clinic, Medical Park, 65520 Bad Camberg, Germany
| | - Heinz Wiendl
- Department of Neurology, Klinik für Allgemeine Neurologie, University of Münster, 48149 Münster, Germany
| | - Brigitte Wildemann
- Department of Neurology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Uwe K. Zettl
- Department of Neurology, University of Rostock, 18147 Rostock, Germany
| | - Ulf Ziemann
- Department of Neurology and Stroke, and Hertie Institute for Clinical Brain Research, Eberhard-Karls-Universität Tübingen, 72076 Tübingen, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neurosciences (FTN) and Research Center for Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Janine Arloth
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Peter Weber
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | | | - Markus O. Scheinhardt
- Institut für Medizinische Biometrie und Statistik, Universität zu Lübeck, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, 23562 Lübeck, Germany
| | - Theresa Dankowski
- Institut für Medizinische Biometrie und Statistik, Universität zu Lübeck, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, 23562 Lübeck, Germany
| | | | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, 81675 Munich, Germany
| | - Darina Czamara
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | | | - Elisabeth B. Binder
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30329, USA
| | - Klaus Berger
- Institut für Epidemiologie und Sozialmedizin der Universität Münster, 48149 Münster, Germany
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Integrative and Experimental Genomics, University of Lübeck, 23562 Lübeck, Germany
- School of Public Health, Faculty of Medicine, Imperial College London, SW7 2AZ London, UK
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, 24105 Kiel, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Stefan Herms
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany
- Department of Biomedicine, Division of Medical Genetics, University of Basel, 4031 Basel, Switzerland
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University and University Medicine Greifswald, 17475 Greifswald, Germany
| | - Marcus Ising
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Karl-Heinz Jöckel
- Institute of Medical Informatics, Biometry, and Epidemiology, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - Tim Kacprowski
- Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University and University Medicine Greifswald, 17475 Greifswald, Germany
| | - Stefan Kloiber
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Matthias Laudes
- Department I of Internal Medicine, Kiel University, 24105 Kiel, Germany
| | - Wolfgang Lieb
- Institute of Epidemiology and Biobank popgen, Kiel University, 24105 Kiel, Germany
| | - Christina M. Lill
- Department of Neurology, Focus Program Translational Neurosciences (FTN) and Research Center for Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Integrative and Experimental Genomics, University of Lübeck, 23562 Lübeck, Germany
| | - Susanne Lucae
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, 81675 Munich, Germany
| | - Susanne Moebus
- Institute of Medical Informatics, Biometry, and Epidemiology, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Medicine I, Ludwig-Maximilians-Universität, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Markus M. Nöthen
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany
| | - Astrid Petersmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Rajesh Rawal
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Ulf Schminke
- Department of Neurology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry, and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Jürgen Wellmann
- Institut für Epidemiologie und Sozialmedizin der Universität Münster, 48149 Münster, Germany
| | - Eleonora Porcu
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, 09042 Cagliari, Italy
| | - Antonella Mulas
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, 09042 Cagliari, Italy
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, 07100 Sassari, Italy
| | - Maristella Pitzalis
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, 09042 Cagliari, Italy
| | - Carlo Sidore
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, 09042 Cagliari, Italy
| | - Ilenia Zara
- Center for Advanced Studies, Research and Development in Sardinia (CRS4), Pula, 09010 Cagliari, Italy
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, 09042 Cagliari, Italy
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, 07100 Sassari, Italy
| | - Magdalena Zoledziewska
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, 09042 Cagliari, Italy
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, 07100 Sassari, Italy
| | - Andreas Ziegler
- Institut für Medizinische Biometrie und Statistik, Universität zu Lübeck, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, 23562 Lübeck, Germany
- Zentrum für Klinische Studien, Universität zu Lübeck, 23562 Lübeck, Germany
- School of Mathematics, Statistics, and Computer Science, University of KwaZulu-Natal, Pietermaritzburg, Scottsville 3209, South Africa
| | - Bernhard Hemmer
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Corresponding author. (B.H.); (B.M.-M.)
| | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK
- Corresponding author. (B.H.); (B.M.-M.)
| |
Collapse
|
510
|
Tannock GW, Lee PS, Wong KH, Lawley B. Why Don't All Infants Have Bifidobacteria in Their Stool? Front Microbiol 2016; 7:834. [PMID: 27303402 PMCID: PMC4886621 DOI: 10.3389/fmicb.2016.00834] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/17/2016] [Indexed: 01/03/2023] Open
Affiliation(s)
- Gerald W Tannock
- Department of Microbiology and Immunology, University of OtagoDunedin, New Zealand; Riddet Centre for Research Excellence, Massey UniversityPalmerston North, New Zealand
| | - Pheng Soon Lee
- Mead Johnson NutritionSingapore, Singapore; Department of Human Nutrition, University of OtagoDunedin, New Zealand
| | | | - Blair Lawley
- Department of Microbiology and Immunology, University of Otago Dunedin, New Zealand
| |
Collapse
|
511
|
Lendvai Á, Deutsch MJ, Plösch T, Ensenauer R. The peroxisome proliferator-activated receptors under epigenetic control in placental metabolism and fetal development. Am J Physiol Endocrinol Metab 2016; 310:E797-810. [PMID: 26860983 DOI: 10.1152/ajpendo.00372.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 02/02/2016] [Indexed: 01/09/2023]
Abstract
The placental metabolism can adapt to the environment throughout pregnancy to both the demands of the fetus and the signals from the mother. Such adaption processes include epigenetic mechanisms, which alter gene expression and may influence the offspring's health. These mechanisms are linked to the diversity of prenatal environmental exposures, including maternal under- or overnutrition or gestational diabetes. The peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that contribute to the developmental plasticity of the placenta by regulating lipid and glucose metabolism pathways, including lipogenesis, steroidogenesis, glucose transporters, and placental signaling pathways, thus representing a link between energy metabolism and reproduction. Among the PPAR isoforms, PPARγ appears to be the main modulator of mammalian placentation. Certain fatty acids and lipid-derived moieties are the natural activating PPAR ligands. By controlling the amounts of maternal nutrients that go across to the fetus, the PPARs play an important regulatory role in placenta metabolism, thereby adapting to the maternal nutritional status. As demonstrated in animal studies, maternal nutrition during gestation can exert long-term influences on the PPAR methylation pattern in offspring organs. This review underlines the current state of knowledge on the relationship between environmental factors and the epigenetic regulation of the PPARs in placenta metabolism and offspring development.
Collapse
Affiliation(s)
- Ágnes Lendvai
- Center for Liver, Digestive, and Metabolic Diseases, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manuel J Deutsch
- Research Center, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Torsten Plösch
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands;
| | - Regina Ensenauer
- Research Center, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany; Experimental Pediatrics, Department of General Pediatrics, Pediatric Cardiology, and Neonatology, Heinrich-Heine-University Düsseldorf, Dusseldorf, Germany
| |
Collapse
|
512
|
Wang L, Shangguan S, Chang S, Yu X, Wang Z, Lu X, Wu L, Zhang T. Determining the association between methylenetetrahydrofolate reductase (MTHFR) gene polymorphisms and genomic DNA methylation level: A meta-analysis. ACTA ACUST UNITED AC 2016; 106:667-74. [PMID: 27173682 DOI: 10.1002/bdra.23511] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND The methylenetetrahydrofolate reductase (MTHFR) polymorphism is a risk factor for neural tube defects. C677T and A1298C MTHFR polymorphisms produce an enzyme with reduced folate-related one carbon metabolism, and this has been associated with aberrant methylation modifications in DNA and protein. METHODS A meta-analysis was conducted to assess the association between MTHFR C677T/A1298C genotypes and global genomic methylation. RESULTS Eleven studies met the inclusion criteria. Of these, 10 were performed on C677T MTHFR genotypes and 6 were performed on A1298C MTHFR genotypes. Our results did not indicate any correlation between global methylation and MTHFR A1298C, C677T polymorphisms. CONCLUSION The results of our study provide evidence to assess the global methylation modification alterations of MTHFR polymorphisms among individuals. However, our data did not found any conceivable proof supporting the hypothesis that common variant of MTHFR A1298C, C677T contributes to methylation modification. Birth Defects Research (Part A) 106:667-674, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Li Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Shaofang Shangguan
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Shaoyan Chang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Xin Yu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Zhen Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Xiaolin Lu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Lihua Wu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Ting Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
513
|
Willermet C. Biological Anthropology in 2015: Open Access, Biocultural Interactions, and Social Change. AMERICAN ANTHROPOLOGIST 2016. [DOI: 10.1111/aman.12529] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Cathy Willermet
- Department of Sociology, Anthropology, and Social Work; Central Michigan University; Mount Pleasant MI 48859
| |
Collapse
|
514
|
Ahmad Najar R, Rahat B, Hussain A, Thakur S, Kaur J, Kaur J, Hamid A. Gene specific epigenetic regulation of hepatic folate transport system is responsible for perturbed cellular folate status during aging and exogenous modulation. Mol Nutr Food Res 2016; 60:1501-13. [PMID: 26990146 DOI: 10.1002/mnfr.201500991] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/05/2016] [Accepted: 03/09/2016] [Indexed: 01/11/2023]
Abstract
SCOPE The present study was designed to identify the molecular mechanism of folate modulation and aging on aberrant liver folate transporter system. METHODS AND RESULTS An in vivo rat model was used, in which weanling, young and adult rats were given folate deficient diet for 3 and 5 months and after 3 months of folate deficiency, one group received physiological folate repletion (2 mg/kg diet) and another group received over supplemented folate diet (8 mg/kg diet) for another 2 months. In adult group, 3 and 5 months of folate deficiency decreased serum and tissue folate levels with decreased uptake of folate, further associated with decreased expression levels of reduced folate carrier (RFC) and increased expression levels of folate exporter (ABCG2) at both mRNA and protein levels, which in turn regulated by promoter hypermethylation of RFC and promoter hypomethylation of ABCG2 gene. CONCLUSION Promoter hypermethylation of RFC and promoter hypomethylation of ABCG2 may be attributed to the down regulation of RFC and up regulation of ABCG2 at mRNA and protein levels in conditions of 3 and 5 months of folate deficiency in the adult group.
Collapse
Affiliation(s)
- Rauf Ahmad Najar
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Department of Biochemistry, Panjab University, Chandigarh, India
| | - Beenish Rahat
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Aashiq Hussain
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Shilpa Thakur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jaspreet Kaur
- University Institute of Engineering and Technology, Panjab University, Chandigarh, India
| | - Jyotdeep Kaur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Abid Hamid
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,CSIR-Academy of Scientific & Innovative Research, New Delhi, India
| |
Collapse
|
515
|
Karimian M, Hosseinzadeh Colagar A. Methionine synthase A2756G transition might be a risk factor for male infertility: Evidences from seven case-control studies. Mol Cell Endocrinol 2016; 425:1-10. [PMID: 26905524 DOI: 10.1016/j.mce.2016.02.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 02/14/2016] [Accepted: 02/15/2016] [Indexed: 12/21/2022]
Abstract
Methionine synthase (MTR) has a crucial role in DNA synthesis and methylation reactions. The aim of this study was to investigate the association of the MTR-A2756G polymorphism with idiopathic male infertility. Blood samples were collected from 217 idiopathic infertile- and 233 healthy-men, and MTR-A2756G genotyping was performed by PCR-RFLP. Meta-analysis was conducted by pooling our data with the data obtained from 6 previous studies. Also, the effects of this substitution on protein structure were evaluated by bioinformatics tools. Our study revealed the association of AG-genotype, GG-genotype, and G-allele with male infertility. Meta-analysis showed a significant association between A2756G transition and male infertility. In addition, structural analysis of the transition effect on protein revealed a significant influence on MTR function (with score: 38; expected accuracy: 66%). These findings suggest that the A2756G substitution might be a genetic risk factor and a potential biomarker for idiopathic male infertility.
Collapse
Affiliation(s)
- Mohammad Karimian
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Abasalt Hosseinzadeh Colagar
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran; Nano and Biotechnology Research Group, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran.
| |
Collapse
|
516
|
Kennedy BE, Hundert AS, Goguen D, Weaver ICG, Karten B. Presymptomatic Alterations in Amino Acid Metabolism and DNA Methylation in the Cerebellum of a Murine Model of Niemann-Pick Type C Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1582-97. [PMID: 27083515 DOI: 10.1016/j.ajpath.2016.02.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 02/08/2016] [Accepted: 02/16/2016] [Indexed: 10/21/2022]
Abstract
The fatal neurodegenerative disorder Niemann-Pick type C (NPC) is caused in most cases by mutations in NPC1, which encodes the late endosomal NPC1 protein. Loss of NPC1 disrupts cholesterol trafficking from late endosomes to the endoplasmic reticulum and plasma membrane, causing cholesterol accumulation in late endosomes/lysosomes. Neurons are particularly vulnerable to this cholesterol trafficking defect, but the pathogenic mechanisms through which NPC1 deficiency causes neuronal dysfunction remain largely unknown. Herein, we have investigated amino acid metabolism in cerebella of NPC1-deficient mice at different stages of NPC disease. Imbalances in amino acid metabolism were evident from increased branched chain amino acid and asparagine levels and altered expression of key enzymes of glutamine/glutamate metabolism in presymptomatic and early symptomatic NPC1-deficient cerebellum. Increased levels of several amino acid intermediates of one-carbon metabolism indicated disturbances in folate and methylation pathways. Alterations in DNA methylation were apparent in decreased expression of DNA methyltransferase 3a and methyl-5'-cytosine-phosphodiester-guanine-domain binding proteins, reduced 5-methylcytosine immunoreactivity in the molecular and Purkinje cell layers, demethylation of genome-wide repetitive LINE-1 elements, and hypermethylation in specific promoter regions of single-copy genes in NPC1-deficient cerebellum at early stages of the disease. Alterations in amino acid metabolism and epigenetic changes in the cerebellum at presymptomatic stages of NPC disease represent previously unrecognized mechanisms of NPC pathogenesis.
Collapse
Affiliation(s)
- Barry E Kennedy
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Amos S Hundert
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Donna Goguen
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ian C G Weaver
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Barbara Karten
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
517
|
Comparative shotgun proteomic analysis of wild and domesticated Opuntia spp. species shows a metabolic adaptation through domestication. J Proteomics 2016; 143:353-364. [PMID: 27072113 DOI: 10.1016/j.jprot.2016.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 01/01/2023]
Abstract
UNLABELLED The Opuntia genus is widely distributed in America, but the highest richness of wild species are found in Mexico, as well as the most domesticated Opuntia ficus-indica, which is the most domesticated species and an important crop in agricultural economies of arid and semiarid areas worldwide. During domestication process, the Opuntia morphological characteristics were favored, such as less and smaller spines in cladodes and less seeds in fruits, but changes at molecular level are almost unknown. To obtain more insights about the Opuntia molecular changes through domestication, a shotgun proteomic analysis and database-dependent searches by homology was carried out. >1000 protein species were identified and by using a label-free quantitation method, the Opuntia proteomes were compared in order to identify differentially accumulated proteins among wild and domesticated species. Most of the changes were observed in glucose, secondary, and 1C metabolism, which correlate with the observed protein, fiber and phenolic compounds accumulation in Opuntia cladodes. Regulatory proteins, ribosomal proteins, and proteins related with response to stress were also observed in differential accumulation. These results provide new valuable data that will help to the understanding of the molecular changes of Opuntia species through domestication. BIOLOGICAL SIGNIFICANCE Opuntia species are well adapted to dry and warm conditions in arid and semiarid regions worldwide, and they are highly productive plants showing considerable promises as an alternative food source. However, there is a gap regarding Opuntia molecular mechanisms that enable them to grow in extreme environmental conditions and how the domestication processes has changed them. In the present study, a shotgun analysis was carried out to characterize the proteomes of five Opuntia species selected by its domestication degree. Our results will help to a better understanding of proteomic features underlying the selection and specialization under evolution and domestication of Opuntia and will provide a platform for basic biology research and gene discovery.
Collapse
|
518
|
Orozco AM, Yeung LF, Guo J, Carriquiry A, Berry RJ. Characteristics of U.S. Adults with Usual Daily Folic Acid Intake above the Tolerable Upper Intake Level: National Health and Nutrition Examination Survey, 2003-2010. Nutrients 2016; 8:195. [PMID: 27043623 PMCID: PMC4848664 DOI: 10.3390/nu8040195] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/11/2016] [Accepted: 03/22/2016] [Indexed: 11/16/2022] Open
Abstract
The Food and Drug Administration mandated that by 1998, all enriched cereal grain products (ECGP) be fortified with folic acid in order to prevent the occurrence of neural tube defects. The Institute of Medicine established the tolerable upper intake level (UL) for folic acid (1000 µg/day for adults) in 1998. We characterized U.S. adults with usual daily folic acid intake exceeding the UL. Using NHANES 2003–2010 data, we estimated the percentage of 18,321 non-pregnant adults with usual daily folic acid intake exceeding the UL, and among them, we calculated the weighted percentage by sex, age, race/ethnicity, sources of folic acid intake, supplement use and median usual daily folic acid intakes. Overall, 2.7% (standard error 0.6%) of participants had usual daily intake exceeding the UL for folic acid; 62.2% were women; 86.3% were non-Hispanic whites; and 98.5% took supplements containing folic acid. When stratified by sex and age groups among those with usual daily folic acid intake exceeding the UL, 20.8% were women aged 19–39 years. Those with usual daily intake exceeding the folic acid UL were more likely to be female, non-Hispanic white, supplement users or to have at least one chronic medical condition compared to those not exceeding the folic acid UL. Among those with usual daily folic acid intake exceeding the UL who also took supplements, 86.6% took on average >400 µg of folic acid/day from supplements. Everyone with usual daily folic acid intake exceeding the UL consumed folic acid from multiple sources. No one in our study population had usual daily folic acid intake exceeding the UL through consumption of mandatorily-fortified enriched cereal grain products alone. Voluntary consumption of supplements containing folic acid is the main factor associated with usual daily intake exceeding the folic acid UL.
Collapse
Affiliation(s)
- Angela M Orozco
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
- David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA 90095, USA.
| | - Lorraine F Yeung
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | - Jing Guo
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | - Alicia Carriquiry
- Department of Statistics, Iowa State University, Ames, IA 50011, USA.
| | - Robert J Berry
- Division of Congenital and Developmental Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| |
Collapse
|
519
|
|
520
|
Liu Y, Guo W, Pu Z, Li X, Lei X, Yao J, Yang X. Developmental changes of Insulin-like growth factors in the liver and muscle of chick embryos. Poult Sci 2016; 95:1396-402. [PMID: 26944971 DOI: 10.3382/ps/pew043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/18/2015] [Indexed: 01/09/2023] Open
Abstract
The insulin-like growth factors ( IGFS: ) are synthesized in tissues and play an important role in embryonic development of avian via autocrine/paracrine mechanisms. In the study, mRNA expression of IGFs were detected by real-time PCR in the muscle and liver from d 10 to 20 of chick embryo ( E10: to E20: ). Methylation of IGF1 promoter in the muscle was analyzed by bisulfite sequencing PCR as well as IGF2 promoter in the liver. These results showed that there was obviously IGF1 expression in liver at E19 and E20. The higher IGF1 expression in muscle was found during E15 to E18 with the peak on E17, and then declined. Correspondingly, the lowest methylation level of IGF1 promoter was detectable on the same embryonic d 17. Expression of IGF2 in muscle increased gradually during embryonic growth and showed higher level in the later stages (E17 to E20) when IGF1 expression began to decrease. IGF2 expression in liver reached the first peak on E14, then declined but gradually elevated from E17. IGF2 promoter methylation in liver showed gradual decline on d 12, 15, 17 and 19 of incubation, meanwhile IGF2 expression of liver increased gradually. These results suggested that IGF1 and IGF2 might separately be more important for muscle and liver growth in chick embryonic development. Variation of IGFs expression during the incubation might be concerned with the methylation of gene promoter. The profile of IGFs expression in chick embryonic tissues may be meaningful for understanding organ growth and embryonic development in chick.
Collapse
Affiliation(s)
- Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China, 712100
| | - Wei Guo
- College of Animal Science and Technology, Northwest A&F University, Yangling, China, 712100
| | - Zhenyu Pu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China, 712100
| | - Xueyuan Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, China, 712100
| | - Xinyu Lei
- College of Animal Science and Technology, Northwest A&F University, Yangling, China, 712100
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, China, 712100
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China, 712100
| |
Collapse
|
521
|
Latino-Martel P, Cottet V, Druesne-Pecollo N, Pierre FH, Touillaud M, Touvier M, Vasson MP, Deschasaux M, Le Merdy J, Barrandon E, Ancellin R. Alcoholic beverages, obesity, physical activity and other nutritional factors, and cancer risk: A review of the evidence. Crit Rev Oncol Hematol 2016; 99:308-23. [DOI: 10.1016/j.critrevonc.2016.01.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/18/2015] [Accepted: 01/07/2016] [Indexed: 02/06/2023] Open
|
522
|
Abstract
As with physical conditions, bipolar disorder is likely to be impacted by diet and nutrition. Patients with bipolar disorder have been noted to have relatively unhealthy diets, which may in part be the reason they also have an elevated risk of metabolic syndrome and obesity. An improvement in the quality of the diet should improve a bipolar patient's overall health risk profile, but it may also improve their psychiatric outcomes. New insights into biological dysfunctions that may be present in bipolar disorder have presented new theoretic frameworks for understanding the relationship between diet and bipolar disorder.
Collapse
Affiliation(s)
- John L Beyer
- Duke University Medical Center, Box 3519 DUMC, Room 4082B, Yellow Zone, Duke South Clinics, Durham, NC 27710, USA.
| | - Martha E Payne
- Office of Research Development, Duke University School of Medicine, Davison Building/Green Zone, Suite 410, Durham, NC 27705, USA
| |
Collapse
|
523
|
Sramek M, Neradil J, Sterba J, Veselska R. Non-DHFR-mediated effects of methotrexate in osteosarcoma cell lines: epigenetic alterations and enhanced cell differentiation. Cancer Cell Int 2016; 16:14. [PMID: 26929741 PMCID: PMC4770555 DOI: 10.1186/s12935-016-0289-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/12/2016] [Indexed: 12/24/2022] Open
Abstract
Background Methotrexate is an important chemotherapeutic drug widely known as an inhibitor of dihydrofolate reductase (DHFR) which inhibits the reduction of folic acid. DHFR-mediated effects are apparently responsible for its primary antineoplastic action. However, other non-DHFR-mediated effects of methotrexate have been recently discovered, which might be very useful in the development of new strategies for the treatment of pediatric malignancies. The principal goal of this study was to analyze the possible impact of clinically achievable methotrexate levels on cell proliferation, mechanisms of epigenetic regulation (DNA methylation and histone acetylation), induced differentiation and the expression of differentiation-related genes in six osteosarcoma cell lines. Methods The Saos-2 reference cell line and five other patient-derived osteosarcoma cell lines were chosen for this study. The MTT assay was used to assess cell proliferation, DNA methylation and histone acetylation were detected using ELISA, and western blotting was used for a detailed analysis of histone acetylation. The expression of differentiation-related genes was quantified using RT-qPCR and the course of cell differentiation was evaluated using Alizarin Red S staining, which detects the level of extracellular matrix mineralization. Results Methotrexate significantly decreased the proliferation of Saos-2 cells exclusively, suggesting that this reference cell line was sensitive to the DHFR-mediated effects of methotrexate. In contrast, other results indicated non-DHFR-mediated effects in patient-derived cell lines. Methotrexate-induced DNA demethylation was detected in almost all of them; methotrexate was able to lower the level of 5-methylcytosine in treated cells, and this effect was similar to the effect of 5-aza-2′-deoxycytidine. Furthermore, methotrexate increased the level of acetylated histone H3 in the OSA-06 cell line. Methotrexate also enhanced all-trans retinoic acid-induced cell differentiation in three patient-derived osteosarcoma cell lines, and the modulation of expression of the differentiation-related genes was also shown. Conclusions Overall non-DHFR-mediated effects of methotrexate were detected in the patient-derived osteosarcoma cell lines. Methotrexate acts as an epigenetic modifier and has a potential impact on cell differentiation and the expression of related genes. Furthermore, the combination of methotrexate and all-trans retinoic acid can be effective as a differentiation therapy for osteosarcoma.
Collapse
Affiliation(s)
- Martin Sramek
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, Kotlarska 2, 611 37 Brno, Czech Republic ; Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Cernopolni 9, 613 00 Brno, Czech Republic
| | - Jakub Neradil
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, Kotlarska 2, 611 37 Brno, Czech Republic ; Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Cernopolni 9, 613 00 Brno, Czech Republic
| | - Jaroslav Sterba
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Cernopolni 9, 613 00 Brno, Czech Republic
| | - Renata Veselska
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, Kotlarska 2, 611 37 Brno, Czech Republic ; Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Cernopolni 9, 613 00 Brno, Czech Republic
| |
Collapse
|
524
|
Geraghty AA, Lindsay KL, Alberdi G, McAuliffe FM, Gibney ER. Nutrition During Pregnancy Impacts Offspring's Epigenetic Status-Evidence from Human and Animal Studies. Nutr Metab Insights 2016; 8:41-7. [PMID: 26917970 PMCID: PMC4758803 DOI: 10.4137/nmi.s29527] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 12/14/2022] Open
Abstract
Pregnancy is a vital time of growth and development during which maternal nutrition significantly influences the future health of both mother and baby. During pregnancy, the fetus experiences a critical period of plasticity. Epigenetics, specifically DNA methylation, plays an important role here. As nutrition is influential for DNA methylation, this review aims to determine if maternal nutrition during pregnancy can modify the offspring's epigenome at birth. Research focuses on micronutrients and methyl donors such as folate and B vitamins. Evidence suggests that maternal nutrition does not largely influence global methylation patterns, particularly in nutrient-replete populations; however, an important impact on gene-specific methylation is observed. A link is shown between maternal nutrition and the methylome of the offspring; however, there remains a paucity of research. With the potential to use DNA methylation patterns at birth to predict health of the child in later life, it is vital that further research be carried out.
Collapse
Affiliation(s)
- Aisling A Geraghty
- Department of Obstetrics & Gynaecology, School of Medicine and Medical Science, National Maternity Hospital, University College Dublin, Dublin, Ireland
| | - Karen L Lindsay
- Department of Obstetrics & Gynaecology, School of Medicine and Medical Science, National Maternity Hospital, University College Dublin, Dublin, Ireland
| | - Goiuri Alberdi
- Department of Obstetrics & Gynaecology, School of Medicine and Medical Science, National Maternity Hospital, University College Dublin, Dublin, Ireland
| | - Fionnuala M McAuliffe
- Department of Obstetrics & Gynaecology, School of Medicine and Medical Science, National Maternity Hospital, University College Dublin, Dublin, Ireland
| | - Eileen R Gibney
- UCD Institute of Food and Health, University College Dublin, Dublin, Ireland
| |
Collapse
|
525
|
Shiao SPK, Yu CH. Meta-Prediction of MTHFR Gene Polymorphism Mutations and Associated Risk for Colorectal Cancer. Biol Res Nurs 2016; 18:357-69. [PMID: 26858257 PMCID: PMC4904378 DOI: 10.1177/1099800415628054] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The methylenetetrahydrofolate reductase (MTHFR) gene is one of
the most investigated of the genes associated with chronic human diseases
because of its associations with hyperhomocysteinemia and toxicity. It has been
proposed as a prototype gene for the prevention of colorectal cancer (CRC). The
major objectives of this meta-analysis were to examine the polymorphism-mutation
patterns of MTHFR and their associations with risk for CRC as
well as potential contributing factors for mutations and disease risks. This
analysis included 33,626 CRC cases and 48,688 controls across 92 studies for
MTHFR 677 and 16,367 cases and 24,874 controls across 54
studies for MTHFR 1298, comprising data for various racial and
ethnic groups, both genders, and multiple cancer sites. MTHFR
677 homozygous TT genotype was protective (p < .05) for CRC
for all included populations; however, with heterogeneity across various
racial–ethnic groups and opposing findings, it was a risk genotype for the
subgroup of Hispanics (p < .01). Additional countries for
which subgroup analyses resulted in 677 TT as a risk genotype included Turkey,
Romania, Croatia, Hungary, Portugal, Mexico, Brazil, U.S. Hawai’i, Taiwan,
India, and Egypt. Countries with the highest mutation rates and risks for both
MTHFR 677 and 1298 genotypes are presented using global
maps to visualize the grouping patterns. Meta-predictive analyses revealed that
air pollution levels were associated with gene polymorphisms for both genotypes.
Future nursing research should be conducted to develop proactive measures to
protect populations in cities where air pollution causes more deaths.
Collapse
Affiliation(s)
- S P K Shiao
- College of Nursing, Augusta University, Augusta, GA, USA
| | - C H Yu
- Department of Psychology, Azusa Pacific University, Azusa CA, USA
| |
Collapse
|
526
|
Pennington KL, DeAngelis MM. Epigenetic Mechanisms of the Aging Human Retina. J Exp Neurosci 2016; 9:51-79. [PMID: 26966390 PMCID: PMC4777243 DOI: 10.4137/jen.s25513] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 01/07/2016] [Accepted: 01/13/2016] [Indexed: 12/20/2022] Open
Abstract
Degenerative retinal diseases, such as glaucoma, age-related macular degeneration, and diabetic retinopathy, have complex etiologies with environmental, genetic, and epigenetic contributions to disease pathology. Much effort has gone into elucidating both the genetic and the environmental risk factors for these retinal diseases. However, little is known about how these genetic and environmental risk factors bring about molecular changes that lead to pathology. Epigenetic mechanisms have received extensive attention of late for their promise of bridging the gap between environmental exposures and disease development via their influence on gene expression. Recent studies have identified epigenetic changes that associate with the incidence and/or progression of each of these retinal diseases. Therefore, these epigenetic modifications may be involved in the underlying pathological mechanisms leading to blindness. Further genome-wide epigenetic studies that incorporate well-characterized tissue samples, consider challenges similar to those relevant to gene expression studies, and combine the genome-wide epigenetic data with genome-wide genetic and expression data to identify additional potentially causative agents of disease are needed. Such studies will allow researchers to create much-needed therapeutics to prevent and/or intervene in disease progression. Improved therapeutics will greatly enhance the quality of life and reduce the burden of disease management for millions of patients living with these potentially blinding conditions.
Collapse
Affiliation(s)
- Katie L Pennington
- Postdoctoral Fellow, Department of Ophthalmology & Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Margaret M DeAngelis
- Associate Professor, Department of Ophthalmology & Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
527
|
Shin C, Baik I. Leukocyte Telomere Length is Associated With Serum Vitamin B12 and Homocysteine Levels in Older Adults With the Presence of Systemic Inflammation. Clin Nutr Res 2016; 5:7-14. [PMID: 26839872 PMCID: PMC4731864 DOI: 10.7762/cnr.2016.5.1.7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 01/12/2016] [Accepted: 01/14/2016] [Indexed: 12/21/2022] Open
Abstract
Folate, vitamin B12, and homocysteine (HCY) are involved in the metabolism of nucleic acid precursors and it has been hypothesized that they also influence telomere length, a biomarker of aging. However, previous studies have reported inconsistent findings, and data for older adults are limited. Our study aimed to evaluate associations between leukocyte telomere length (LTL) and serum folate, vitamin B12, and HCY levels among adults aged 55 years and over. In a cross-sectional study in 798 men and women aged 55-79 years, serum folate, vitamin B12, and HCY levels were measured using chemiluminescent immunometric assays, and relative LTL was assessed using quantitative real-time polymerase chain reaction. To evaluate associations between LTL and serum folate, vitamin B12, and HCY levels, multiple linear regression models were used. In multiple models adjusted for age, sex, serum high sensitive C-reactive protein (hs-CRP) levels, and other potential confounding factors, we found no association between LTL and serum folate, vitamin B12, and HCY levels. However, we did find a significant inverse association between HCY levels and LTL in participants with serum hs-CRP levels of ≥ 2 mg/L (p < 0.05). Moreover, there was a trend toward an association between HCY and vitamin B12 levels in these individuals (p = 0.08). In those with serum hs-CRP levels of < 2 mg/L, HCY was inversely associated with vitamin B12 levels (p < 0.001) and had no association with LTL. Our findings suggest that increased serum HCY levels, when combined with the presence of systemic inflammation, may play a role in accelerating biological aging.
Collapse
Affiliation(s)
- Chol Shin
- Department of Internal Medicine, Korea University Ansan Hospital, Ansan, 15355 Korea
| | - Inkyung Baik
- Department of Foods and Nutrition, College of Natural Sciences, Kookmin University, Seoul, 02707 Korea
| |
Collapse
|
528
|
Hsu HC, Chang WM, Wu JY, Huang CC, Lu FJ, Chuang YW, Chang PJ, Chen KH, Hong CZ, Yeh RH, Liu TZ, Chen CH. Folate Deficiency Triggered Apoptosis of Synoviocytes: Role of Overproduction of Reactive Oxygen Species Generated via NADPH Oxidase/Mitochondrial Complex II and Calcium Perturbation. PLoS One 2016; 11:e0146440. [PMID: 26771387 PMCID: PMC4714898 DOI: 10.1371/journal.pone.0146440] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 12/17/2015] [Indexed: 12/31/2022] Open
Abstract
Despite a plethora of literature has documented that osteoarthritis (OA) is veritably associated with oxidative stress-mediated chondrocyte death and matrix degradation, yet the possible involvement of synoviocyte abnormality as causative factor of OA has not been thoroughly investigated. For this reason, we conduct the current studies to insight into how synoviocytes could respond to an episode of folate-deprived (FD) condition. First, when HIG-82 synoviocytes were cultivated under FD condition, a time-dependent growth impediment was observed and the demise of these cells was demonstrated to be apoptotic in nature mediated through FD-evoked overproduction of reactive oxygen species (ROS) and drastically released of cytosolic calcium (Ca2+) concentrations. Next, we uncovered that FD-evoked ROS overproduction could only be strongly suppressed by either mitochondrial complex II inhibitors (TTFA and carboxin) or NADPH oxidase (NOX) inhibitors (AEBSF and apocynin), but not by mitochondrial complex I inhibitor (rotenone) and mitochondrial complex III inhibitor (antimycin A). Interestingly, this selective inhibition of FD-evoked ROS by mitochondrial complex II and NOX inhibitors was found to correlate excellently with the suppression of cytosolic Ca2+ release and reduced the magnitude of the apoptotic TUNEL-positive cells. Taken together, we present the first evidence here that FD-triggered ROS overproduction in synoviocytes is originated from mitochondrial complex II and NOX. Both elevated ROS in tandem with cytosolic Ca2+ overload serve as final arbitrators for apoptotic lethality of synoviocytes cultivated under FD condition. Thus, folate supplementation may be beneficial to patients with OA.
Collapse
Affiliation(s)
- Hung-Chih Hsu
- Department of Physical Medicine and Rehabilitation, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan
- Department of Nursing, Chang-Gung University of Science and Technology, Chia-Yi, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Center of Advanced Integrative Sports Medicine, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan
| | - Wen-Ming Chang
- Department of Physical Medicine and Rehabilitation, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan
- Department of Nursing, Chang-Gung University of Science and Technology, Chia-Yi, Taiwan
| | - Jin-Yi Wu
- Department of Microbiology, Immunology and Biopharmaceuticals, Collage of Life Sciences, National Chiayi University, Chiayi City 60004, Taiwan
| | - Chin-Chin Huang
- Department of Microbiology, Immunology and Biopharmaceuticals, Collage of Life Sciences, National Chiayi University, Chiayi City 60004, Taiwan
| | - Fung-Jou Lu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Wen Chuang
- Department of Physical Medicine and Rehabilitation, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan
- Department of Nursing, Chang-Gung University of Science and Technology, Chia-Yi, Taiwan
| | - Pey-Jium Chang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kai-Hua Chen
- Department of Physical Medicine and Rehabilitation, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan
- Department of Nursing, Chang-Gung University of Science and Technology, Chia-Yi, Taiwan
| | - Chang-Zern Hong
- Department of Physical therapy, Hung Kuang University, Taichung, Taiwan
| | - Rang-Hui Yeh
- Department of Physical Medicine and Rehabilitation, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan
| | - Tsan-Zon Liu
- Translational Research Laboratory, Cancer Center, Taipei Medical University and Hospital, Taipei, Taiwan
- * E-mail: (TZL); (CHC)
| | - Ching-Hsein Chen
- Department of Microbiology, Immunology and Biopharmaceuticals, Collage of Life Sciences, National Chiayi University, Chiayi City 60004, Taiwan
- * E-mail: (TZL); (CHC)
| |
Collapse
|
529
|
Jia L, Li J, He B, Jia Y, Niu Y, Wang C, Zhao R. Abnormally activated one-carbon metabolic pathway is associated with mtDNA hypermethylation and mitochondrial malfunction in the oocytes of polycystic gilt ovaries. Sci Rep 2016; 6:19436. [PMID: 26758245 PMCID: PMC4725837 DOI: 10.1038/srep19436] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/14/2015] [Indexed: 02/07/2023] Open
Abstract
Polycystic ovarian syndrome (PCOS) is associated with hyperhomocysteinemia and polycystic ovaries (PCO) usually produce oocytes of poor quality. However, the intracellular mechanism linking hyperhomocysteinemia and oocyte quality remains elusive. In this study, the quality of the oocytes isolated from healthy and polycystic gilt ovaries was evaluated in vitro in association with one-carbon metabolism, mitochondrial DNA (mtDNA) methylation, and mitochondrial function. PCO oocytes demonstrated impaired polar body extrusion, and significantly decreased cleavage and blastocyst rates. The mitochondrial distribution was disrupted in PCO oocytes, together with decreased mitochondrial membrane potential and deformed mitochondrial structure. The mtDNA copy number and the expression of mtDNA-encoded genes were significantly lower in PCO oocytes. Homocysteine concentration in follicular fluid was significantly higher in PCO group, which was associated with significantly up-regulated one-carbon metabolic enzymes betaine homocysteine methyltransferase (BHMT), glycine N-methyltransferase (GNMT) and the DNA methyltransferase DNMT1. Moreover, mtDNA sequences coding for 12S, 16S rRNA and ND4, as well as the D-loop region were significantly hypermethylated in PCO oocytes. These results indicate that an abnormal activation of one-carbon metabolism and hypermethylation of mtDNA may contribute, largely, to the mitochondrial malfunction and decreased quality of PCO-derived oocytes in gilts.
Collapse
Affiliation(s)
- Longfei Jia
- Key Laboratory of Animal Physiology &Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Juan Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Bin He
- Key Laboratory of Animal Physiology &Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Yimin Jia
- Key Laboratory of Animal Physiology &Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Yingjie Niu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Chenfei Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology &Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China.,Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing 210095, P. R. China
| |
Collapse
|
530
|
François M, Leifert WR, Tellam R, Fenech MF. Folate deficiency and DNA-methyltransferase inhibition modulate G-quadruplex frequency. Mutagenesis 2016; 31:409-16. [DOI: 10.1093/mutage/gev088] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
531
|
Vergote IB, Marth C, Coleman RL. Role of the folate receptor in ovarian cancer treatment: evidence, mechanism, and clinical implications. Cancer Metastasis Rev 2016; 34:41-52. [PMID: 25564455 DOI: 10.1007/s10555-014-9539-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Folate can be transported into the cell by the reduced folate carrier (RFC), the proton-coupled folate transporter (PCFT), or the folate receptor (FR), of which various isoforms exist. While the RFC and PCFT are expressed by many normal cells, the FR is present only in a small proportion of normal tissues. In these tissues, the FR expression level is often low and restricted to the apical surface of polarized epithelial cells. In contrast, FR is expressed on the blood-accessible basal and lateral membranes of many types of epithelial cancer. Considering that FR is expressed in few nonmalignant cell types on luminal membranes generally not accessible for molecules transported in the blood, FR is considered a promising antitumor target. As FR expression seems associated with tumor progression and prognosis, anticancer therapies targeting FR are currently being developed, such as farletuzumab (Morphotek, Exton, PA, USA), IMGN853 (ImmunoGen, Waltham, MA, USA), vintafolide, and EC1456 (both Endocyte Inc., West Lafayette, IN, USA). FR expression could be used as a response-predictive biomarker for these treatments. The ability to identify patients and treat them with an effective therapy based on the known expression of the tumor marker would, indeed, be the next step in predictive medicine for these patients. This review summarizes the role of FR in ovarian cancer and the value of FR as a prognostic biomarker for ovarian cancer and a response-predictive biomarker for folate-targeted therapeutics.
Collapse
Affiliation(s)
- Ignace B Vergote
- Department of Obstetrics and Gynaecolog, Leuven Cancer Institute, University Hospital Leuven, Herestraat 49, 3000, Leuven, Belgium,
| | | | | |
Collapse
|
532
|
ZHANG R, WU K, ZHAN C, LIU X, GONG Z. Folic Acid Supplementation Reduces the Mutagenicity and Genotoxicity Caused by Benzo(a)pyrene. J Nutr Sci Vitaminol (Tokyo) 2016; 62:26-31. [DOI: 10.3177/jnsv.62.26] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Rong ZHANG
- Institute of Food Science and Engineering, WuHan Polytechnic University
| | - Kejia WU
- Institute of Food Science and Engineering, WuHan Polytechnic University
| | - Caigui ZHAN
- Institute of Food Science and Engineering, WuHan Polytechnic University
| | - Xin LIU
- Institute of Food Science and Engineering, WuHan Polytechnic University
| | - Zhiyong GONG
- Institute of Food Science and Engineering, WuHan Polytechnic University
| |
Collapse
|
533
|
Breinig M, Klein FA, Huber W, Boutros M. A chemical-genetic interaction map of small molecules using high-throughput imaging in cancer cells. Mol Syst Biol 2015; 11:846. [PMID: 26700849 PMCID: PMC4704494 DOI: 10.15252/msb.20156400] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Small molecules often affect multiple targets, elicit off-target effects, and induce genotype-specific responses. Chemical genetics, the mapping of the genotype dependence of a small molecule's effects across a broad spectrum of phenotypes can identify novel mechanisms of action. It can also reveal unanticipated effects and could thereby reduce high attrition rates of small molecule development pipelines. Here, we used high-content screening and image analysis to measure effects of 1,280 pharmacologically active compounds on complex phenotypes in isogenic cancer cell lines which harbor activating or inactivating mutations in key oncogenic signaling pathways. Using multiparametric chemical-genetic interaction analysis, we observed phenotypic gene-drug interactions for more than 193 compounds, with many affecting phenotypes other than cell growth. We created a resource termed the Pharmacogenetic Phenome Compendium (PGPC), which enables exploration of drug mode of action, detection of potential off-target effects, and the generation of hypotheses on drug combinations and synergism. For example, we demonstrate that MEK inhibitors amplify the viability effect of the clinically used anti-alcoholism drug disulfiram and show that the EGFR inhibitor tyrphostin AG555 has off-target activity on the proteasome. Taken together, this study demonstrates how combining multiparametric phenotyping in different genetic backgrounds can be used to predict additional mechanisms of action and to reposition clinically used drugs.
Collapse
Affiliation(s)
- Marco Breinig
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany Department of Cell and Molecular Biology, Heidelberg University, Heidelberg, Germany
| | - Felix A Klein
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Wolfgang Huber
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany Department of Cell and Molecular Biology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
534
|
van Mil NH, Bouwland-Both MI, Stolk L, Verbiest MMPJ, Hofman A, Jaddoe VWV, Verhulst FC, Eilers PHC, Uitterlinden AG, Steegers EAP, Tiemeier H, Steegers-Theunissen RPM. Determinants of maternal pregnancy one-carbon metabolism and newborn human DNA methylation profiles. Reproduction 2015; 148:581-92. [PMID: 25392189 DOI: 10.1530/rep-14-0260] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Maternal one-carbon (1-C) metabolism provides methylgroups for fetal development and programing by DNA methylation as one of the underlying epigenetic mechanisms. We aimed to investigate maternal 1-C biomarkers, folic acid supplement use, and MTHFR C677T genotype as determinants of 1-C metabolism in early pregnancy in association with newborn DNA methylation levels of fetal growth and neurodevelopment candidate genes. The participants were 463 mother-child pairs of Dutch national origin from a large population-based birth cohort in Rotterdam, The Netherlands. In early pregnancy (median 13.0 weeks, 90% range 10.4-17.1), we assessed the maternal folate and homocysteine blood concentrations, folic acid supplement use, and the MTHFR C677T genotype in mothers and newborns. In newborns, DNA methylation was measured in umbilical cord blood white blood cells at 11 regions of the seven genes: NR3C1, DRD4, 5-HTT, IGF2DMR, H19, KCNQ1OT1, and MTHFR. The associations between the 1-C determinants and DNA methylation were examined using linear mixed models. An association was observed between maternal folate deficiency and lower newborn DNA methylation, which attenuated after adjustment for potential confounders. The maternal MTHFR TT genotype was significantly associated with lower DNA methylation. However, maternal homocysteine and folate concentrations, folic acid supplement use, and the MTHFR genotype in the newborn were not associated with newborn DNA methylation. The maternal MTHFR C677T genotype, as a determinant of folate status and 1-C metabolism, is associated with variations in the epigenome of a selection of genes in newborns. Research on the implications of these variations in methylation on gene expression and health is recommended.
Collapse
Affiliation(s)
- Nina H van Mil
- The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Marieke I Bouwland-Both
- The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Lisette Stolk
- The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Michael M P J Verbiest
- The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Albert Hofman
- The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Frank C Verhulst
- The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Paul H C Eilers
- The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Andre G Uitterlinden
- The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Eric A P Steegers
- The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Henning Tiemeier
- The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Régine P M Steegers-Theunissen
- The Generation R Study GroupDepartment of Child and Adolescent PsychiatryDepartment of Obstetrics and GynecologyDepartment of Internal MedicineDepartment of EpidemiologyDepartment of PaediatricsDepartment of BiostatisticsDepartment of PsychiatryErasmus MC, University Medical Centre Rotterdam, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| |
Collapse
|
535
|
Kiekens F, Daele JV, Blancquaert D, Van Der Straeten D, Lambert WE, Stove CP. Determination of five folate monoglutamates in rodent diets. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:10089-10095. [PMID: 26501433 DOI: 10.1021/acs.jafc.5b04075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
A method for the quantitative determination of folates in rodent diets is very important for correct interpretation of folate intake during feeding trials, given the possible discrepancy between the actual folate concentration in the diet and that mentioned on the product sheet. Liquid chromatography tandem-mass spectrometry is the method of choice to differentiate and quantify the individual folate species present. This discrepancy may be accounted for by, e.g., inaccurate folic acid supplementation and/or the presence of endogenous reduced and substituted folates. We developed a method, validated based on FDA guidelines, that allows the measurement of added and endogenous folates by quantitative determination of 5 folate monoglutamates with linear ranges from 8 μg to 2 mg/kg feed. This information, combined with feed intake data, allows insight into the actual folate intake in animal feeding studies. The relevance of this method was illustrated by the analysis of several feed samples of varying composition, by the investigation of the effect of casein incorporation, and by evaluating the variability of the folate content between pellets and production batches.
Collapse
Affiliation(s)
- Filip Kiekens
- Laboratory of Toxicology, Ghent University , Ottergemsesteenweg 460, B-9000 Gent, Belgium
| | - Jeroen Van Daele
- Laboratory of Toxicology, Ghent University , Ottergemsesteenweg 460, B-9000 Gent, Belgium
| | - Dieter Blancquaert
- Laboratory of Functional Plant Biology, Department of Physiology, Ghent University , K.L. Ledeganckstraat 35, B-9000 Gent, Belgium
| | - Dominique Van Der Straeten
- Laboratory of Functional Plant Biology, Department of Physiology, Ghent University , K.L. Ledeganckstraat 35, B-9000 Gent, Belgium
| | - Willy E Lambert
- Laboratory of Toxicology, Ghent University , Ottergemsesteenweg 460, B-9000 Gent, Belgium
| | - Christophe P Stove
- Laboratory of Toxicology, Ghent University , Ottergemsesteenweg 460, B-9000 Gent, Belgium
| |
Collapse
|
536
|
Nilsson E, Matte A, Perfilyev A, de Mello VD, Käkelä P, Pihlajamäki J, Ling C. Epigenetic Alterations in Human Liver From Subjects With Type 2 Diabetes in Parallel With Reduced Folate Levels. J Clin Endocrinol Metab 2015; 100:E1491-501. [PMID: 26418287 PMCID: PMC4702449 DOI: 10.1210/jc.2015-3204] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Epigenetic variation may contribute to the development of complex metabolic diseases such as type 2 diabetes (T2D). Hepatic insulin resistance is a hallmark of T2D. However, it remains unknown whether epigenetic alterations take place in the liver from diabetic subjects. Therefore, we investigated the genome-wide DNA methylation pattern in the liver from subjects with T2D and nondiabetic controls and related epigenetic alterations to gene expression and circulating folate levels. RESEARCH DESIGN AND METHODS Liver biopsies were obtained from 35 diabetic and 60 nondiabetic subjects, which are part of the Kuopio Obesity Surgery Study. The genome-wide DNA methylation pattern was analyzed in the liver using the HumanMethylation450 BeadChip. RNA expression was analyzed from a subset of subjects using the HumanHT-12 Expression BeadChip. RESULTS After correction for multiple testing, we identified 251 individual CpG sites that exhibit differential DNA methylation in liver obtained from T2D compared with nondiabetic subjects (Q < .05). These include CpG sites annotated to genes that are biologically relevant to the development of T2D such as GRB10, ABCC3, MOGAT1, and PRDM16. The vast majority of the significant CpG sites (94%) displayed decreased DNA methylation in liver from subjects with T2D. The hypomethylation found in liver from diabetic subjects may be explained by reduced folate levels. Indeed, subjects with T2D had significantly reduced erythrocyte folate levels compared with nondiabetic subjects. We further identified 29 genes that displayed both differential DNA methylation and gene expression in human T2D liver including the imprinted gene H19. CONCLUSIONS Our study highlights the importance of epigenetic and transcriptional changes in the liver from subjects with T2D. Reduced circulating folate levels may provide an explanation for hypomethylation in the human diabetic liver.
Collapse
|
537
|
Sobiak B, Graczyk‐Jarzynka A, Leśniak W. Comparison of DNA Methylation and Expression Pattern of S100 and Other Epidermal Differentiation Complex Genes in Differentiating Keratinocytes. J Cell Biochem 2015; 117:1092-8. [DOI: 10.1002/jcb.25392] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/05/2015] [Indexed: 02/05/2023]
Affiliation(s)
- Barbara Sobiak
- Department of Molecular and Cellular NeurobiologyNencki Institute of Experimental Biology, 3 Pasteur StreetWarsaw02‐093Poland
| | - Agnieszka Graczyk‐Jarzynka
- Department of Molecular and Cellular NeurobiologyNencki Institute of Experimental Biology, 3 Pasteur StreetWarsaw02‐093Poland
| | - Wiesława Leśniak
- Department of Molecular and Cellular NeurobiologyNencki Institute of Experimental Biology, 3 Pasteur StreetWarsaw02‐093Poland
| |
Collapse
|
538
|
Encyclopedia of bacterial gene circuits whose presence or absence correlate with pathogenicity--a large-scale system analysis of decoded bacterial genomes. BMC Genomics 2015; 16:773. [PMID: 26459834 PMCID: PMC4603813 DOI: 10.1186/s12864-015-1957-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 09/28/2015] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Bacterial infections comprise a global health challenge as the incidences of antibiotic resistance increase. Pathogenic potential of bacteria has been shown to be context dependent, varying in response to environment and even within the strains of the same genus. RESULTS We used the KEGG repository and extensive literature searches to identify among the 2527 bacterial genomes in the literature those implicated as pathogenic to the host, including those which show pathogenicity in a context dependent manner. Using data on the gene contents of these genomes, we identified sets of genes highly abundant in pathogenic but relatively absent in commensal strains and vice versa. In addition, we carried out genome comparison within a genus for the seventeen largest genera in our genome collection. We projected the resultant lists of ortholog genes onto KEGG bacterial pathways to identify clusters and circuits, which can be linked to either pathogenicity or synergy. Gene circuits relatively abundant in nonpathogenic bacteria often mediated biosynthesis of antibiotics. Other synergy-linked circuits reduced drug-induced toxicity. Pathogen-abundant gene circuits included modules in one-carbon folate, two-component system, type-3 secretion system, and peptidoglycan biosynthesis. Antibiotics-resistant bacterial strains possessed genes modulating phagocytosis, vesicle trafficking, cytoskeletal reorganization, and regulation of the inflammatory response. Our study also identified bacterial genera containing a circuit, elements of which were previously linked to Alzheimer's disease. CONCLUSIONS Present study produces for the first time, a signature, in the form of a robust list of gene circuitry whose presence or absence could potentially define the pathogenicity of a microbiome. Extensive literature search substantiated a bulk majority of the commensal and pathogenic circuitry in our predicted list. Scanning microbiome libraries for these circuitry motifs will provide further insights into the complex and context dependent pathogenicity of bacteria.
Collapse
|
539
|
Bahado-Singh RO, Zaffra R, Albayarak S, Chelliah A, Bolinjkar R, Turkoglu O, Radhakrishna U. Epigenetic markers for newborn congenital heart defect (CHD). J Matern Fetal Neonatal Med 2015; 29:1881-7. [PMID: 26429603 DOI: 10.3109/14767058.2015.1069811] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Our objective was to determine whether there were significant differences in genome-wide DNA methylation in newborns with major congenital heart defect (CHD) compared to controls. We also evaluated methylation of cytosines in CpG motifs for the detection of these CHDs. METHODS Genome-wide DNA methylation analysis was performed on DNA from 60 newborns with various CHDs, including hypoplastic left heart syndrome, ventricular septal deficit, atrial septal defect, pulmonary stenosis, coarctation of the aorta and Tetralogy of Fallot, and 32 controls. RESULTS Highly significant differences in cytosine methylation were seen in a large number of genes throughout the genome for all CHD categories. Gene ontology analysis of CHD overall indicated over-represented biological processes involving cell development and differentiation, and anatomical structure morphogenesis. Methylation of individual cytosines in CpG motifs had high diagnostic accuracy for the detection of CHD. For example, for coarctation one predictive model based on levels of particular cytosine nucleotides achieved a sensitivity of 100% and specificity of 93.8% (AUC = 0.974, p < 0.00001). CONCLUSION Profound differences in cytosine methylation were observed in hundreds of genes in newborns with different types of CHD. There appears to be the potential for development of accurate genetic biomarkers for CHD detection in newborns.
Collapse
Affiliation(s)
- Ray O Bahado-Singh
- a Department of Obstetrics and Gynecology , William Beaumont School of Medicine, Oakland University , Royal Oak , MI , USA and
| | - Rita Zaffra
- b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Samet Albayarak
- b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Anushka Chelliah
- b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Rashmi Bolinjkar
- b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Onur Turkoglu
- a Department of Obstetrics and Gynecology , William Beaumont School of Medicine, Oakland University , Royal Oak , MI , USA and
| | - Uppala Radhakrishna
- a Department of Obstetrics and Gynecology , William Beaumont School of Medicine, Oakland University , Royal Oak , MI , USA and
| |
Collapse
|
540
|
Morgado J, Sanches B, Anjos R, Coelho C. Programming of Essential Hypertension: What Pediatric Cardiologists Need to Know. Pediatr Cardiol 2015; 36:1327-37. [PMID: 26015087 DOI: 10.1007/s00246-015-1204-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/14/2015] [Indexed: 01/11/2023]
Abstract
Hypertension is recognized as one of the major contributing factors to cardiovascular disease, but its etiology remains incompletely understood. Known genetic and environmental influences can only explain a small part of the variability in cardiovascular disease risk. The missing heritability is currently one of the most important challenges in blood pressure and hypertension genetics. Recently, some promising approaches have emerged that move beyond the DNA sequence and focus on identification of blood pressure genes regulated by epigenetic mechanisms such as DNA methylation, histone modification and microRNAs. This review summarizes information on gene-environmental interactions that lead toward the developmental programming of hypertension with specific reference to epigenetics and provides pediatricians and pediatric cardiologists with a more complete understanding of its pathogenesis.
Collapse
Affiliation(s)
- Joana Morgado
- Pediatrics Department, Hospital do Espírito Santo de Évora, Largo Senhor da Pobreza, 7000-811, Évora, Portugal.
| | - Bruno Sanches
- Pediatrics Department, Hospital Garcia de Orta, Almada, Portugal
| | - Rui Anjos
- Pediatric Cardiology Department, Hospital Santa Cruz, Lisbon, Portugal
| | - Constança Coelho
- Genetics Laboratory, Environmental Health Institute, Lisbon Medical School, Lisbon, Portugal
| |
Collapse
|
541
|
Singh MD, Thomas P, Owens J, Hague W, Fenech M. Potential role of folate in pre-eclampsia. Nutr Rev 2015; 73:694-722. [PMID: 26359215 DOI: 10.1093/nutrit/nuv028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Dietary deficiencies of folate and other B vitamin cofactors involved in one-carbon metabolism, together with genetic polymorphisms in key folate-methionine metabolic pathway enzymes, are associated with increases in circulating plasma homocysteine, reduction in DNA methylation patterns, and genome instability events. All of these biomarkers have also been associated with pre-eclampsia. The aim of this review was to explore the literature and identify potential knowledge gaps in relation to the role of folate at the genomic level in either the etiology or the prevention of pre-eclampsia. A systematic search strategy was designed to identify citations in electronic databases for the following terms: folic acid supplementation AND pre-eclampsia, folic acid supplementation AND genome stability, folate AND genome stability AND pre-eclampsia, folic acid supplementation AND DNA methylation, and folate AND DNA methylation AND pre-eclampsia. Forty-three articles were selected according to predefined selection criteria. The studies included in the present review were not homogeneous, which made pooled analysis of the data very difficult. The present review highlights associations between folate deficiency and certain biomarkers observed in various tissues of women at risk of pre-eclampsia. Further investigation is required to understand the role of folate in either the etiology or the prevention of pre-eclampsia.
Collapse
Affiliation(s)
- Mansi Dass Singh
- M.D. Singh, J. Owens, and W. Hague are with the School of Pediatrics and Reproductive Health, Discipline of Obstetrics and Gynecology, Faculty of Health Sciences, Robinson Institute, Australian Research Centre for Health of Women and Babies, The University of Adelaide, Adelaide, South Australia, Australia. M.D. Singh, P. Thomas and M. Fenech are with the Genome Health and Personalized Nutrition Laboratory Commonwealth Scientific and Industrial Research Organization (CSIRO), Food and Nutrition Flagship, Adelaide, South Australia, Australia
| | - Philip Thomas
- M.D. Singh, J. Owens, and W. Hague are with the School of Pediatrics and Reproductive Health, Discipline of Obstetrics and Gynecology, Faculty of Health Sciences, Robinson Institute, Australian Research Centre for Health of Women and Babies, The University of Adelaide, Adelaide, South Australia, Australia. M.D. Singh, P. Thomas and M. Fenech are with the Genome Health and Personalized Nutrition Laboratory Commonwealth Scientific and Industrial Research Organization (CSIRO), Food and Nutrition Flagship, Adelaide, South Australia, Australia
| | - Julie Owens
- M.D. Singh, J. Owens, and W. Hague are with the School of Pediatrics and Reproductive Health, Discipline of Obstetrics and Gynecology, Faculty of Health Sciences, Robinson Institute, Australian Research Centre for Health of Women and Babies, The University of Adelaide, Adelaide, South Australia, Australia. M.D. Singh, P. Thomas and M. Fenech are with the Genome Health and Personalized Nutrition Laboratory Commonwealth Scientific and Industrial Research Organization (CSIRO), Food and Nutrition Flagship, Adelaide, South Australia, Australia
| | - William Hague
- M.D. Singh, J. Owens, and W. Hague are with the School of Pediatrics and Reproductive Health, Discipline of Obstetrics and Gynecology, Faculty of Health Sciences, Robinson Institute, Australian Research Centre for Health of Women and Babies, The University of Adelaide, Adelaide, South Australia, Australia. M.D. Singh, P. Thomas and M. Fenech are with the Genome Health and Personalized Nutrition Laboratory Commonwealth Scientific and Industrial Research Organization (CSIRO), Food and Nutrition Flagship, Adelaide, South Australia, Australia
| | - Michael Fenech
- M.D. Singh, J. Owens, and W. Hague are with the School of Pediatrics and Reproductive Health, Discipline of Obstetrics and Gynecology, Faculty of Health Sciences, Robinson Institute, Australian Research Centre for Health of Women and Babies, The University of Adelaide, Adelaide, South Australia, Australia. M.D. Singh, P. Thomas and M. Fenech are with the Genome Health and Personalized Nutrition Laboratory Commonwealth Scientific and Industrial Research Organization (CSIRO), Food and Nutrition Flagship, Adelaide, South Australia, Australia.
| |
Collapse
|
542
|
Aarabi M, San Gabriel MC, Chan D, Behan NA, Caron M, Pastinen T, Bourque G, MacFarlane AJ, Zini A, Trasler J. High-dose folic acid supplementation alters the human sperm methylome and is influenced by the MTHFR C677T polymorphism. Hum Mol Genet 2015; 24:6301-13. [PMID: 26307085 DOI: 10.1093/hmg/ddv338] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/17/2015] [Indexed: 12/24/2022] Open
Abstract
Dietary folate is a major source of methyl groups required for DNA methylation, an epigenetic modification that is actively maintained and remodeled during spermatogenesis. While high-dose folic acid supplementation (up to 10 times the daily recommended dose) has been shown to improve sperm parameters in infertile men, the effects of supplementation on the sperm epigenome are unknown. To assess the impact of 6 months of high-dose folic acid supplementation on the sperm epigenome, we studied 30 men with idiopathic infertility. Blood folate concentrations increased significantly after supplementation with no significant improvements in sperm parameters. Methylation levels of the differentially methylated regions of several imprinted loci (H19, DLK1/GTL2, MEST, SNRPN, PLAGL1, KCNQ1OT1) were normal both before and after supplementation. Reduced representation bisulfite sequencing (RRBS) revealed a significant global loss of methylation across different regions of the sperm genome. The most marked loss of DNA methylation was found in sperm from patients homozygous for the methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism, a common polymorphism in a key enzyme required for folate metabolism. RRBS analysis also showed that most of the differentially methylated tiles were located in DNA repeats, low CpG-density and intergenic regions. Ingenuity Pathway Analysis revealed that methylation of promoter regions was altered in several genes involved in cancer and neurobehavioral disorders including CBFA2T3, PTPN6, COL18A1, ALDH2, UBE4B, ERBB2, GABRB3, CNTNAP4 and NIPA1. Our data reveal alterations of the human sperm epigenome associated with high-dose folic acid supplementation, effects that were exacerbated by a common polymorphism in MTHFR.
Collapse
Affiliation(s)
- Mahmoud Aarabi
- Department of Human Genetics, Montreal Children's Hospital and Research Institute of the McGill University Health Centre, Montreal, QC, Canada H4A 3J1
| | - Maria C San Gabriel
- Division of Urology, Department of Surgery and, Research Institute of the McGill University Health Centre, Montreal, QC, Canada H4A 3J1
| | - Donovan Chan
- Montreal Children's Hospital and Research Institute of the McGill University Health Centre, Montreal, QC, Canada H4A 3J1
| | - Nathalie A Behan
- Nutrition Research Division, Health Canada, Ottawa, ON, Canada K1A 0K9 and
| | - Maxime Caron
- Department of Human Genetics, McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada H3A 1A4
| | - Tomi Pastinen
- Department of Human Genetics, McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada H3A 1A4
| | - Guillaume Bourque
- Department of Human Genetics, McGill University and Genome Quebec Innovation Centre, Montreal, QC, Canada H3A 1A4
| | | | - Armand Zini
- Division of Urology, Department of Surgery and, Research Institute of the McGill University Health Centre, Montreal, QC, Canada H4A 3J1
| | - Jacquetta Trasler
- Department of Human Genetics, Departments of Pediatrics and Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada H4A 3J1, Montreal Children's Hospital and Research Institute of the McGill University Health Centre, Montreal, QC, Canada H4A 3J1,
| |
Collapse
|
543
|
Moen EL, Mariani CJ, Zullow H, Jeff-Eke M, Litwin E, Nikitas JN, Godley LA. New themes in the biological functions of 5-methylcytosine and 5-hydroxymethylcytosine. Immunol Rev 2015; 263:36-49. [PMID: 25510270 DOI: 10.1111/imr.12242] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
5-methylcytosine (5-mC) and 5-hydroxymethylcytosine (5-hmC) play a critical role in development and normal physiology. Alterations in 5-mC and 5-hmC patterns are common events in hematopoietic neoplasms. In this review, we begin by emphasizing the importance of 5-mC, 5-hmC, and their enzymatic modifiers in hematological malignancies. Then, we discuss the functions of 5-mC and 5-hmC at distinct genic contexts, including promoter regions, gene bodies, intron-exon boundaries, alternative promoters, and intragenic microRNAs. Recent advances in technology have allowed for the study of 5-mC and 5-hmC independently and specifically permitting distinction between the bases that show them to have transcriptional effects that vary by their location relative to gene structure. We extend these observations to their functions at enhancers and transcription factor binding sites. We discuss dietary influences on 5-mC and 5-hmC levels and summarize the literature on the effects of folate and vitamin C on 5-mC and 5-hmC, respectively. Finally, we discuss how these new themes in the functions of 5-mC and 5-hmC will likely influence the broader research field of epigenetics.
Collapse
Affiliation(s)
- Erika L Moen
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA; Committee on Cancer Biology, The University of Chicago, Chicago, IL, USA
| | | | | | | | | | | | | |
Collapse
|
544
|
Geng Y, Gao R, Chen X, Liu X, Liao X, Li Y, Liu S, Ding Y, Wang Y, He J. Folate deficiency impairs decidualization and alters methylation patterns of the genome in mice. Mol Hum Reprod 2015; 21:844-56. [PMID: 26246607 DOI: 10.1093/molehr/gav045] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/28/2015] [Indexed: 01/09/2023] Open
Abstract
Existing evidence suggests that adverse pregnancy outcomes are closely related with dietary factors. Previous studies in mice have focused on the harm of folate deficiency (FD) on development of embryo, while the effect of low maternal folate levels on maternal intrauterine environment during early pregnancy remains unclear. Since our previous study found that FD treatment of mice causes no apparent defects in embryo implantation but is accompanied by female subfertility, we next chose to investigate a potential role of FD on molecular events after implantation. We observed that the decidual bulges began to be stunted on pregnancy day 6. The results of functional experiments in vivo and in vitro showed that FD inhibited the process of endometrial decidualization. It has been confirmed that DNA methylation participates in decidualization, and folate as a methyl donor could change the methylation patterns of genes. Thus, we hypothesized that FD impairs maternal endometrial decidualization by altering the methylation profiles of related genes. Reduced representation bisulphite sequencing was carried out to detect the methylation profiles of endometrium on pregnancy day 6-8, which is equivalent to the decidualization period in mice. The results confirmed that FD changes the methylation patterns of genome, and GO analysis of the differentially methylated regions revealed that the associated genes mainly participate in biological adhesion, biological regulation, cell proliferation, development, metabolism and signalling. In addition, we found some candidates for regulators of decidual transformation, such as Nr1h3 and Nr5a1. The data indicate that FD inhibits decidualization, possibly by altering methylation patterns of the genome in mice.
Collapse
Affiliation(s)
- Yanqing Geng
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Box 197, No.1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, PR China
| | - Rufei Gao
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Box 197, No.1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, PR China
| | - Xuemei Chen
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Box 197, No.1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, PR China
| | - Xueqing Liu
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Box 197, No.1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, PR China
| | - Xinggui Liao
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Box 197, No.1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, PR China
| | - Yanli Li
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Box 197, No.1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, PR China
| | - Shangjing Liu
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Box 197, No.1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, PR China
| | - Yubin Ding
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Box 197, No.1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, PR China
| | - Yingxiong Wang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Box 197, No.1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, PR China
| | - Junlin He
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Box 197, No.1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, PR China
| |
Collapse
|
545
|
Farias N, Ho N, Butler S, Delaney L, Morrison J, Shahrzad S, Coomber BL. The effects of folic acid on global DNA methylation and colonosphere formation in colon cancer cell lines. J Nutr Biochem 2015; 26:818-26. [PMID: 25804133 DOI: 10.1016/j.jnutbio.2015.02.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 02/18/2015] [Accepted: 02/20/2015] [Indexed: 02/07/2023]
Abstract
Folate and its synthetic form, folic acid (FA), are essential vitamins for the regeneration of S-adenosyl methionine molecules, thereby maintaining adequate cellular methylation. The deregulation of DNA methylation is a contributing factor to carcinogenesis, as alterations in genetic methylation may contribute to stem cell reprogramming and dedifferentiation processes that lead to a cancer stem cell (CSC) phenotype. Here, we investigate the potential effects of FA exposure on DNA methylation and colonosphere formation in cultured human colorectal cancer (CRC) cell lines. We show for the first time that HCT116, LS174T, and SW480 cells grown without adequate FA demonstrate significantly impaired colonosphere forming ability with limited changes in CD133, CD166, and EpCAM surface expression. These differences were accompanied by concomitant changes to DNA methyltransferase (DNMT) enzyme expression and DNA methylation levels, which varied depending on cell line. Taken together, these results demonstrate an interaction between FA metabolism and CSC phenotype in vitro and help elucidate a connection between supplemental FA intake and CRC development.
Collapse
Affiliation(s)
- Nathan Farias
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada N1G 2W1.
| | - Nelson Ho
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada N1G 2W1.
| | - Stacey Butler
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada N1G 2W1.
| | - Leanne Delaney
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada N1G 2W1.
| | - Jodi Morrison
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada N1G 2W1.
| | | | - Brenda L Coomber
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada N1G 2W1.
| |
Collapse
|
546
|
Agodi A, Barchitta M, Quattrocchi A, Maugeri A, Canto C, Marchese AE, Vinciguerra M. Low fruit consumption and folate deficiency are associated with LINE-1 hypomethylation in women of a cancer-free population. GENES AND NUTRITION 2015; 10:480. [PMID: 26183162 PMCID: PMC4504850 DOI: 10.1007/s12263-015-0480-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/02/2015] [Indexed: 01/05/2023]
Abstract
Several dietary agents, such as micronutrient and non-nutrient components, the so-called bioactive food components, have been shown to display anticancer properties and influence genetic processes. The most common epigenetic change is DNA methylation. Hypomethylation of long interspersed elements (LINE-1) has been associated with an increased risk of several cancers, although conflicting findings have also been observed. The aim of the present study was to test the hypothesis that a low adherence to the Mediterranean diet (MD) and folate deficiency may cause LINE-1 hypomethylation in blood leukocytes of healthy women, and thus genomic instability. One hundred and seventy-seven non-pregnant women were enrolled. Mediterranean diet score (MDS) and folate intake were calculated using a food frequency questionnaire. LINE-1 methylation level was measured by pyrosequencing analysis in three CpG sites of LINE-1 promoter. According to MDS, only 9.6 % of subjects achieved a high adherence to MD. Taking into account the use of supplements, there was a high prevalence of folate deficiency (73.4 %). Women whose consumption of fruit was below the median value (i.e., <201 gr/day) were 3.7 times more likely to display LINE-1 hypomethylation than women whose consumption was above the median value (OR 3.7; 95 % CI 1.4–9.5). Similarly, women with folate deficiency were 3.6 times more likely to display LINE-1 hypomethylation than women with no folate deficiency (OR 3.6; 95 % CI 1.1–12.1). A dietary pattern characterized by low fruit consumption and folate deficiency is associated with LINE-1 hypomethylation and with cancer risk.
Collapse
Affiliation(s)
- Antonella Agodi
- Department of Medical and Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, Via S. Sofia 87, 95121, Catania, Italy,
| | | | | | | | | | | | | |
Collapse
|
547
|
Lee JA, Shinn P, Jaken S, Oliver S, Willard FS, Heidler S, Peery RB, Oler J, Chu S, Southall N, Dexheimer TS, Smallwood J, Huang R, Guha R, Jadhav A, Cox K, Austin CP, Simeonov A, Sittampalam GS, Husain S, Franklin N, Wild DJ, Yang JJ, Sutherland JJ, Thomas CJ. Novel Phenotypic Outcomes Identified for a Public Collection of Approved Drugs from a Publicly Accessible Panel of Assays. PLoS One 2015; 10:e0130796. [PMID: 26177200 PMCID: PMC4503722 DOI: 10.1371/journal.pone.0130796] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/26/2015] [Indexed: 12/17/2022] Open
Abstract
Phenotypic assays have a proven track record for generating leads that become first-in-class therapies. Whole cell assays that inform on a phenotype or mechanism also possess great potential in drug repositioning studies by illuminating new activities for the existing pharmacopeia. The National Center for Advancing Translational Sciences (NCATS) pharmaceutical collection (NPC) is the largest reported collection of approved small molecule therapeutics that is available for screening in a high-throughput setting. Via a wide-ranging collaborative effort, this library was analyzed in the Open Innovation Drug Discovery (OIDD) phenotypic assay modules publicly offered by Lilly. The results of these tests are publically available online at www.ncats.nih.gov/expertise/preclinical/pd2 and via the PubChem Database (https://pubchem.ncbi.nlm.nih.gov/) (AID 1117321). Phenotypic outcomes for numerous drugs were confirmed, including sulfonylureas as insulin secretagogues and the anti-angiogenesis actions of multikinase inhibitors sorafenib, axitinib and pazopanib. Several novel outcomes were also noted including the Wnt potentiating activities of rotenone and the antifolate class of drugs, and the anti-angiogenic activity of cetaben.
Collapse
Affiliation(s)
- Jonathan A. Lee
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Paul Shinn
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Susan Jaken
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Sarah Oliver
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Francis S. Willard
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Steven Heidler
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Robert B. Peery
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Jennifer Oler
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Shaoyou Chu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Noel Southall
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thomas S. Dexheimer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jeffrey Smallwood
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ajit Jadhav
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Karen Cox
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Christopher P. Austin
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - G. Sitta Sittampalam
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Saba Husain
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Natalie Franklin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - David J. Wild
- Indiana University School of Informatics and Computing, Bloomington, Indiana, United States of America
| | - Jeremy J. Yang
- Indiana University School of Informatics and Computing, Bloomington, Indiana, United States of America
| | - Jeffrey J. Sutherland
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
- * E-mail: (JJS); (CJT)
| | - Craig J. Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (JJS); (CJT)
| |
Collapse
|
548
|
Bailey LB, Stover PJ, McNulty H, Fenech MF, Gregory JF, Mills JL, Pfeiffer CM, Fazili Z, Zhang M, Ueland PM, Molloy AM, Caudill MA, Shane B, Berry RJ, Bailey RL, Hausman DB, Raghavan R, Raiten DJ. Biomarkers of Nutrition for Development-Folate Review. J Nutr 2015; 145:1636S-1680S. [PMID: 26451605 PMCID: PMC4478945 DOI: 10.3945/jn.114.206599] [Citation(s) in RCA: 325] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/11/2014] [Accepted: 04/14/2015] [Indexed: 12/13/2022] Open
Abstract
The Biomarkers of Nutrition for Development (BOND) project is designed to provide evidence-based advice to anyone with an interest in the role of nutrition in health. Specifically, the BOND program provides state-of-the-art information and service with regard to selection, use, and interpretation of biomarkers of nutrient exposure, status, function, and effect. To accomplish this objective, expert panels are recruited to evaluate the literature and to draft comprehensive reports on the current state of the art with regard to specific nutrient biology and available biomarkers for assessing nutrients in body tissues at the individual and population level. Phase I of the BOND project includes the evaluation of biomarkers for 6 nutrients: iodine, iron, zinc, folate, vitamin A, and vitamin B-12. This review represents the second in the series of reviews and covers all relevant aspects of folate biology and biomarkers. The article is organized to provide the reader with a full appreciation of folate's history as a public health issue, its biology, and an overview of available biomarkers (serum folate, RBC folate, and plasma homocysteine concentrations) and their interpretation across a range of clinical and population-based uses. The article also includes a list of priority research needs for advancing the area of folate biomarkers related to nutritional health status and development.
Collapse
Affiliation(s)
- Lynn B Bailey
- Department of Foods and Nutrition, University of Georgia, Athens, GA;
| | - Patrick J Stover
- Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | - Helene McNulty
- Northern Ireland Centre for Food and Health, Biomedical Sciences Research Institute, University of Ulster, Londonderry, United Kingdom
| | - Michael F Fenech
- Genome Health Nutrigenomics Laboratory, Food, Nutrition, and Bioproducts Flagship, Commonwealth Scientific and Industrial Research Organization, Adelaide, Australia
| | - Jesse F Gregory
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL
| | - James L Mills
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD
| | | | - Zia Fazili
- National Center for Environmental Health, CDC, Atlanta, GA
| | - Mindy Zhang
- National Center for Environmental Health, CDC, Atlanta, GA
| | - Per M Ueland
- Department of Clinical Science, Univeristy of Bergen, Bergen, Norway
| | - Anne M Molloy
- Institute of Molecular Medicine, Trinity College, Dublin, Ireland
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | - Barry Shane
- Department of Nutritional Sciences and Toxicology, University of California-Berkeley, Berkeley, CA
| | - Robert J Berry
- National Center on Birth Defects and Developmental Disabilities, CDC, Atlanta, GA; and
| | | | - Dorothy B Hausman
- Department of Foods and Nutrition, University of Georgia, Athens, GA
| | - Ramkripa Raghavan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD
| | - Daniel J Raiten
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD;
| |
Collapse
|
549
|
Monteiro JP, Kussmann M, Kaput J. The genomics of micronutrient requirements. GENES & NUTRITION 2015; 10:466. [PMID: 25981693 PMCID: PMC4434349 DOI: 10.1007/s12263-015-0466-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/22/2015] [Indexed: 01/04/2023]
Abstract
Healthy nutrition is accepted as a cornerstone of public health strategies for reducing the risk of noncommunicable conditions such as obesity, cardiovascular disease, and related morbidities. However, many research studies continue to focus on single or at most a few factors that may elicit a metabolic effect. These reductionist approaches resulted in: (1) exaggerated claims for nutrition as a cure or prevention of disease; (2) the wide use of empirically based dietary regimens, as if one fits all; and (3) frequent disappointment of consumers, patients, and healthcare providers about the real impact nutrition can make on medicine and health. Multiple factors including environment, host and microbiome genetics, social context, the chemical form of the nutrient, its (bio)availability, and chemical and metabolic interactions among nutrients all interact to result in nutrient requirement and in health outcomes. Advances in laboratory methodologies, especially in analytical and separation techniques, are making the chemical dissection of foods and their availability in physiological tissues possible in an unprecedented manner. These omics technologies have opened opportunities for extending knowledge of micronutrients and of their metabolic and endocrine roles. While these technologies are crucial, more holistic approaches to the analysis of physiology and environment, novel experimental designs, and more sophisticated computational methods are needed to advance our understanding of how nutrition influences health of individuals.
Collapse
Affiliation(s)
- Jacqueline Pontes Monteiro
- />Department of Pediatrics, Faculty of Medicine, Nutrition and Metabolism, University of São Paulo, Bandeirantes Avenue, HCFMRP Campus USP, 3900, Ribeirão Preto, SP 14049-900 Brazil
| | - Martin Kussmann
- />Nestlé Institute of Health Sciences, Innovation Square, EPFL Campus, 1015 Lausanne, Switzerland
- />Ecole Polytechnique Fédérale Lausanne, Lausanne, Switzerland
| | - Jim Kaput
- />Nestlé Institute of Health Sciences, Innovation Square, EPFL Campus, 1015 Lausanne, Switzerland
- />Service d’endorcrinologie, diabetologie et metabolosime du CHUV, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
550
|
Molecular transitions from papillomavirus infection to cervical precancer and cancer: Role of stromal estrogen receptor signaling. Proc Natl Acad Sci U S A 2015; 112:E3255-64. [PMID: 26056290 DOI: 10.1073/pnas.1509322112] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
To study the multistep process of cervical cancer development, we analyzed 128 frozen cervical samples spanning normalcy, increasingly severe cervical intraepithelial neoplasia (CIN1- CIN3), and cervical cancer (CxCa) from multiple perspectives, revealing a cascade of progressive changes. Compared with normal tissue, expression of many DNA replication/repair and cell proliferation genes was increased in CIN1/CIN2 lesions and further sustained in CIN3, consistent with high-risk human papillomavirus (HPV)-induced tumor suppressor inactivation. The CIN3-to-CxCa transition showed metabolic shifts, including decreased expression of mitochondrial electron transport complex components and ribosomal protein genes. Significantly, despite clinical, epidemiological, and animal model results linking estrogen and estrogen receptor alpha (ERα) to CxCa, ERα expression declined >15-fold from normalcy to cancer, showing the strongest inverse correlation of any gene with the increasing expression of p16, a marker for HPV-linked cancers. This drop in ERα in CIN and tumor cells was confirmed at the protein level. However, ERα expression in stromal cells continued throughout CxCa development. Our further studies localized stromal ERα to FSP1+, CD34+, SMA- precursor fibrocytes adjacent to normal and precancerous CIN epithelium, and FSP1-, CD34-, SMA+ activated fibroblasts in CxCas. Moreover, rank correlations with ERα mRNA identified IL-8, CXCL12, CXCL14, their receptors, and other angiogenesis and immune cell infiltration and inflammatory factors as candidates for ERα-induced stroma-tumor signaling pathways. The results indicate that estrogen signaling in cervical cancer has dramatic differences from ERα+ breast cancers, and imply that estrogen signaling increasingly proceeds indirectly through ERα in tumor-associated stromal fibroblasts.
Collapse
|