151
|
Yokouchi Y, Imaoka M, Niino N, Kiyosawa N, Kai K. Comprehensive Evaluation of (+)-Usnic Acid–induced Cardiotoxicity in Rats by Sequential Cross-omics Analysis. Toxicol Pathol 2017; 45:481-492. [DOI: 10.1177/0192623317707074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Two-week administration of (+)-usnic acid (UA) induces mitochondrial swelling of cardiomyocytes, and toxicogenomic analysis of the heart revealed upregulation of oxidative stress, amino acid limitation, and endoplasmic reticulum stress–related genes in rats. To analyze the pathogenesis, UA was orally administrated to rats for 1, 4, 7, and 14 days, and sequential histopathological, genomic, and metabolomic analyses were performed on the heart, liver, and plasma. As a result, mitochondrial swelling of cardiomyocytes was observed on day 15 preceded by genomic upregulation on days 5 and 8. Of the focused gene groups, amino acid limitation–related genes represented by Mthfd2 showed numerically higher values or upregulation from day 5, which was sustained through the experimental period. On the contrary, oxidative stress–related genes were upregulated temporally on day 5. In metabolomic analysis, amino acids such as taurocholate and their metabolites fluctuated in concert with the upregulation of amino acid limitation–related genes in the heart, liver, and plasma. Moreover, accumulations of bile acids were manifested in all the tested tissues, while no histopathological change was seen in the liver. Increased bile acids might have an indirect effect on the myocardium; however, more detailed analysis is required. In conclusion, amino acid limitation was suggested as the pivotal toxic trigger of UA-induced cardiotoxicity.
Collapse
Affiliation(s)
- Y. Yokouchi
- Translational Medicine and Clinical Pharmacology Department, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - M. Imaoka
- Medicinal Safety Research Laboratories, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - N. Niino
- Medicinal Safety Research Laboratories, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - N. Kiyosawa
- Translational Medicine and Clinical Pharmacology Department, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - K. Kai
- Medicinal Safety Research Laboratories, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| |
Collapse
|
152
|
Bruemmer KJ, Brewer TF, Chang CJ. Fluorescent probes for imaging formaldehyde in biological systems. Curr Opin Chem Biol 2017; 39:17-23. [PMID: 28527906 DOI: 10.1016/j.cbpa.2017.04.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/13/2017] [Accepted: 04/13/2017] [Indexed: 12/13/2022]
Abstract
Formaldehyde (FA) is a common environmental toxin but is also endogenously produced through a diverse array of essential biological processes, including mitochondrial one-carbon metabolism, metabolite oxidation, and nuclear epigenetic modifications. Its high electrophilicity enables reactivity with a wide variety of biological nucleophiles, which can be beneficial or detrimental to cellular function depending on the context. New methods that enable detection of FA in living systems can help disentangle the signal/stress dichotomy of this simplest reactive carbonyl species (RCS), and fluorescent probes for FA with high selectivity and sensitivity have emerged as promising chemical tools in this regard.
Collapse
Affiliation(s)
- Kevin J Bruemmer
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | - Thomas F Brewer
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | | |
Collapse
|
153
|
Bae S, Kamynina E, Farinola AF, Caudill MA, Stover PJ, Cassano PA, Berry R, Peña-Rosas JP. Provision of folic acid for reducing arsenic toxicity in arsenic-exposed children and adults. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2017. [DOI: 10.1002/14651858.cd012649] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Sajin Bae
- Cornell University; Division of Nutritional Sciences; 324 Savage Hall 244 Garden Avenue Ithaca NY USA 14853
| | - Elena Kamynina
- Cornell University; Division of Nutritional Sciences; 324 Savage Hall 244 Garden Avenue Ithaca NY USA 14853
| | - Adetutu F Farinola
- University of Ibadan; Faculty of Public Health, Department of Human Nutrition and Dietetics; Seat of Wisdom Chapel Ibadan Oyo State Nigeria 200282
| | - Marie A Caudill
- Cornell University; Division of Nutritional Sciences; 324 Savage Hall 244 Garden Avenue Ithaca NY USA 14853
| | - Patrick J Stover
- Cornell University; Division of Nutritional Sciences; 324 Savage Hall 244 Garden Avenue Ithaca NY USA 14853
| | - Patricia A Cassano
- Cornell University; Division of Nutritional Sciences; 324 Savage Hall 244 Garden Avenue Ithaca NY USA 14853
| | - Robert Berry
- Independent cosultant; 1376 N Decatur Rd NE Atlanta Georgia USA 30306
| | - Juan Pablo Peña-Rosas
- World Health Organization; Evidence and Programme Guidance, Department of Nutrition for Health and Development; 20 Avenue Appia Geneva GE Switzerland 1211
| |
Collapse
|
154
|
Agrawal A, Khan MJ, Graugnard DE, Vailati-Riboni M, Rodriguez-Zas SL, Osorio JS, Loor JJ. Prepartal Energy Intake Alters Blood Polymorphonuclear Leukocyte Transcriptome During the Peripartal Period in Holstein Cows. Bioinform Biol Insights 2017; 11:1177932217704667. [PMID: 28579762 PMCID: PMC5414586 DOI: 10.1177/1177932217704667] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/13/2017] [Indexed: 12/18/2022] Open
Abstract
In the dairy industry, cow health and farmer profits depend on the balance between diet (ie, nutrient composition, daily intake) and metabolism. This is especially true during the transition period, where dramatic physiological changes foster vulnerability to immunosuppression, negative energy balance, and clinical and subclinical disorders. Using an Agilent microarray platform, this study examined changes in the transcriptome of bovine polymorphonuclear leukocytes (PMNLs) due to prepartal dietary intake. Holstein cows were fed a high-straw, control-energy diet (CON; NEL = 1.34 Mcal/kg) or overfed a moderate-energy diet (OVE; NEL = 1.62 Mcal/kg) during the dry period. Blood for PMNL isolation and metabolite analysis was collected at −14 and +7 days relative to parturition. At an analysis of variance false discovery rate <0.05, energy intake (OVE vs CON) influenced 1806 genes. Dynamic Impact Approach bioinformatics analysis classified treatment effects on Kyoto Encyclopedia of Genes and Genomes pathways, including activated oxidative phosphorylation and biosynthesis of unsaturated fatty acids and inhibited RNA polymerase, proteasome, and toll-like receptor signaling pathway. This analysis indicates that processes critical for energy metabolism and cellular and immune function were affected with mixed results. However, overall interpretation of the transcriptome data agreed in part with literature documenting a potentially detrimental, chronic activation of PMNL in response to overfeeding. The widespread, transcriptome-level changes captured here confirm the importance of dietary energy adjustments around calving on the immune system.
Collapse
Affiliation(s)
- A Agrawal
- Division of Nutritional Sciences, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - M J Khan
- Division of Nutritional Sciences, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - D E Graugnard
- Division of Nutritional Sciences, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - M Vailati-Riboni
- Division of Nutritional Sciences, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - S L Rodriguez-Zas
- Division of Nutritional Sciences, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - J S Osorio
- Department of Dairy Science, South Dakota State University, Brookings, SD, USA
| | - J J Loor
- Division of Nutritional Sciences, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
155
|
Singh G, Singh V. Functional elucidation of hypothetical proteins for their indispensable roles toward drug designing targets from Helicobacter pylori strain HPAG1. J Biomol Struct Dyn 2017; 36:906-918. [DOI: 10.1080/07391102.2017.1302361] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Gagandeep Singh
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, TAB, Shahpur 176206, India
| | - Vikram Singh
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, TAB, Shahpur 176206, India
| |
Collapse
|
156
|
Mills EL, Kelly B, O'Neill LAJ. Mitochondria are the powerhouses of immunity. Nat Immunol 2017; 18:488-498. [PMID: 28418387 DOI: 10.1038/ni.3704] [Citation(s) in RCA: 707] [Impact Index Per Article: 88.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/02/2017] [Indexed: 12/14/2022]
Abstract
Recent evidence indicates that mitochondria lie at the heart of immunity. Mitochondrial DNA acts as a danger-associated molecular pattern (DAMP), and the mitochondrial outer membrane is a platform for signaling molecules such as MAVS in RIG-I signaling, and for the NLRP3 inflammasome. Mitochondrial biogenesis, fusion and fission have roles in aspects of immune-cell activation. Most important, Krebs cycle intermediates such as succinate, fumarate and citrate engage in processes related to immunity and inflammation, in both innate and adaptive immune cells. These discoveries are revealing mitochondrial targets that could potentially be exploited for therapeutic gain in inflammation and cancer.
Collapse
Affiliation(s)
- Evanna L Mills
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Beth Kelly
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
157
|
Sadahiro S, Suzuki T, Tanaka A, Okada K, Saito G, Miyakita H, Ogimi T, Nagase H. Gene expression levels of gamma-glutamyl hydrolase in tumor tissues may be a useful biomarker for the proper use of S-1 and tegafur-uracil/leucovorin in preoperative chemoradiotherapy for patients with rectal cancer. Cancer Chemother Pharmacol 2017; 79:1077-1085. [PMID: 28417167 PMCID: PMC5438825 DOI: 10.1007/s00280-017-3295-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/24/2017] [Indexed: 12/18/2022]
Abstract
Purpose Preoperative chemoradiotherapy (CRT) using 5-fluorouracil (5-FU)-based chemotherapy is the standard of care for rectal cancer. The effect of additional chemotherapy during the period between the completion of radiotherapy and surgery remains unclear. Predictive factors for CRT may differ between combination chemotherapy with S-1 and with tegafur-uracil/leucovorin (UFT/LV). Methods The subjects were 54 patients with locally advanced rectal cancer who received preoperative CRT with S-1 or UFT/LV. The pathological tumor response was assessed according to the tumor regression grade (TRG). The expression levels of 18 CRT-related genes were determined using RT-PCR assay. Results A pathological response (TRG 1-2) was observed in 23 patients (42.6%). In a multivariate logistic regression analysis for pathological response, the overall expression levels of four genes, HIF1A, MTHFD1, GGH and TYMS, were significant, and the accuracy rate of the predictive model was 83.3%. The effects of the gene expression levels of GGH on the response differed significantly according to the treatment regimen. The total pathological response rate of both high-GGH patients in the S-1 group and low-GGH patients in the UFT/LV group was 58.3%. Conclusion Additional treatment with 5-FU-based chemotherapy during the interval between radiotherapy and surgery is not beneficial in patients who have received 5-FU-based CRT. The expression levels of four genes, HIF1A, MTHFD1, GGH and TYMS, in tumor tissues can predict the response to preoperative CRT including either S-1 or UFT/LV. In particular, the gene expression level of GGH in tumor tissues may be a useful biomarker for the appropriate use of S-1 and UFT/LV in CRT.
Collapse
Affiliation(s)
- Sotaro Sadahiro
- Department of Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan.
| | - T Suzuki
- Department of Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - A Tanaka
- Department of Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - K Okada
- Department of Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - G Saito
- Department of Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - H Miyakita
- Department of Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - T Ogimi
- Department of Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - H Nagase
- Applied Pharmacology Lab., Taiho Pharmaceutical Co., Ltd., 224-2 Ebisuno Hiraishi, Kawauchi-cho, Tokushima, 771-0194, Japan
| |
Collapse
|
158
|
Misselbeck K, Marchetti L, Field MS, Scotti M, Priami C, Stover PJ. A hybrid stochastic model of folate-mediated one-carbon metabolism: Effect of the common C677T MTHFR variant on de novo thymidylate biosynthesis. Sci Rep 2017; 7:797. [PMID: 28400561 PMCID: PMC5429759 DOI: 10.1038/s41598-017-00854-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/13/2017] [Indexed: 11/19/2022] Open
Abstract
Folate-mediated one-carbon metabolism (FOCM) is an interconnected network of metabolic pathways, including those required for the de novo synthesis of dTMP and purine nucleotides and for remethylation of homocysteine to methionine. Mouse models of folate-responsive neural tube defects (NTDs) indicate that impaired de novo thymidylate (dTMP) synthesis through changes in SHMT expression is causative in folate-responsive NTDs. We have created a hybrid computational model comprised of ordinary differential equations and stochastic simulation. We investigated whether the de novo dTMP synthesis pathway was sensitive to perturbations in FOCM that are known to be associated with human NTDs. This computational model shows that de novo dTMP synthesis is highly sensitive to the common MTHFR C677T polymorphism and that the effect of the polymorphism on FOCM is greater in folate deficiency. Computational simulations indicate that the MTHFR C677T polymorphism and folate deficiency interact to increase the stochastic behavior of the FOCM network, with the greatest instability observed for reactions catalyzed by serine hydroxymethyltransferase (SHMT). Furthermore, we show that de novo dTMP synthesis does not occur in the cytosol at rates sufficient for DNA replication, supporting empirical data indicating that impaired nuclear de novo dTMP synthesis results in uracil misincorporation into DNA.
Collapse
Affiliation(s)
- Karla Misselbeck
- The Microsoft Research - University of Trento Centre for Computational and Systems Biology (COSBI), Piazza Manifattura, 1, 38068, Rovereto (TN), Italy
- Department of Mathematics, University of Trento, Trento, Italy
| | - Luca Marchetti
- The Microsoft Research - University of Trento Centre for Computational and Systems Biology (COSBI), Piazza Manifattura, 1, 38068, Rovereto (TN), Italy
| | - Martha S Field
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, 14853, USA
| | - Marco Scotti
- GEOMAR Helmholtz Centre for Ocean Research Kiel, Düsternbrooker Weg 20, 24105, Kiel, Germany
| | - Corrado Priami
- The Microsoft Research - University of Trento Centre for Computational and Systems Biology (COSBI), Piazza Manifattura, 1, 38068, Rovereto (TN), Italy.
- Department of Mathematics, University of Trento, Trento, Italy.
| | - Patrick J Stover
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, 14853, USA.
| |
Collapse
|
159
|
Bae S, Chon J, Field MS, Stover PJ. Alcohol Dehydrogenase 5 Is a Source of Formate for De Novo Purine Biosynthesis in HepG2 Cells. J Nutr 2017; 147:499-505. [PMID: 28228507 PMCID: PMC5368588 DOI: 10.3945/jn.116.244467] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/18/2016] [Accepted: 01/30/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Formate provides one-carbon units for de novo purine and thymidylate (dTMP) synthesis and is produced via both folate-dependent and folate-independent pathways. Folate-independent pathways are mediated by cytosolic alcohol dehydrogenase 5 (ADH5) and mitochondrial aldehyde dehydrogenase 2 (ALDH2), which generate formate by oxidizing formaldehyde. Formate is a potential biomarker of B-vitamin-dependent one-carbon metabolism.Objective: This study investigated the contributions of ADH5 and ALDH2 to formate production and folate-dependent de novo purine and dTMP synthesis in HepG2 cells.Methods:ADH5 knockout and ALDH2 knockdown HepG2 cells were cultured in folate-deficient [0 nM (6S) 5-formyltetrahydrofolate] or folate-sufficient [25 nM (6S) 5-formyltetrahydrofolate] medium. Purine biosynthesis was quantified as the ratio of [14C]-formate to [3H]-hypoxanthine incorporated into genomic DNA, which indicates the contribution of the de novo purine synthesis pathway relative to salvage synthesis. dTMP synthesis was quantified as the ratio of [14C]-deoxyuridine to [3H]-thymidine incorporation into genomic DNA, which indicates the capacity of de novo dTMP synthesis relative to salvage synthesis.Results: The [14C]-formate-to-[3H]-hypoxanthine ratio was greater in ADH5 knockout than in wild-type HepG2 cells, under conditions of both folate deficiency (+30%; P < 0.001) and folate sufficiency (+22%; P = 0.02). These data indicate that ADH5 deficiency increases the use of exogenous formate for de novo purine biosynthesis. The [14C]-deoxyuridine-to-[3H]-thymidine ratio did not differ between ADH5 knockout and wild-type cells, indicating that ADH5 deficiency does not affect de novo dTMP synthesis capacity relative to salvage synthesis. Under folate deficiency, ALDH2 knockdown cells exhibited a 37% lower ratio of [14C]-formate to [3H]-hypoxanthine (P < 0.001) compared with wild-type HepG2 cells, indicating decreased use of exogenous formate, or increased endogenous formate synthesis, for de novo purine biosynthesis.Conclusions: In HepG2 cells, ADH5 is a source of formate for de novo purine biosynthesis, especially during folate deficiency when folate-dependent formate production is limited. Formate is also shown to be limiting in the growth of HepG2 cells.
Collapse
Affiliation(s)
- Sajin Bae
- Division of Nutritional Sciences and
| | - James Chon
- Graduate Field of Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY
| | | | - Patrick J Stover
- Division of Nutritional Sciences and .,Graduate Field of Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY
| |
Collapse
|
160
|
Kamynina E, Lachenauer ER, DiRisio AC, Liebenthal RP, Field MS, Stover PJ. Arsenic trioxide targets MTHFD1 and SUMO-dependent nuclear de novo thymidylate biosynthesis. Proc Natl Acad Sci U S A 2017; 114:E2319-E2326. [PMID: 28265077 PMCID: PMC5373342 DOI: 10.1073/pnas.1619745114] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Arsenic exposure increases risk for cancers and is teratogenic in animal models. Here we demonstrate that small ubiquitin-like modifier (SUMO)- and folate-dependent nuclear de novo thymidylate (dTMP) biosynthesis is a sensitive target of arsenic trioxide (As2O3), leading to uracil misincorporation into DNA and genome instability. Methylenetetrahydrofolate dehydrogenase 1 (MTHFD1) and serine hydroxymethyltransferase (SHMT) generate 5,10-methylenetetrahydrofolate for de novo dTMP biosynthesis and translocate to the nucleus during S-phase, where they form a multienzyme complex with thymidylate synthase (TYMS) and dihydrofolate reductase (DHFR), as well as the components of the DNA replication machinery. As2O3 exposure increased MTHFD1 SUMOylation in cultured cells and in in vitro SUMOylation reactions, and increased MTHFD1 ubiquitination and MTHFD1 and SHMT1 degradation. As2O3 inhibited de novo dTMP biosynthesis in a dose-dependent manner, increased uracil levels in nuclear DNA, and increased genome instability. These results demonstrate that MTHFD1 and SHMT1, which are key enzymes providing one-carbon units for dTMP biosynthesis in the form of 5,10-methylenetetrahydrofolate, are direct targets of As2O3-induced proteolytic degradation, providing a mechanism for arsenic in the etiology of cancer and developmental anomalies.
Collapse
Affiliation(s)
- Elena Kamynina
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853
| | - Erica R Lachenauer
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853
- Graduate Field of Biology and Biomedical Sciences, Cornell University, Ithaca, NY 14853
| | - Aislyn C DiRisio
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853
| | | | - Martha S Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853
| | - Patrick J Stover
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853;
- Graduate Field of Biology and Biomedical Sciences, Cornell University, Ithaca, NY 14853
- Graduate Field of Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853
| |
Collapse
|
161
|
McKee SE, Grissom NM, Herdt CT, Reyes TM. Methyl donor supplementation alters cognitive performance and motivation in female offspring from high-fat diet-fed dams. FASEB J 2017; 31:2352-2363. [PMID: 28209774 DOI: 10.1096/fj.201601172r] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/30/2017] [Indexed: 12/18/2022]
Abstract
During gestation, fetal nutrition is entirely dependent on maternal diet. Maternal consumption of excess fat during pregnancy has been linked to an increased risk of neurologic disorders in offspring, including attention deficit/hyperactivity disorder, autism, and schizophrenia. In a mouse model, high-fat diet (HFD)-fed offspring have cognitive and executive function deficits as well as whole-genome DNA and promoter-specific hypomethylation in multiple brain regions. Dietary methyl donor supplementation during pregnancy or adulthood has been used to alter DNA methylation and behavior. Given that extensive brain development occurs during early postnatal life-particularly within the prefrontal cortex (PFC), a brain region critical for executive function-we examined whether early life methyl donor supplementation (e.g., during adolescence) could ameliorate executive function deficits observed in offspring that were exposed to maternal HFD. By using operant testing, progressive ratio, and the PFC-dependent 5-choice serial reaction timed task (5-CSRTT), we determined that F1 female offspring (B6D2F1/J) from HFD-fed dams have decreased motivation (decreased progressive ratio breakpoint) and require a longer stimulus length to complete the 5-CSRTT task successfully, whereas early life methyl donor supplementation increased motivation and shortened the minimum stimulus length required for a correct response in the 5-CSRTT. Of interest, we found that expression of 2 chemokines, CCL2 and CXCL10, correlated with the median stimulus length in the 5-CSRTT. Furthermore, we found that acute adult supplementation of methyl donors increased motivation in HFD-fed offspring and those who previously received supplementation with methyl donors. These data point to early life as a sensitive time during which dietary methyl donor supplementation can alter PFC-dependent cognitive behaviors.-McKee, S. E., Grissom, N. M., Herdt, C. T., Reyes, T. M. Methyl donor supplementation alters cognitive performance and motivation in female offspring from high-fat diet-fed dams.
Collapse
Affiliation(s)
- Sarah E McKee
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicola M Grissom
- Department of Psychology, College of Liberal Arts, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christopher T Herdt
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Teresa M Reyes
- Department of Psychiatry and Behavioral Neurosciences, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
162
|
Folate nutrition and blood-brain barrier dysfunction. Curr Opin Biotechnol 2017; 44:146-152. [PMID: 28189938 DOI: 10.1016/j.copbio.2017.01.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 01/30/2023]
Abstract
Mammals require essential nutrients from dietary sources to support normal metabolic, physiological and neuronal functions, to prevent diseases of nutritional deficiency as well as to prevent chronic disease. Disease and/or its treatment can modify fundamental biological processes including cellular nutrient accretion, stability and function in cells. These effects can be isolated to a specific diseased organ in the absence of whole-body alterations in nutrient status or biochemistry. Loss of blood-brain barrier function, which occurs in in-born errors of metabolism and in chronic disease, can cause brain-specific folate deficiency and contribute to disease co-morbidity. The role of brain folate deficiency in neuropsychiatric disorders is reviewed, as well as emerging diagnostic and nutritional strategies to identify and address brain folate deficiency in blood-brain barrier dysfunction.
Collapse
|
163
|
van der Plas E, Schachar RJ, Hitzler J, Crosbie J, Guger SL, Spiegler BJ, Ito S, Nieman BJ. Brain structure, working memory and response inhibition in childhood leukemia survivors. Brain Behav 2017; 7:e00621. [PMID: 28239531 PMCID: PMC5318374 DOI: 10.1002/brb3.621] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 11/09/2016] [Accepted: 11/11/2016] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION Survival rates for children with acute lymphoblastic leukemia (ALL) approach 95%. At the same time, there is growing concern that chemotherapy causes alterations in brain development and cognitive abilities. We performed MRI measurements of white and gray matter volume to explore how variation in brain structure may be related to cognitive abilities in ALL survivors and healthy controls. METHODS The sample included 24 male ALL survivors who had completed contemporary treatment 3-11 years prior, and 21 age- and sex-matched controls. Participants were between 8 and 18 years old. Working memory and motor response inhibition were measured with the N-Back and Stop Signal Tasks (SST), respectively. Participants underwent 3T structural MRI to assess white and gray matter volumes overall, lobe-wise, and in cortical and atlas-identified subcortical structures. Mental health was assessed with the Child Behavioral Checklist. RESULTS ALL survivors performed more poorly on measures of working memory and response inhibition than controls. Frontal and parietal white matter, temporal and occipital gray matter volume, and volumes of subcortical white and gray matter structures were significantly reduced in ALL survivors compared with controls. Significant structure-function correlations were observed between working memory performance and volume of the amygdala, thalamus, striatum, and corpus callosum. Response inhibition was correlated with frontal white matter volume. No differences were found in psychopathology. CONCLUSIONS Compared with controls, a reduction in volume across brain regions and tissue types, was detectable in ALL survivors years after completion of therapy. These structural alterations were correlated with neurocognitive performance, particularly in working memory. Confirming these observations in a larger, more representative sample of the population is necessary. Additionally, establishing the time course of these changes-and the treatment, genetic, and environmental factors that influence them-may provide opportunities to identify at-risk patients, inform the design of treatment modifications, and minimize adverse cognitive outcomes.
Collapse
Affiliation(s)
- Ellen van der Plas
- Physiology and Experimental Medicine The Hospital for Sick Children Research Institute Toronto ON Canada; Psychiatry Research The Hospital for Sick Children Toronto ON Canada
| | - Russell J Schachar
- Psychiatry Research The Hospital for Sick Children Toronto ON Canada; Department of Psychiatry Faculty of Medicine The University of Toronto Toronto ON Canada
| | - Johann Hitzler
- Department of Pediatrics Faculty of Medicine The University of Toronto Toronto ON Canada; Department of Haematology/Oncology The Hospital for Sick Children Toronto ON Canada
| | - Jennifer Crosbie
- Psychiatry Research The Hospital for Sick Children Toronto ON Canada
| | - Sharon L Guger
- Department of Psychology The Hospital for Sick Children Toronto ON Canada
| | - Brenda J Spiegler
- Department of Pediatrics Faculty of Medicine The University of Toronto Toronto ON Canada; Department of Psychology The Hospital for Sick Children Toronto ON Canada
| | - Shinya Ito
- Physiology and Experimental Medicine The Hospital for Sick Children Research Institute Toronto ON Canada; Clinical Pharmacology and Toxicology The Hospital for Sick Children Toronto ON Canada; Pharmacology and Pharmacy Faculty of Medicine The University of Toronto Toronto ON Canada
| | - Brian J Nieman
- Physiology and Experimental Medicine The Hospital for Sick Children Research Institute Toronto ON Canada; Mouse Imaging Centre (MICe) The Hospital for Sick Children Toronto ON Canada; Ontario Institute for Cancer Research Toronto ON Canada; Department of Medical Biophysics The University of Toronto Toronto ON Canada
| |
Collapse
|
164
|
Dhobale M. Neurotrophic Factors and Maternal Nutrition During Pregnancy. VITAMINS AND HORMONES 2017; 104:343-366. [DOI: 10.1016/bs.vh.2016.10.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
165
|
Hazra S, Ghosh S, Hazra B. Phytochemicals With Antileishmanial Activity. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2017. [DOI: 10.1016/b978-0-444-63931-8.00008-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
166
|
Troesch B, Weber P, Mohajeri MH. Potential Links between Impaired One-Carbon Metabolism Due to Polymorphisms, Inadequate B-Vitamin Status, and the Development of Alzheimer's Disease. Nutrients 2016; 8:E803. [PMID: 27973419 PMCID: PMC5188458 DOI: 10.3390/nu8120803] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/06/2016] [Accepted: 12/07/2016] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) is the major cause of dementia and no preventive or effective treatment has been established to date. The etiology of AD is poorly understood, but genetic and environmental factors seem to play a role in its onset and progression. In particular, factors affecting the one-carbon metabolism (OCM) are thought to be important and elevated homocysteine (Hcy) levels, indicating impaired OCM, have been associated with AD. We aimed at evaluating the role of polymorphisms of key OCM enzymes in the etiology of AD, particularly when intakes of relevant B-vitamins are inadequate. Our review indicates that a range of compensatory mechanisms exist to maintain a metabolic balance. However, these become overwhelmed if the activity of more than one enzyme is reduced due to genetic factors or insufficient folate, riboflavin, vitamin B6 and/or vitamin B12 levels. Consequences include increased Hcy levels and reduced capacity to synthetize, methylate and repair DNA, and/or modulated neurotransmission. This seems to favor the development of hallmarks of AD particularly when combined with increased oxidative stress e.g., in apolipoprotein E (ApoE) ε4 carriers. However, as these effects can be compensated at least partially by adequate intakes of B-vitamins, achieving optimal B-vitamin status for the general population should be a public health priority.
Collapse
Affiliation(s)
- Barbara Troesch
- DSM Nutritional Products Ltd., Wurmisweg 576, Kaiseraugst 4303, Switzerland.
| | - Peter Weber
- DSM Nutritional Products Ltd., Wurmisweg 576, Kaiseraugst 4303, Switzerland.
| | - M Hasan Mohajeri
- DSM Nutritional Products Ltd., Wurmisweg 576, Kaiseraugst 4303, Switzerland.
| |
Collapse
|
167
|
Ding Y, Pedersen ER, Svingen GF, Helgeland Ø, Gregory JF, Løland KH, Meyer K, Tell GS, Ueland PM, Nygård OK. Methylenetetrahydrofolate Dehydrogenase 1 Polymorphisms Modify the Associations of Plasma Glycine and Serine With Risk of Acute Myocardial Infarction in Patients With Stable Angina Pectoris in WENBIT (Western Norway B Vitamin Intervention Trial). ACTA ACUST UNITED AC 2016; 9:541-547. [DOI: 10.1161/circgenetics.116.001483] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 10/21/2016] [Indexed: 02/07/2023]
Abstract
Background—
Serine and glycine interconversion and methylenetetrahydrofolate dehydrogenase 1 (MTHFD1)–mediated 1-carbon transfer are the major sources of methyl groups for 1-carbon metabolism. Recently, plasma glycine and a common polymorphism in MTHFD1 have been associated with risk of acute myocardial infarction (AMI). It is, therefore, of interest to explore if these 2 pathways interact in relation to AMI.
Methods and Results—
A total of 2571 participants in the WENBIT (Western Norway B Vitamin Intervention Trial) undergoing coronary angiography for stable angina pectoris were studied. Associations of plasma serine and glycine concentrations with risk of AMI across 2 common and functional MTHFD1 polymorphisms (
rs2236225
and
rs1076991
) were explored in Cox regression models. During a median follow-up of 4.7 years, 212 patients (8.2%) experienced an AMI. In age- and sex-adjusted analyses, plasma glycine (
P
<0.01), but not serine (
P
=0.52), showed an overall association with AMI. However, interactions of MTHFD1
rs2236225
polymorphism with both plasma serine and glycine were observed (
P
interaction
=0.03 for both). Low plasma serine and glycine were associated with an increased risk of AMI among patients carrying the
rs2236225
minor A allele. Similarly, low plasma glycine showed stronger risk relationship with AMI in the
rs1076991
CC genotype carriers but weaker associations in patients carrying the minor T allele (
P
interaction
=0.02).
Conclusions—
Our results showed that 2 common and functional polymorphisms in the
MTHFD1
gene modulate the risk associations of plasma serine and glycine with AMI. These findings emphasize the possible role of the MTHFD1 in regulating serine and glycine metabolism in relation to atherosclerotic complications.
Clinical Trial Registration—
URL:
http://www.clinicaltrials.gov
. Unique Identifier: NCT00354081.
Collapse
Affiliation(s)
- Yunpeng Ding
- From the Department of Clinical Science (Y.D., E.R.P., P.M.U., O.K.N.), KG Jebsen Center for Diabetes Research, Department of Clinical Science (Ø.H., O.K.N.), and Department of Global Public Health and Primary Care (G.S.T.), University of Bergen, Norway; Department of Heart Disease (G.F.T.S., K.H.L., O.K.N.) and Department of Pediatrics (Ø.H.), Haukeland University Hospital, Bergen, Norway; Laboratory of Clinical Biochemistry, Bergen, Norway (P.M.U.); Food Science and Human Nutrition Department,
| | - Eva R. Pedersen
- From the Department of Clinical Science (Y.D., E.R.P., P.M.U., O.K.N.), KG Jebsen Center for Diabetes Research, Department of Clinical Science (Ø.H., O.K.N.), and Department of Global Public Health and Primary Care (G.S.T.), University of Bergen, Norway; Department of Heart Disease (G.F.T.S., K.H.L., O.K.N.) and Department of Pediatrics (Ø.H.), Haukeland University Hospital, Bergen, Norway; Laboratory of Clinical Biochemistry, Bergen, Norway (P.M.U.); Food Science and Human Nutrition Department,
| | - Gard F.T. Svingen
- From the Department of Clinical Science (Y.D., E.R.P., P.M.U., O.K.N.), KG Jebsen Center for Diabetes Research, Department of Clinical Science (Ø.H., O.K.N.), and Department of Global Public Health and Primary Care (G.S.T.), University of Bergen, Norway; Department of Heart Disease (G.F.T.S., K.H.L., O.K.N.) and Department of Pediatrics (Ø.H.), Haukeland University Hospital, Bergen, Norway; Laboratory of Clinical Biochemistry, Bergen, Norway (P.M.U.); Food Science and Human Nutrition Department,
| | - Øyvind Helgeland
- From the Department of Clinical Science (Y.D., E.R.P., P.M.U., O.K.N.), KG Jebsen Center for Diabetes Research, Department of Clinical Science (Ø.H., O.K.N.), and Department of Global Public Health and Primary Care (G.S.T.), University of Bergen, Norway; Department of Heart Disease (G.F.T.S., K.H.L., O.K.N.) and Department of Pediatrics (Ø.H.), Haukeland University Hospital, Bergen, Norway; Laboratory of Clinical Biochemistry, Bergen, Norway (P.M.U.); Food Science and Human Nutrition Department,
| | - Jesse F. Gregory
- From the Department of Clinical Science (Y.D., E.R.P., P.M.U., O.K.N.), KG Jebsen Center for Diabetes Research, Department of Clinical Science (Ø.H., O.K.N.), and Department of Global Public Health and Primary Care (G.S.T.), University of Bergen, Norway; Department of Heart Disease (G.F.T.S., K.H.L., O.K.N.) and Department of Pediatrics (Ø.H.), Haukeland University Hospital, Bergen, Norway; Laboratory of Clinical Biochemistry, Bergen, Norway (P.M.U.); Food Science and Human Nutrition Department,
| | - Kjetil H. Løland
- From the Department of Clinical Science (Y.D., E.R.P., P.M.U., O.K.N.), KG Jebsen Center for Diabetes Research, Department of Clinical Science (Ø.H., O.K.N.), and Department of Global Public Health and Primary Care (G.S.T.), University of Bergen, Norway; Department of Heart Disease (G.F.T.S., K.H.L., O.K.N.) and Department of Pediatrics (Ø.H.), Haukeland University Hospital, Bergen, Norway; Laboratory of Clinical Biochemistry, Bergen, Norway (P.M.U.); Food Science and Human Nutrition Department,
| | - Klaus Meyer
- From the Department of Clinical Science (Y.D., E.R.P., P.M.U., O.K.N.), KG Jebsen Center for Diabetes Research, Department of Clinical Science (Ø.H., O.K.N.), and Department of Global Public Health and Primary Care (G.S.T.), University of Bergen, Norway; Department of Heart Disease (G.F.T.S., K.H.L., O.K.N.) and Department of Pediatrics (Ø.H.), Haukeland University Hospital, Bergen, Norway; Laboratory of Clinical Biochemistry, Bergen, Norway (P.M.U.); Food Science and Human Nutrition Department,
| | - Grethe S. Tell
- From the Department of Clinical Science (Y.D., E.R.P., P.M.U., O.K.N.), KG Jebsen Center for Diabetes Research, Department of Clinical Science (Ø.H., O.K.N.), and Department of Global Public Health and Primary Care (G.S.T.), University of Bergen, Norway; Department of Heart Disease (G.F.T.S., K.H.L., O.K.N.) and Department of Pediatrics (Ø.H.), Haukeland University Hospital, Bergen, Norway; Laboratory of Clinical Biochemistry, Bergen, Norway (P.M.U.); Food Science and Human Nutrition Department,
| | - Per M. Ueland
- From the Department of Clinical Science (Y.D., E.R.P., P.M.U., O.K.N.), KG Jebsen Center for Diabetes Research, Department of Clinical Science (Ø.H., O.K.N.), and Department of Global Public Health and Primary Care (G.S.T.), University of Bergen, Norway; Department of Heart Disease (G.F.T.S., K.H.L., O.K.N.) and Department of Pediatrics (Ø.H.), Haukeland University Hospital, Bergen, Norway; Laboratory of Clinical Biochemistry, Bergen, Norway (P.M.U.); Food Science and Human Nutrition Department,
| | - Ottar K. Nygård
- From the Department of Clinical Science (Y.D., E.R.P., P.M.U., O.K.N.), KG Jebsen Center for Diabetes Research, Department of Clinical Science (Ø.H., O.K.N.), and Department of Global Public Health and Primary Care (G.S.T.), University of Bergen, Norway; Department of Heart Disease (G.F.T.S., K.H.L., O.K.N.) and Department of Pediatrics (Ø.H.), Haukeland University Hospital, Bergen, Norway; Laboratory of Clinical Biochemistry, Bergen, Norway (P.M.U.); Food Science and Human Nutrition Department,
| |
Collapse
|
168
|
Abstract
Thymidylate (dTMP) biosynthesis plays an essential and exclusive function in DNA synthesis and proper cell division, and therefore has been an attractive therapeutic target. Folate analogs, known as antifolates, and nucleotide analogs that inhibit the enzymatic action of the de novo thymidylate biosynthesis pathway and are commonly used in cancer treatment. In this review, we examine the mechanisms by which the antifolate 5-fluorouracil, as well as other dTMP synthesis inhibitors, function in cancer treatment in light of emerging evidence that dTMP synthesis occurs in the nucleus. Nuclear localization of the de novo dTMP synthesis pathway requires modification of the pathway enzymes by the small ubiquitin-like modifier (SUMO) protein. SUMOylation is required for nuclear localization of the de novo dTMP biosynthesis pathway, and disruption in the SUMO pathway inhibits cell proliferation in several cancer models. We summarize evidence that the nuclear localization of the dTMP biosynthesis pathway is a critical factor in the efficacy of antifolate-based therapies that target dTMP synthesis.
Collapse
|
169
|
Denk MK, Milutinović NS, Marczenko KM, Sadowski NM, Paschos A. Nature's hydrides: rapid reduction of halocarbons by folate model compounds. Chem Sci 2016; 8:1883-1887. [PMID: 28553478 PMCID: PMC5424806 DOI: 10.1039/c6sc04314c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/10/2016] [Indexed: 11/21/2022] Open
Abstract
Methylenetetrahydrofolate models (green substructure) reduce organohalides to the respective hydrocarbons under biomimetic conditions and mimic the activity of dehalohydrogenases.
Halocarbons R–X are reduced to hydrocarbons R–H by folate model compounds under biomimetic conditions. The reactions correspond to a halide–hydride exchange with the methylenetetrahydrofolate (MTHF) models acting as hydride donors. The MTHF models are also functional equivalents of dehalohydrogenases but, unlike these enzymes, do not require a metal cofactor. The reactions suggest that halocarbons have the potential to act as endocrinological disruptors of biochemical pathways involving MTHF. As a case in point, we observe the rapid reaction of the MTHF models with the inhalation anaesthetic halothane. The ready synthetic accessibility of the MTHF models as well as their dehalogenation activity in the presence of air and moisture allow for the remediation of toxic, halogenated hydrocarbons.
Collapse
Affiliation(s)
- Michael K Denk
- Department of Chemistry , University of Guelph , 50 Stone Road East , Guelph , Ontario N1G 2W1 , Canada
| | - Nicholas S Milutinović
- Department of Chemistry , University of Guelph , 50 Stone Road East , Guelph , Ontario N1G 2W1 , Canada
| | - Katherine M Marczenko
- Department of Chemistry , University of Guelph , 50 Stone Road East , Guelph , Ontario N1G 2W1 , Canada
| | - Natalie M Sadowski
- Department of Chemistry , University of Guelph , 50 Stone Road East , Guelph , Ontario N1G 2W1 , Canada
| | - Athanasios Paschos
- Department of Biology , McMaster University , 1280 Main Street West , Hamilton , Ontario L8S 4K1 , Canada.,Mohawk College of Applied Arts and Technology , Department of Chemical and Environmental Technology , 135 Fennell Ave West , Hamilton , Ontario L9C 1E9 , Canada
| |
Collapse
|
170
|
Kalhan SC. One carbon metabolism in pregnancy: Impact on maternal, fetal and neonatal health. Mol Cell Endocrinol 2016; 435:48-60. [PMID: 27267668 PMCID: PMC5014566 DOI: 10.1016/j.mce.2016.06.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 06/02/2016] [Accepted: 06/02/2016] [Indexed: 02/07/2023]
Abstract
One carbon metabolism or methyl transfer, a crucial component of metabolism in all cells and tissues, supports the critical function of synthesis of purines, thymidylate and methylation via multiple methyl transferases driven by the ubiquitous methyl donor s-adenosylmethionine. Serine is the primary methyl donor to the one carbon pool. Intracellular folates and methionine metabolism are the critical components of one carbon transfer. Methionine metabolism requires vitamin B12, B6 as cofactors and is modulated by endocrine signals and is responsive to nutrient intake. Perturbations in one carbon transfer can have profound effects on cell proliferation, growth and function. Epidemiological studies in humans and experimental model have established a strong relationship between impaired fetal growth and the immediate and long term consequences to the health of the offspring. It is speculated that during development, maternal environmental and nutrient influences by their effects on one carbon transfer can impact the health of the mother, impair growth and reprogram metabolism of the fetus, and cause long term morbidity in the offspring. The potential for such effects is underscored by the unique responses in methionine metabolism in the human mother during pregnancy, the absence of transsulfuration activity in the fetus, ontogeny of methionine metabolism in the placenta and the unique metabolism of serine and glycine in the fetus. Dietary protein restriction in animals and marginal protein intake in humans causes characteristic changes in one carbon metabolism. The impact of perturbations in one carbon metabolism on the health of the mother during pregnancy, on fetal growth and the neonate are discussed and their possible mechanism explored.
Collapse
Affiliation(s)
- Satish C Kalhan
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Department of Pathobiology, Lerner Research Institute, NE-40, Cleveland Clinic, 9500 Euclid Av, Cleveland, OH, 44195, USA.
| |
Collapse
|
171
|
Jackson RA, Nguyen ML, Barrett AN, Tan YY, Choolani MA, Chen ES. Synthetic combinations of missense polymorphic genetic changes underlying Down syndrome susceptibility. Cell Mol Life Sci 2016; 73:4001-17. [PMID: 27245382 PMCID: PMC11108497 DOI: 10.1007/s00018-016-2276-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 02/08/2023]
Abstract
Single nucleotide polymorphisms (SNPs) are important biomolecular markers in health and disease. Down syndrome, or Trisomy 21, is the most frequently occurring chromosomal abnormality in live-born children. Here, we highlight associations between SNPs in several important enzymes involved in the one-carbon folate metabolic pathway and the elevated maternal risk of having a child with Down syndrome. Our survey highlights that the combination of SNPs may be a more reliable predictor of the Down syndrome phenotype than single SNPs alone. We also describe recent links between SNPs in p53 and its related pathway proteins and Down syndrome, as well as highlight several proteins that help to associate apoptosis and p53 signaling with the Down syndrome phenotype. In addition to a comprehensive review of the literature, we also demonstrate that several SNPs reside within the same regions as these Down syndrome-linked SNPs, and propose that these closely located nucleotide changes may provide new candidates for future exploration.
Collapse
Affiliation(s)
- Rebecca A Jackson
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, #05-05, MD7, 8 Medical Drive, Singapore, 117597, Singapore
| | - Mai Linh Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, #05-05, MD7, 8 Medical Drive, Singapore, 117597, Singapore
| | - Angela N Barrett
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, #05-05, MD7, 8 Medical Drive, Singapore, 117597, Singapore
| | - Yuan Yee Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, #05-05, MD7, 8 Medical Drive, Singapore, 117597, Singapore
| | - Mahesh A Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, #05-05, MD7, 8 Medical Drive, Singapore, 117597, Singapore.
- National University Health System, Singapore, Singapore.
| | - Ee Sin Chen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, #05-05, MD7, 8 Medical Drive, Singapore, 117597, Singapore.
- National University Health System, Singapore, Singapore.
- NUS Graduate School of Science and Engineering, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
172
|
Sarmah S, Muralidharan P, Marrs JA. Common congenital anomalies: Environmental causes and prevention with folic acid containing multivitamins. ACTA ACUST UNITED AC 2016; 108:274-286. [PMID: 27718306 DOI: 10.1002/bdrc.21138] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 09/23/2016] [Indexed: 12/13/2022]
Abstract
Congenital anomalies, congenital defects, or birth defects are significant causes of death in infants. The most common congenital defects are congenital heart defects (CHDs) and neural tube defects (NTDs). Defects induced by genetic mutations, environmental exposure to toxins, or a combination of these effects can result in congenital malformations, leading to infant death or long-term disabilities. These defects produce significant mortality and morbidity in the affected individuals, and families are affected emotional and financially. Also, society is impacted on many levels. Congenital anomalies may be reduced by dietary supplements of folic acid and other vitamins. Here, we review the evidence for specific roles of toxins (alcohol, cigarette smoke) in causing common severe congenital anomalies like CHDs, NTDs, and ocular defects. We also review the evidence for beneficial effects for dietary supplementation, and highlight gaps in our knowledge, where research may contribute to additional benefits of intervention that can reduce birth defects. Extensive discussion of common severe congenital anomalies (CHDs, NTDs, and ocular defects) illustrates the effects of diet on the frequency and severity of these defects. Birth Defects Research (Part C) 108:274-286, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Swapnalee Sarmah
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202.
| | - Pooja Muralidharan
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202
| | - James A Marrs
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202.
| |
Collapse
|
173
|
Association between methylenetetrahydrofolate reductase (MTHFR) C677T gene polymorphism and risk of ischemic stroke in North Indian population: A hospital based case–control study. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2016. [DOI: 10.1016/j.ejmhg.2016.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
174
|
Kim BG, Park HW. Tetrahydrofolate increases suspension growth of dihydrofolate reductase-deficient chinese hamster ovary DG44 cells in chemically defined media. Biotechnol Prog 2016; 32:1539-1546. [PMID: 27578320 DOI: 10.1002/btpr.2351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/17/2016] [Indexed: 11/09/2022]
Abstract
Adaptation of dihydrofolate reductase (DHFR)-deficient Chinese hamster ovary (CHO) DG44 cells to chemically defined suspension culture conditions is a time-consuming and labor-intensive process because nonadapted DHFR-deficient CHO DG44 cells normally show poor growth in chemically defined medium (CDM). We examined the effects of folate derivatives, ribonucleotides, and nucleobases on the growth of suspension-adapted DHFR-deficient CHO DG44 cells in CDM. Among the tested additives, tetrahydrofolate (THF) was identified as an effective component for increasing cell growth. THF supplementation in the range of 0.2-359 μM enhanced cell growth in in-house CDM. Addition of 3.6 μM THF to in-house CDM resulted in a more than 2.5-fold increase in maximum viable cell density. Moreover, supplementation of six different commercial CDMs with 3.6 μM THF yielded up to 2.9-fold enhancement of maximum viable cell density. An anchorage- and serum-dependent DHFR-deficient CHO DG44 cell line was adapted within two consecutive passages to suspension growth in in-house CDM supplemented with 3.6 μM THF. These data indicate that supplementation of chemically defined cell culture media with greater than 0.2 μM THF can help achieve a high density of suspension-adapted DHFR-deficient CHO DG44 cells and may facilitate rapid adaptation of nonadapted DHFR-deficient CHO DG44 cells to suspension culture. © 2016 American Institute of Chemical Engineers Biotechnol. Prog., 32:1539-1546, 2016.
Collapse
Affiliation(s)
- Bong Gyun Kim
- Dept. of Chemical Engineering, Hanyang University, Seoul, 133-791, South Korea
| | - Hong Woo Park
- Dept. of Chemical Engineering, Hanyang University, Seoul, 133-791, South Korea
| |
Collapse
|
175
|
Measurement of Histone Methylation Dynamics by One-Carbon Metabolic Isotope Labeling and High-energy Collisional Dissociation Methylation Signature Ion Detection. Sci Rep 2016; 6:31537. [PMID: 27530234 PMCID: PMC4987619 DOI: 10.1038/srep31537] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 07/21/2016] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence suggests that cellular metabolites and nutrition levels control epigenetic modifications, including histone methylation. However, it is not currently possible to measure the metabolic control of histone methylation. Here we report a novel detection method to monitor methyl transfer from serine to histones through the one-carbon metabolic pathway, using stable-isotope labeling and detection of lysine methylation signature ions generated in high-energy-dissociation (HCD) tandem mass spectrometry. This method is a long-needed tool to study the metabolic control of histone methylation.
Collapse
|
176
|
Corbin JM, Ruiz-Echevarría MJ. One-Carbon Metabolism in Prostate Cancer: The Role of Androgen Signaling. Int J Mol Sci 2016; 17:E1208. [PMID: 27472325 PMCID: PMC5000606 DOI: 10.3390/ijms17081208] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/16/2016] [Accepted: 07/18/2016] [Indexed: 01/06/2023] Open
Abstract
Cancer cell metabolism differs significantly from the metabolism of non-transformed cells. This altered metabolic reprogramming mediates changes in the uptake and use of nutrients that permit high rates of proliferation, growth, and survival. The androgen receptor (AR) plays an essential role in the establishment and progression of prostate cancer (PCa), and in the metabolic adaptation that takes place during this progression. In its role as a transcription factor, the AR directly affects the expression of several effectors and regulators of essential catabolic and biosynthetic pathways. Indirectly, as a modulator of the one-carbon metabolism, the AR can affect epigenetic processes, DNA metabolism, and redox balance, all of which are important factors in tumorigenesis. In this review, we focus on the role of AR-signaling on one-carbon metabolism in tumorigenesis. Clinical implications of one-carbon metabolism and AR-targeted therapies for PCa are discussed in this context.
Collapse
Affiliation(s)
- Joshua M Corbin
- Department of Pathology, Oklahoma University Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Maria J Ruiz-Echevarría
- Department of Pathology, Oklahoma University Health Sciences Center and Stephenson Cancer Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
177
|
Khaldoun Oularbi H, Richeval C, Lebaili N, Zerrouki-Daoudi N, Baha M, Djennas N, Allorge D. Ameliorative effect of vitamin C against hepatotoxicity induced by emamectin benzoate in rats. Hum Exp Toxicol 2016; 36:709-717. [PMID: 27461011 DOI: 10.1177/0960327116661022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In the present study, we aimed to assess the potential protective effect of ascorbic acid (AA) against emamectin benzoate (EMB)-induced hepatotoxicity. For this purpose, biochemical, histopathological and analytical investigations were performed. Male Wistar rats were distributed into three groups, that is, a control group, an EMB group given 10 mg EMB/kg body weight (BW) by gavage and an EMB + AA group given 10 mg EMB/kg BW and vitamin C intraperitoneally (200 mg/kg). The duration of the treatment was 28 days and the duration of the study was 42 days. There was a statistically significant increase of all hepatic biomarkers, that is, aspartate aminotransferase, alanine aminotransferase and gamma-glutamyltransferase activities, and glycemia, in EMB-treated group when compared with the control group. Light microscopic observations revealed variable signs of hepatotoxicity in the EMB group, which were represented by alteration of normal hepatic architecture, inflammatory cell infiltration, hepatocellular steatosis and foci of necrosis at 28 and 42 days post-treatment. However, co-treatment with vitamin C reduced EMB-related liver toxicity and diminished the abnormal biochemical and architectural damage. Emamectin B1a and B1b residues were detectable in all plasma samples of treated rats at 14, 21 and 28 days of treatment. The drug liver tissue concentration was significantly lower in EMB + AA group compared with EMB group at 28 and 42 days. In conclusion, the findings of the present study clearly indicate a significant protective action of vitamin C against EMB hepatotoxicity.
Collapse
Affiliation(s)
- H Khaldoun Oularbi
- 1 Département de Biologie et Physiologie cellulaire, faculté des Sciences de la Nature et de la Vie, Université Blida 1, BP 270, route Soumaa, Blida, Algeria.,2 Laboratoire de Recherche d'Éco-Biologie Animale, École Normale Supérieure de Kouba Bachir El Ibrahimi, Algiers, Algeria
| | - C Richeval
- 3 CHU Lille, Unité Fonctionnelle de Toxicologie, F-59000 Lille, France
| | - N Lebaili
- 2 Laboratoire de Recherche d'Éco-Biologie Animale, École Normale Supérieure de Kouba Bachir El Ibrahimi, Algiers, Algeria
| | - N Zerrouki-Daoudi
- 4 Laboratoire des Ressorces Naturelles, Universite Mouloud Mammeri, Tizi-Ouzou, Algeria
| | - M Baha
- 2 Laboratoire de Recherche d'Éco-Biologie Animale, École Normale Supérieure de Kouba Bachir El Ibrahimi, Algiers, Algeria
| | - N Djennas
- 5 Laboratoire D'anatomie Pathologie CHRU Parnet, Alger, Algeria
| | - D Allorge
- 2 Laboratoire de Recherche d'Éco-Biologie Animale, École Normale Supérieure de Kouba Bachir El Ibrahimi, Algiers, Algeria.,6 Univ. Lille, EA 4483 - IMPECS - IMPact de l'Environnement Chimique sur la Santé humaine, F-59000 Lille, France
| |
Collapse
|
178
|
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: 6.6] [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
|
179
|
Ron-Harel N, Santos D, Ghergurovich JM, Sage PT, Reddy A, Lovitch SB, Dephoure N, Satterstrom FK, Sheffer M, Spinelli JB, Gygi S, Rabinowitz JD, Sharpe AH, Haigis MC. Mitochondrial Biogenesis and Proteome Remodeling Promote One-Carbon Metabolism for T Cell Activation. Cell Metab 2016; 24:104-17. [PMID: 27411012 PMCID: PMC5330619 DOI: 10.1016/j.cmet.2016.06.007] [Citation(s) in RCA: 290] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/12/2016] [Accepted: 06/10/2016] [Indexed: 01/06/2023]
Abstract
Naive T cell stimulation activates anabolic metabolism to fuel the transition from quiescence to growth and proliferation. Here we show that naive CD4(+) T cell activation induces a unique program of mitochondrial biogenesis and remodeling. Using mass spectrometry, we quantified protein dynamics during T cell activation. We identified substantial remodeling of the mitochondrial proteome over the first 24 hr of T cell activation to generate mitochondria with a distinct metabolic signature, with one-carbon metabolism as the most induced pathway. Salvage pathways and mitochondrial one-carbon metabolism, fed by serine, contribute to purine and thymidine synthesis to enable T cell proliferation and survival. Genetic inhibition of the mitochondrial serine catabolic enzyme SHMT2 impaired T cell survival in culture and antigen-specific T cell abundance in vivo. Thus, during T cell activation, mitochondrial proteome remodeling generates specialized mitochondria with enhanced one-carbon metabolism that is critical for T cell activation and survival.
Collapse
Affiliation(s)
- Noga Ron-Harel
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel Santos
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Jonathan M Ghergurovich
- The Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Peter T Sage
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Anita Reddy
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Scott B Lovitch
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Noah Dephoure
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - F Kyle Satterstrom
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Michal Sheffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jessica B Spinelli
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Joshua D Rabinowitz
- The Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Marcia C Haigis
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
180
|
Raz S, Stark M, Assaraf YG. Folylpoly-γ-glutamate synthetase: A key determinant of folate homeostasis and antifolate resistance in cancer. Drug Resist Updat 2016; 28:43-64. [PMID: 27620954 DOI: 10.1016/j.drup.2016.06.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 06/10/2016] [Accepted: 06/16/2016] [Indexed: 01/26/2023]
Abstract
Mammalians are devoid of autonomous biosynthesis of folates and hence must obtain them from the diet. Reduced folate cofactors are B9-vitamins which play a key role as donors of one-carbon units in the biosynthesis of purine nucleotides, thymidylate and amino acids as well as in a multitude of methylation reactions including DNA, RNA, histone and non-histone proteins, phospholipids, as well as intermediate metabolites. The products of these S-adenosylmethionine (SAM)-dependent methylations are involved in the regulation of key biological processes including transcription, translation and intracellular signaling. Folate-dependent one-carbon metabolism occurs in several subcellular compartments including the cytoplasm, mitochondria, and nucleus. Since folates are essential for DNA replication, intracellular folate cofactors play a central role in cancer biology and inflammatory autoimmune disorders. In this respect, various folate-dependent enzymes catalyzing nucleotide biosynthesis have been targeted by specific folate antagonists known as antifolates. Currently, antifolates are used in drug treatment of multiple human cancers, non-malignant chronic inflammatory disorders as well as bacterial and parasitic infections. An obligatory key component of intracellular folate retention and intracellular homeostasis is (anti)folate polyglutamylation, mediated by the unique enzyme folylpoly-γ-glutamate synthetase (FPGS), which resides in both the cytoplasm and mitochondria. Consistently, knockout of the FPGS gene in mice results in embryonic lethality. FPGS catalyzes the addition of a long polyglutamate chain to folates and antifolates, hence rendering them polyanions which are efficiently retained in the cell and are now bound with enhanced affinity by various folate-dependent enzymes. The current review highlights the crucial role that FPGS plays in maintenance of folate homeostasis under physiological conditions and delineates the plethora of the molecular mechanisms underlying loss of FPGS function and consequent antifolate resistance in cancer.
Collapse
Affiliation(s)
- Shachar Raz
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Michal Stark
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
181
|
Ganz AB, Shields K, Fomin VG, Lopez YS, Mohan S, Lovesky J, Chuang JC, Ganti A, Carrier B, Yan J, Taeswuan S, Cohen VV, Swersky CC, Stover JA, Vitiello GA, Malysheva OV, Mudrak E, Caudill MA. Genetic impairments in folate enzymes increase dependence on dietary choline for phosphatidylcholine production at the expense of betaine synthesis. FASEB J 2016; 30:3321-3333. [PMID: 27342765 DOI: 10.1096/fj.201500138rr] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 06/14/2016] [Indexed: 01/15/2023]
Abstract
Although single nucleotide polymorphisms (SNPs) in folate-mediated pathways predict susceptibility to choline deficiency during severe choline deprivation, it is unknown if effects persist at recommended intakes. Thus, we used stable isotope liquid chromatography-mass spectrometry (LC-MS) methodology to examine the impact of candidate SNPs on choline metabolism in a long-term, randomized, controlled feeding trial among pregnant, lactating, and nonpregnant (NP) women consuming 480 or 930 mg/d choline (22% as choline-d9, with d9 indicating a deuterated trimethyl amine group) and meeting folate-intake recommendations. Variants impairing folate metabolism, methylenetetrahydrofolate reductase (MTHFR) rs1801133, methionine synthase (MTR) rs1805087 [wild-type (WT)], MTR reductase (MTRR) rs1801394, and methylenetetrahydrofolate dehydrogenase-methenyltetrahydrofolate cyclohydrolase-formyltetrahydrofolate synthetase (MTHFD1) rs2236225, influenced choline dynamics, frequently through interactions with reproductive state and choline intake, with fewer genotypic alterations observed among pregnant women. Women with these variants partitioned more dietary choline toward phosphatidylcholine (PC) biosynthesis via the cytidine diphosphate (CDP)-choline pathway at the expense of betaine synthesis even when use of betaine as a methyl donor was increased. Choline intakes of 930 mg/d restored partitioning of dietary choline between betaine and CDP-PC among NP (MTHFR rs1801133 and MTR rs1805087 WT) and lactating (MTHFD1 rs2236225) women with risk genotypes. Overall, our findings indicate that loss-of-function variants in folate-metabolizing enzymes strain cellular PC production, possibly via impaired folate-dependent phosphatidylethanolamine-N-methyltransferase (PEMT)-PC synthesis, and suggest that women with these risk genotypes may benefit from choline intakes exceeding current recommendations.-Ganz, A. B., Shields, K., Fomin, V. G., Lopez, Y. S., Mohan, S., Lovesky, J., Chuang, J. C., Ganti, A., Carrier, B., Yan, J., Taeswuan, S., Cohen, V. V., Swersky, C. C., Stover, J. A., Vitiello, G. A., Malysheva, O. V., Mudrak, E., Caudill, M. A. Genetic impairments in folate enzymes increase dependence on dietary choline for phosphatidylcholine production at the expense of betaine synthesis.
Collapse
Affiliation(s)
- Ariel B Ganz
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Kelsey Shields
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Vlad G Fomin
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Yusnier S Lopez
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Sanjay Mohan
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Jessica Lovesky
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Jasmine C Chuang
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Anita Ganti
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Bradley Carrier
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Jian Yan
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Siraphat Taeswuan
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Vanessa V Cohen
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Camille C Swersky
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Julie A Stover
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Gerardo A Vitiello
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Olga V Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| | - Erika Mudrak
- Statistical Consulting Unit, Cornell University, Ithaca, New York, USA
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA; and
| |
Collapse
|
182
|
Krushkal J, Zhao Y, Hose C, Monks A, Doroshow JH, Simon R. Concerted changes in transcriptional regulation of genes involved in DNA methylation, demethylation, and folate-mediated one-carbon metabolism pathways in the NCI-60 cancer cell line panel in response to cancer drug treatment. Clin Epigenetics 2016; 8:73. [PMID: 27347216 PMCID: PMC4919895 DOI: 10.1186/s13148-016-0240-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Aberrant patterns of DNA methylation are abundant in cancer, and epigenetic pathways are increasingly being targeted in cancer drug treatment. Genetic components of the folate-mediated one-carbon metabolism pathway can affect DNA methylation and other vital cell functions, including DNA synthesis, amino acid biosynthesis, and cell growth. RESULTS We used a bioinformatics tool, the Transcriptional Pharmacology Workbench, to analyze temporal changes in gene expression among epigenetic regulators of DNA methylation and demethylation, and one-carbon metabolism genes in response to cancer drug treatment. We analyzed gene expression information from the NCI-60 cancer cell line panel after treatment with five antitumor agents, 5-azacytidine, doxorubicin, vorinostat, paclitaxel, and cisplatin. Each antitumor agent elicited concerted changes in gene expression of multiple pathway components across the cell lines. Expression changes of FOLR2, SMUG1, GART, GADD45A, MBD1, MTR, MTHFD1, and CTH were significantly correlated with chemosensitivity to some of the agents. Among many genes with concerted expression response to individual antitumor agents were genes encoding DNA methyltransferases DNMT1, DNMT3A, and DNMT3B, epigenetic and DNA repair factors MGMT, GADD45A, and MBD1, and one-carbon metabolism pathway members MTHFD1, TYMS, DHFR, MTR, MAT2A, SLC19A1, ATIC, and GART. CONCLUSIONS These transcriptional changes are likely to influence vital cellular functions of DNA methylation and demethylation, cellular growth, DNA biosynthesis, and DNA repair, and some of them may contribute to cytotoxic and apoptotic action of the drugs. This concerted molecular response was observed in a time-dependent manner, which may provide future guidelines for temporal selection of genetic drug targets for combination drug therapy treatment regimens.
Collapse
Affiliation(s)
- Julia Krushkal
- />Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD 20850 USA
| | - Yingdong Zhao
- />Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD 20850 USA
| | - Curtis Hose
- />Molecular Pharmacology Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Anne Monks
- />Molecular Pharmacology Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - James H. Doroshow
- />Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD 20892 USA
| | - Richard Simon
- />Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD 20850 USA
| |
Collapse
|
183
|
Development of ic-ELISA and lateral-flow immunochromatographic assay strip for the detection of folic acid in energy drinks and milk samples. FOOD AGR IMMUNOL 2016. [DOI: 10.1080/09540105.2016.1183600] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
184
|
French E, Kim BS, Iyer-Pascuzzi AS. Mechanisms of quantitative disease resistance in plants. Semin Cell Dev Biol 2016; 56:201-208. [PMID: 27212254 DOI: 10.1016/j.semcdb.2016.05.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/14/2016] [Accepted: 05/18/2016] [Indexed: 11/29/2022]
Abstract
Quantitative disease resistance (QDR) causes the reduction, but not absence, of disease, and is a major type of disease resistance for many crop species. QDR results in a continuous distribution of disease scores across a segregating population, and is typically due to many genes with small effects. It may also be a source of durable resistance. The past decade has seen significant progress in cloning genes underlying QDR. In this review, we focus on these recently cloned genes and identify new themes of QDR emerging from these studies.
Collapse
Affiliation(s)
- Elizabeth French
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN 47907, United States
| | - Bong-Suk Kim
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN 47907, United States
| | - Anjali S Iyer-Pascuzzi
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
185
|
Matone A, Scott-Boyer MP, Carayol J, Fazelzadeh P, Lefebvre G, Valsesia A, Charon C, Vervoort J, Astrup A, Saris WHM, Morine M, Hager J. Network Analysis of Metabolite GWAS Hits: Implication of CPS1 and the Urea Cycle in Weight Maintenance. PLoS One 2016; 11:e0150495. [PMID: 26938218 PMCID: PMC4777532 DOI: 10.1371/journal.pone.0150495] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 02/15/2016] [Indexed: 01/09/2023] Open
Abstract
Background and Scope Weight loss success is dependent on the ability to refrain from regaining the lost weight in time. This feature was shown to be largely variable among individuals, and these differences, with their underlying molecular processes, are diverse and not completely elucidated. Altered plasma metabolites concentration could partly explain weight loss maintenance mechanisms. In the present work, a systems biology approach has been applied to investigate the potential mechanisms involved in weight loss maintenance within the Diogenes weight-loss intervention study. Methods and Results A genome wide association study identified SNPs associated with plasma glycine levels within the CPS1 (Carbamoyl-Phosphate Synthase 1) gene (rs10206976, p-value = 4.709e-11 and rs12613336, p-value = 1.368e-08). Furthermore, gene expression in the adipose tissue showed that CPS1 expression levels were associated with successful weight maintenance and with several SNPs within CPS1 (cis-eQTL). In order to contextualize these results, a gene-metabolite interaction network of CPS1 and glycine has been built and analyzed, showing functional enrichment in genes involved in lipid metabolism and one carbon pool by folate pathways. Conclusions CPS1 is the rate-limiting enzyme for the urea cycle, catalyzing carbamoyl phosphate from ammonia and bicarbonate in the mitochondria. Glycine and CPS1 are connected through the one-carbon pool by the folate pathway and the urea cycle. Furthermore, glycine could be linked to metabolic health and insulin sensitivity through the betaine osmolyte. These considerations, and the results from the present study, highlight a possible role of CPS1 and related pathways in weight loss maintenance, suggesting that it might be partly genetically determined in humans.
Collapse
Affiliation(s)
- Alice Matone
- The Microsoft Research—University of Trento Centre for Computational Systems Biology (COSBI), Rovereto, Italy
| | - Marie-Pier Scott-Boyer
- The Microsoft Research—University of Trento Centre for Computational Systems Biology (COSBI), Rovereto, Italy
| | - Jerome Carayol
- Nestlé Institute of Health Sciences SA, Lausanne, Switzerland
| | - Parastoo Fazelzadeh
- Nutrition, Metabolism & Genomics group, University of Wageningen, Wageningen, Netherlands
| | | | - Armand Valsesia
- Nestlé Institute of Health Sciences SA, Lausanne, Switzerland
| | - Celine Charon
- CEA-Genomics Institute- National Genotyping Center, Evry, France
| | - Jacques Vervoort
- Nutrition, Metabolism & Genomics group, University of Wageningen, Wageningen, Netherlands
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Wim H. M. Saris
- Dept of Human Biology Medical and Health Science Faculty, University of Maastricht, Maastricht, Netherlands
| | - Melissa Morine
- The Microsoft Research—University of Trento Centre for Computational Systems Biology (COSBI), Rovereto, Italy
| | - Jörg Hager
- Nestlé Institute of Health Sciences SA, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
186
|
Rijpma SR, van der Velden M, Bilos A, Jansen RS, Mahakena S, Russel FGM, Sauerwein RW, van de Wetering K, Koenderink JB. MRP1 mediates folate transport and antifolate sensitivity in Plasmodium falciparum. FEBS Lett 2016; 590:482-92. [PMID: 26900081 DOI: 10.1002/1873-3468.12079] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 01/12/2016] [Accepted: 01/18/2016] [Indexed: 11/05/2022]
Abstract
Multidrug resistance-associated proteins (MRP) of Plasmodium falciparum have been associated with altered drug sensitivity. Knowledge on MRP substrate specificity is indispensible for the characterization of resistance mechanisms and identifying its physiological roles. An untargeted metabolomics approach detected decreased folate concentrations in red blood cells infected with schizont stage parasites lacking expression of MRP1. Furthermore, a tenfold decrease in sensitivity toward the folate analog methotrexate was detected for parasites lacking MRP1. PfMRP1 is involved in the export of folate from parasites into red blood cells and is therefore a relevant factor for efficient malaria treatment through the folate pathway.
Collapse
Affiliation(s)
- Sanna R Rijpma
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maarten van der Velden
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Albert Bilos
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Robert S Jansen
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sunny Mahakena
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Koen van de Wetering
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jan B Koenderink
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
187
|
Joubert BR, den Dekker HT, Felix JF, Bohlin J, Ligthart S, Beckett E, Tiemeier H, van Meurs JB, Uitterlinden AG, Hofman A, Håberg SE, Reese SE, Peters MJ, Andreassen BK, Steegers EAP, Nilsen RM, Vollset SE, Midttun Ø, Ueland PM, Franco OH, Dehghan A, de Jongste JC, Wu MC, Wang T, Peddada SD, Jaddoe VWV, Nystad W, Duijts L, London SJ. Maternal plasma folate impacts differential DNA methylation in an epigenome-wide meta-analysis of newborns. Nat Commun 2016; 7:10577. [PMID: 26861414 PMCID: PMC4749955 DOI: 10.1038/ncomms10577] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 12/31/2015] [Indexed: 12/13/2022] Open
Abstract
Folate is vital for fetal development. Periconceptional folic acid supplementation and food fortification are recommended to prevent neural tube defects. Mechanisms whereby periconceptional folate influences normal development and disease are poorly understood: epigenetics may be involved. We examine the association between maternal plasma folate during pregnancy and epigenome-wide DNA methylation using Illumina's HumanMethyl450 Beadchip in 1,988 newborns from two European cohorts. Here we report the combined covariate-adjusted results using meta-analysis and employ pathway and gene expression analyses. Four-hundred forty-three CpGs (320 genes) are significantly associated with maternal plasma folate levels during pregnancy (false discovery rate 5%); 48 are significant after Bonferroni correction. Most genes are not known for folate biology, including APC2, GRM8, SLC16A12, OPCML, PRPH, LHX1, KLK4 and PRSS21. Some relate to birth defects other than neural tube defects, neurological functions or varied aspects of embryonic development. These findings may inform how maternal folate impacts the developing epigenome and health outcomes in offspring.
Collapse
Affiliation(s)
- Bonnie R Joubert
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| | - Herman T den Dekker
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Pediatrics, Division of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Jon Bohlin
- Norwegian Institute of Public Health, Oslo 0403, Norway
| | - Symen Ligthart
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Emma Beckett
- Department of Applied Sciences, School of Environmental and Life Sciences, University of Newcastle, Ourimbah, New South Wales 2258, Australia.,Food and Nutrition Flagship, CSIRO, North Ryde, New South Wales 2113, Australia
| | - Henning Tiemeier
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Psychiatry, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Joyce B van Meurs
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Siri E Håberg
- Norwegian Institute of Public Health, Oslo 0403, Norway
| | - Sarah E Reese
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| | - Marjolein J Peters
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Bettina Kulle Andreassen
- Department of Clinical Molecular Biology, Institute of Clinical Medicine, University of Oslo, Oslo 0316, Norway
| | - Eric A P Steegers
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Roy M Nilsen
- Department of Research and Development, Centre for Clinical Research, Haukeland University Hospital, Bergen 5021, Norway
| | - Stein E Vollset
- Norwegian Institute of Public Health, Oslo 0403, Norway.,Department of Global Public Health and Primary Care, University of Bergen, Bergen 5018, Norway
| | | | - Per M Ueland
- Department of Clinical Science, University of Bergen, Bergen 5018, Norway.,Laboratory of Clinical Biochemistry, Haukeland University Hospital, Bergen 5018, Norway
| | - Oscar H Franco
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Johan C de Jongste
- Department of Pediatrics, Division of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Michael C Wu
- Public Health Sciences Program, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Tianyuan Wang
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| | - Shyamal D Peddada
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Wenche Nystad
- Norwegian Institute of Public Health, Oslo 0403, Norway
| | - Liesbeth Duijts
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Pediatrics, Division of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3000 CA, Netherlands
| | - Stephanie J London
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
188
|
Field MS, Kamynina E, Stover PJ. MTHFD1 regulates nuclear de novo thymidylate biosynthesis and genome stability. Biochimie 2016; 126:27-30. [PMID: 26853819 DOI: 10.1016/j.biochi.2016.02.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 02/02/2016] [Indexed: 11/16/2022]
Abstract
Disruptions in folate-mediated one-carbon metabolism (FOCM) are associated with risk for several pathologies including developmental anomalies such as neural tube defects and congenital heart defects, diseases of aging including cognitive decline, neurodegeneration and epithelial cancers, and hematopoietic disorders including megaloblastic anemia. However, the causal pathways and mechanisms that underlie these pathologies remain unresolved. Because folate-dependent anabolic pathways are tightly interconnected and best described as a metabolic network, the identification of causal pathways and associated mechanisms of pathophysiology remains a major challenge in identifying the contribution of individual pathways to disease phenotypes. Investigations of genetic mouse models and human inborn errors of metabolism enable a more precise dissection of the pathways that constitute the FOCM network and enable elucidation of causal pathways associated with NTDs. In this overview, we summarize recent evidence that the enzyme MTHFD1 plays an essential role in FOCM in humans and in mice, and that it determines the partitioning of folate-activated one carbon units between the folate-dependent de novo thymidylate and homocysteine remethylation pathways through its regulated nuclear localization. We demonstrate that impairments in MTHFD1 activity compromise both homocysteine remethylation and de novo thymidylate biosynthesis, and provide evidence that MTHFD1-associated disruptions in de novo thymidylate biosynthesis lead to genome instability that may underlie folate-associated immunodeficiency and birth defects.
Collapse
Affiliation(s)
- Martha S Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Elena Kamynina
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Patrick J Stover
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; Graduate Field of Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
189
|
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: 1.3] [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
|
190
|
Cook LS, Leung ACY, Swenerton K, Gallagher RP, Magliocco A, Steed H, Koebel M, Nation J, Eshragh S, Brooks-Wilson A, Le ND. Adult lifetime alcohol consumption and invasive epithelial ovarian cancer risk in a population-based case-control study. Gynecol Oncol 2015; 140:277-84. [PMID: 26691218 DOI: 10.1016/j.ygyno.2015.12.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 12/04/2015] [Accepted: 12/11/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Meta-analyses report a null association between recent alcohol consumption and ovarian cancer risk. However, because few studies investigated different types of alcohol over adult ages, we investigated adult lifetime and type (beer, wine, spirits) of consumption and risk. METHODS Consumption after age 20years was ascertained in 1144 invasive epithelial ovarian cancer cases and 2513 controls in a population-based case-control study (Alberta and British Columbia, Canada, 2001-2012). Non-drinkers consumed any types of alcohol <12 times per year on average. Logistic regression was use to estimate adjusted odds ratios [aOR] and 95% confidence intervals [CIs]. RESULTS Wine consumption was associated with a risk reduction (aOR=0.67, 95% CI: 0.50-0.88) relative to non-drinkers, but not beer (aOR=1.06, 95% CI: 0.71-1.58) or spirits (aOR=0.98, 95% CI: 0.69-1.39). The reduced risk was stronger for exclusive red wine drinkers (aOR=0.44, 95% CI: 0.19-0.92) than white wine drinkers (aOR=0.79, 95% CI: 0.46-1.34), although most women drank both types of wine. Risk decreased with increasing cumulative consumption of any wine (P-trend<0.05) and was evident for the serous histotype. Wine consumption initiated prior to age 50 was associated with a risk reduction (e.g., at 40-49years, aOR=0.58, 95% CI: 0.42-0.78), but not drinking initiated after 50years of age. For any type, level, or age at initiation of alcohol consumption, we found no increased risks. CONCLUSIONS For the moderate consumption in this study, higher levels of wine consumption were generally associated with risk reductions; reductions may be stronger for red wine. Our results suggest that alcohol consumption that is guideline concordant will not increase epithelial ovarian cancer risk.
Collapse
Affiliation(s)
- Linda S Cook
- Internal Medicine, University of New Mexico and UNM Comprehensive Cancer Center, Albuquerque, NM, USA; Community Health Sciences, University of Calgary, Calgary, AB, Canada; Alberta Health Services, Calgary, AB, Canada.
| | - Andy C Y Leung
- Cancer Control Research, BC Cancer Research Centre, Vancouver, BC, Canada.
| | | | | | - Anthony Magliocco
- Department of Anatomic Pathology, H Lee Moffitt Cancer Center, Tampa, FL, USA.
| | - Helen Steed
- Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada.
| | - Martin Koebel
- Pathology & Laboratory Medicine, University of Calgary, Calgary, AB, Canada.
| | - Jill Nation
- Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Sima Eshragh
- Pathology, Vancouver General Hospital, Vancouver, BC, Canada.
| | - Angela Brooks-Wilson
- Cancer Control Research, BC Cancer Research Centre, Vancouver, BC, Canada; Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, Canada; Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada.
| | - Nhu D Le
- Cancer Control Research, BC Cancer Research Centre, Vancouver, BC, Canada.
| |
Collapse
|
191
|
Effect of in ovo feeding of folic acid on the folate metabolism, immune function and epigenetic modification of immune effector molecules of broiler. Br J Nutr 2015; 115:411-21. [DOI: 10.1017/s0007114515004511] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
AbstractThis study was conducted to investigate the effect of in ovo feeding (IOF) of folic acid on the folate metabolism, immune function and the involved epigenetic modification of broilers. A total of 400 (Cobb) hatching eggs were randomly divided into four groups (0, 50, 100 and 150 µg injection of folic acid at embryonic age 11 d), and chicks hatched from each treatment were randomly divided into six replicates with 12 broilers/replicate after incubation. The results indicated that, in ovo, 100- and 150-µg folic acid injections improved the hatchability. The average daily gain and feed conversion ratio increased in the 150-µg group during the late growth stage. Simultaneously, in the 100- and 150-µg groups, an increase was observed in hepatic folate content and the expression of methylenetetrahydrofolate reductase (d1 and 42) and methionine synthase reductase (d21). IgG and IgM concentrations, as well as plasma lysozyme activity of broilers, showed a marked increase along with increasing folic acid levels. The splenic expression levels of IL-2 and IL-4 were up-regulated, whereas that of IL-6 was down-regulated, in the 100- and 150-µg folic acid treatment groups. In addition, histone methylation in IL-2 and IL-4 promoters exhibited an enrichment of H3K4m2 but a loss of H3K9me2 with the increased amount of folic acid additive. In contrast, a decrease in H3K4m2 and an increase in H3K9me2 were observed in the IL-6 promoter in folic acid treatments. Furthermore, in ovo, the 150-µg folic acid injection improved the chromatin tightness of the IL-2 and IL-4 promoter regions. Our findings suggest that IOF of 150 µg of folic acid can improve the growth performance and folate metabolism of broilers, and enhance the relationship between immune function and epigenetic regulation of immune genes, which are involved with the alterations in chromatin conformation and histone methylation in their promoters.
Collapse
|
192
|
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: 0.9] [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
|
193
|
Zhang X, Diao R, Zhu X, Li Z, Cai Z. Metabolic characterization of asthenozoospermia using nontargeted seminal plasma metabolomics. Clin Chim Acta 2015; 450:254-61. [DOI: 10.1016/j.cca.2015.09.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/27/2015] [Accepted: 09/01/2015] [Indexed: 12/26/2022]
|
194
|
Walker MC, van der Donk WA. The many roles of glutamate in metabolism. J Ind Microbiol Biotechnol 2015; 43:419-30. [PMID: 26323613 DOI: 10.1007/s10295-015-1665-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 07/25/2015] [Indexed: 12/20/2022]
Abstract
The amino acid glutamate is a major metabolic hub in many organisms and as such is involved in diverse processes in addition to its role in protein synthesis. Nitrogen assimilation, nucleotide, amino acid, and cofactor biosynthesis, as well as secondary natural product formation all utilize glutamate in some manner. Glutamate also plays a role in the catabolism of certain amines. Understanding glutamate's role in these various processes can aid in genome mining for novel metabolic pathways or the engineering of pathways for bioremediation or chemical production of valuable compounds.
Collapse
Affiliation(s)
- Mark C Walker
- Department of Chemistry and Howard Hughes Medical Institute, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL, 61801, USA
| | - Wilfred A van der Donk
- Department of Chemistry and Howard Hughes Medical Institute, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, IL, 61801, USA.
| |
Collapse
|
195
|
Burda P, Kuster A, Hjalmarson O, Suormala T, Bürer C, Lutz S, Roussey G, Christa L, Asin-Cayuela J, Kollberg G, Andersson BA, Watkins D, Rosenblatt DS, Fowler B, Holme E, Froese DS, Baumgartner MR. Characterization and review of MTHFD1 deficiency: four new patients, cellular delineation and response to folic and folinic acid treatment. J Inherit Metab Dis 2015; 38:863-72. [PMID: 25633902 DOI: 10.1007/s10545-015-9810-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/15/2014] [Accepted: 01/06/2015] [Indexed: 01/15/2023]
Abstract
In the folate cycle MTHFD1, encoded by MTHFD1, is a trifunctional enzyme containing 5,10-methylenetetrahydrofolate dehydrogenase, 5,10-methenyltetrahydrofolate cyclohydrolase and 10-formyltetrahydrofolate synthetase activity. To date, only one patient with MTHFD1 deficiency, presenting with hyperhomocysteinemia, megaloblastic anaemia, hemolytic uremic syndrome (HUS) and severe combined immunodeficiency, has been identified (Watkins et al J Med Genet 48:590-2, 2011). We now describe four additional patients from two different families. The second patient presented with hyperhomocysteinemia, megaloblastic anaemia, HUS, microangiopathy and retinopathy; all except the retinopathy resolved after treatment with hydroxocobalamin, betaine and folinic acid. The third patient developed megaloblastic anaemia, infection, autoimmune disease and moderate liver fibrosis but not hyperhomocysteinemia, and was successfully treated with a regime that included and was eventually reduced to folic acid. The other two, elder siblings of the third patient, died at 9 weeks of age with megaloblastic anaemia, infection and severe acidosis and had MTFHD1 deficiency diagnosed retrospectively. We identified a missense mutation (c.806C > T, p.Thr296Ile) and a splice site mutation (c.1674G > A) leading to exon skipping in the second patient, while the other three harboured a missense mutation (c.146C > T, p.Ser49Phe) and a premature stop mutation (c.673G > T, p.Glu225*), all of which were novel. Patient fibroblast studies revealed severely reduced methionine formation from [(14)C]-formate, which did not increase in cobalamin supplemented culture medium but was responsive to folic and folinic acid. These additional cases increase the clinical spectrum of this intriguing defect, provide in vitro evidence of disturbed methionine synthesis and substantiate the effectiveness of folic or folinic acid treatment.
Collapse
Affiliation(s)
- P Burda
- Division of Metabolism and Children's Research Center, University Children's Hospital, Steinwiesstrasse 75, 8032, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Altered folate metabolism modifies cell proliferation and progesterone secretion in human placental choriocarcinoma JEG-3 cells. Br J Nutr 2015; 114:844-52. [PMID: 26299783 DOI: 10.1017/s0007114515002688] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Folate is an essential B vitamin required for de novo purine and thymidylate synthesis, and for the remethylation of homocysteine to form methionine. Folate deficiency has been associated with placenta-related pregnancy complications, as have SNP in genes of the folate-dependent enzymes, methionine synthase (MTR) and methylenetetrahydrofolate dehydrogenase 1 (MTHFD1). We aimed to determine the effect of altered folate metabolism on placental cell proliferation, viability and invasive capacity and on progesterone and human chorionic gonadotropin (hCG) secretion. Human placental choriocarcinoma (JEG-3) cells cultured in low folic acid (FA) (2 nM) demonstrated 13% (P<0.001) and 26% (P<0.001) lower proliferation, 5.5% (P=0.025) and 7.5% (P=0.004) lower invasion capacity, and 5 to 7.5% (P=0.004-0.025) lower viability compared with control (20 nM) or supplemented (100 nM) cells, respectively. FA concentration had no effect on progesterone or hCG secretion. Small interfering RNA (siRNA) knockdown of MTR gene and protein expression resulted in 17.7% (P<0.0001) lower proliferation and 61% (P=0.014) higher progesterone secretion, but had no effect on cell invasion and hCG secretion. siRNA knockdown of MTHFD1 gene expression in the absence of detectable changes in protein expression resulted in 10.3% (P=0.001) lower cell proliferation, but had no effect on cell invasion and progesterone or hCG secretion. Our data indicate that impaired folate metabolism can result in lower trophoblast proliferation, and could alter viability, invasion capacity and progesterone secretion, which may explain in part the observed associations between folate and placenta-related complications.
Collapse
|
197
|
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: 331] [Impact Index Per Article: 33.1] [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
|
198
|
Zinck JW, de Groh M, MacFarlane AJ. Genetic modifiers of folate, vitamin B-12, and homocysteine status in a cross-sectional study of the Canadian population. Am J Clin Nutr 2015; 101:1295-304. [PMID: 25948668 DOI: 10.3945/ajcn.115.107219] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/23/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Genetic variation can cause variable responses to environmental stimuli. A number of single-nucleotide polymorphisms (SNPs) have been associated with B vitamin status or chronic diseases related to vitamin B-12 and folate metabolism. OBJECTIVE Our objective was to identify associations between common SNPs in genes related to folate and vitamin B-12 metabolism or associated with B vitamin-related chronic diseases and biomarkers of nutrient status in a population exposed to folic acid fortification. DESIGN A panel of 116 SNPs was sequenced by using the Sequenom iPLEX Gold platform in a sample of 3114 adults aged 20-79 y from the Canadian Health Measures Survey, cycle 1. Associations between these SNPs and red blood cell (RBC) folate, serum vitamin B-12, and plasma total homocysteine were determined. RESULTS Twenty-one SNPs and 6 haplotype blocks were associated with RBC folate, serum vitamin B-12, and/or plasma homocysteine concentrations. Vitamin status was associated mainly with SNPs in genes directly involved in vitamin absorption/uptake (CUBN, CD320), transport (TCN1, TCN2), or metabolism (BHMT2, CBS, MTHFR, MUT, SHMT1). Other SNPs included those in the DNMT2, DPEP1, FUT2, NOX4, and PON1 genes. CONCLUSIONS We identified novel associations between SNPs in CD320 and DNMT2, which had been previously associated with neural tube defects, and vitamin B-12 status, as well as between SNPs in SHMT1, which had been previously associated with colorectal cancer and cardiovascular disease risk, and RBC folate status. These novel associations provide a plausible metabolic rationale for the association of these SNPs with B vitamin-related diseases. We also observed a novel association between an SNP in CUBN with RBC folate and confirmed the association of a number of SNPs with B vitamin status in this large cross-sectional study.
Collapse
Affiliation(s)
- John Wr Zinck
- From the Science Integration Division, Public Health Agency of Canada, Ottawa, ON, Canada (JWRZ and MdG), and Nutrition Research Division, Health Canada, Ottawa, ON, Canada (JWRZ and AJM)
| | - Margaret de Groh
- From the Science Integration Division, Public Health Agency of Canada, Ottawa, ON, Canada (JWRZ and MdG), and Nutrition Research Division, Health Canada, Ottawa, ON, Canada (JWRZ and AJM)
| | - Amanda J MacFarlane
- From the Science Integration Division, Public Health Agency of Canada, Ottawa, ON, Canada (JWRZ and MdG), and Nutrition Research Division, Health Canada, Ottawa, ON, Canada (JWRZ and AJM).
| |
Collapse
|
199
|
Washburn SE, Caudill MA, Malysheva O, MacFarlane AJ, Behan NA, Harnett B, MacMillan L, Pongnopparat T, Brosnan JT, Brosnan ME. Formate metabolism in fetal and neonatal sheep. Am J Physiol Endocrinol Metab 2015; 308:E921-7. [PMID: 25805190 PMCID: PMC4436996 DOI: 10.1152/ajpendo.00046.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/17/2015] [Indexed: 01/23/2023]
Abstract
By virtue of its role in nucleotide synthesis, as well as the provision of methyl groups for vital methylation reactions, one-carbon metabolism plays a crucial role in growth and development. Formate, a critical albeit neglected component of one-carbon metabolism, occurs extracellularly and may provide insights into cellular events. We examined formate metabolism in chronically cannulated fetal sheep (gestation days 119-121, equivalent to mid-third trimester in humans) and in their mothers as well as in normal full-term lambs. Plasma formate levels were much higher in fetal lamb plasma and in amniotic fluid (191 ± 62 and 296 ± 154 μM, respectively) than in maternal plasma (33 ± 13 μM). Measurements of folate, vitamin B12, and homocysteine showed that these high formate levels could not be due to vitamin deficiencies. Elevated formate levels were also found in newborn lambs and persisted to about 8 wk of age. Formate was also found in sheep milk. Potential precursors of one-carbon groups were also measured in fetal and maternal plasma and in amniotic fluid. There were very high concentrations of serine in the fetus (∼1.6 mM in plasma and 3.5 mM in the amniotic fluid) compared with maternal plasma (0.19 mM), suggesting increased production of formate; however, we cannot rule out decreased formate utilization. Dimethylglycine, a choline metabolite, was also 30 times higher in the fetus than in the mother.
Collapse
Affiliation(s)
- Shannon E Washburn
- Department of Veterinary Physiology and Pharmacology and Michael DeBakey Institute, College of Veterinary Medicine and Biomedical Sciences, Texas A & M University, College Station, Texas
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Olga Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | | | - Nathalie A Behan
- Nutrition Research Division, Health Canada, Ottawa, Ontario, Canada; and
| | - Brian Harnett
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Luke MacMillan
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Theerawat Pongnopparat
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - John T Brosnan
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Margaret E Brosnan
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
200
|
Yin K. Positive correlation between expression level of mitochondrial serine hydroxymethyltransferase and breast cancer grade. Onco Targets Ther 2015; 8:1069-74. [PMID: 25999742 PMCID: PMC4437609 DOI: 10.2147/ott.s82433] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Metabolic reprogramming plays an essential role in supporting the survival and proliferation of cancer cells. Serine hydroxymethyltransferase (SHMT) directs serine to the metabolism of one-carbon unit and the synthesis of thymidilate as a key factor in this metabolic shift. Although the mitochondrial isoform of SHMT (SHMT2) has been proven to be a crucial factor in the serine/glycine metabolism in several cancer cell types, the expression pattern of SHMT2 and the correlation of expression level of SHMT2 and other clinicopathological parameters in clinical breast cancer remain to be explored. In this research, 76 breast cancer patients who underwent modified radical mastectomy were enrolled for immunohistochemical analysis of the expression level of SHMT2 in their cancerous breast tissues for comparison with that in matching, distant noncancerous tissues. The results showed that SHMT2 was not expressed in the distant noncancerous cells. In contrast, SHMT2 protein could be stained in all breast cancer samples at varying degrees. Higher level of SHMT2 was expressed in grade III breast cancer cells than that those in grade I–II (P<0.05). In conclusion, SHMT2 was highly expressed in breast cancer cells, and the expression level of SHMT2 was positively correlated with breast cancer grade, suggesting that SHMT2 could be a target for anticancer therapies.
Collapse
Affiliation(s)
- Ke Yin
- Department of Thyroid and Breast Surgery, Ningbo First Hospital, Ningbo, People's Republic of China
| |
Collapse
|