1
|
Dong J, Shelp GV, Poole EM, Cook WJJ, Michaud J, Cho CE. Prenatal choline supplementation enhances metabolic outcomes with differential impact on DNA methylation in Wistar rat offspring and dams. J Nutr Biochem 2024:109806. [PMID: 39547266 DOI: 10.1016/j.jnutbio.2024.109806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/21/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Choline is an essential nutrient required for proper functioning of organs and serves as a methyl donor. In liver where choline metabolism primarily occurs, glucose homeostasis is regulated through insulin receptor substrates (IRS) 1 and 2. Here, we determined the role of prenatal choline as a modulator of metabolic health and DNA methylation in liver of offspring and dams. Pregnant Wistar rat dams were fed an AIN-93G diet and received drinking water either with supplemented 0.25% choline (w/w) as choline bitartrate or untreated control. All offspring were weaned to a high-fat diet for 12 weeks. Prenatal choline supplementation led to higher insulin sensitivity in female offspring at weaning as well as lower body weight and food intake and higher insulin sensitivity in female and male adult offspring compared to offspring from untreated dams. Higher hepatic betaine concentrations were observed in dams and female offspring of choline-supplemented dams at weaning and higher glycerophosphocholine in female and male offspring at post-weaning compared to the untreated control, suggestive of sustaining different choline pathways. Hepatic gene expression of Irs2 was higher in dams at weaning and female offspring at weaning and post-weaning, whereas Irs1 was lower in male offspring at post-weaning. Gene-specific DNA methylation of Irs2 was lower in female offspring at post-weaning and Irs1 methylation was higher in male offspring at post-weaning that exhibited an inverse relationship between methylation and gene expression. In conclusion, prenatal choline supplementation contributes to improved parameters of insulin signaling but these effects varied across time and offspring sex.
Collapse
Affiliation(s)
- Jianzhang Dong
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Gia V Shelp
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Elizabeth M Poole
- Department of Family Relations and Applied Nutrition, University of Guelph, Guelph, Ontario, Canada
| | - William J J Cook
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Jana Michaud
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Clara E Cho
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
2
|
Shelp GV, Dong J, Orlov NO, Malysheva OV, Bender E, Shoveller AK, Bakovic M, Cho CE. Exposure to prenatal excess or imbalanced micronutrients leads to long-term perturbations in one-carbon metabolism, trimethylamine-N-oxide and DNA methylation in Wistar rat offspring. FASEB J 2024; 38:e70032. [PMID: 39212230 DOI: 10.1096/fj.202401018rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/05/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Prenatal multivitamins, including folic acid, are commonly consumed in excess, whereas choline, an essential nutrient and an important source of labile methyl groups, is underconsumed. Here, we characterized profiles of one-carbon metabolism and related pathways and patterns of DNA methylation in offspring exposed to excess or imbalanced micronutrients prenatally. Pregnant Wistar rats were fed either recommended 1× vitamins (RV), high 10× vitamins (HV), high 10× folic acid with recommended choline (HFolRC), or high 10× folic acid with no choline (HFolNC). Offspring were weaned to a high-fat diet for 12 weeks. Circulating metabolites were analyzed with a focus on the hypothalamus, an area known to be under epigenetic regulation. HV, HFolRC, and HFolNC males had higher body weight (BW) and lower plasma choline and methionine consistent with lower hypothalamic S-adenosylmethionine (SAM):S-adenosylhomocysteine (SAH) and global DNA methylation compared with RV. HV and HFolNC females had higher BW and lower plasma 5-methyltetrahydrofolate and methionine consistent with lower hypothalamic global DNA methylation compared with RV. Plasma dimethylglycine (DMG) and methionine were higher as with hypothalamic SAM:SAH and global DNA methylation in HFolRC females without changes in BW compared with RV. Plasma trimethylamine and trimethylamine-N-oxide were higher in males but lower in females from HFolRC compared with RV. Network modeling revealed a link between the folate-dependent pathway and SAH, with most connections through DMG. Final BW was negatively correlated with choline, DMG, and global DNA methylation. In conclusion, prenatal intake of excess or imbalanced micronutrients induces distinct metabolic and epigenetic perturbations in offspring that reflect long-term nutritional programming of health.
Collapse
Affiliation(s)
- Gia V Shelp
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Jianzhang Dong
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Nikolai O Orlov
- Department of Chemistry, University of Guelph, Guelph, Ontario, Canada
| | - Olga V Malysheva
- Division of Nutritional Sciences, Human Metabolic Research Unit, Cornell University, Ithaca, New York, USA
| | - Erica Bender
- Division of Nutritional Sciences, Human Metabolic Research Unit, Cornell University, Ithaca, New York, USA
| | - Anna K Shoveller
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario, Canada
| | - Marica Bakovic
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Clara E Cho
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
3
|
Amarsi R, Furse S, Cleaton MAM, Maurel S, Mitchell AL, Ferguson-Smith AC, Cenac N, Williamson C, Koulman A, Charalambous M. A co-ordinated transcriptional programme in the maternal liver supplies long chain polyunsaturated fatty acids to the conceptus using phospholipids. Nat Commun 2024; 15:6767. [PMID: 39117683 PMCID: PMC11310303 DOI: 10.1038/s41467-024-51089-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
The long and very long chain polyunsaturated fatty acids (LC-PUFAs) are preferentially transported by the mother to the fetus. Failure to supply LC-PUFAs is strongly linked with stillbirth, fetal growth restriction, and impaired neurodevelopmental outcomes. However, dietary supplementation during pregnancy is unable to simply reverse these outcomes, suggesting imperfectly understood interactions between dietary fatty acid intake and the molecular mechanisms of maternal supply. Here we employ a comprehensive approach combining untargeted and targeted lipidomics with transcriptional profiling of maternal and fetal tissues in mouse pregnancy. Comparison of wild-type mice with genetic models of impaired lipid metabolism allows us to describe maternal hepatic adaptations required to provide LC-PUFAs to the developing fetus. A late pregnancy-specific, selective activation of the Liver X Receptor signalling pathway dramatically increases maternal supply of LC-PUFAs within circulating phospholipids. Crucially, genetic ablation of this pathway in the mother reduces LC-PUFA accumulation by the fetus, specifically of docosahexaenoic acid (DHA), a critical nutrient for brain development.
Collapse
Affiliation(s)
- Risha Amarsi
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, SE19RT, UK
- Pregnancy Physiology Laboratory, Francis Crick Institute, 1 Midland Road, NW1 1AT, London, UK
| | - Samuel Furse
- Biological chemistry group, Jodrell laboratory, Royal Botanic Gardens Kew, Kew Road, Richmond, Surrey, TW9 3DS, UK
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK
| | - Mary A M Cleaton
- Department of Genetics, Downing Street, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Sarah Maurel
- IRSD, Université de Toulouse-Paul Sabatier, INSERM, INRAe, ENVT, UPS, Toulouse, France
| | - Alice L Mitchell
- Department of Women and Children's Health, King's College London, Guy's Campus, London, UK
| | - Anne C Ferguson-Smith
- Department of Genetics, Downing Street, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Nicolas Cenac
- IRSD, Université de Toulouse-Paul Sabatier, INSERM, INRAe, ENVT, UPS, Toulouse, France
| | - Catherine Williamson
- Department of Women and Children's Health, King's College London, Guy's Campus, London, UK
| | - Albert Koulman
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK
| | - Marika Charalambous
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, SE19RT, UK.
- Pregnancy Physiology Laboratory, Francis Crick Institute, 1 Midland Road, NW1 1AT, London, UK.
| |
Collapse
|
4
|
Su GM, Guo QW, Shen YL, Cai JJ, Chen X, Lin J, Fang DZ. Association between PEMT rs7946 and blood pressure levels in Chinese adolescents. Blood Press Monit 2024; 29:180-187. [PMID: 38502043 DOI: 10.1097/mbp.0000000000000703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
OBJECTIVES This study was to explore blood pressure levels in Chinese adolescents with different genotypes of phosphatidylethanolamine N-methyltransferase (PEMT) gene ( PEMT ) rs7946, as well as effects of dietary intake on blood pressure levels with different genders and different genotypes of PEMT rs7946. METHODS PEMT rs7946 genotypes were identified by PCR-restriction fragment length polymorphism and verified by DNA sequencing. Blood pressure was measured using a standard mercury sphygmomanometer. Dietary intakes were analyzed based on a 3-day diet diary, and dietary components were calculated using computer software. RESULTS A total of 721 high school students (314 males and 407 females) at the age of 16.86 ± 0.59 years were included. The A allele carriers of PEMT rs7946 had increased levels of SBP, DBP, mean arterial pressure (MAP) and pulse pressure (PP) than the GG homozygotes in the female subjects. There were significant interactions between PEMT rs7946 and gender on SBP and MAP levels, regardless of whether an unadjusted or adjusted model was used. When dietary intake was taken into account, fat intake was positively associated with SBP and PP in the male GG homozygotes, while protein intake was positively associated with PP in the female A allele carriers of PEMT rs7946. CONCLUSION This study suggests that PEMT rs7946 is significantly associated with blood pressure levels in human being. There might be interactions among PEMT rs7946, gender, and dietary intake on blood pressure levels in the adolescent population.
Collapse
Affiliation(s)
- Guo Ming Su
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | | | | | | | | | | | | |
Collapse
|
5
|
Asantewaa G, Tuttle ET, Ward NP, Kang YP, Kim Y, Kavanagh ME, Girnius N, Chen Y, Rodriguez K, Hecht F, Zocchi M, Smorodintsev-Schiller L, Scales TQ, Taylor K, Alimohammadi F, Duncan RP, Sechrist ZR, Agostini-Vulaj D, Schafer XL, Chang H, Smith ZR, O'Connor TN, Whelan S, Selfors LM, Crowdis J, Gray GK, Bronson RT, Brenner D, Rufini A, Dirksen RT, Hezel AF, Huber AR, Munger J, Cravatt BF, Vasiliou V, Cole CL, DeNicola GM, Harris IS. Glutathione synthesis in the mouse liver supports lipid abundance through NRF2 repression. Nat Commun 2024; 15:6152. [PMID: 39034312 PMCID: PMC11271484 DOI: 10.1038/s41467-024-50454-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
Cells rely on antioxidants to survive. The most abundant antioxidant is glutathione (GSH). The synthesis of GSH is non-redundantly controlled by the glutamate-cysteine ligase catalytic subunit (GCLC). GSH imbalance is implicated in many diseases, but the requirement for GSH in adult tissues is unclear. To interrogate this, we have developed a series of in vivo models to induce Gclc deletion in adult animals. We find that GSH is essential to lipid abundance in vivo. GSH levels are highest in liver tissue, which is also a hub for lipid production. While the loss of GSH does not cause liver failure, it decreases lipogenic enzyme expression, circulating triglyceride levels, and fat stores. Mechanistically, we find that GSH promotes lipid abundance by repressing NRF2, a transcription factor induced by oxidative stress. These studies identify GSH as a fulcrum in the liver's balance of redox buffering and triglyceride production.
Collapse
Affiliation(s)
- Gloria Asantewaa
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Emily T Tuttle
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Nathan P Ward
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Yun Pyo Kang
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Yumi Kim
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Madeline E Kavanagh
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
- Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Nomeda Girnius
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Katherine Rodriguez
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Fabio Hecht
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Marco Zocchi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Leonid Smorodintsev-Schiller
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - TashJaé Q Scales
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Kira Taylor
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Fatemeh Alimohammadi
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Renae P Duncan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Zachary R Sechrist
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Diana Agostini-Vulaj
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Xenia L Schafer
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA
| | - Hayley Chang
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Zachary R Smith
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas N O'Connor
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Sarah Whelan
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Laura M Selfors
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Jett Crowdis
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - G Kenneth Gray
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Dirk Brenner
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Alessandro Rufini
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Aram F Hezel
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Aaron R Huber
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Joshua Munger
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Benjamin F Cravatt
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Calvin L Cole
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Gina M DeNicola
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Isaac S Harris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
6
|
Kubo Y, Fukuoka H, Shoji K, Mori C, Sakurai K, Nishikawa M, Oshida K, Yamashiro Y, Kawabata T. Longitudinal Analysis of One-Carbon Metabolism-Related Metabolites in Maternal and Cord Blood of Japanese Pregnant Women. Nutrients 2024; 16:1765. [PMID: 38892698 PMCID: PMC11174998 DOI: 10.3390/nu16111765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
One-carbon metabolism (OCM) is a complex and interconnected network that undergoes drastic changes during pregnancy. In this study, we investigated the longitudinal distribution of OCM-related metabolites in maternal and cord blood and explored their relationships. Additionally, we conducted cross-sectional analyses to examine the interrelationships among these metabolites. This study included 146 healthy pregnant women who participated in the Chiba Study of Mother and Child Health. Maternal blood samples were collected during early pregnancy, late pregnancy, and delivery, along with cord blood samples. We analyzed 18 OCM-related metabolites in serum using stable isotope dilution liquid chromatography/tandem mass spectrometry. We found that serum S-adenosylmethionine (SAM) concentrations in maternal blood remained stable throughout pregnancy. Conversely, S-adenosylhomocysteine (SAH) concentrations increased, and the total homocysteine/total cysteine ratio significantly increased with advancing gestational age. The betaine/dimethylglycine ratio was negatively correlated with total homocysteine in maternal blood for all sampling periods, and this correlation strengthened with advances in gestational age. Most OCM-related metabolites measured in this study showed significant positive correlations between maternal blood at delivery and cord blood. These findings suggest that maternal OCM status may impact fetal development and indicate the need for comprehensive and longitudinal evaluations of OCM during pregnancy.
Collapse
Affiliation(s)
- Yoshinori Kubo
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (K.S.); (T.K.)
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu 520-2192, Shiga, Japan
| | - Hideoki Fukuoka
- Department of Perinatal Mesenchymal Stem Cell Research, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Fukushima, Japan;
| | - Kumiko Shoji
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (K.S.); (T.K.)
| | - Chisato Mori
- Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Chiba, Japan;
- Department of Sustainable Health Science, Center for Preventive Medical Sciences, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Chiba, Japan
| | - Kenichi Sakurai
- Department of Nutrition and Metabolic Medicine, Center for Preventive Medical Sciences, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Chiba, Japan;
| | - Masazumi Nishikawa
- Department of Food Management, School of Food, Agricultural and Environmental Sciences, Miyagi University, 2-2-1 Hatadate, Taihaku-ku, Sendai 982-0215, Miyagi, Japan;
| | - Kyoichi Oshida
- Faculty of Beauty & Wellness, Professional University of Beauty & Wellness, 3-9-3 Ushikubo, Tsuzuki-ku, Yokohama 224-0012, Kanagawa, Japan;
| | - Yuichiro Yamashiro
- Probiotics Research Laboratory, Graduate School of Medicine, Juntendo University, 2-9-8-3F, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan;
| | - Terue Kawabata
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (K.S.); (T.K.)
| |
Collapse
|
7
|
Vallianou NG, Kounatidis D, Psallida S, Panagopoulos F, Stratigou T, Geladari E, Karampela I, Tsilingiris D, Dalamaga M. The Interplay Between Dietary Choline and Cardiometabolic Disorders: A Review of Current Evidence. Curr Nutr Rep 2024; 13:152-165. [PMID: 38427291 PMCID: PMC11133147 DOI: 10.1007/s13668-024-00521-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE OF REVIEW Choline is an essential nutrient for human health and cellular homeostasis as it is necessary for the synthesis of lipid cell membranes, lipoproteins, and the synthesis of the neurotransmitter acetylcholine. The aim of this review is to analyze the beneficial effects of choline and its significance in cellular metabolism and various inflammatory pathways, such as the inflammasome. We will discuss the significance of dietary choline in cardiometabolic disorders, such as non-alcoholic fatty liver disease (NAFLD), cardiovascular disease (CVD), and chronic kidney disease (CKD) as well as in cognitive function and associated neuropsychiatric disorders. RECENT FINDINGS Choline deficiency has been related to the development of NAFLD and cognitive disability in the offspring as well as in adulthood. In sharp contrast, excess dietary intake of choline mediated via the increased production of trimethylamine by the gut microbiota and increased trimethylamine-N-oxide (TMAO) levels has been related to atherosclerosis in most studies. In this context, CVD and CKD through the accumulation of TMAO, p-Cresyl-sulfate (pCS), and indoxyl-sulfate (IS) in serum may be the result of the interplay between excess dietary choline, the increased production of TMAO by the gut microbiota, and the resulting activation of inflammatory responses and fibrosis. A balanced diet, with no excess nor any deficiency in dietary choline, is of outmost importance regarding the prevention of cardiometabolic disorders as well as cognitive function. Large-scale studies with the use of next-generation probiotics, especially Akkermansia muciniphila and Faecalibacterium prausnitzii, should further examine their therapeutic potential in this context.
Collapse
Affiliation(s)
- Natalia G Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, Athens, Greece.
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, Athens, Greece.
| | - Dimitris Kounatidis
- Department of Internal Medicine, Hippokration General Hospital, 114 Vassilissis Sofias str, Athens, Greece
| | - Sotiria Psallida
- Department of Microbiology, KAT General Hospital of Attica, 2 Nikis str, Athens, Greece
| | - Fotis Panagopoulos
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, Athens, Greece
| | - Theodora Stratigou
- Department of Endocrinology and Metabolism, Evangelismos General Hospital, 45-47 Ipsilantou str, Athens, Greece
| | - Eleni Geladari
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, Athens, Greece
| | - Irene Karampela
- 2nd Department of Critical Care, Medical School, University of Athens, Attikon General University Hospital, 1 Rimini str, Athens, Greece
| | - Dimitrios Tsilingiris
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, 68100, Alexandroupoli, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, Athens, Greece.
| |
Collapse
|
8
|
Yazdanpanah M, Yazdanpanah N, Gamache I, Ong K, Perry JRB, Manousaki D. Metabolome-wide Mendelian randomization for age at menarche and age at natural menopause. Genome Med 2024; 16:69. [PMID: 38802955 PMCID: PMC11131236 DOI: 10.1186/s13073-024-01322-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/22/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND The role of metabolism in the variation of age at menarche (AAM) and age at natural menopause (ANM) in the female population is not entirely known. We aimed to investigate the causal role of circulating metabolites in AAM and ANM using Mendelian randomization (MR). METHODS We combined MR with genetic colocalization to investigate potential causal associations between 658 metabolites and AAM and between 684 metabolites and ANM. We extracted genetic instruments for our exposures from four genome-wide association studies (GWAS) on circulating metabolites and queried the effects of these variants on the outcomes in two large GWAS from the ReproGen consortium. Additionally, we assessed the mediating role of the body mass index (BMI) in these associations, identified metabolic pathways implicated in AAM and ANM, and sought validation for selected metabolites in the Avon Longitudinal Study of Parents and Children (ALSPAC). RESULTS Our analysis identified 10 candidate metabolites for AAM, but none of them colocalized with AAM. For ANM, 76 metabolites were prioritized (FDR-adjusted MR P-value ≤ 0.05), with 17 colocalizing, primarily in the glycerophosphocholines class, including the omega-3 fatty acid and phosphatidylcholine (PC) categories. Pathway analyses and validation in ALSPAC mothers also highlighted the role of omega and polyunsaturated fatty acids levels in delaying age at menopause. CONCLUSIONS Our study suggests that metabolites from the glycerophosphocholine and fatty acid families play a causal role in the timing of both menarche and menopause. This underscores the significance of specific metabolic pathways in the biology of female reproductive longevity.
Collapse
Affiliation(s)
- Mojgan Yazdanpanah
- Research Center of the Sainte-Justine University Hospital, Université de Montréal, 3175 Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada
| | - Nahid Yazdanpanah
- Research Center of the Sainte-Justine University Hospital, Université de Montréal, 3175 Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada
| | - Isabel Gamache
- Research Center of the Sainte-Justine University Hospital, Université de Montréal, 3175 Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada
| | - Ken Ong
- MRC Epidemiology Unit, School of Clinical Medicine, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - John R B Perry
- MRC Epidemiology Unit, School of Clinical Medicine, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
- Metabolic Research Laboratory, School of Clinical Medicine, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Despoina Manousaki
- Research Center of the Sainte-Justine University Hospital, Université de Montréal, 3175 Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada.
- Departments of Pediatrics, Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Canada.
| |
Collapse
|
9
|
Sfakianoudis K, Zikopoulos A, Grigoriadis S, Seretis N, Maziotis E, Anifandis G, Xystra P, Kostoulas C, Giougli U, Pantos K, Simopoulou M, Georgiou I. The Role of One-Carbon Metabolism and Methyl Donors in Medically Assisted Reproduction: A Narrative Review of the Literature. Int J Mol Sci 2024; 25:4977. [PMID: 38732193 PMCID: PMC11084717 DOI: 10.3390/ijms25094977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
One-carbon (1-C) metabolic deficiency impairs homeostasis, driving disease development, including infertility. It is of importance to summarize the current evidence regarding the clinical utility of 1-C metabolism-related biomolecules and methyl donors, namely, folate, betaine, choline, vitamin B12, homocysteine (Hcy), and zinc, as potential biomarkers, dietary supplements, and culture media supplements in the context of medically assisted reproduction (MAR). A narrative review of the literature was conducted in the PubMed/Medline database. Diet, ageing, and the endocrine milieu of individuals affect both 1-C metabolism and fertility status. In vitro fertilization (IVF) techniques, and culture conditions in particular, have a direct impact on 1-C metabolic activity in gametes and embryos. Critical analysis indicated that zinc supplementation in cryopreservation media may be a promising approach to reducing oxidative damage, while female serum homocysteine levels may be employed as a possible biomarker for predicting IVF outcomes. Nonetheless, the level of evidence is low, and future studies are needed to verify these data. One-carbon metabolism-related processes, including redox defense and epigenetic regulation, may be compromised in IVF-derived embryos. The study of 1-C metabolism may lead the way towards improving MAR efficiency and safety and ensuring the lifelong health of MAR infants.
Collapse
Affiliation(s)
- Konstantinos Sfakianoudis
- Centre for Human Reproduction, Genesis Athens Clinic, 14-16, Papanikoli, 15232 Athens, Greece; (K.S.); (K.P.)
| | - Athanasios Zikopoulos
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
- Obstetrics and Gynecology, Royal Cornwall Hospital, Treliske, Truro TR1 3LJ, UK
| | - Sokratis Grigoriadis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (E.M.); (P.X.)
| | - Nikolaos Seretis
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
| | - Evangelos Maziotis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (E.M.); (P.X.)
| | - George Anifandis
- Department of Obstetrics and Gynecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41222 Larisa, Greece;
| | - Paraskevi Xystra
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (E.M.); (P.X.)
| | - Charilaos Kostoulas
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
| | - Urania Giougli
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
| | - Konstantinos Pantos
- Centre for Human Reproduction, Genesis Athens Clinic, 14-16, Papanikoli, 15232 Athens, Greece; (K.S.); (K.P.)
| | - Mara Simopoulou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (E.M.); (P.X.)
| | - Ioannis Georgiou
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
| |
Collapse
|
10
|
Hoffman MC, Hunter SJ, D'Alessandro A, Christians U, Law AJ, Freedman R. Maternal Plasma Choline during Gestation and Small for Gestational Age Infants. Am J Perinatol 2024; 41:e939-e948. [PMID: 36584689 PMCID: PMC11407527 DOI: 10.1055/s-0042-1759775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Small for gestational age (SGA) infants are at increased risk for neonatal morbidity and developmental problems in childhood. No current interventions during human pregnancy address this problem. This study investigated the possible relationship between maternal choline concentration during pregnancy and SGA infants. STUDY DESIGN Maternal plasma choline concentrations were sampled at 16 and 28 weeks' gestation from women in a public prenatal clinic. Additional factors assessed were maternal age, body mass index, infection, C-reactive protein, hair cortisol, and compliance with prenatal vitamins and folate. Infants below the 10th percentile for gestational age were classified as SGA. Binary logistic regression was used to identify significant associated factors in pregnancies resulting in SGA infants compared with pregnancies resulting in non-SGA infants. RESULTS Thirteen (8%) of 159 women had SGA infants. Maternal plasma choline concentrations were low for pregnant participants whose infants were SGA, with the 28-week concentration significantly lower compared with other participants. Plasma choline concentrations ≥7 μM at 28 weeks, consistent with a minimally adequate dietary intake of choline-containing foods, were achieved by only 2 (15%) of mothers with SGA infants, compared with 51% of mothers whose infants were not SGA. Choline concentrations <7 μM at 28 weeks' gestation were associated with an odds ratio for SGA of 16.6 (95% confidence interval: 1.5-189.2, p = 0.023). Other significant factors were female sex and maternal C-reactive protein plasma concentration during gestation. CONCLUSION This observational study suggests that higher maternal choline levels may influence the risk for SGA. Maternal plasma choline concentrations are not routinely available in clinical laboratories. However, plasma choline levels can be increased by the mothers' intake of choline or phosphatidylcholine supplements. No nutritional intervention is currently recommended to prevent SGA, but the evidence from this study suggests that further consideration of the role of maternal choline may be warranted. KEY POINTS · More females are small for gestational age.. · Low maternal choline is related to small infants.. · Maternal choline ≥7 μM at 28 weeks appears optimal..
Collapse
Affiliation(s)
- Maria C Hoffman
- Division of Maternal and Fetal Medicine, Departments of Obstetrics and Gynecology and Psychiatry, University of Colorado School of Medicine, Aurora, Colorado
| | - Sharon J Hunter
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, Colorado
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Uwe Christians
- Department of Anesthesiology, iC42 Clinical Research and Development, University of Colorado School of Medicine, Aurora, Colorado
| | - Amanda J Law
- Department of Psychiatry, Cell and Developmental Biology, and Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Robert Freedman
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
11
|
Nguyen HT, Oktayani PPI, Lee SD, Huang LC. Choline in pregnant women: a systematic review and meta-analysis. Nutr Rev 2024:nuae026. [PMID: 38607338 DOI: 10.1093/nutrit/nuae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Abstract
CONTEXT Choline is a critical nutrient. Inadequate choline intake during pregnancy increases the risk of adverse maternal and offspring health. OBJECTIVE A systematic review and meta-analysis were conducted to examine the current recommendations for choline intake by pregnant women, estimate the overall prevalence of pregnant women with adequate choline intake, and explore associations between maternal choline level and adverse pregnancy outcomes (APOs). METHODS Choline recommendations for pregnant women were assessed from eight nutrient guidelines of the United States, United Kingdom, Canada, Australia, Asia, International Federation of Gynecology and Obstetrics, and World Health Organization. Data on the prevalence of pregnant women with adequate choline intake and the association between maternal choline level and APOs were collected from 5 databases up to May 2023. Meta-analyses with random effects and subgroup analyses were performed for the pooled estimate of prevalence and association. RESULTS Five recent nutrition guidelines from the United States (United States Department of Agriculture), United States (Food and Drug Administration), Canada, Australia, and the International Federation of Gynecology and Obstetrics have emphasized the importance of adequate choline intake for pregnant women. Of 27 publications, 19 articles explored the prevalence and 8 articles explored the association. Meta-analysis of 12 prevalence studies revealed a concerning 11.24% (95% confidence interval, 6.34-17.26) prevalence of pregnant women with adequate choline intake recommendations. A meta-analysis of 6 studies indicated a significant association between high maternal choline levels and a reduced risk of developing APOs, with an odds ratio of 0.51 (95% confidence interval, 0.40-0.65). CONCLUSION The existing guidelines highlight the importance of choline in supporting maternal health and fetal development during pregnancy. Furthermore, a high maternal choline level was likely to be associated with a lower risk of APOs. However, 88.76% of pregnant women do not achieve the optimal choline intake. Therefore, specific policies and actions may be necessary to improve choline intake in pregnant women's care and support the well-being of pregnant women. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CDR42023410561.
Collapse
Affiliation(s)
- Hoan Thi Nguyen
- College of Health Care Science, China Medical University, Taichung, Taiwan
- Nursing and Medical Technology, University of Medicine and Pharmacy, Ho Chi Minh City, VietNam
| | | | - Shin-Da Lee
- College of Health Care Science, China Medical University, Taichung, Taiwan
- Department of Physical Therapy, China Medical University, Taichung, Taiwan
| | - Li-Chi Huang
- College of Health Care Science, China Medical University, Taichung, Taiwan
- School of Nursing, China Medical University, Taichung, Taiwan
- Department of Nursing, China Medical University Children Hospital, Taichung, Taiwan
| |
Collapse
|
12
|
Yue Q, Huang C, Zhou R, Zhang Y, Wang D, Zhang Z, Chen H. Integrated transcriptomic and metabolomic analyses reveal potential regulatory pathways regulating bone metabolism pre- and postsexual maturity in hens. Poult Sci 2024; 103:103555. [PMID: 38417334 PMCID: PMC10907858 DOI: 10.1016/j.psj.2024.103555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 03/01/2024] Open
Abstract
At the onset of sexual maturity, the increasing circulating estrogen stimulates the formation of medullary bone, which provides available calcium for eggshell formation. The bone loss of laying hens is caused by the continuous dynamic changes of structure bone leading to bone fragility and susceptibility. The degree of medullary bone mineralization in sexual maturity is positively correlated with bone quality in the late laying stage. This study aimed to explore the molecular regulation mechanism of bone metabolism pre- and postsexual maturity in hens based on the joint analysis of transcriptome and metabolome. A total of 50 Hy-line Sonia pullets with comparable body weight at 13 wk were selected. Eight pullets were killed at 15 wk (juvenile hens, JH) and 19 wk (laying hens, LH), and LHs were killed within 3 h after oviposition. Differentially expressed genes and metabolites in tibia were analyzed based on transcriptome and metabolome, and then combined to construct the relevant metabolisms and hub genes. In the LH hens, plasma levels of estrogen and tartrate-resistant acid phosphatase were significantly elevated by 1.7 and 1.3 times. In addition, the midpoint diameter, bone mineral density and bone mineral content of the tibia and femur were higher at 19 wk of age. A total of 580 differentially expressed genes were found between the JH and LH group in the tibia, including 280 up-regulated, and 300 down-regulated genes in the LH group. Gene set enrichment analysis (GSEA) showed that the intracellular biosynthesis and secretion of matrix vesicles were significantly enrichment in the LH hens. A total of 21 differential metabolites were identified between JH and LH group. Estradiol valerate positively correlated with L-theanine, tryptophan betaine, dopamine, and perindopril. Joint analysis showed that the top 20 hub genes were enriched in cholesterol biosynthesis and phospholipid metabolism, which played a key regulatory role in bone metabolism during pre- and postsexual maturity. These results provide a theoretical foundation for maintaining efficient egg production and reducing bone health problems in laying hens.
Collapse
Affiliation(s)
- Qiaoxian Yue
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Chenxuan Huang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Rongyan Zhou
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China.
| | - Yinlang Zhang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Dehe Wang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Zhenhong Zhang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Hui Chen
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| |
Collapse
|
13
|
Sheyn D, Momotaz H, Hijaz A, Zeleznik O, Minassian V, Penney KL. Effect of Dietary Choline Consumption on the Development of Urinary Urgency Incontinence in a Longitudinal Cohort of Women. Int Urogynecol J 2024; 35:667-676. [PMID: 38334759 DOI: 10.1007/s00192-024-05740-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/09/2024] [Indexed: 02/10/2024]
Abstract
INTRODUCTION AND HYPOTHESIS The objective of this study was to determine whether differences in the cumulative dietary intake of choline, is associated with the risk of developing urge urinary incontinence (UUI). METHODS This was an analysis within the Nurses' Health Study (NHS) I and II. The main exposure was the cumulative daily intake for each choline-containing compound obtained from a detailed daily food frequency questionnaire. The primary outcome was UUI, defined as urine loss with a sudden feeling of bladder fullness or when a toilet is inaccessible, occurring >1/month. Cox proportional hazards regression models were used to calculate multivariate-adjusted relative risks and 95% confidence intervals (CIs) for the association between total choline and choline derivatives and risk of UUI. Fixed effects meta-analyses of results from NHSI and NHSII were performed for postmenopausal women only to obtain a pooled estimate of the impact of choline consumption on UUI. RESULTS There were 33,273 participants in NHSI and 38,732 in NHSII who met all the criteria for inclusion in the analysis. The incidence of UUI was 9.41% (n=3,139) in NHSI and 4.25% (n=1,646) in NHSII. After adjusting for confounders choline was not found to be associated with UUI in postmenopausal women. However, in premenopausal women, relative to the lowest quartile, the highest quartile of consumption of total choline (aRR = 0.79, 95% CI: 0.64-0.99), free choline (aRR = 0.74, 95% CI: 0.58-0.94), and phosphocholine (aRR = 0.77, 95% CI: 0.61-0.96) were associated with a reduced risk of UUI. CONCLUSIONS Increased dietary choline consumption was associated with a reduced risk of UUI among premenopausal women.
Collapse
Affiliation(s)
- David Sheyn
- Department of Urology, University Hospitals System, Cleveland, OH, 44104, USA.
- Case Western Reserve University, Cleveland, OH, USA.
| | | | - Adonis Hijaz
- Department of Urology, University Hospitals System, Cleveland, OH, 44104, USA
- Case Western Reserve University, Cleveland, OH, USA
| | - Oana Zeleznik
- Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Vatche Minassian
- Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Kathryn L Penney
- Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Harvard TH Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
14
|
Socha MW, Flis W, Wartęga M. Epigenetic Genome Modifications during Pregnancy: The Impact of Essential Nutritional Supplements on DNA Methylation. Nutrients 2024; 16:678. [PMID: 38474806 PMCID: PMC10934520 DOI: 10.3390/nu16050678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Pregnancy is an extremely stressful period in a pregnant woman's life. Currently, women's awareness of the proper course of pregnancy and its possible complications is constantly growing. Therefore, a significant percentage of women increasingly reach for various dietary supplements during gestation. Some of the most popular substances included in multi-ingredient supplements are folic acid and choline. Those substances are associated with positive effects on fetal intrauterine development and fewer possible pregnancy-associated complications. Recently, more and more attention has been paid to the impacts of specific environmental factors, such as diet, stress, physical activity, etc., on epigenetic modifications, understood as changes occurring in gene expression without the direct alteration of DNA sequences. Substances such as folic acid and choline may participate in epigenetic modifications by acting via a one-carbon cycle, leading to the methyl-group donor formation. Those nutrients may indirectly impact genome phenotype by influencing the process of DNA methylation. This review article presents the current state of knowledge on the use of folic acid and choline supplementation during pregnancy, taking into account their impacts on the maternal-fetal unit and possible pregnancy outcomes, and determining possible mechanisms of action, with particular emphasis on their possible impacts on epigenetic modifications.
Collapse
Affiliation(s)
- Maciej W. Socha
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland;
- Department of Obstetrics and Gynecology, St. Adalbert’s Hospital in Gdańsk, Copernicus Healthcare Entity, Jana Pawła II 50, 80-462 Gdańsk, Poland
| | - Wojciech Flis
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland;
- Department of Obstetrics and Gynecology, St. Adalbert’s Hospital in Gdańsk, Copernicus Healthcare Entity, Jana Pawła II 50, 80-462 Gdańsk, Poland
| | - Mateusz Wartęga
- Department of Pathophysiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland;
| |
Collapse
|
15
|
Hunter SK, Hoffman MC, D’Alessandro A, Freedman R. Developmental Windows for Effects of Choline and Folate on Excitatory and Inhibitory Neurotransmission During Human Gestation. Dev Psychobiol 2024; 66:e22453. [PMID: 38646069 PMCID: PMC11031125 DOI: 10.1002/dev.22453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/10/2023] [Indexed: 04/23/2024]
Abstract
Choline and folate are critical nutrients for fetal brain development, but the timing of their influence during gestation has not been previously characterized. At different periods during gestation, choline stimulation of α7-nicotinic receptors facilitates conversion of γ-aminobutyric acid (GABA) receptors from excitatory to inhibitory and recruitment of GluR1-R2 receptors for faster excitatory responses to glutamate. The outcome of the fetal development of inhibition and excitation was assessed in 159 newborns by P50 cerebral auditory-evoked responses. Paired stimuli, S1, S2, were presented 500 msec apart. Higher P50 amplitude in response to S1 (P50S1microV) assesses excitation, and lower P50S2microV assesses inhibition in this paired-stimulus paradigm. Development of inhibition was related solely to maternal choline plasma concentration and folate supplementation at 16 weeks' gestation. Development of excitation was related only to maternal choline at 28 weeks. Higher maternal choline concentrations later in gestation did not compensate for earlier lower concentrations. At 4 years of age, increased behavior problems on the Child Behavior Checklist 1½-5yrs were related to both newborn inhibition and excitation. Incomplete development of inhibition and excitation associated with lower choline and folate during relatively brief periods of gestation thus has enduring effects on child development.
Collapse
Affiliation(s)
- Sharon K. Hunter
- Department of Psychiatry, University of Colorado School of Medicine
| | - M. Camille Hoffman
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine
| | - Robert Freedman
- Department Pharmacology, University of Colorado School of Medicine
| |
Collapse
|
16
|
Obeid R, Schön C, Derbyshire E, Jiang X, Mellott TJ, Blusztajn JK, Zeisel SH. A Narrative Review on Maternal Choline Intake and Liver Function of the Fetus and the Infant; Implications for Research, Policy, and Practice. Nutrients 2024; 16:260. [PMID: 38257153 PMCID: PMC10820518 DOI: 10.3390/nu16020260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Dietary choline is needed to maintain normal health, including normal liver function in adults. Fatty liver induced by a choline-deficient diet has been consistently observed in human and animal studies. The effect of insufficient choline intake on hepatic fat accumulation is specific and reversible when choline is added to the diet. Choline requirements are higher in women during pregnancy and lactation than in young non-pregnant women. We reviewed the evidence on whether choline derived from the maternal diet is necessary for maintaining normal liver function in the fetus and breastfed infants. Studies have shown that choline from the maternal diet is actively transferred to the placenta, fetal liver, and human milk. This maternal-to-child gradient can cause depletion of maternal choline stores and increase the susceptibility of the mother to fatty liver. Removing choline from the diet of pregnant rats causes fatty liver both in the mother and the fetus. The severity of fatty liver in the offspring was found to correspond to the severity of fatty liver in the respective mothers and to the duration of feeding the choline-deficient diet to the mother. The contribution of maternal choline intake in normal liver function of the offspring can be explained by the role of phosphatidylcholine in lipid transport and as a component of cell membranes and the function of choline as a methyl donor that enables synthesis of phosphatidylcholine in the liver. Additional evidence is needed on the effect of choline intake during pregnancy and lactation on health outcomes in the fetus and infant. Most pregnant and lactating women are currently not achieving the adequate intake level of choline through the diet. Therefore, public health policies are needed to ensure sufficient choline intake through adding choline to maternal multivitamin supplements.
Collapse
Affiliation(s)
- Rima Obeid
- Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, D-66420 Homburg, Germany
| | - Christiane Schön
- BioTeSys GmbH, Nutritional CRO, Schelztorstrasse 54-56, D-73728 Esslingen, Germany
| | | | - Xinyin Jiang
- Department of Health and Nutrition Sciences, Brooklyn College, City University of New York, 4110C Ingersoll Hall, 2900 Bedford Ave., Brooklyn, NY 11210, USA
| | - Tiffany J. Mellott
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Jan Krzysztof Blusztajn
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Steven H. Zeisel
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27514, USA
| |
Collapse
|
17
|
Saadat N, Ciarelli J, Pallas B, Padmanabhan V, Vyas AK. Sex-Specific Perturbation of Systemic Lipidomic Profile in Newborn Lambs Impacted by Prenatal Testosterone Excess. Endocrinology 2023; 165:bqad187. [PMID: 38060679 PMCID: PMC10750263 DOI: 10.1210/endocr/bqad187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Indexed: 12/27/2023]
Abstract
Gestational hyperandrogenism adversely impacts offspring health. Using an ovine model, we found that prenatal testosterone (T) excess adversely affects growth and cardiometabolic outcomes in female offspring and produces sex-specific effects on fetal myocardium. Since lipids are essential to cardiometabolic function, we hypothesized that prenatal T excess leads to sex-specific disruptions in lipid metabolism at birth. Shotgun lipidomics was performed on the plasma samples collected 48 hours after birth from female (F) and male (M) lambs of control (C) and (T) sheep (CF = 4, TF = 7, CM = 5, TM = 10) and data were analyzed by univariate analysis, multivariate dimensionality reduction modeling followed by functional enrichment, and pathway analyses. Biosynthesis of phosphatidylserine was the major pathway responsible for sex differences in controls. Unsupervised and supervised models showed separation between C and T in both sexes with glycerophospholipids and glycerolipids classes being responsible for the sex differences between C and T. T excess increased cholesterol in females while decreasing phosphatidylcholine levels in male lambs. Specifically, T excess: 1) suppressed the phosphatidylethanolamine N-methyltransferase (PEMT) phosphatidylcholine synthesis pathway overall and in TM lambs as opposed to suppression of carnitine levels overall and TF lambs; and 2) activated biosynthesis of ether-linked (O-)phosphatidylethanolamine and O-phosphatidylcholine from O-diacylglycerol overall and in TF lambs. Higher cholesterol levels could underlie adverse cardiometabolic outcomes in TF lambs, whereas suppressed PEMT pathway in TM lambs could lead to endoplasmic reticulum stress and defective lipid transport. These novel findings point to sex-specific effects of prenatal T excess on lipid metabolism in newborn lambs, a precocial ovine model of translational relevance.
Collapse
Affiliation(s)
- Nadia Saadat
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Joseph Ciarelli
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Brooke Pallas
- Unit Lab Animal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Arpita Kalla Vyas
- Department of Pediatrics, Washington University St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
18
|
Obeid R, Karlsson T. Choline - a scoping review for Nordic Nutrition Recommendations 2023. Food Nutr Res 2023; 67:10359. [PMID: 38187796 PMCID: PMC10770654 DOI: 10.29219/fnr.v67.10359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/15/2022] [Accepted: 11/10/2023] [Indexed: 01/09/2024] Open
Abstract
Choline is an essential nutrient with metabolic roles as a methyl donor in one carbon metabolism and as a precursor for membrane phospholipids and the neurotransmitter acetylcholine. Choline content is particularly high in liver, eggs, and wheat germ, although it is present in a variety of foods. The main dietary sources of choline in the Nordic and Baltic countries are meat, dairy, eggs, and grain. A diet that is devoid of choline causes liver and muscle dysfunction within 3 weeks. Choline requirements are higher during pregnancy and lactation than in non-pregnant women. Although no randomized controlled trials are available, observational studies in human, supported by coherence from interventional studies with neurodevelopmental outcomes and experimental studies in animals, strongly suggest that sufficient intake of choline during pregnancy is necessary for normal brain development and function in the child. Observational studies suggested that adequate intake of choline could have positive effects on cognitive function in older people. However, prospective data are lacking, and no intervention studies are available in the elderly.
Collapse
Affiliation(s)
- Rima Obeid
- Department of Clinical Chemistry and Laboratory Medicine, University Hospital of the Saarland, Homburg, Germany
| | - Therese Karlsson
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
19
|
Smiriglia A, Lorito N, Serra M, Perra A, Morandi A, Kowalik MA. Sex difference in liver diseases: How preclinical models help to dissect the sex-related mechanisms sustaining NAFLD and hepatocellular carcinoma. iScience 2023; 26:108363. [PMID: 38034347 PMCID: PMC10682354 DOI: 10.1016/j.isci.2023.108363] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023] Open
Abstract
Only a few preclinical findings are confirmed in the clinic, posing a critical issue for clinical development. Therefore, identifying the best preclinical models can help to dissect molecular and mechanistic insights into liver disease pathogenesis while being clinically relevant. In this context, the sex relevance of most preclinical models has been only partially considered. This is particularly significant in NAFLD and HCC, which have a higher prevalence in men when compared to pre-menopause women but not to those in post-menopausal status, suggesting a role for sex hormones in the pathogenesis of the diseases. This review gathers the sex-relevant findings and the available preclinical models focusing on both in vitro and in vivo studies and discusses the potential implications and perspectives of introducing the sex effect in the selection of the best preclinical model. This is a critical aspect that would help to tailor personalized therapies based on sex.
Collapse
Affiliation(s)
- Alfredo Smiriglia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Nicla Lorito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Marina Serra
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Andrea Perra
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Marta Anna Kowalik
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| |
Collapse
|
20
|
Zelows MM, Cady C, Dharanipragada N, Mead AE, Kipp ZA, Bates EA, Varadharajan V, Banerjee R, Park SH, Shelman NR, Clarke HA, Hawkinson TR, Medina T, Sun RC, Lydic TA, Hinds TD, Brown JM, Softic S, Graf GA, Helsley RN. Loss of carnitine palmitoyltransferase 1a reduces docosahexaenoic acid-containing phospholipids and drives sexually dimorphic liver disease in mice. Mol Metab 2023; 78:101815. [PMID: 37797918 PMCID: PMC10568566 DOI: 10.1016/j.molmet.2023.101815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND AND AIMS Genome and epigenome wide association studies identified variants in carnitine palmitoyltransferase 1a (CPT1a) that associate with lipid traits. The goal of this study was to determine the role of liver-specific CPT1a on hepatic lipid metabolism. APPROACH AND RESULTS Male and female liver-specific knockout (LKO) and littermate controls were placed on a low-fat or high-fat diet (60% kcal fat) for 15 weeks. Mice were necropsied after a 16 h fast, and tissues were collected for lipidomics, matrix-assisted laser desorption ionization mass spectrometry imaging, kinome analysis, RNA-sequencing, and protein expression by immunoblotting. Female LKO mice had increased serum alanine aminotransferase levels which were associated with greater deposition of hepatic lipids, while male mice were not affected by CPT1a deletion relative to male control mice. Mice with CPT1a deletion had reductions in DHA-containing phospholipids at the expense of monounsaturated fatty acids (MUFA)-containing phospholipids in whole liver and at the level of the lipid droplet (LD). Male and female LKO mice increased RNA levels of genes involved in LD lipolysis (Plin2, Cidec, G0S2) and in polyunsaturated fatty acid metabolism (Elovl5, Fads1, Elovl2), while only female LKO mice increased genes involved in inflammation (Ly6d, Mmp12, Cxcl2). Kinase profiling showed decreased protein kinase A activity, which coincided with increased PLIN2, PLIN5, and G0S2 protein levels and decreased triglyceride hydrolysis in LKO mice. CONCLUSIONS Liver-specific deletion of CPT1a promotes sexually dimorphic steatotic liver disease (SLD) in mice, and here we have identified new mechanisms by which females are protected from HFD-induced liver injury.
Collapse
Affiliation(s)
- Mikala M Zelows
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, USA
| | - Corissa Cady
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Nikitha Dharanipragada
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Anna E Mead
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Zachary A Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Evelyn A Bates
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA
| | | | - Rakhee Banerjee
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Se-Hyung Park
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA; Department of Pediatrics and Gastroenterology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Nathan R Shelman
- Department of Pathology and Laboratory Medicine, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Harrison A Clarke
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida College of Medicine, Gainesville, FL, USA
| | - Tara R Hawkinson
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida College of Medicine, Gainesville, FL, USA
| | - Terrymar Medina
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ramon C Sun
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida College of Medicine, Gainesville, FL, USA
| | - Todd A Lydic
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA; Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, KY, USA; Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - J Mark Brown
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Samir Softic
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA; Department of Pediatrics and Gastroenterology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Gregory A Graf
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA; Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Robert N Helsley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA; Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, KY, USA; Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, USA; Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA; Department of Internal Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Kentucky College of Medicine, Lexington, KY, USA.
| |
Collapse
|
21
|
Minarski M, Maas C, Heinrich C, Böckmann KA, Bernhard W, Shunova A, Poets CF, Franz AR. Choline and Betaine Levels in Plasma Mirror Choline Intake in Very Preterm Infants. Nutrients 2023; 15:4758. [PMID: 38004152 PMCID: PMC10675502 DOI: 10.3390/nu15224758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Choline is essential for cell membrane formation and methyl transfer reactions, impacting parenchymal and neurological development. It is therefore enriched via placental transfer, and fetal plasma concentrations are high. In spite of the greater needs of very low birth weight infants (VLBWI), choline content of breast milk after preterm delivery is lower (median (p25-75): 158 mg/L (61-360 mg/L) compared to term delivery (258 mg/L (142-343 mg/L)). Even preterm formula or fortified breast milk currently provide insufficient choline to achieve physiological plasma concentrations. This secondary analysis of a randomized controlled trial comparing growth of VLBWI with different levels of enteral protein supply aimed to investigate whether increased enteral choline intake results in increased plasma choline, betaine and phosphatidylcholine concentrations. We measured total choline content of breast milk from 33 mothers of 34 VLBWI. Enteral choline intake from administered breast milk, formula and fortifier was related to the respective plasma choline, betaine and phosphatidylcholine concentrations. Plasma choline and betaine levels in VLBWI correlated directly with enteral choline intake, but administered choline was insufficient to achieve physiological (fetus-like) concentrations. Hence, optimizing maternal choline status, and the choline content of milk and fortifiers, is suggested to increase plasma concentrations of choline, ameliorate the choline deficit and improve growth and long-term development of VLBWI.
Collapse
Affiliation(s)
- Michaela Minarski
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Christoph Maas
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Christine Heinrich
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Katrin A. Böckmann
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Wolfgang Bernhard
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Anna Shunova
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Christian F. Poets
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
| | - Axel R. Franz
- Department of Neonatology, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany (W.B.)
- Center for Pediatric Clinical Studies, University Children’s Hospital, Tübingen University Hospital, 72076 Tübingen, Germany
| |
Collapse
|
22
|
Li J, Huang J, Lv Y, Ji H. Association between dietary intakes of B vitamins and nonalcoholic fatty liver disease in postmenopausal women: a cross-sectional study. Front Nutr 2023; 10:1272321. [PMID: 37927496 PMCID: PMC10621796 DOI: 10.3389/fnut.2023.1272321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is increasingly common globally, particularly among postmenopausal women. Diet plays a fundamental role in the treatment of NAFLD. However, clinical research on the dietary intakes of B vitamins, specifically in postmenopausal women, is scant. Hence, it is imperative to study the impact of B vitamin dietary intake in postmenopausal women. Methods This study utilized National Health and Nutrition Examination Survey (NHANES) data for 668 postmenopausal women. Logistic regression analysis was conducted to investigate the association of the intakes of B vitamins with hepatic steatosis and liver fibrosis prevalence. The analysis accounted for various covariates and employed restricted cubic spline analysis to examine potential nonlinear relationships. Additionally, interactions among age, diabetes, and B-vitamin intakes, as well as the interaction between folate and vitamin B12 intake, were explored. Results Higher intakes of folate [0.30 (0.10-0.88)], choline [0.26 (0.07-0.95)], vitamin B1, and vitamin B2 were associated with a reduced risk of hepatic steatosis in postmenopausal women. The associations of niacin (P-nonlinear = 0.0003), vitamin B1 (P-nonlinear = 0.036), and vitamin B2 (P-nonlinear<0.0001) intakes with hepatic steatosis showed a nonlinear pattern. However, no significant associations were observed between the intakes of niacin, vitamin B6 and vitamin B12 and hepatic steatosis. Furthermore, there were no significant associations between B-vitamin intakes and liver fibrosis. No interaction effects were observed. Conclusion Dietary intakes of folate, choline, vitamin B1, and vitamin B2 may be associated with liver steatosis in postmenopausal women, these results suggest that optimizing the intake of these specific B vitamins may have a protective effect against liver steatosis in postmenopausal women, offering valuable insights into potential dietary strategies to promote their well-being.
Collapse
Affiliation(s)
- Jiajie Li
- Department of Hepatobiliary and Pancreatic Medicine, Infectious Disease. and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jingda Huang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yanqing Lv
- Department of Hepatobiliary and Pancreatic Medicine, Infectious Disease. and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Huifan Ji
- Department of Hepatobiliary and Pancreatic Medicine, Infectious Disease. and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
23
|
Smith TJS, Navas-Acien A, Baker S, Kok C, Kruczynski K, Avolio LN, Pisanic N, Randad PR, Fry RC, Goessler W, van Geen A, Buckley JP, Rahman MH, Ali H, Haque R, Shaikh S, Siddiqua TJ, Schulze K, West KP, Labrique AB, Heaney CD. Anthropometric measures and arsenic methylation among pregnant women in rural northern Bangladesh. ENVIRONMENTAL RESEARCH 2023; 234:116453. [PMID: 37343752 PMCID: PMC10518461 DOI: 10.1016/j.envres.2023.116453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/02/2023] [Accepted: 06/17/2023] [Indexed: 06/23/2023]
Abstract
INTRODUCTION Arsenic methylation converts inorganic arsenic (iAs) to monomethyl (MMA) and dimethyl (DMA) arsenic compounds. Body mass index (BMI) has been positively associated with arsenic methylation efficiency (higher DMA%) in adults, but evidence in pregnancy is inconsistent. We estimated associations between anthropometric measures and arsenic methylation among pregnant women in rural northern Bangladesh. METHODS We enrolled pregnant women (n = 784) (median [IQR] gestational week: 14 [13, 15]) in Gaibandha District, Bangladesh from 2018 to 2019. Anthropometric measures were BMI, subscapular and triceps skinfold thicknesses, and mid-upper arm circumference (MUAC), fat area (MUAFA), and muscle area (MUAMA). Arsenic methylation measures were urinary iAs, MMA, and DMA divided by their sum and multiplied by 100 (iAs%, MMA%, and DMA%), primary methylation index (MMA/iAs; PMI), and secondary methylation index (DMA/MMA; SMI). In complete cases (n = 765 [97.6%]), we fitted linear, beta, and Dirichlet regression models to estimate cross-sectional differences in iAs%, MMA%, DMA%, PMI, and SMI per IQR-unit difference in each anthropometric measure, adjusting for drinking water arsenic, age, gestational age, education, living standards index, and plasma folate, vitamin B12, and homocysteine. RESULTS Median (IQR) BMI, subscapular skinfold thickness, triceps skinfold thickness, MUAC, MUAFA, and MUAMA were 21.5 (19.4, 23.8) kg/m2, 17.9 (13.2, 24.2) mm, 14.2 (10.2, 18.7) mm, 25.9 (23.8, 28.0) cm, 15.3 (10.5, 20.3) cm2, and 29.9 (25.6, 34.2) cm2, respectively. Median (IQR) iAs%, MMA%, DMA%, PMI, and SMI were 12.0 (9.3, 15.2)%, 6.6 (5.3, 8.3)%, 81.0 (77.1, 84.6)%, 0.6 (0.4, 0.7), and 12.2 (9.3, 15.7), respectively. In both unadjusted and adjusted linear models, all anthropometric measures were negatively associated with iAs%, MMA%, and PMI and positively associated with DMA% and SMI. For example, fully adjusted mean differences (95% CI) in DMA% per IQR-unit difference in BMI, subscapular skinfolds thickness, triceps skinfold thickness, MUAC, MUAFA, and MUAMA were 1.72 (1.16, 2.28), 1.58 (0.95, 2.21), 1.74 (1.11, 2.37), 1.45 (0.85, 2.06), 1.70 (1.08, 2.31), and 0.70 (0.13, 1.27) pp, respectively. CONCLUSIONS Anthropometric measures were positively associated with arsenic methylation efficiency among pregnant women in the early second trimester.
Collapse
Affiliation(s)
- Tyler J S Smith
- Department of Environmental Health & Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Sarah Baker
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Caryn Kok
- Department of Environmental Health & Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kate Kruczynski
- Department of Environmental Health & Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Lindsay N Avolio
- Department of Environmental Health & Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nora Pisanic
- Department of Environmental Health & Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Pranay R Randad
- Department of Environmental Health & Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Rebecca C Fry
- Department of Environmental Sciences & Engineering, University of North Carolina at Chapel Hill Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Walter Goessler
- Institute of Chemistry - Analytical Chemistry, University of Graz, Graz, Austria
| | - Alexander van Geen
- Lamont-Doherty Earth Observatory, Columbia University, Palisades, NY, USA
| | - Jessie P Buckley
- Department of Environmental Health & Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Md Hafizur Rahman
- JiVitA Maternal and Child Health and Nutrition Research Project, Rangpur, Bangladesh
| | - Hasmot Ali
- JiVitA Maternal and Child Health and Nutrition Research Project, Rangpur, Bangladesh
| | - Rezwanul Haque
- JiVitA Maternal and Child Health and Nutrition Research Project, Rangpur, Bangladesh
| | - Saijuddin Shaikh
- JiVitA Maternal and Child Health and Nutrition Research Project, Rangpur, Bangladesh
| | - Towfida J Siddiqua
- JiVitA Maternal and Child Health and Nutrition Research Project, Rangpur, Bangladesh
| | - Kerry Schulze
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Keith P West
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Alain B Labrique
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christopher D Heaney
- Department of Environmental Health & Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
24
|
Zelows MM, Cady C, Dharanipragada N, Mead AE, Kipp ZA, Bates EA, Varadharajan V, Banerjee R, Park SH, Shelman NR, Clarke HA, Hawkinson TR, Medina T, Sun RC, Lydic TA, Hinds TD, Brown JM, Softic S, Graf GA, Helsley RN. Loss of Carnitine Palmitoyltransferase 1a Reduces Docosahexaenoic Acid-Containing Phospholipids and Drives Sexually Dimorphic Liver Disease in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.17.553705. [PMID: 37645721 PMCID: PMC10462091 DOI: 10.1101/2023.08.17.553705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Background and Aims Genome and epigenome wide association studies identified variants in carnitine palmitoyltransferase 1a (CPT1a) that associate with lipid traits. The goal of this study was to determine the impact by which liver-specific CPT1a deletion impacts hepatic lipid metabolism. Approach and Results Six-to-eight-week old male and female liver-specific knockout (LKO) and littermate controls were placed on a low-fat or high-fat diet (HFD; 60% kcal fat) for 15 weeks. Mice were necropsied after a 16 hour fast, and tissues were collected for lipidomics, matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI), kinome analysis, RNA-sequencing, and protein expression by immunoblotting. Female LKO mice had increased serum alanine aminotransferase (ALT) levels which were associated with greater deposition of hepatic lipids, while male mice were not affected by CPT1a deletion relative to male control mice. Mice with CPT1a deletion had reductions in DHA-containing phospholipids at the expense of monounsaturated fatty acids (MUFA)-containing phospholipids in both whole liver and at the level of the lipid droplet (LD). Male and female LKO mice increased RNA levels of genes involved in LD lipolysis ( Plin2 , Cidec , G0S2 ) and in polyunsaturated fatty acid (PUFA) metabolism ( Elovl5, Fads1, Elovl2 ), while only female LKO mice increased genes involved in inflammation ( Ly6d, Mmp12, Cxcl2 ). Kinase profiling showed decreased protein kinase A (PKA) activity, which coincided with increased PLIN2, PLIN5, and G0S2 protein levels and decreased triglyceride hydrolysis in LKO mice. Conclusions Liver-specific deletion of CPT1a promotes sexually dimorphic steatotic liver disease (SLD) in mice, and here we have identified new mechanisms by which females are protected from HFD-induced liver injury. Graphical Summary
Collapse
|
25
|
Weston WC, Hales KH, Hales DB. Flaxseed Reduces Cancer Risk by Altering Bioenergetic Pathways in Liver: Connecting SAM Biosynthesis to Cellular Energy. Metabolites 2023; 13:945. [PMID: 37623888 PMCID: PMC10456508 DOI: 10.3390/metabo13080945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
This article illustrates how dietary flaxseed can be used to reduce cancer risk, specifically by attenuating obesity, type 2 diabetes, and non-alcoholic fatty liver disease (NAFLD). We utilize a targeted metabolomics dataset in combination with a reanalysis of past work to investigate the "metabo-bioenergetic" adaptations that occur in White Leghorn laying hens while consuming dietary flaxseed. Recently, we revealed how the anti-vitamin B6 effects of flaxseed augment one-carbon metabolism in a manner that accelerates S-adenosylmethionine (SAM) biosynthesis. Researchers recently showed that accelerated SAM biosynthesis activates the cell's master energy sensor, AMP-activated protein kinase (AMPK). Our paper provides evidence that flaxseed upregulates mitochondrial fatty acid oxidation and glycolysis in liver, concomitant with the attenuation of lipogenesis and polyamine biosynthesis. Defatted flaxseed likely functions as a metformin homologue by upregulating hepatic glucose uptake and pyruvate flux through the pyruvate dehydrogenase complex (PDC) in laying hens. In contrast, whole flaxseed appears to attenuate liver steatosis and body mass by modifying mitochondrial fatty acid oxidation and lipogenesis. Several acylcarnitine moieties indicate Randle cycle adaptations that protect mitochondria from metabolic overload when hens consume flaxseed. We also discuss a paradoxical finding whereby flaxseed induces the highest glycated hemoglobin percentage (HbA1c%) ever recorded in birds, and we suspect that hyperglycemia is not the cause. In conclusion, flaxseed modifies bioenergetic pathways to attenuate the risk of obesity, type 2 diabetes, and NAFLD, possibly downstream of SAM biosynthesis. These findings, if reproducible in humans, can be used to lower cancer risk within the general population.
Collapse
Affiliation(s)
- William C. Weston
- Department of Molecular, Cellular & Systemic Physiology, School of Medicine, Southern Illinois University, Carbondale, IL 62901, USA;
| | - Karen H. Hales
- Department of Obstetrics & Gynecology, School of Medicine, Southern Illinois University, Carbondale, IL 62901, USA;
| | - Dale B. Hales
- Department of Molecular, Cellular & Systemic Physiology, School of Medicine, Southern Illinois University, Carbondale, IL 62901, USA;
- Department of Obstetrics & Gynecology, School of Medicine, Southern Illinois University, Carbondale, IL 62901, USA;
| |
Collapse
|
26
|
Wu CH, Chang TY, Chen YC, Huang RFS. PEMT rs7946 Polymorphism and Sex Modify the Effect of Adequate Dietary Choline Intake on the Risk of Hepatic Steatosis in Older Patients with Metabolic Disorders. Nutrients 2023; 15:3211. [PMID: 37513629 PMCID: PMC10383596 DOI: 10.3390/nu15143211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/10/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
In humans, PEMT rs7946 polymorphism exerts sex-specific effects on choline requirement and hepatic steatosis (HS) risk. Few studies have explored the interaction effect of the PEMT rs7946 polymorphism and sex on the effect of adequate choline intake on HS risk. In this cross-sectional study, we investigated the association between PEMT polymorphism and adequate choline intake on HS risk. We enrolled 250 older patients with metabolic disorders with (n = 152) or without (n = 98; control) ultrasonically diagnosed HS. An elevated PEMT rs7946 A allele level was associated with a lower HS risk and body mass index in both men and women. Dietary choline intake-assessed using a semiquantitative food frequency questionnaire-was associated with reduced obesity in men only (p for trend < 0.05). ROC curve analysis revealed that the cutoff value of energy-adjusted choline intake for HS diagnosis was 448 mg/day in women (AUC: 0.62; 95% CI: 0.57-0.77) and 424 mg/day in men (AUC: 0.63, 95% CI: 0.57-0.76). In women, GG genotype and high choline intake (>448 mg/day) were associated with a 79% reduction in HS risk (adjusted OR: 0.21; 95% CI: 0.05-0.82); notably, GA or AA genotype was associated with a reduced HS risk regardless of choline intake (p < 0.05). In men, GG genotype and high choline intake (>424 mg/day) were associated with a 3.7-fold increase in HS risk (OR: 3.7; 95% CI: 1.19-11.9). Further adjustments for a high-density lipoprotein level and body mass index mitigated the effect of choline intake on HS risk. Current dietary choline intake may be inadequate for minimizing HS risk in postmenopausal Taiwanese women carrying the PEMT rs7946 GG genotype. Older men consuming more than the recommended amount of choline may have an increased risk of nonalcoholic fatty liver disease; this risk is mediated by a high-density lipoprotein level and obesity.
Collapse
Affiliation(s)
- Chien-Hsien Wu
- Ph.D. Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
- Department of Gastroenterology and Hepatology, Taipei Hospital, Ministry of Health and Welfare, New Taipei City 242033, Taiwan
| | - Ting-Yu Chang
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| | - Yen-Chu Chen
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| | - Rwei-Fen S Huang
- Ph.D. Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| |
Collapse
|
27
|
DiStefano JK. The Role of Choline, Soy Isoflavones, and Probiotics as Adjuvant Treatments in the Prevention and Management of NAFLD in Postmenopausal Women. Nutrients 2023; 15:2670. [PMID: 37375574 DOI: 10.3390/nu15122670] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a prevalent condition among postmenopausal women that can lead to severe liver dysfunction and increased mortality. In recent years, research has focused on identifying potential lifestyle dietary interventions that may prevent or treat NAFLD in this population. Due to the complex and multifactorial nature of NAFLD in postmenopausal women, the disease can present as different subtypes, with varying levels of clinical presentation and variable treatment responses. By recognizing the significant heterogeneity of NAFLD in postmenopausal women, it may be possible to identify specific subsets of individuals who may benefit from targeted nutritional interventions. The purpose of this review was to examine the current evidence supporting the role of three specific nutritional factors-choline, soy isoflavones, and probiotics-as potential nutritional adjuvants in the prevention and treatment of NAFLD in postmenopausal women. There is promising evidence supporting the potential benefits of these nutritional factors for NAFLD prevention and treatment, particularly in postmenopausal women, and further research is warranted to confirm their effectiveness in alleviating hepatic steatosis in this population.
Collapse
Affiliation(s)
- Johanna K DiStefano
- Diabetes and Metabolic Disease Research Unit, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| |
Collapse
|
28
|
Li J, Xin Y, Li J, Chen H, Li H. Phosphatidylethanolamine N-methyltransferase: from Functions to Diseases. Aging Dis 2023; 14:879-891. [PMID: 37191416 PMCID: PMC10187709 DOI: 10.14336/ad.2022.1025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022] Open
Abstract
Locating on endoplasmic reticulum and mitochondria associated membrane, Phosphatidylethanolamine N-methyltransferase (PEMT), catalyzes phosphatidylethanolamine methylation to phosphatidylcholine. As the only endogenous pathway for choline biosynthesis in mammals, the dysregulation of PEMT can lead to imbalance of phospholipid metabolism. Dysregulation of phospholipid metabolism in the liver or heart can lead to deposition of toxic lipid species that adversely result in dysfunction of hepatocyte/cardiomyocyte. Studies have shown that PEMT-/- mice increased susceptibility of diet-induced fatty liver and steatohepatitis. However, knockout of PEMT protects against diet-induced atherosclerosis, diet-induced obesity, and insulin resistance. Thus, novel insights to the function of PEMT in various organs should be summarized. Here, we reviewed the structural and functional properties of PEMT, highlighting its role in the pathogenesis of obesity, liver diseases, cardiovascular diseases, and other conditions.
Collapse
Affiliation(s)
- Jiayu Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Yanguo Xin
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Jingye Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Hui Chen
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Hongwei Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Beijing, China.
| |
Collapse
|
29
|
Pregnancy homocysteine and cobalamin status predict childhood metabolic health in the offspring. Pediatr Res 2023; 93:633-642. [PMID: 35641553 DOI: 10.1038/s41390-022-02117-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/08/2022] [Accepted: 05/08/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Inadequate pregnancy cobalamin status has been associated with adverse offspring metabolic health in Indian and Nepalese studies. Studies of pregnancy cobalamin status and mid-childhood health outside of Asia are scarce. METHODS Associations between pregnancy fasting plasma total homocysteine (tHcy), cobalamin status (plasma cobalamin, holotranscobalamin (holoTC), methylmalonic acid (MMA)) and mid-childhood metabolic score (MetSco) ((including fat mass index (zFMI), homeostatic model assessment of insulin resistance (zHOMA-IR) and dyslipidemia (zTG - zHDLc)/2) z-scores)) were investigated in a prospective study of 293 mother-child dyads. RESULTS Highest versus low-mid pregnancy tHcy tertile was associated with higher mid-childhood MetSco, specifically with higher child zFMI. Stratifying by sex, the maternal tHcy-child MetSco association was limited to boys and confirmed for zFMI and zHOMA-IR. The maternal tHcy-child zFMI association was not mediated by birth weight z-score. First trimester plasma cobalamin was not associated with child outcomes, but other indicators of cobalamin status were. Lowest versus mid-high plasma holoTC tertile was associated with MetSco (specifically zFMI and zHOMA-IR) and highest versus low-mid plasma MMA tertile with higher MetSco and dyslipidemia in boys. CONCLUSIONS Moderately elevated pregnancy tHcy and low cobalamin status were associated with mid-childhood metabolic score in boys. The pregnancy tHcy-child zFMI association was not mediated by birth weight. IMPACT Fasting plasma total homocysteine (tHcy) during pregnancy and low cobalamin status during early pregnancy are associated with mid-childhood metabolic score and its components in the offspring. These findings were only significant in male offspring. The study provides new evidence that impaired one carbon metabolism during pregnancy is associated with negative health outcomes in the offspring, in a population with low prevalence of cobalamin deficiency. The maternal-offspring associations were observed in the functional markers of cobalamin status (holotranscobalamin and methylmalonic acid) and tHcy, not with plasma cobalamin concentration. Screening for low pregnancy cobalamin status should be considered.
Collapse
|
30
|
Vasconcellos C, Ferreira O, Lopes MF, Ribeiro AF, Vasques J, Guerreiro CS. Nutritional Genomics in Nonalcoholic Fatty Liver Disease. Biomedicines 2023; 11:biomedicines11020319. [PMID: 36830856 PMCID: PMC9953045 DOI: 10.3390/biomedicines11020319] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common chronic condition associated with genetic and environmental factors in which fat abnormally accumulates in the liver. NAFLD is epidemiologically associated with obesity, type 2 diabetes, and dyslipidemia. Environmental factors, such as physical inactivity and an unbalanced diet, interact with genetic factors, such as epigenetic mechanisms and polymorphisms for the genesis and development of the condition. Different genetic polymorphisms seem to be involved in this context, including variants in PNPLA3, TM6SF2, PEMT, and CHDH genes, playing a role in the disease's susceptibility, development, and severity. From carbohydrate intake and weight loss to omega-3 supplementation and caloric restriction, different dietary and nutritional factors appear to be involved in controlling the onset and progression of NAFLD conditions influencing metabolism, gene, and protein expression. The polygenic risk score represents a sum of trait-associated alleles carried by an individual and seems to be associated with NAFLD outcomes depending on the dietary context. Understanding the exact extent to which lifestyle interventions and genetic predispositions can play a role in the prevention and management of NAFLD can be crucial for the establishment of a personalized and integrative approach to patients.
Collapse
Affiliation(s)
- Carolina Vasconcellos
- Laboratório de Nutrição, Faculdade de Medicina, Centro Académico de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Oureana Ferreira
- Laboratório de Nutrição, Faculdade de Medicina, Centro Académico de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Marta Filipa Lopes
- Laboratório de Nutrição, Faculdade de Medicina, Centro Académico de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - André Filipe Ribeiro
- Laboratório de Nutrição, Faculdade de Medicina, Centro Académico de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - João Vasques
- Laboratório de Nutrição, Faculdade de Medicina, Centro Académico de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Catarina Sousa Guerreiro
- Laboratório de Nutrição, Faculdade de Medicina, Centro Académico de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Correspondence:
| |
Collapse
|
31
|
Zhao M, Ma L, Honda T, Kato A, Ohshiro T, Yokoyama S, Yamamoto K, Ito T, Imai N, Ishizu Y, Nakamura M, Kawashima H, Tsuji NM, Ishigami M, Fujishiro M. Astaxanthin Attenuates Nonalcoholic Steatohepatitis with Downregulation of Osteoprotegerin in Ovariectomized Mice Fed Choline-Deficient High-Fat Diet. Dig Dis Sci 2023; 68:155-163. [PMID: 35397697 DOI: 10.1007/s10620-022-07489-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/14/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Postmenopausal estrogen decline increases the risk of developing nonalcoholic steatohepatitis (NASH), and it might accelerate progression to cirrhosis and hepatocellular carcinoma. AIMS This study aimed to investigate a novel therapy for postmenopausal women who are diagnosed with NASH. METHODS Seven-week-old female C57BL/6 J mice were divided into three experimental groups as follows: (1) sham operation (SHAM group), (2) ovariectomy (OVX group), and (3) ovariectomy + 0.02% astaxanthin (OVX + ASTX group). These three groups of mice were fed a choline-deficient high-fat (CDHF) diet for 8 weeks. Blood serum and liver tissues were collected to examine liver injury, histological changes, and hepatic genes associated with NASH. An in vitro study was performed with the hepatic stellate cell line LX-2. RESULTS The administration of ASTX significantly improved pathological NASH with suppressed steatosis, inflammation, and fibrosis, in comparison with those in the OVX-induced estrogen deficiency group. As a result, liver injury was also attenuated with reduced levels of alanine aminotransferase and aspartate transaminase. In addition, our study found that ASTX supplementation decreased hepatic osteoprotegerin (OPG) in vivo, a possible factor that contributes to NASH development. In vitro, this study further confirmed that ASTX has an inhibitory effect on the secretion of OPG in LX-2 human hepatic stellate cells. CONCLUSIONS Our findings suggest that ASTX alleviates CDHF-OVX-induced pathohistological NASH with downregulated OPG, possibly via suppression of the transforming growth factor beta pathway. ASTX could has promise for use in postmenopausal women diagnosed with NASH.
Collapse
Affiliation(s)
- Meng Zhao
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Lingyun Ma
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Takashi Honda
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Asuka Kato
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan.,ITOCHU Collaborative Research-Molecular Targeted Cancer Treatment for Next Generation, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Taichi Ohshiro
- ITOCHU Collaborative Research-Molecular Targeted Cancer Treatment for Next Generation, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Shinya Yokoyama
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kenta Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Norihiro Imai
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yoji Ishizu
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Masanao Nakamura
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Noriko M Tsuji
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan.,Division of Immune Homeostasis, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan.,Department of Food Science, Jumonji University, Saitama, Japan
| | - Masatoshi Ishigami
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550, Japan.,Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
32
|
Obeid R, Derbyshire E, Schön C. Association between Maternal Choline, Fetal Brain Development, and Child Neurocognition: Systematic Review and Meta-Analysis of Human Studies. Adv Nutr 2022; 13:2445-2457. [PMID: 36041182 PMCID: PMC9776654 DOI: 10.1093/advances/nmac082] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/01/2021] [Accepted: 07/25/2022] [Indexed: 01/29/2023] Open
Abstract
We studied associations between prenatal and early postnatal choline intake, brain development, and neurocognitive function of children. We conducted a systematic review followed by a meta-analysis and critical appraisal of human studies published from 1997 to 2021. Thirty publications were identified. The meta-analysis included 5 of 7 case-control studies studying neural tube defects (NTDs) in relation to maternal choline intakes/circulating concentrations. Low maternal choline intake/circulating concentrations were associated with a higher OR for NTDs among 1131 mothers of newborns with NTDs and 4439 control mothers (pooled estimate = 1.36; 95% CI: 1.11, 1.67). The 95% prediction intervals were 0.78, 2.36. Findings and critical evaluation of 10 publications with interventional designs showed that higher maternal choline intakes during the second half of pregnancy and early postnatal period (550 mg up to 1 g/d on top of the diet) or a child intake of 513 to 625 mg/d from supplements were safe and likely to demonstrate favorable effects on several domains of child neurocognition, such as memory, attention, and visuospatial learning versus the comparators. Findings from observational studies (n = 13) partly supported the association between maternal choline intake/serum concentrations and child neurocognition, but there was low confidence in the use of plasma choline concentrations as a choline intake marker. In conclusion, low maternal choline intakes were associated with a higher OR for NTDs. The risk could be up to 2.36-fold in some populations. Despite limitations of available trials and observational studies, higher maternal choline intake was likely to be associated with better child neurocognition/neurodevelopment. The results should be used to guide choline intake recommendations in pregnancy and lactation, especially because most young women are not achieving the reference intake of choline. This meta-analysis is registered at PROSPERO as CRD42021233790.
Collapse
|
33
|
Morita SY, Ikeda Y. Regulation of membrane phospholipid biosynthesis in mammalian cells. Biochem Pharmacol 2022; 206:115296. [DOI: 10.1016/j.bcp.2022.115296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/05/2022] [Accepted: 10/05/2022] [Indexed: 11/02/2022]
|
34
|
Petkevicius K, Palmgren H, Glover MS, Ahnmark A, Andréasson AC, Madeyski-Bengtson K, Kawana H, Allman EL, Kaper D, Uhrbom M, Andersson L, Aasehaug L, Forsström J, Wallin S, Ahlstedt I, Leke R, Karlsson D, González-King H, Löfgren L, Nilsson R, Pellegrini G, Kono N, Aoki J, Hess S, Sienski G, Pilon M, Bohlooly-Y M, Maresca M, Peng XR. TLCD1 and TLCD2 regulate cellular phosphatidylethanolamine composition and promote the progression of non-alcoholic steatohepatitis. Nat Commun 2022; 13:6020. [PMID: 36241646 PMCID: PMC9568529 DOI: 10.1038/s41467-022-33735-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 09/29/2022] [Indexed: 11/29/2022] Open
Abstract
The fatty acid composition of phosphatidylethanolamine (PE) determines cellular metabolism, oxidative stress, and inflammation. However, our understanding of how cells regulate PE composition is limited. Here, we identify a genetic locus on mouse chromosome 11, containing two poorly characterized genes Tlcd1 and Tlcd2, that strongly influences PE composition. We generated Tlcd1/2 double-knockout (DKO) mice and found that they have reduced levels of hepatic monounsaturated fatty acid (MUFA)-containing PE species. Mechanistically, TLCD1/2 proteins act cell intrinsically to promote the incorporation of MUFAs into PEs. Furthermore, TLCD1/2 interact with the mitochondria in an evolutionarily conserved manner and regulate mitochondrial PE composition. Lastly, we demonstrate the biological relevance of our findings in dietary models of metabolic disease, where Tlcd1/2 DKO mice display attenuated development of non-alcoholic steatohepatitis compared to controls. Overall, we identify TLCD1/2 proteins as key regulators of cellular PE composition, with our findings having broad implications in understanding and treating disease. The regulation of cellular phosphatidylethanolamine (PE) acyl chain composition is poorly understood. Here, the authors show that TLCD1 and TLCD2 proteins mediate the formation of monounsaturated fatty acid-containing PE species and promote the progression of non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Kasparas Petkevicius
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden. .,Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Henrik Palmgren
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Matthew S Glover
- Dynamic Omics, Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Andrea Ahnmark
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anne-Christine Andréasson
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Hiroki Kawana
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Advanced Research & Development Programs for Medical Innovation (AMED-LEAP), Tokyo, Japan
| | - Erik L Allman
- Dynamic Omics, Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Delaney Kaper
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Martin Uhrbom
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Liselotte Andersson
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Leif Aasehaug
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Johan Forsström
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Simonetta Wallin
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ingela Ahlstedt
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Renata Leke
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Daniel Karlsson
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Hernán González-King
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lars Löfgren
- Translational Science and Experimental Medicine, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ralf Nilsson
- Translational Science and Experimental Medicine, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Giovanni Pellegrini
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Nozomu Kono
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Advanced Research & Development Programs for Medical Innovation (AMED-LEAP), Tokyo, Japan
| | - Sonja Hess
- Dynamic Omics, Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Grzegorz Sienski
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Marc Pilon
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | | | - Marcello Maresca
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Xiao-Rong Peng
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
35
|
Genetic Variants in One-Carbon Metabolism and Their Effects on DHA Biomarkers in Pregnant Women: A Post-Hoc Analysis. Nutrients 2022; 14:nu14183801. [PMID: 36145177 PMCID: PMC9506554 DOI: 10.3390/nu14183801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
The delivery of docosahexanoic acid (DHA) to the fetus is dependent on maternal one-carbon metabolism, as the latter supports the hepatic synthesis and export of a DHA-enriched phosphatidylcholine molecule via the phosphatidylethanolamine N-methyltransferase (PEMT) pathway. The following is a post-hoc analysis of a choline intervention study that sought to investigate whether common variants in one-carbon metabolizing genes associate with maternal and/or fetal blood biomarkers of DHA status. Pregnant women entering their second trimester were randomized to consume, until delivery, either 25 (n = 15) or 550 (n = 15) mg choline/d, and the effects of genetic variants in the PEMT, BHMT, MTHFD1, and MTHFR genes on DHA status were examined. Variant (vs. non-variant) maternal PEMT rs4646343 genotypes tended to have lower maternal RBC DHA (% total fatty acids) throughout gestation (6.9% vs. 7.4%; main effect, p = 0.08) and lower cord RBC DHA at delivery (7.6% vs. 8.4%; main effect, p = 0.09). Conversely, variant (vs. non-variant) maternal MTHFD1 rs2235226 genotypes exhibited higher cord RBC DHA (8.3% vs. 7.3%; main effect, p = 0.0003) and higher cord plasma DHA (55 vs. 41 μg/mL; main effect, p = 0.05). Genotype tended to interact with maternal choline intake (p < 0.1) to influence newborn DHA status for PEMT rs4646343 and PEMT rs7946. These data support the need to consider variants in one-carbon metabolic genes in studies assessing DHA status and requirements during pregnancy.
Collapse
|
36
|
Piras IS, Raju A, Don J, Schork NJ, Gerhard GS, DiStefano JK. Hepatic PEMT Expression Decreases with Increasing NAFLD Severity. Int J Mol Sci 2022; 23:ijms23169296. [PMID: 36012560 PMCID: PMC9409182 DOI: 10.3390/ijms23169296] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Choline deficiency causes hepatic fat accumulation, and is associated with a higher risk of nonalcoholic fatty liver disease (NAFLD) and more advanced NAFLD-related hepatic fibrosis. Reduced expression of hepatic phosphatidylethanolamine N-methyltransferase (PEMT), which catalyzes the production of phosphatidylcholine, causes steatosis, inflammation, and fibrosis in mice. In humans, common PEMT variants impair phosphatidylcholine synthesis, and are associated with NAFLD risk. We investigated hepatic PEMT expression in a large cohort of patients representing the spectrum of NAFLD, and examined the relationship between PEMT genetic variants and gene expression. Hepatic PEMT expression was reduced in NAFLD patients with inflammation and fibrosis (i.e., nonalcoholic steatohepatitis or NASH) compared to participants with normal liver histology (β = −1.497; p = 0.005). PEMT levels also declined with increasing severity of fibrosis with cirrhosis < incomplete cirrhosis < bridging fibrosis (β = −1.185; p = 0.011). Hepatic PEMT expression was reduced in postmenopausal women with NASH compared to those with normal liver histology (β = −3.698; p = 0.030). We detected a suggestive association between rs7946 and hepatic fibrosis (p = 0.083). Although none of the tested variants were associated with hepatic PEMT expression, computational fine mapping analysis indicated that rs4646385 may impact PEMT levels in the liver. Hepatic PEMT expression decreases with increasing severity of NAFLD in obese individuals and postmenopausal women, and may contribute to disease pathogenesis in a subset of NASH patients.
Collapse
Affiliation(s)
- Ignazio S. Piras
- Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Anish Raju
- Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Janith Don
- Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | | | - Glenn S. Gerhard
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19122, USA
| | - Johanna K. DiStefano
- Translational Genomics Research Institute, Phoenix, AZ 85004, USA
- Correspondence:
| |
Collapse
|
37
|
Han YH, Choi H, Kim HJ, Lee MO. Chemotactic cytokines secreted from Kupffer cells contribute to the sex-dependent susceptibility to non-alcoholic fatty liver diseases in mice. Life Sci 2022; 306:120846. [PMID: 35914587 DOI: 10.1016/j.lfs.2022.120846] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/15/2022]
Abstract
AIMS The global prevalence of non-alcoholic fatty liver disease (NAFLD) has rapidly increased over the last decade due to an elevated occurrence of metabolic syndromes. Importantly, the prevalence and severity of NAFLD is higher in men than in women. Therefore, in the present study we endeavored to identify the mechanistic disparity between male and female mice. MAIN METHODS Global gene transcriptomics analysis was done with the high-fat diet (HFD)-induced NAFLD model of male, female, and ovariectomized (OVX) female mice. The expression of CCL2, CXCL2, and CXCL10 in mRNA level and serum protein level was done by qPCR and ELISA each. Immunohistochemistry staining was used to observe hepatic immune cell infiltration. To analyzing portion of immune cells, flow cytometry was done with isolated liver cells from HFD-fed male and female mice. Primary mouse liver cells were isolated from male and female mice for in vitro studies. KEY FINDINGS We identified sex differences in inflammatory chemokines, CCL2, CXCL2, and CXCL10, with the expression of these chemokines enhanced in male and OVX, but not in female, mice after HFD feeding. Resident Kupffer cells (KCs) were identified as the major source of production of CCL2, CXCL2, and CXCL10 in the mouse NAFLD model. Notably, KCs obtained from male mice expressed higher levels of chemokines than those from female mice, indicating that KCs may mediate the sex discrepancy in NAFLD progression. SIGNIFICANCE Our findings offer new insights into the pathology of sex-specific differences in NAFLD, involving chemokines and KCs.
Collapse
Affiliation(s)
- Yong-Hyun Han
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; College of Pharmacy, Kangwon National University, Chuncheon, Republic of Korea.
| | - Haena Choi
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea.
| | - Hyeon-Ji Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea; Bio-MAX institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Saigo Y, Sasase T, Uno K, Shinozaki Y, Maekawa T, Sano R, Toriniwa Y, Miyajima K, Ohta T. Establishment of a new nonalcoholic steatohepatitis model; Ovariectomy exacerbates nonalcoholic steatohepatitis-like pathology in diabetic rats. J Pharmacol Toxicol Methods 2022; 116:107190. [DOI: 10.1016/j.vascn.2022.107190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022]
|
39
|
Carrieri L, Osella AR, Ciccacci F, Giannelli G, Scavo MP. Premenopausal Syndrome and NAFLD: A New Approach Based on Gender Medicine. Biomedicines 2022; 10:1184. [PMID: 35625920 PMCID: PMC9138606 DOI: 10.3390/biomedicines10051184] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifactorial condition that affects 25% of the world's population. There is a clear difference in both geographical distribution and sex in childbearing age. These differences are reduced when women become older and senescence begins. The factors that affect the likelihood of developing NAFLD in a premenopausal woman are an imbalance of sex hormones (especially in estradiol and androgen), microbiome dysregulation, insulin resistance, early menarche, the length of time that the woman breastfeeds for and polycystic ovarian syndrome (PCOS). The aim of this review is to identify various physical ailments that may not appear to be serious to young women but that then affect the onset of NAFLD in perimenopause and can degenerate into NASH. These conditions should also be considered in future clinical management, as well as in research opportunities, in order to customize the monitoring and treatment of NAFLD, considering gender medicine for those women who had early metabolic symptoms that were not considered to be significant at the time.
Collapse
Affiliation(s)
- Livianna Carrieri
- Personalized Medicine Laboratory, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Via Turi 27, 70013 Castellana Grotte, Italy;
| | - Alberto Ruben Osella
- Laboratory of Epidemiology and Biostatistics, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Via Turi 27, 70013 Castellana Grotte, Italy;
| | - Fausto Ciccacci
- UniCamillus Saint Camillus International, University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy;
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Via Turi 27, 70013 Castellana Grotte, Italy;
| | - Maria Principia Scavo
- Personalized Medicine Laboratory, National Institute of Gastroenterology IRCCS “S. de Bellis” Research Hospital, Via Turi 27, 70013 Castellana Grotte, Italy;
| |
Collapse
|
40
|
Klatt KC, McDougall MQ, Malysheva OV, Taesuwan S, Loinard-González A(AP, Nevins JEH, Beckman K, Bhawal R, Anderson E, Zhang S, Bender E, Jackson KH, King DJ, Dyer RA, Devapatla S, Vidavalur R, Brenna JT, Caudill MA. Prenatal choline supplementation improves biomarkers of maternal docosahexaenoic acid (DHA) status among pregnant participants consuming supplemental DHA: a randomized controlled trial. Am J Clin Nutr 2022; 116:820-832. [PMID: 35575618 PMCID: PMC9437984 DOI: 10.1093/ajcn/nqac147] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/01/2022] [Accepted: 06/10/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Dietary methyl donors (e.g., choline) support the activity of the phosphatidylethanolamine N-methyltransferase (PEMT) pathway, which generates phosphatidylcholine (PC) molecules enriched in DHA that are exported from the liver and made available to extrahepatic tissues. OBJECTIVES This study investigated the effect of prenatal choline supplementation on biomarkers of DHA status among pregnant participants consuming supplemental DHA. METHODS Pregnant participants (n = 30) were randomly assigned to receive supplemental choline intakes of 550 mg/d [500 mg/d d0-choline + 50 mg/d deuterium-labeled choline (d9-choline); intervention] or 25 mg/d (25 mg/d d9-choline; control) from gestational week (GW) 12-16 until delivery. All participants received a daily 200-mg DHA supplement and consumed self-selected diets. Fasting blood samples were obtained at baseline, GW 20-24, and GW 28-32; maternal/cord blood was obtained at delivery. Mixed-effects linear models were used to assess the impact of prenatal choline supplementation on maternal and newborn DHA status. RESULTS Choline supplementation (550 vs. 25 mg/d) did not achieve a statistically significant intervention × time interaction for RBC PC-DHA (P = 0.11); a significant interaction was observed for plasma PC-DHA and RBC total DHA, with choline supplementation yielding higher levels (+32-38% and +8-11%, respectively) at GW 28-32 (P < 0.05) and delivery (P < 0.005). A main effect of choline supplementation on plasma total DHA was also observed (P = 0.018); its interaction with time was not significant (P = 0.068). Compared with controls, the intervention group exhibited higher (P = 0.007; main effect) plasma enrichment of d3-PC (d3-PC/total PC). Moreover, the ratio of d3-PC to d9-PC was higher (+50-67%; P < 0.001) in the choline intervention arm (vs. control) at GW 20-24, GW 28-32, and delivery. CONCLUSIONS Prenatal choline supplementation improves hepatic DHA export and biomarkers of DHA status by bolstering methyl group supply for PEMT activity among pregnant participants consuming supplemental DHA. This trial is registered at www.clinicaltrials.gov as NCT03194659.
Collapse
Affiliation(s)
| | | | - Olga V Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Siraphat Taesuwan
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA,Faculty of Agro-Industry, Chiang Mai University, Chiang Mai, Thailand
| | | | - Julie E H Nevins
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Kara Beckman
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Ruchika Bhawal
- Proteomics and Metabolomics Facility, Cornell University, Ithaca, NY, USA
| | - Elizabeth Anderson
- Proteomics and Metabolomics Facility, Cornell University, Ithaca, NY, USA
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Cornell University, Ithaca, NY, USA
| | - Erica Bender
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | | | - D Janette King
- The Analytical Core for Metabolomics and Nutrition, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Roger A Dyer
- The Analytical Core for Metabolomics and Nutrition, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - J Thomas Brenna
- Department of Pediatrics, University of Texas, Austin, TX, USA
| | | |
Collapse
|
41
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) can develop in lean individuals. Despite a better metabolic profile, the risk of disease progression to hepatic inflammation, fibrosis, and decompensated cirrhosis in the lean is similar to that in obesity-related NAFLD and lean individuals may experience more severe hepatic consequences and higher mortality relative to those with a higher body mass index (BMI). In the absence of early symptoms and abnormal laboratory findings, lean individuals are not likely to be screened for NAFLD or related comorbidities; however, given the progressive nature of the disease and the increased risk of morbidity and mortality, a clearer understanding of the natural history of NAFLD in lean individuals, as well as efforts to raise awareness of the potential health risks of NAFLD in lean individuals, are warranted. In this review, we summarize available data on NAFLD prevalence, clinical characteristics, outcomes, and mortality in lean individuals and discuss factors that may contribute to the development of NAFLD in this population, including links between dietary and genetic factors, menopausal status, and ethnicity. We also highlight the need for greater representation of lean individuals in NAFLD-related clinical trials, as well as more studies to better characterize lean NAFLD, develop improved screening algorithms, and determine specific treatment strategies based on underlying etiology.
Collapse
Affiliation(s)
- Johanna K. DiStefano
- Diabetes and Fibrotic Disease Research Unit, Translational Genomics Research Institute, Phoenix, USA
| | - Glenn S. Gerhard
- Lewis Katz School of Medicine, Temple University School of Medicine, Philadelphia, PA 19140 USA
| |
Collapse
|
42
|
Verri Hernandes V, Dordevic N, Hantikainen EM, Sigurdsson BB, Smárason SV, Garcia-Larsen V, Gögele M, Caprioli G, Bozzolan I, Pramstaller PP, Rainer J. Age, Sex, Body Mass Index, Diet and Menopause Related Metabolites in a Large Homogeneous Alpine Cohort. Metabolites 2022; 12:metabo12030205. [PMID: 35323648 PMCID: PMC8955763 DOI: 10.3390/metabo12030205] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/19/2022] Open
Abstract
Metabolomics in human serum samples provide a snapshot of the current metabolic state of an individuum. Metabolite concentrations are influenced by both genetic and environmental factors. Concentrations of certain metabolites can further depend on age, sex, menopause, and diet of study participants. A better understanding of these relationships is pivotal for the planning of metabolomics studies involving human subjects and interpretation of their results. We generated one of the largest single-site targeted metabolomics data sets consisting of 175 quantified metabolites in 6872 study participants. We identified metabolites significantly associated with age, sex, body mass index, diet, and menopausal status. While most of our results agree with previous large-scale studies, we also found novel associations including serotonin as a sex and BMI-related metabolite and sarcosine and C2 carnitine showing significantly higher concentrations in post-menopausal women. Finally, we observed strong associations between higher consumption of food items and certain metabolites, mostly phosphatidylcholines and lysophosphatidylcholines. Most, and the strongest, relationships were found for habitual meat intake while no significant relationships were found for most fruits, vegetables, and grain products. Summarizing, our results reconfirm findings from previous population-based studies on an independent cohort. Together, these findings will ultimately enable the consolidation of sets of metabolites which are related to age, sex, BMI, and menopause as well as to participants’ diet.
Collapse
Affiliation(s)
- Vinicius Verri Hernandes
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Nikola Dordevic
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Essi Marjatta Hantikainen
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Baldur Bragi Sigurdsson
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
- Department of Clinical Biochemistry, Landspitali—University Hospital, 108 Reykjavik, Iceland
| | - Sigurður Vidir Smárason
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
- BASF SE, 67063 Ludwigshafen, Germany
| | - Vanessa Garcia-Larsen
- Program in Human Nutrition, Department of International Health, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | - Martin Gögele
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Giulia Caprioli
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Ilaria Bozzolan
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Peter P. Pramstaller
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
| | - Johannes Rainer
- Institute for Biomedicine (Affiliated to the University of Lübeck), Eurac Research, 39100 Bozen, Italy; (V.V.H.); (N.D.); (E.M.H.); (B.B.S.); (S.V.S.); (M.G.); (G.C.); (I.B.); (P.P.P.)
- Correspondence:
| |
Collapse
|
43
|
Lower plasma glutathione, choline, and betaine concentrations are associated with fatty liver in postmenopausal women. Nutr Res 2022; 101:23-30. [DOI: 10.1016/j.nutres.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 11/18/2022]
|
44
|
Lee C, Kim J, Han J, Oh D, Kim M, Jeong H, Kim TJ, Kim SW, Kim JN, Seo YS, Suzuki A, Kim JH, Jung Y. Formyl peptide receptor 2 determines sex-specific differences in the progression of nonalcoholic fatty liver disease and steatohepatitis. Nat Commun 2022; 13:578. [PMID: 35102146 PMCID: PMC8803937 DOI: 10.1038/s41467-022-28138-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/12/2022] [Indexed: 12/21/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an important health concern worldwide and progresses into nonalcoholic steatohepatitis (NASH). Although prevalence and severity of NAFLD/NASH are higher in men than premenopausal women, it remains unclear how sex affects NAFLD/NASH pathophysiology. Formyl peptide receptor 2 (FPR2) modulates inflammatory responses in several organs; however, its role in the liver is unknown. Here we show that FPR2 mediates sex-specific responses to diet-induced NAFLD/NASH. NASH-like liver injury was induced in both sexes during choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) feeding, but compared with females, male mice had more severe hepatic damage. Fpr2 was more highly expressed in hepatocytes and healthy livers from females than males, and FPR2 deletion exacerbated liver damage in CDAHFD-fed female mice. Estradiol induced Fpr2 expression, which protected hepatocytes and the liver from damage. In conclusion, our results demonstrate that FPR2 mediates sex-specific responses to diet-induced NAFLD/NASH, suggesting a novel therapeutic target for NAFLD/NASH.
Collapse
Affiliation(s)
- Chanbin Lee
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Jieun Kim
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Jinsol Han
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Dayoung Oh
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Minju Kim
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Hayeong Jeong
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Tae-Jin Kim
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
- Department of Biological Sciences, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Sang-Woo Kim
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
- Department of Biological Sciences, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Jeong Nam Kim
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
- Department of Microbiology, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Young-Su Seo
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
- Department of Microbiology, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea
| | - Ayako Suzuki
- Division of Gastroenterology and Hepatology, Duke University, Durham, NC, USA
| | - Jae Ho Kim
- Department of Physiology, Pusan National University School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Youngmi Jung
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea.
- Department of Biological Sciences, College of Natural Science, Pusan National University, Pusan, 46241, Republic of Korea.
| |
Collapse
|
45
|
Korsmo HW, Dave B, Trasino S, Saxena A, Liu J, Caviglia JM, Edwards K, Dembitzer M, Sheeraz S, Khaldi S, Jiang X. Maternal Choline Supplementation and High-Fat Feeding Interact to Influence DNA Methylation in Offspring in a Time-Specific Manner. Front Nutr 2022; 9:841787. [PMID: 35165655 PMCID: PMC8837519 DOI: 10.3389/fnut.2022.841787] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/07/2022] [Indexed: 11/18/2022] Open
Abstract
Maternal methyl donor supplementation during pregnancy has demonstrated lasting influence on offspring DNA methylation. However, it is unknown whether an adverse postnatal environment, such as high-fat (HF) feeding, overrides the influence of prenatal methyl donor supplementation on offspring epigenome. In this study, we examined whether maternal supplementation of choline (CS), a methyl donor, interacts with prenatal and postnatal HF feeding to alter global and site-specific DNA methylation in offspring. We fed wild-type C57BL/6J mouse dams a HF diet with or without CS throughout gestation. After weaning, the offspring were exposed to HF feeding for 6 weeks resembling a continued obesogenic environment. Our results suggest that maternal CS under the HF condition (HFCS) increased global DNA methylation and DNA methyltransferase 1 (Dnmt1) expression in both fetal liver and brain. However, during the postnatal period, HFCS offspring demonstrated lower global DNA methylation and Dnmt1 expression was unaltered in both the liver and visceral adipose tissue. Site-specific DNA methylation analysis during both fetal and postnatal periods demonstrated that HFCS offspring had higher methylation of CpGs in the promoter of Srebf1, a key mediator of de novo lipogenesis. In conclusion, the influence of maternal CS on offspring DNA methylation is specific to HF feeding status during prenatal and postnatal periods. Without continued CS during the postnatal period, global DNA methylation enhanced by prenatal CS in the offspring was overridden by postnatal HF feeding.
Collapse
Affiliation(s)
- Hunter W. Korsmo
- Department of Biochemistry, The Graduate Center, City University of New York (CUNY), New York, NY, United States
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Bhoomi Dave
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Steven Trasino
- School of Urban Public Health, Hunter College of the CUNY, New York, NY, United States
| | - Anjana Saxena
- Department of Biochemistry, The Graduate Center, City University of New York (CUNY), New York, NY, United States
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Jia Liu
- Advanced Science Research Center at the Graduate Center of the CUNY, New York, NY, United States
| | - Jorge Matias Caviglia
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Kaydine Edwards
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Moshe Dembitzer
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Shameera Sheeraz
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Sarah Khaldi
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Xinyin Jiang
- Department of Biochemistry, The Graduate Center, City University of New York (CUNY), New York, NY, United States
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| |
Collapse
|
46
|
Fuchs HE, O’Connell K, Du M, Navarro SL, Brasky TM, Kantor ED. Vitamin B 12 Supplementation and Vitamin B 12 Blood Serum Levels: Evaluation of Effect Modification by Gender and Smoking Status. Nutr Cancer 2022; 74:2373-2383. [PMID: 34817305 PMCID: PMC9761981 DOI: 10.1080/01635581.2021.2007271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Research suggests that high intake of supplemental vitamin B12 may be associated with increased risk of cancer, with some evidence that this association may vary by gender and smoking status. This investigation evaluates if similar patterns in association are observed for data for 11,757 adults from the National Health and Nutrition Examination Survey (1999-2006). Survey-weighted multivariable-adjusted linear regression was used to evaluate the association between regular B12 supplement use and log-transformed serum B12 levels. Persons taking vitamin B12 through a multivitamin/multimineral (MVMM) had a median supplemental intake of 12 mcg/day (Q1: 6, Q3: 25), compared to 100 mcg/day (Q1: 22, Q3: 500) for persons reporting supplemental B12 intake through a MVMM-exclusive source. MVMM users had a geometric mean serum B12 26% (95% CI: 23%-30%) higher than nonusers, whereas MVMM-exclusive users' geometric mean was 61% (95% CI: 53%-70%) higher than nonusers (p-trend < 0.001). Although a positive trend (p-trend < 0.001) was observed for both men and women, the association was stronger among women (p-interaction < 0.001). No interaction was observed for smoking status (p-interaction = 0.45). B12 supplementation is associated with higher levels of serum B12, with significant interaction by gender but not smoking. Further work is needed to better understand the interplay of B12 and gender.
Collapse
Affiliation(s)
- Hannah E. Fuchs
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Kelli O’Connell
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mengmeng Du
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sandi L. Navarro
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Theodore M. Brasky
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Elizabeth D. Kantor
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Elizabeth D. Kantor, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 485 Lexington Avenue, 2nd Floor, New York, NY 10017, Phone: 646.888.8247,
| |
Collapse
|
47
|
OUP accepted manuscript. Nutr Rev 2022; 80:1985-2001. [DOI: 10.1093/nutrit/nuac015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
48
|
Kim R, Nijhout HF, Reed MC. One-carbon metabolism during the menstrual cycle and pregnancy. PLoS Comput Biol 2021; 17:e1009708. [PMID: 34914693 PMCID: PMC8741061 DOI: 10.1371/journal.pcbi.1009708] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 01/07/2022] [Accepted: 12/01/2021] [Indexed: 11/18/2022] Open
Abstract
Many enzymes in one-carbon metabolism (OCM) are up- or down-regulated by the sex hormones which vary diurnally and throughout the menstrual cycle. During pregnancy, estradiol and progesterone levels increase tremendously to modulate physiological changes in the reproductive system. In this work, we extend and improve an existing mathematical model of hepatic OCM to understand the dynamic metabolic changes that happen during the menstrual cycle and pregnancy due to estradiol variation. In particular, we add the polyamine drain on S-adenosyl methionine and the direct effects of estradiol on the enzymes cystathionine β-synthase (CBS), thymidylate synthase (TS), and dihydrofolate reductase (DHFR). We show that the homocysteine concentration varies inversely with estradiol concentration, discuss the fluctuations in 14 other one-carbon metabolites and velocities throughout the menstrual cycle, and draw comparisons with the literature. We then use the model to study the effects of vitamin B12, vitamin B6, and folate deficiencies and explain why homocysteine is not a good biomarker for vitamin deficiencies. Additionally, we compute homocysteine throughout pregnancy, and compare the results with experimental data. Our mathematical model explains how numerous homeostatic mechanisms in OCM function and provides new insights into how homocysteine and its deleterious effects are influenced by estradiol. The mathematical model can be used by others for further in silico experiments on changes in one-carbon metabolism during the menstrual cycle and pregnancy.
Collapse
Affiliation(s)
- Ruby Kim
- Department of Mathematics, Duke University, Durham, North Carolina, United States of America
| | - H. Frederik Nijhout
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Michael C. Reed
- Department of Mathematics, Duke University, Durham, North Carolina, United States of America
| |
Collapse
|
49
|
Bortz J, Klatt KC, Wallace TC. Perspective: Estrogen and the Risk of Cognitive Decline: A Missing Choline(rgic) Link? Adv Nutr 2021; 13:S2161-8313(22)00068-0. [PMID: 34849527 PMCID: PMC8970832 DOI: 10.1093/advances/nmab145] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Factors that influence the risk of neurocognitive decline and Alzheimer's disease (AD) may provide insight into therapies for both disease treatment and prevention. While age is the most striking risk factor for AD, it is notable that the prevalence of AD is higher in women, representing two-thirds of cases. To explore potential underlying biological underpinnings of this observation, the intent of this article is to explore the interplay between cognitive aging and sex hormones, the cholinergic system, and novel hypotheses related to the essential nutrient, choline. Mechanistic evidence points toward estrogen's neuroprotective effects being strongly dependent on its interactions with the cholinergic system, a modulator of attentional functioning, learning, and memory. Estrogen has been shown to attenuate anticholinergic-induced impairments in verbal memory and normalize patterns of frontal and occipital cortex activation, resulting in a more "young adult" phenotype. However, similar to estrogen replacement's effect in cardiovascular diseases, its putative protective effects may be restricted to early postmenopausal women only, supportive of the "critical window hypothesis." Estrogen's impact on the cholinergic system may act both locally in the brain but also through peripheral tissues. Estrogen is critical for inducing endogenous choline synthesis via the phosphatidylethanolamine N-methyltransferase (PEMT) pathway of phosphatidylcholine (PC) synthesis. PEMT is dramatically induced in response to estrogen, producing not only a PC molecule and source of choline for the brain but also a key source of the long-chain omega-3 fatty acid, DHA. Herein, we highlight novel hypotheses related to hormone replacement therapy and nutrient metabolism aimed at directing future preclinical and clinical investigation.
Collapse
|
50
|
Glade MJ, Crook MA. Choline deficiency: Is it being recognized? Nutrition 2021; 94:111509. [PMID: 34862116 DOI: 10.1016/j.nut.2021.111509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 11/15/2022]
Affiliation(s)
| | - Martin A Crook
- Department of Clinical Biochemistry and Metabolic Medicine, Guy's & St Thomas' Hospitals, London, UK.
| |
Collapse
|