1
|
King C, Plakke B. Maternal choline supplementation in neurodevelopmental disorders: mechanistic insights from animal models and future directions. Nutr Neurosci 2025; 28:405-424. [PMID: 39046330 DOI: 10.1080/1028415x.2024.2377084] [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] [Indexed: 07/25/2024]
Abstract
OBJECTIVES To synthesize evidence from animal models of neurodevelopmental disorders (NDD) using maternal choline supplementation, to characterize current knowledge on the mechanisms of choline's protective effects against NDD, and to identify gaps in knowledge for future study. METHODS A literature review was conducted in PubMed to identify studies using prenatal choline supplementation interventions in rodent models of neurodevelopmental disorders. 24 studies were identified, and behavioral and biological findings were extracted from each. Studies examining both genetic and environmental risk factors were included. RESULTS Maternal choline supplementation during gestation is protective against both genetic and environmental NDD risk factors. Maternal choline supplementation improves both cognitive and affective outcomes throughout the lifespan in NDD models. Prenatal choline improved these outcomes through its participation in processes like neurogenesis, epigenetic regulation, and anti-inflammatory signaling. DISCUSSION Maternal choline supplementation improves behavioral and neurobiological outcomes in animal models of NDD, paralleling findings in humans. Animal models provide a unique opportunity to study the mechanisms by which gestational choline improves neurodevelopmental outcomes. This is especially important since nearly 90% of pregnant people in the United States are deficient in choline intake. However, much is still unknown about the mechanisms through which choline and its derivatives act. Further research into this topic, especially mechanistic studies in animal models, is critical to modernize maternal choline intake guidelines and to develop interventions to increase maternal choline intake in vulnerable populations.
Collapse
Affiliation(s)
- Cole King
- Psychological Sciences, Kansas State University, Manhattan, KS, USA
| | - Bethany Plakke
- Psychological Sciences, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
2
|
Wang W, Sun Y, Cao R, Luo W, Beng S, Zhang J, Wang X, Peng C. Illustrate the metabolic regulatory effects of Ganoderma Lucidum polysaccharides on cognitive dysfunction in formaldehyde-exposed mouse brain by mass spectrometry imaging. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118060. [PMID: 40120485 DOI: 10.1016/j.ecoenv.2025.118060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
Long-term formaldehyde (FA) exposure causes cognitive dysfunction, often associated with metabolic disorders. While some studies suggest Ganoderma lucidum polysaccharides (GLP) can improve cognitive function, such as Alzheimer's disease. However, the effects of GLP on FA-exposed cognitive dysfunction and the regulation of GLP on brain metabolic disturbances caused by FA remain unclear. In our study, we revealed that GLP significantly reversed FA-exposed spatial cognitive deficits in mice by using Morris Water Maze and Histological analysis. Furthermore, desorption electrospray ionization mass spectrometry imaging (DESI-MSI) found that exposure of FA can caused dysregulated expression of 35 metabolites. Following GLP treatment, there was a significant restoration of the imbalance of choline and acetylcholine, carnitine and acetylcarnitine, and spermidine and spermine, which were all involved in choline metabolism, carnitine metabolism, and polyamine metabolism. Our results suggested that GLP alleviated FA-exposed cognitive dysfunction, likely through modulation of metabolic pathways, providing a potential therapeutic approach for FA-related cognitive dysfunction.
Collapse
Affiliation(s)
- Wen Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei 230012, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Yuanyuan Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Renting Cao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Wenhui Luo
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei 230012, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Shujuan Beng
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei 230012, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Jing Zhang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei 230012, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Xiaoqun Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Can Peng
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei 230012, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Generic Technology Research center for Anhui TCM Industry, Anhui University of Chinese Medicine, Hefei 230012, China; Rural Revitalization Collaborative Technical Service Center of Anhui Province, Anhui University of Chinese Medicine, Hefei 230012, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
3
|
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 2025; 136:109806. [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] [MESH Headings] [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. The objective of this research was to determine 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 postweaning 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 postweaning, whereas Irs1 was lower in male offspring at postweaning. Gene-specific DNA methylation of Irs2 was lower in female offspring at postweaning and Irs1 methylation was higher in male offspring at postweaning 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
|
4
|
Ibrahim M, Bednarczyk M, Stadnicka K, Grochowska E. Inter- and Transgenerational Effects of In Ovo Stimulation with Bioactive Compounds on Cecal Tonsils and Cecal Mucosa Transcriptomes in a Chicken Model. Int J Mol Sci 2025; 26:1174. [PMID: 39940944 PMCID: PMC11817890 DOI: 10.3390/ijms26031174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
Exploring how early-life nutritional interventions may impact future generations, this study examines the inter- and transgenerational effects of in ovo injection of bioactive compounds on gene expression in the cecal tonsils and cecal mucosa using a chicken model. Synbiotic PoultryStar® (Biomin) and choline were injected in ovo on the 12th day of egg incubation. Three experimental groups were established in the generation F1: (1) a control group (C) receiving 0.9% physiological saline (NaCl), (2) a synbiotic group (SYN) receiving 2 mg/embryo, and (3) a combined synbiotic and choline group (SYNCH) receiving 2 mg synbiotic and 0.25 mg choline per embryo. For the generations F2 and F3, the SYN and SYNCH groups were each divided into two subgroups: (A) those injected solely in F1 (SYNs and SYNCHs) and (B) those injected in each generation (SYNr and SYNCHr). At 21 weeks posthatching, cecal tonsil and cecal mucosa samples were collected from F1, F2, and F3 birds for transcriptomic analysis. Gene expression profiling revealed distinct intergenerational and transgenerational patterns in both tissues. In cecal tonsils, a significant transgenerational impact on gene expression was noted in the generation F3, following a drop in F2. In contrast, cecal mucosa showed more gene expression changes in F2, indicating intergenerational effects. While some effects carried into F3, they were less pronounced, except in the SYNs group, which experienced an increase compared to F2. The study highlights that transgenerational effects of epigenetic modifications are dynamic and unpredictable, with effects potentially re-emerging in later generations under certain conditions or fading or intensifying over time. This study provides valuable insights into how epigenetic nutritional stimulation during embryonic development may regulate processes in the cecal tonsils and cecal mucosa across multiple generations. Our findings provide evidence supporting the phenomenon of epigenetic dynamics in a chicken model.
Collapse
Affiliation(s)
- Mariam Ibrahim
- Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland; (M.I.); (K.S.)
- PBS Doctoral School, Bydgoszcz University of Science and Technology, Aleje prof. S. Kaliskiego 7, 85-796 Bydgoszcz, Poland
| | - Marek Bednarczyk
- Department of Animal Biotechnology and Genetics, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland
| | - Katarzyna Stadnicka
- Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland; (M.I.); (K.S.)
| | - Ewa Grochowska
- Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland; (M.I.); (K.S.)
| |
Collapse
|
5
|
Chen L, Liu Q, Li J, Zhang Y, Yang C, Zhao Y. Peripheral blood ABCG1 gene DNA methylation: mediating the relationship between dietary intake of methyl donor nutrients and stroke risk. Nutr Res 2025; 133:54-63. [PMID: 39675233 DOI: 10.1016/j.nutres.2024.10.004] [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: 08/09/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 12/17/2024]
Abstract
Dysregulation of methyl donor nutrients interferes with DNA methylation and is associated with neurological diseases. ABCG1 gene regulates cholesterol to HDL-C, maintains lipid homeostasis, and has been linked to both methyl nutrition and neurological risks. The aim was to investigate whether there is an effect of ABCG1 DNA methylation on the relationship between intake of methyl donor nutrients and the risk of stroke occurrence. We hypothesize that the intake of methyl donor nutrients may influence stroke occurrence by modulating the methylation status of ABCG1. This study utilized a case-control design and selected 52 stroke patients along with 52 healthy controls from Northwest China. Dietary information was collected using a FFQ, and methylation levels were measured at 29 CpG sites of the ABCG1 gene. A significant linear trend was found between dietary intake of the methyl donor nutrient choline and CpG_19.20 methylation of the ABCG1 gene (β = -0.037, P = 0.033). Additionally, a significant association was observed between CpG_19.20 methylation and the risk of stroke (OR 2.325, 95% CI 1.210-4.466). Mediation analysis revealed that choline intake indirectly influenced stroke occurrence through its effect on CpG_19.20 methylation levels in the ABCG1 gene (β = -0.015, SE = 0.013, 95% CI = [-0.053, -0.001]). We found that DNA methylation at specific CpG sites of the peripheral blood ABCG1 gene mediates the association between dietary methyl donor nutrient intake and stroke risk in an adult population from Northwest China. New insights are provided on the prevention and treatment of stroke.
Collapse
Affiliation(s)
- Li Chen
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Qianru Liu
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Juan Li
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yuhong Zhang
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Chan Yang
- School of Nursing, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yi Zhao
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
6
|
Gautier MK, Kelley CM, Lee SH, Mufson EJ, Ginsberg SD. Maternal choline supplementation rescues early endosome pathology in basal forebrain cholinergic neurons in the Ts65Dn mouse model of Down syndrome and Alzheimer's disease. Neurobiol Aging 2024; 144:30-42. [PMID: 39265450 PMCID: PMC11490376 DOI: 10.1016/j.neurobiolaging.2024.09.002] [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: 01/17/2024] [Revised: 07/27/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024]
Abstract
Individuals with DS develop Alzheimer's disease (AD) neuropathology, including endosomal-lysosomal system abnormalities and degeneration of basal forebrain cholinergic neurons (BFCNs). We investigated whether maternal choline supplementation (MCS) affects early endosome pathology within BFCNs using the Ts65Dn mouse model of DS/AD. Ts65Dn and disomic (2N) offspring from dams administered MCS were analyzed for endosomal pathology at 3-4 months or 10-12 months. Morphometric analysis of early endosome phenotype was performed on individual BFCNs using Imaris. The effects of MCS on the endosomal interactome were interrogated by relative co-expression (RCE) analysis. MCS effectively reduced age- and genotype-associated increases in early endosome number in Ts65Dn and 2N offspring, and prevented increases in early endosome size in Ts65Dn offspring. RCE revealed a loss of interactome cooperativity among endosome genes in Ts65Dn offspring that was restored by MCS. These findings demonstrate MCS rescues early endosome pathology, a driver of septohippocampal circuit dysfunction. The genotype-independent benefits of MCS on endosomal phenotype indicate translational applicability as an early-life therapy for DS as well as other neurodevelopmental/neurodegenerative disorders involving endosomal pathology.
Collapse
Affiliation(s)
- Megan K Gautier
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Pathobiology and Translational Medicine Program, New York University Grossman School of Medicine, New York, NY, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Christy M Kelley
- Complex Adaptive Systems Initiative, Arizona State University, Tempe, AZ, USA; Institute for Future Health, Scottsdale, AZ, USA
| | - Sang Han Lee
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Elliott J Mufson
- Departments of Translational Neuroscience and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA; Department of Neuroscience & Physiology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Tobolski D, Zwierzchowski G, Wójcik R, Haxhiaj K, Wishart DS, Ametaj BN. Metabolic Fingerprinting of Blood and Urine of Dairy Cows Affected by Bovine Leukemia Virus: A Mass Spectrometry Approach. Metabolites 2024; 14:624. [PMID: 39590860 PMCID: PMC11596772 DOI: 10.3390/metabo14110624] [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: 10/22/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
OBJECTIVES This study investigated metabolic changes associated with bovine leukemia virus (BLV) infection in dairy cows, focusing on pre-parturition alterations. METHODS Metabolite identification in serum and urine samples was performed using a targeted metabolomics method, employing the TMIC Prime kit in combination with flow injection analysis and liquid chromatography-tandem mass spectrometry. RESULTS Of 145 cows examined, 42 (28.9%) were BLV-seropositive. Around 38% of infected cows showed high somatic cell counts indicative of subclinical mastitis, with 15 experiencing additional health issues such as ketosis, milk fever, and lameness. Despite these conditions, no significant differences in milk yield or composition were observed between the infected and control groups. Metabolomic analysis conducted at -8 and -4 weeks prepartum revealed significant metabolic differences between BLV-infected and healthy cows. At -8 weeks, 30 serum metabolites were altered, including sphingomyelins, lysophosphatidylcholines, amino acids, and acylcarnitines, suggesting disruptions in membrane integrity, energy metabolism, and immune function indicative of early neoplastic transformations. By -4 weeks, the number of altered metabolites decreased to 17, continuing to reflect metabolic disruptions in cows with leukemia. Multivariate analysis highlighted distinct metabolic profiles between infected and control cows, identifying key discriminating metabolites such as choline, aspartic acid, phenylalanine, and arginine. Urine metabolomics revealed significant prepartum shifts in metabolites related to glucose, asymmetric dimethylarginine, and pyruvic acid, among others. CONCLUSIONS The research confirmed metabolomics' efficacy in defining a BLV infection metabolic profile, elucidating leukosis-associated metabolic disruptions. This approach facilitates the identification of BLV-infected cows and enhances understanding of infection pathophysiology, providing a foundation for advanced management and intervention strategies in dairy herds. The study underscores the profound impact of leukosis on metabolic processes and highlights urine metabolomics' utility in non-invasively detecting BLV infection, offering the potential for improved herd health management.
Collapse
Affiliation(s)
- Dawid Tobolski
- Department of Large Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
| | - Grzegorz Zwierzchowski
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; (G.Z.); (K.H.)
- Faculty of Biology and Biotechnology, University of Warmia and Mazury, 1a Oczapowskiego Str., 10-719 Olsztyn, Poland
| | - Roman Wójcik
- Faculty of Veterinary Medicine, University of Warmia and Mazury, 1a Oczapowskiego Str., 10-719 Olsztyn, Poland;
| | - Klevis Haxhiaj
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; (G.Z.); (K.H.)
| | - David S. Wishart
- Departments of Biological and Computer Sciences, University of Alberta, Edmonton, AB T6G 2P5, Canada;
| | - Burim N. Ametaj
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; (G.Z.); (K.H.)
| |
Collapse
|
8
|
Abstract
AIMS High diet quality is related to better health outcomes in general. During pregnancy, a high-quality diet is of paramount importance to promote optimal maternal and neonatal outcomes. This is a scoping review of research available on diet quality indexes (DQIs) for use during pregnancy that summarizes the DQIs in terms of development, country of origin, population used, components, scoring and weighting of components, and evaluation. Furthermore, the DQIs are discussed narratively to inform and direct the development of improved and country-specific DQIs for pregnancy. METHODS The EBSCOhost database was used to identify English-language, peer-reviewed articles published between 2000 and 2023, from which 11 publications were identified that describe the development of pregnancy-specific DQIs. This review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Extension for Scoping Reviews model. RESULTS Almost all DQIs (n = 9 of 11) were developed in high-income countries, using dietary intake data from food frequency questionnaires. Several DQIs (n = 5 of 11) used the US Healthy Eating Index as basis and modified it in various ways. Almost all DQIs included both foods and nutrients as components (n = 9 of 11), with vegetables being the most commonly included component alone (n = 8 of 11) or combined with fruit (n = 2 of 11). CONCLUSION Because most DQIs were developed using dietary guidelines, recommendations, and dietary intake data from high-income countries, it is recommended that pregnancy-specific DQIs be developed and validated to reflect the nutrition guidelines for lower-income and culturally diverse countries.
Collapse
Affiliation(s)
- Liska Robb
- Department of Nutrition and Dietetics, School of Health and Rehabilitation Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein, Republic of South Africa
| | - Gina Joubert
- Department of Biostatistics, School of Biomedical Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein, Republic of South Africa
| | - Corinna May Walsh
- Department of Nutrition and Dietetics, School of Health and Rehabilitation Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein, Republic of South Africa
| |
Collapse
|
9
|
Klement L, Jansakun C, Yan B, Staffer S, Tuma-Kellner S, Altamura S, Muckenthaler M, Merle U, Chamulitrat W. Myeloid-specific deletion of group VIA calcium-independent phospholipase A2 induces pro-inflammatory LPS response predominantly in male mice via MIP-1α activation. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167016. [PMID: 38198970 DOI: 10.1016/j.bbadis.2024.167016] [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: 06/29/2023] [Revised: 12/15/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024]
Abstract
Polymorphisms of group VIA calcium-independent phospholipase A2 (PLA2G6) are associated with blood C-reactive protein suggesting its role in inflammation. We showed that myeloid-specific Pla2g6-deficiency in Pla2g6M-/- mice led to exaggerated inflammation and fibrosis in a lean fatty liver model. We here investigated whether these mutants display alteration in immune response after treatment with E. coli lipopolysaccharides (LPS) under acute (a single dose) and persistent (four doses) conditions. Without LPS treatment, male Pla2g6M-/- (but not Flox) mice at 12 months of age exhibited splenomegaly and hepatic necrosis, and ~ 30 % of them exhibited autoimmune hepatitis showing lymphoplasma cells with CD3(+) and CD45R(+) staining. Under acute LPS, male mutants showed an elevation of plasma MIP-1α and immunoglobulinA as well as upregulation of hepatic apoptosis and fibrosis PARP-1, Bax, MCP-1, α-SMA, and collagen I proteins. Their bone-marrow-derived macrophages also showed an elevation of MIP-1α release upon LPS stimulation in vitro. Female mutants under acute LPS showed a moderate increase in plasma KC/CXCL1, MCP-1, and IL10, and they showed no remarkable increase in hepatic fibrosis under acute or persistent LPS. Male mutants under persistent LPS displayed an elevation of aspartate aminotransferase, blood eosinophils, and hepatic apoptosis. Moreover, ~30 % of these mutants exhibited eosinophilic sclerosing portal hepatitis associated with an upregulated protein expression of hepatic CD8α, CD68, eosinophilic cationic protein, and Ly6G. Thus, myeloid-PLA2G6 deficiency led to an autoimmune and LPS-induced inflammatory liver disease via MIP-1α in a male-predominant manner. Our results may be applicable to patients with PLA2G6 mutations who undergo bacterial infection and sepsis.
Collapse
Affiliation(s)
- Lukas Klement
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Chutima Jansakun
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80161, Thailand
| | - Bin Yan
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Simone Staffer
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Sabine Tuma-Kellner
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Sandro Altamura
- Department of Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany
| | - Martina Muckenthaler
- Department of Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), German Centre for Cardiovascular Research, Partner Site, University of Heidelberg, Germany
| | - Uta Merle
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Walee Chamulitrat
- Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| |
Collapse
|
10
|
Bellio TA, Laguna-Torres JY, Campion MS, Chou J, Yee S, Blusztajn JK, Mellott TJ. Perinatal choline supplementation prevents learning and memory deficits and reduces brain amyloid Aβ42 deposition in AppNL-G-F Alzheimer's disease model mice. PLoS One 2024; 19:e0297289. [PMID: 38315685 PMCID: PMC10843108 DOI: 10.1371/journal.pone.0297289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 01/02/2024] [Indexed: 02/07/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by cognitive and memory impairments and neuropathological abnormalities. AD has no cure, inadequate treatment options, and a limited understanding of possible prevention measures. Previous studies have demonstrated that AD model mice that received a diet high in the essential nutrient choline had reduced amyloidosis, cholinergic deficits, and gliosis, and increased neurogenesis. In this study, we investigated the lifelong effects of perinatal choline supplementation on behavior, cognitive function, and amyloidosis in AppNL-G-F AD model mice. Pregnant and lactating mice were given a diet containing either 1.1 g/kg (control) or 5 g/kg (supplemented) of choline chloride until weaning and subsequently, all offspring received the control diet throughout their life. At 3, 6, 9, and 12 months of age, animals were behaviorally tested in the Open Field Test, Elevated Plus Maze, Barnes Maze, and in a contextual fear conditioning paradigm. Immunohistochemical analysis of Aβ42 was also conducted on the brains of these mice. AppNL-G-F mice displayed hippocampal-dependent spatial learning deficits starting at 3-months-old that persisted until 12-months-old. These spatial learning deficits were fully prevented by perinatal choline supplementation at young ages (3 and 6 months) but not in older mice (12 months). AppNL-G-F mice also had impaired fearful learning and memory at 9- and 12-months-old that were diminished by choline supplementation. Perinatal choline supplementation reduced Aβ42 deposition in the amygdala, cortex, and hippocampus of AppNL-G-F mice. Together, these results demonstrate that perinatal choline supplementation is capable of preventing cognitive deficits and dampening amyloidosis in AppNL-G-F mice and suggest that ensuring adequate choline consumption during early life may be a valuable method to prevent or reduce AD dementia and neuropathology.
Collapse
Affiliation(s)
- Thomas A. Bellio
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Jessenia Y. Laguna-Torres
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Mary S. Campion
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Jay Chou
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Sheila Yee
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Jan K. Blusztajn
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Tiffany J. Mellott
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
11
|
Lin Y, Wu J, Zhuo Y, Feng B, Fang Z, Xu S, Li J, Zhao H, Wu D, Hua L, Che L. Effects of maternal methyl donor intake during pregnancy on ileum methylation and function in an intrauterine growth restriction pig model. J Anim Sci Biotechnol 2024; 15:19. [PMID: 38310243 PMCID: PMC10838427 DOI: 10.1186/s40104-023-00970-w] [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: 07/21/2023] [Accepted: 12/04/2023] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Intrauterine growth retardation (IUGR) affects intestinal growth, morphology, and function, which leads to poor growth performance and high mortality. The present study explored whether maternal dietary methyl donor (MET) supplementation alleviates IUGR and enhances offspring's growth performance by improving intestinal growth, function, and DNA methylation of the ileum in a porcine IUGR model. METHODS Forty multiparous sows were allocated to the control or MET diet groups from mating until delivery. After farrowing, 8 pairs of IUGR and normal birth weight piglets from 8 litters were selected for sampling before suckling colostrum. RESULTS The results showed that maternal MET supplementation tended to decrease the IUGR incidence and increased the average weaning weight of piglets. Moreover, maternal MET supplementation significantly reduced the plasma concentrations of isoleucine, cysteine, urea, and total amino acids in sows and newborn piglets. It also increased lactase and sucrase activity in the jejunum of newborn piglets. MET addition resulted in lower ileal methionine synthase activity and increased betaine homocysteine S-methyltransferase activity in the ileum of newborn piglets. DNA methylation analysis of the ileum showed that MET supplementation increased the methylation level of DNA CpG sites in the ileum of newborn piglets. Down-regulated differentially methylated genes were enriched in folic acid binding, insulin receptor signaling pathway, and endothelial cell proliferation. In contrast, up-regulated methylated genes were enriched in growth hormone receptor signaling pathway and nitric oxide biosynthetic process. CONCLUSIONS Maternal MET supplementation can reduce the incidence of IUGR and increase the weaning litter weight of piglets, which may be associated with better intestinal function and methylation status.
Collapse
Affiliation(s)
- Yan Lin
- Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Jiangnan Wu
- Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yong Zhuo
- Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Bin Feng
- Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Zhengfeng Fang
- Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Shengyu Xu
- Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Jian Li
- Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Hua Zhao
- Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - De Wu
- Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Lun Hua
- Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Lianqiang Che
- Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
12
|
Ullah A, Sarwar I, Suheryani I, Ahmad S, Andlib S, Buzdar JA, Kakar MU, Arain MA. Role of dietary lecithin as an emulsifying agent in poultry nutrition: efficacy and feasibility. WORLD POULTRY SCI J 2024; 80:187-206. [DOI: 10.1080/00439339.2023.2268584] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Affiliation(s)
- Aftab Ullah
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Imran Sarwar
- Department of Electrical Engineering, Balochistan University of Engineering and Technology Khuzdar, Balochistan, Pakistan
| | - Imran Suheryani
- Department of Pharmaceutics, Faculty of Pharmacy, University of Sindh Jamshoro, Jamshoro, Pakistan
| | - Sajjad Ahmad
- Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture,Water and Marine Sciences, Uthal, Pakistan
| | - Shaista Andlib
- Department of Microbiology, Faculty of Biological Sciences, Quaid-i-Azam University Islamabad
| | - Jameel Ahmed Buzdar
- Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture,Water and Marine Sciences, Uthal, Pakistan
| | - Mohib Ullah Kakar
- Faculty of Marine Sciences, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, Pakistan
| | - Muhammad Asif Arain
- Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture,Water and Marine Sciences, Uthal, Pakistan
| |
Collapse
|
13
|
Choi SW, Park S, Duah A, Kim K, Park M. Consequences of the 2019 Public Charge Rule Announcement and Publication on Prenatal WIC Participation Among Immigrant Families: Evidence of Spillover Effects. J Immigr Minor Health 2023; 25:1229-1238. [PMID: 37530903 DOI: 10.1007/s10903-023-01523-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2023] [Indexed: 08/03/2023]
Abstract
This study analyzed the effects of the announcement and publication of the 2019 Public Charge Rule on participation of the special supplemental nutrition program for women, infants, and children (WIC) among pregnant immigrants. A difference-in-differences approach was used to analyze the changes in prenatal WIC participation before and after the 2019 Public Charge Rule announcement and publication among immigrants relative to US natives. We identified 17,623,683 live singletons born in a hospital from 2015 to 2019. Compared to US natives, the odds of prenatal WIC participation among immigrants were 11.4% lower after the 2019 Public Charge Rule announcement, and 19% lower after the final rule was published. The results of this study suggest that pregnant immigrants may decide not to participate in the WIC program due to the fear of jeopardizing their immigration status after the announcement and publication of the 2019 Public Charge Rule.
Collapse
Affiliation(s)
- Sung W Choi
- School of Public Affairs, Pennsylvania State University - Harrisburg, 777 West Harrisburg Pike, Middletown, PA, 17057, USA.
| | - Sujeong Park
- School of Public Affairs, Pennsylvania State University - Harrisburg, 777 West Harrisburg Pike, Middletown, PA, 17057, USA
| | - Abena Duah
- School of Public Affairs, Pennsylvania State University - Harrisburg, 777 West Harrisburg Pike, Middletown, PA, 17057, USA
| | - Kyungha Kim
- Department of Practice, Sciences, and Health Outcomes Research, University of Maryland - Baltimore, 220 Arch Street, Baltimore, MD, 21201, USA
| | - Mingean Park
- School of Public Affairs, Pennsylvania State University - Harrisburg, 777 West Harrisburg Pike, Middletown, PA, 17057, USA
| |
Collapse
|
14
|
Jiang X, Hu Z, Qiu X, Wu L, Zhou R, Yang Y, Xiang X. Poria cocos (Schw.) Wolf, a Traditional Chinese Edible Medicinal Herb, Promotes Neuronal Differentiation, and the Morphological Maturation of Newborn Neurons in Neural Stem/Progenitor Cells. Molecules 2023; 28:7480. [PMID: 38005201 PMCID: PMC10672746 DOI: 10.3390/molecules28227480] [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: 09/08/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Neurogenesis in the adult brain comprises the entire set of events of neuronal development. It begins with the division of precursor cells to form a mature, integrated, and functioning neuronal network. Adult neurogenesis is believed to play an important role in animals' cognitive abilities, including learning and memory. In the present study, significant neuronal differentiation-promoting activity of 80% (v/v) ethanol extract of P. cocos (EEPC) was found in Neuro-2a cells and mouse cortical neural stem/progenitor cells (NSPCs). Subsequently, a total of 97 compounds in EEPC were identified by UHPLC-Q-Exactive-MS/MS. Among them, four major compounds-Adenosine; Choline; Ethyl palmitoleate; and L-(-)-arabinitol-were further studied for their neuronal differentiation-promoting activity. Of which, choline has the most significant neuronal differentiation-promoting activity, indicating that choline, as the main bioactive compound in P. cocos, may have a positive effect on learning and memory functions. Compared with similar research literature, this is the first time that the neuronal differentiation-promoting effects of P. cocos extract have been studied.
Collapse
Affiliation(s)
- Xia Jiang
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, College of Biological and Food Engineering, Huaihua University, Huaihua 418008, China; (X.J.); (Z.H.); (X.Q.); (L.W.); (R.Z.); (Y.Y.)
- College of Chemistry and Materials Engineering, Huaihua University, Huaihua 418008, China
| | - Zhaotun Hu
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, College of Biological and Food Engineering, Huaihua University, Huaihua 418008, China; (X.J.); (Z.H.); (X.Q.); (L.W.); (R.Z.); (Y.Y.)
| | - Xiaoyan Qiu
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, College of Biological and Food Engineering, Huaihua University, Huaihua 418008, China; (X.J.); (Z.H.); (X.Q.); (L.W.); (R.Z.); (Y.Y.)
| | - Liming Wu
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, College of Biological and Food Engineering, Huaihua University, Huaihua 418008, China; (X.J.); (Z.H.); (X.Q.); (L.W.); (R.Z.); (Y.Y.)
| | - Rong Zhou
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, College of Biological and Food Engineering, Huaihua University, Huaihua 418008, China; (X.J.); (Z.H.); (X.Q.); (L.W.); (R.Z.); (Y.Y.)
| | - Yaoyao Yang
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, College of Biological and Food Engineering, Huaihua University, Huaihua 418008, China; (X.J.); (Z.H.); (X.Q.); (L.W.); (R.Z.); (Y.Y.)
| | - Xiaoliang Xiang
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, College of Biological and Food Engineering, Huaihua University, Huaihua 418008, China; (X.J.); (Z.H.); (X.Q.); (L.W.); (R.Z.); (Y.Y.)
- Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, Huaihua University, Huaihua 418008, China
| |
Collapse
|
15
|
Gautier MK, Kelley CM, Lee SH, Alldred MJ, McDaid J, Mufson EJ, Stutzmann GE, Ginsberg SD. Maternal choline supplementation protects against age-associated cholinergic and GABAergic basal forebrain neuron degeneration in the Ts65Dn mouse model of Down syndrome and Alzheimer's disease. Neurobiol Dis 2023; 188:106332. [PMID: 37890559 PMCID: PMC10752300 DOI: 10.1016/j.nbd.2023.106332] [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: 08/07/2023] [Revised: 10/02/2023] [Accepted: 10/22/2023] [Indexed: 10/29/2023] Open
Abstract
Down syndrome (DS) is a genetic disorder caused by triplication of human chromosome 21. In addition to intellectual disability, DS is defined by a premature aging phenotype and Alzheimer's disease (AD) neuropathology, including septohippocampal circuit vulnerability and degeneration of basal forebrain cholinergic neurons (BFCNs). The Ts65Dn mouse model recapitulates key aspects of DS/AD pathology, namely age-associated atrophy of BFCNs and cognitive decline in septohippocampal-dependent behavioral tasks. We investigated whether maternal choline supplementation (MCS), a well-tolerated treatment modality, protects vulnerable BFCNs from age- and genotype-associated degeneration in trisomic offspring. We also examined the effect of trisomy, and MCS, on GABAergic basal forebrain parvalbumin neurons (BFPNs), an unexplored neuronal population in this DS model. Unbiased stereological analyses of choline acetyltransferase (ChAT)-immunoreactive BFCNs and parvalbumin-immunoreactive BFPNs were conducted using confocal z-stacks of the medial septal nucleus and the vertical limb of the diagonal band (MSN/VDB) in Ts65Dn mice and disomic (2N) littermates at 3-4 and 10-12 months of age. MCS trisomic offspring displayed significant increases in ChAT-immunoreactive neuron number and density compared to unsupplemented counterparts, as well as increases in the area of the MSN/VDB occupied by ChAT-immunoreactive neuropil. MCS also rescued BFPN number and density in Ts65Dn offspring, a novel rescue of a non-cholinergic cell population. Furthermore, MCS prevented age-associated loss of BFCNs and MSN/VDB regional area in 2N offspring, indicating genotype-independent neuroprotective benefits. These findings demonstrate MCS provides neuroprotection of vulnerable BFCNs and non-cholinergic septohippocampal BFPNs, indicating this modality has translational value as an early life therapy for DS, as well as extending benefits to the aging population at large.
Collapse
Affiliation(s)
- Megan K Gautier
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Pathobiology and Translational Medicine Program, New York University Grossman School of Medicine, New York, NY, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Christy M Kelley
- Complex Adaptive Systems Initiative, Arizona State University, Tempe, AZ, USA; Institute for Future Health, Scottsdale, AZ, USA
| | - Sang Han Lee
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - John McDaid
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University/The Chicago Medical School, North Chicago, IL, USA
| | - Elliott J Mufson
- Departments of Translational Neuroscience and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Grace E Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University/The Chicago Medical School, North Chicago, IL, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA; Department of Neuroscience & Physiology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
16
|
Jeong S, Chokkalla AK, Davis CK, Vemuganti R. Post-stroke depression: epigenetic and epitranscriptomic modifications and their interplay with gut microbiota. Mol Psychiatry 2023; 28:4044-4055. [PMID: 37188778 PMCID: PMC10646155 DOI: 10.1038/s41380-023-02099-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 04/21/2023] [Accepted: 05/02/2023] [Indexed: 05/17/2023]
Abstract
Epigenetic and epitranscriptomic modifications that regulate physiological processes of an organism at the DNA and RNA levels, respectively, are novel therapeutic candidates for various neurological diseases. Gut microbiota and its metabolites are known to modulate DNA methylation and histone modifications (epigenetics), as well as RNA methylation especially N6-methyladenosine (epitranscriptomics). As gut microbiota as well as these modifications are highly dynamic across the lifespan of an organism, they are implicated in the pathogenesis of stroke and depression. The lack of specific therapeutic interventions for managing post-stroke depression emphasizes the need to identify novel molecular targets. This review highlights the interaction between the gut microbiota and epigenetic/epitranscriptomic pathways and their interplay in modulating candidate genes that are involved in post-stroke depression. This review further focuses on the three candidates, including brain-derived neurotrophic factor, ten-eleven translocation family proteins, and fat mass and obesity-associated protein based on their prevalence and pathoetiologic role in post-stroke depression.
Collapse
Affiliation(s)
- Soomin Jeong
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA.
- William S. Middleton Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
17
|
Chen M, Fan L, Wu G, Wang H, Gu S. Histone methyltransferase enzyme enhancer of zeste homolog 2 counteracts ischemic brain injury via H3K27me3-mediated regulation of PI3K/AKT/mTOR signaling pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:2240-2255. [PMID: 37334851 DOI: 10.1002/tox.23863] [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: 04/14/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND Epigenetic histone methylation plays a crucial role in cerebral ischemic injury, particularly in the context of ischemic stroke. However, the complete understanding of regulators involved in histone methylation, such as Enhancer of Zeste Homolog 2 (EZH2), along with their functional effects and underlying mechanisms, remains incomplete. METHODS Here, we employed a rat model of MCAO (Middle cerebral artery occlusion) and an OGD (Oxygen-Glucose Deprivation) model of primary cortical neurons to study the role of EZH2 and H3K27me3 in cerebral ischemia-reperfusion injury. The infarct volume was measured through TTC staining, while cell apoptosis was detected using TUNEL staining. The mRNA expression levels were quantified through quantitative real-time polymerase chain reaction (qPCR), whereas protein expressions were evaluated via western blotting and immunofluorescence experiments. RESULTS The expression levels of EZH2 and H3K27me3 were upregulated in OGD; these expression levels were further enhanced by GSK-J4 but reduced by EPZ-6438 and AKT inhibitor (LY294002) under OGD conditions. Similar trends were observed for mTOR, AKT, and PI3K while contrasting results were noted for UTX and JMJD3. The phosphorylation levels of mTOR, AKT, and PI3K were activated by OGD, further stimulated by GSK-J4, but inhibited by EPZ-6438 and AKT inhibitor. Inhibition of EZH2 or AKT effectively counteracted OGD-/MCAO-induced cell apoptosis. Additionally, inhibition of EZH2 or AKT mitigated MCAO-induced infarct size and neurological deficit in vivo. CONCLUSIONS Collectively, our results demonstrate that EZH2 inhibition exerts a protective effect against ischemic brain injury by modulating the H3K27me3/PI3K/AKT/mTOR signaling pathway. The results provide novel insights into potential therapeutic mechanisms for stroke treatment.
Collapse
Affiliation(s)
- Miao Chen
- Department of Emergency, The First Affiliated Hospital of Hainan Medical University, Haikou, People's Republic of China
| | - Limin Fan
- The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Guoping Wu
- Department of Emergency, Sansha People's Hospital, Sansha, People's Republic of China
| | - Hairong Wang
- Department of Emergency, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Shuo Gu
- Department of Pediatric Neurosurgery, The First Affiliated Hospital of Hainan Medical University, Haikou, People's Republic of China
| |
Collapse
|
18
|
Derbyshire E, Maes M. The Role of Choline in Neurodevelopmental Disorders-A Narrative Review Focusing on ASC, ADHD and Dyslexia. Nutrients 2023; 15:2876. [PMID: 37447203 DOI: 10.3390/nu15132876] [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: 06/02/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Neurodevelopmental disorders appear to be rising in prevalence, according to the recent Global Burden of Disease Study. This rise is likely to be multi-factorial, but the role of certain nutrients known to facilitate neurodevelopment should be considered. One possible contributing factor could be attributed to deficits in choline intake, particularly during key stages of neurodevelopment, which includes the first 1000 days of life and childhood. Choline, a key micronutrient, is crucial for optimal neurodevelopment and brain functioning of offspring. The present narrative review discusses the main research, describing the effect of choline in neurodevelopmental disorders, to better understand its role in the etiology and management of these disorders. In terms of findings, low choline intakes and reduced or altered choline status have been reported in relevant population subgroups: pregnancy (in utero), children with autism spectrum disorders, people with attention deficit hyperactivity disorder and those with dyslexia. In conclusion, an optimal choline provision may offer some neuronal protection in early life and help to mitigate some cognitive effects in later life attributed to neurodevelopmental conditions. Research indicates that choline may act as a modifiable risk factor for certain neurodevelopmental conditions. Ongoing research is needed to unravel the mechanisms and explanations.
Collapse
Affiliation(s)
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok 4002, Thailand
- Research Institute, Medical University of Plovdiv, 10330 Plovdiv, Bulgaria
| |
Collapse
|
19
|
Paules EM, Silva-Gomez JA, Friday WB, Zeisel SH, Trujillo-Gonzalez I. Choline Regulates SOX4 through miR-129-5p and Modifies H3K27me3 in the Developing Cortex. Nutrients 2023; 15:2774. [PMID: 37375678 PMCID: PMC10304412 DOI: 10.3390/nu15122774] [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: 05/22/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Choline availability regulates neural progenitor cell proliferation and differentiation in the developing cerebral cortex. Here, we investigated the molecular mechanism underlying this process and demonstrated that choline regulates the transcription factor SOX4 in neural progenitor cells. Specifically, we found that low choline intake during neurogenesis reduces SOX4 protein levels, causing the downregulation of EZH2, a histone methyltransferase. Importantly, we demonstrate that low choline is not involved in SOX4 protein degradation rate and established that protein reduction is caused by aberrant expression of a microRNA (miR-129-5p). To confirm the role of miR-129-5p, we conducted gain-of-function and loss-of-function assays in neural progenitor cells and demonstrated that directly altering miR-129-5p levels could affect SOX4 protein levels. We also observed that the reduction in SOX4 and EZH2 led to decreased global levels of H3K27me3 in the developing cortex, contributing to reduced proliferation and precocious differentiation. For the first time, to our knowledge, we demonstrate that a nutrient, choline, regulates a master transcription factor and its downstream targets, providing a novel insight into the role of choline in brain development.
Collapse
Affiliation(s)
- Evan M. Paules
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; (E.M.P.); (J.A.S.-G.); (W.B.F.); (S.H.Z.)
| | - Jorge A. Silva-Gomez
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; (E.M.P.); (J.A.S.-G.); (W.B.F.); (S.H.Z.)
| | - Walter B. Friday
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; (E.M.P.); (J.A.S.-G.); (W.B.F.); (S.H.Z.)
| | - Steve H. Zeisel
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; (E.M.P.); (J.A.S.-G.); (W.B.F.); (S.H.Z.)
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| | - Isis Trujillo-Gonzalez
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; (E.M.P.); (J.A.S.-G.); (W.B.F.); (S.H.Z.)
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| |
Collapse
|
20
|
Bragg MG, Prado EL, Caswell BL, Arnold CD, George M, Oakes LM, Beckner AG, DeBolt MC, Bennett BJ, Maleta KM, Stewart CP. The association between plasma choline, growth and neurodevelopment among Malawian children aged 6-15 months enroled in an egg intervention trial. MATERNAL & CHILD NUTRITION 2023; 19:e13471. [PMID: 36567549 PMCID: PMC10019050 DOI: 10.1111/mcn.13471] [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: 07/28/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/27/2022]
Abstract
Choline is an essential micronutrient that may influence growth and development; however, few studies have examined postnatal choline status and children's growth and development in low- and middle-income countries. The aim of this observational analysis was to examine associations of plasma choline with growth and development among Malawian children aged 6-15 months enrolled in an egg intervention trial. Plasma choline and related metabolites (betaine, dimethylglycine and trimethylamine N-oxide) were measured at baseline and 6-month follow-up, along with anthropometric (length, weight, head circumference) and developmental assessments (the Malawi Developmental Assessment Tool [MDAT], the Infant Orienting with Attention task [IOWA], a visual paired comparison [VPC] task and an elicited imitation [EI] task). In cross-sectional covariate-adjusted models, each 1 SD higher plasma choline was associated with lower length-for-age z-score (-0.09 SD [95% confidence interval, CI -0.17 to -0.01]), slower IOWA response time (8.84 ms [1.66-16.03]) and faster processing speed on the VPC task (-203.5 ms [-366.2 to -40.7]). In predictive models, baseline plasma choline was negatively associated with MDAT fine motor z-score at 6-month follow-up (-0.13 SD [-0.22 to -0.04]). There were no other significant associations of plasma choline with child measures. Similarly, associations of choline metabolites with growth and development were null except higher trimethylamine N-oxide was associated with slower information processing on the VPC task and higher memory scores on the EI task. In this cohort of children with low dietary choline intake, we conclude that there were no strong or consistent associations between plasma choline and growth and development.
Collapse
Affiliation(s)
- Megan G. Bragg
- Department of NutritionUniversity of California DavisDavisCaliforniaUSA
- AJ Drexel Autism InstituteDrexel UniversityPhiladelphiaPennsylvaniaUSA
| | | | - Bess L. Caswell
- USDA Western Human Nutrition Research CenterDavisCaliforniaUSA
| | - Charles D. Arnold
- Department of NutritionUniversity of California DavisDavisCaliforniaUSA
| | - Matthews George
- School of Public Health and Family MedicineKamuzu University of Health SciencesBlantyreMalawi
| | - Lisa M. Oakes
- Center for Mind and BrainUniversity of California DavisDavisCaliforniaUSA
| | - Aaron G. Beckner
- Center for Mind and BrainUniversity of California DavisDavisCaliforniaUSA
| | - Michaela C. DeBolt
- Center for Mind and BrainUniversity of California DavisDavisCaliforniaUSA
| | | | - Kenneth M. Maleta
- School of Public Health and Family MedicineKamuzu University of Health SciencesBlantyreMalawi
| | | |
Collapse
|
21
|
Korsmo HW, Kadam I, Reaz A, Bretter R, Saxena A, Johnson CH, Caviglia JM, Jiang X. Prenatal Choline Supplement in a Maternal Obesity Model Modulates Offspring Hepatic Lipidomes. Nutrients 2023; 15:965. [PMID: 36839327 PMCID: PMC9963284 DOI: 10.3390/nu15040965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/09/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023] Open
Abstract
Maternal obesity during pregnancy adversely impacts offspring health, predisposing them to chronic metabolic diseases characterized by insulin resistance, dysregulated macronutrient metabolism, and lipid overload, such as metabolic-associated fatty liver disease (MAFLD). Choline is a semi-essential nutrient involved in lipid and one-carbon metabolism that is compromised during MAFLD progression. Here, we investigated under high-fat (HF) obesogenic feeding how maternal choline supplementation (CS) influenced the hepatic lipidome of mouse offspring. Our results demonstrate that maternal HF+CS increased relative abundance of a subclass of phospholipids called plasmalogens in the offspring liver at both embryonic day 17.5 and after 6 weeks of postnatal HF feeding. Consistent with the role of plasmalogens as sacrificial antioxidants, HF+CS embryos were presumably protected with lower oxidative stress. After postnatal HF feeding, the maternal HF+CS male offspring also had higher relative abundance of both sphingomyelin d42:2 and its side chain, nervonic acid (FA 24:1). Nervonic acid is exclusively metabolized in the peroxisome and is tied to plasmalogen synthesis. Altogether, this study demonstrates that under the influence of obesogenic diet, maternal CS modulates the fetal and postnatal hepatic lipidome of male offspring, favoring plasmalogen synthesis, an antioxidative response that may protect the mouse liver from damages due to HF feeding.
Collapse
Affiliation(s)
- Hunter W. Korsmo
- PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA
| | - Isma’il Kadam
- PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA
| | - Aziza Reaz
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA
| | - Rachel Bretter
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA
| | - Anjana Saxena
- Department of Biology, Brooklyn College of the City University of New York, New York, NY 11210, USA
| | | | - Jorge Matias Caviglia
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA
| | - Xinyin Jiang
- PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA
| |
Collapse
|
22
|
Shanmuganathan M, Bogert M, Kroezen Z, Britz-McKibbin P, Atkinson SA. Dynamic Metabolic Signatures of Choline and Carnitine across Healthy Pregnancy and in Cord Blood: Association with Maternal Dietary Protein. J Nutr 2023; 153:999-1007. [PMID: 36780943 DOI: 10.1016/j.tjnut.2023.02.013] [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: 09/15/2022] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/13/2023] Open
Abstract
BACKGROUND In pregnancy, choline is deemed an essential nutrient and carnitine needs are increased, but amounts remain undefined. OBJECTIVES We aimed to measure choline and total dietary protein and dairy protein intake from food and supplements across pregnancy and the response to intake by profiling choline and carnitine metabolites across pregnancy and in cord blood. METHODS An exploratory analysis of choline and protein intake from 3-d diet records and measures of 36 serum choline and carnitine metabolites in early (12-17 wk) and late (36-38 wk) pregnancy was conducted in participants from the Be Healthy in Pregnancy study randomized to high dairy protein+walking exercise or usual care. Metabolites were measured in fasted maternal and cord serum using multisegment injection-capillary electrophoresis-mass spectrometry. Mixed ANOVA adjusted for body mass index was performed for comparison of metabolites across pregnancy and between intervention and control. RESULTS In 104 participants, the median (Q1, Q3) total choline intake was 347 (263, 427) mg/d in early and 322 (270, 437) mg/d in late pregnancy. Only ∼20% of participants achieved the recommended adequate intake (450 mg/d) and ∼10% consumed supplemental choline (8-200 mg/d). Serum-free choline (μmol/L) was higher in late compared with early pregnancy [12.9 (11.4, 15.1) compared with 9.68 (8.25, 10.61), P < 0.001], but choline downstream metabolites were similar across pregnancy. Serum carnitine [10.3 (9.01, 12.2) compared with 15.9 (14.1, 17.9) μmol/L, P < 0.001] and acetylcarnitine [2.35 (1.92, 2.68) compared with 3.0 (2.56, 3.59), P < 0.001] were significantly lower in late pregnancy. High cord:maternal serum metabolite ratios were found in most measured metabolites. CONCLUSIONS Despite inadequate choline intake, serum-free choline was elevated in late pregnancy and enriched in cord blood compared with maternal serum. Serum carnitine declined in late pregnancy despite a high protein diet. The higher cord:maternal concentrations in choline and carnitine metabolites suggest active uptake in late pregnancy, reflecting the importance of these circulating metabolites in fetal development. This trial was registered at clinicaltrials.gov as NCT01689961.
Collapse
Affiliation(s)
- Meera Shanmuganathan
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Meghan Bogert
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Zachary Kroezen
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
23
|
Inadequate Choline Intake in Pregnant Women in Germany. Nutrients 2022; 14:nu14224862. [PMID: 36432547 PMCID: PMC9696170 DOI: 10.3390/nu14224862] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Choline is an essential nutrient that is involved in various developmental processes during pregnancy. While the general adequate choline intake (AI) for adults has been set at 400 mg/day by the European Food Safety Authority (EFSA), an AI of 480 mg/day has been derived for pregnant women. To date, the choline intake of pregnant women in Germany has not been investigated yet. Therefore, in this survey, the total choline intake from dietary and supplementary sources in pregnant women was estimated using an online questionnaire. A total of 516 pregnant women participated in the survey, of which 283 met the inclusion criteria (13 to 41 weeks of gestational age, 19−45 years). 224 (79%) of the participants followed an omnivorous diet, 59 (21%) were vegetarian or vegan. Median choline intake was 260.4 (±141.4) mg/day, and only 19 women (7%) achieved the adequate choline intake. The median choline intake of omnivores was significantly higher than that of vegetarians/vegans (269.5 ± 141.5 mg/day vs. 205.2 ± 101.2 mg/day; p < 0.0001). 5% (13/283) of pregnant women took choline-containing dietary supplements. In these women, dietary supplements provided 19% of the total choline intake. Due to the importance of choline for the developmental processes during pregnancy, the study results prove the urgent need for an improved choline supply for pregnant women.
Collapse
|
24
|
Schapovalova O, Gorlova A, de Munter J, Sheveleva E, Eropkin M, Gorbunov N, Sicker M, Umriukhin A, Lyubchyk S, Lesch KP, Strekalova T, Schroeter CA. Immunomodulatory effects of new phytotherapy on human macrophages and TLR4- and TLR7/8-mediated viral-like inflammation in mice. Front Med (Lausanne) 2022; 9:952977. [PMID: 36091684 PMCID: PMC9450044 DOI: 10.3389/fmed.2022.952977] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022] Open
Abstract
Background While all efforts have been undertaken to propagate the vaccination and develop remedies against SARS-CoV-2, no satisfactory management of this infection is available yet. Moreover, poor availability of any preventive and treatment measures of SARS-CoV-2 in economically disadvantageous communities aggravates the course of the pandemic. Here, we studied a new immunomodulatory phytotherapy (IP), an extract of blackberry, chamomile, garlic, cloves, and elderberry as a potential low-cost solution for these problems given the reported efficacy of herbal medicine during the previous SARS virus outbreak. Methods The key feature of SARS-CoV-2 infection, excessive inflammation, was studied in in vitro and in vivo assays under the application of the IP. First, changes in tumor-necrosis factor (TNF) and lnteurleukin-1 beta (IL-1β) concentrations were measured in a culture of human macrophages following the lipopolysaccharide (LPS) challenge and treatment with IP or prednisolone. Second, chronically IP-pre-treated CD-1 mice received an agonist of Toll-like receptors (TLR)-7/8 resiquimod and were examined for lung and spleen expression of pro-inflammatory cytokines and blood formula. Finally, chronically IP-pre-treated mice challenged with LPS injection were studied for "sickness" behavior. Additionally, the IP was analyzed using high-potency-liquid chromatography (HPLC)-high-resolution-mass-spectrometry (HRMS). Results LPS-induced in vitro release of TNF and IL-1β was reduced by both treatments. The IP-treated mice displayed blunted over-expression of SAA-2, ACE-2, CXCL1, and CXCL10 and decreased changes in blood formula in response to an injection with resiquimod. The IP-treated mice injected with LPS showed normalized locomotion, anxiety, and exploration behaviors but not abnormal forced swimming. Isoquercitrin, choline, leucine, chlorogenic acid, and other constituents were identified by HPLC-HRMS and likely underlie the IP immunomodulatory effects. Conclusions Herbal IP-therapy decreases inflammation and, partly, "sickness behavior," suggesting its potency to combat SARS-CoV-2 infection first of all via its preventive effects.
Collapse
Affiliation(s)
- Olesia Schapovalova
- Caparica Faculdade de Ciencias e Tecnologia da Universidade Nova de Lisboa, NOVA Lisbon University, Lisbon, Portugal
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
| | - Anna Gorlova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Cognitive Dysfunctions, Federal Budgetary Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Johannes de Munter
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
| | - Elisaveta Sheveleva
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Cognitive Dysfunctions, Federal Budgetary Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Mikhail Eropkin
- Department of Etiology and Epidemiology, Smorodintsev Research Institute of Influenza, St. Petersburg State University, Saint Petersburg, Russia
| | - Nikita Gorbunov
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Michail Sicker
- Rehabilitation Research Unit of Clinic of Bad Kreuzbach, Bad Kreuzbach, Germany
| | - Aleksei Umriukhin
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sergiy Lyubchyk
- Caparica Faculdade de Ciencias e Tecnologia da Universidade Nova de Lisboa, NOVA Lisbon University, Lisbon, Portugal
- EIGES Center, Universidade Lusofona, Lisboa, Portugal
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University and Neuroplast BV, Maastricht, Netherlands
- Laboratory of Cognitive Dysfunctions, Federal Budgetary Institute of General Pathology and Pathophysiology, Moscow, Russia
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
25
|
Prenatal Iron Deficiency and Choline Supplementation Interact to Epigenetically Regulate Jarid1b and Bdnf in the Rat Hippocampus into Adulthood. Nutrients 2021; 13:nu13124527. [PMID: 34960080 PMCID: PMC8706459 DOI: 10.3390/nu13124527] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 11/24/2022] Open
Abstract
Early-life iron deficiency (ID) causes long-term neurocognitive impairments and gene dysregulation that can be partially mitigated by prenatal choline supplementation. The long-term gene dysregulation is hypothesized to underlie cognitive dysfunction. However, mechanisms by which iron and choline mediate long-term gene dysregulation remain unknown. In the present study, using a well-established rat model of fetal-neonatal ID, we demonstrated that ID downregulated hippocampal expression of the gene encoding JmjC-ARID domain-containing protein 1B (JARID1B), an iron-dependent histone H3K4 demethylase, associated with a higher histone deacetylase 1 (HDAC1) enrichment and a lower enrichment of acetylated histone H3K9 (H3K9ac) and phosphorylated cAMP response element-binding protein (pCREB). Likewise, ID reduced transcriptional capacity of the gene encoding brain-derived neurotrophic factor (BDNF), a target of JARID1B, associated with repressive histone modifications such as lower H3K9ac and pCREB enrichments at the Bdnf promoters in the adult rat hippocampus. Prenatal choline supplementation did not prevent the ID-induced chromatin modifications at these loci but induced long-lasting repressive chromatin modifications in the iron-sufficient adult rats. Collectively, these findings demonstrated that the iron-dependent epigenetic mechanism mediated by JARID1B accounted for long-term Bdnf dysregulation by early-life ID. Choline supplementation utilized a separate mechanism to rescue the effect of ID on neural gene regulation. The negative epigenetic effects of choline supplementation in the iron-sufficient rat hippocampus necessitate additional investigations prior to its use as an adjunctive therapeutic agent.
Collapse
|
26
|
Derbyshire E, Obeid R, Schön C. Habitual Choline Intakes across the Childbearing Years: A Review. Nutrients 2021; 13:nu13124390. [PMID: 34959942 PMCID: PMC8709092 DOI: 10.3390/nu13124390] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 11/20/2022] Open
Abstract
Choline is an important nutrient during the first 1000 days post conception due to its roles in brain function. An increasing number of studies have measured choline intakes at the population level. We collated the evidence focusing on habitual choline intakes in the preconceptual, pregnancy, and lactation life stages. We conducted a review including studies published from 2004 to 2021. Twenty-six relevant publications were identified. After excluding studies with a high choline intake (>400 mg/day; two studies) or low choline intake (<200 mg/day; one study), average choline intake in the remaining 23 studies ranged from 233 mg/day to 383 mg/day, even with the inclusion of choline from supplements. Intakes were not higher in studies among pregnant and lactating women compared with studies in nonpregnant women. To conclude, during the childbearing years and across the globe, habitual intakes of choline from foods alone and foods and supplements combined appear to be consistently lower than the estimated adequate intakes for this target group. Urgent measures are needed to (1) improve the quality of choline data in global food composition databases, (2) encourage the reporting of choline intakes in dietary surveys, (3) raise awareness about the role(s) of choline in foetal–maternal health, and (4) consider formally advocating the use of choline supplements in women planning a pregnancy, pregnant, or lactating.
Collapse
Affiliation(s)
| | - Rima Obeid
- Department of Clinical Chemistry and Laboratory Medicine, University Hospital of the Saarland, D-66420 Homburg, Germany;
| | - Christiane Schön
- BioTeSys GmbH, Schelztorstrasse 54-56, D-73728 Esslingen, Germany;
| |
Collapse
|
27
|
Phosphatidylcholine restores neuronal plasticity of neural stem cells under inflammatory stress. Sci Rep 2021; 11:22891. [PMID: 34819604 PMCID: PMC8613233 DOI: 10.1038/s41598-021-02361-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/08/2021] [Indexed: 12/22/2022] Open
Abstract
The balances between NSCs growth and differentiation, and between glial and neuronal differentiation play a key role in brain regeneration after any pathological conditions. It is well known that the nervous tissue shows a poor recovery after injury due to the factors present in the wounded microenvironment, particularly inflammatory factors, that prevent neuronal differentiation. Thus, it is essential to generate a favourable condition for NSCs and conduct them to differentiate towards functional neurons. Here, we show that neuroinflammation has no effect on NSCs proliferation but induces an aberrant neuronal differentiation that gives rise to dystrophic, non-functional neurons. This is perhaps the initial step of brain failure associated to many neurological disorders. Interestingly, we demonstrate that phosphatidylcholine (PtdCho)-enriched media enhances neuronal differentiation even under inflammatory stress by modifying the commitment of post-mitotic cells. The pro-neurogenic effect of PtdCho increases the population of healthy normal neurons. In addition, we provide evidences that this phospholipid ameliorates the damage of neurons and, in consequence, modulates neuronal plasticity. These results contribute to our understanding of NSCs behaviour under inflammatory conditions, opening up new venues to improve neurogenic capacity in the brain.
Collapse
|
28
|
Rubini E, Baijens IMM, Horánszky A, Schoenmakers S, Sinclair KD, Zana M, Dinnyés A, Steegers-Theunissen RPM, Rousian M. Maternal One-Carbon Metabolism during the Periconceptional Period and Human Foetal Brain Growth: A Systematic Review. Genes (Basel) 2021; 12:1634. [PMID: 34681028 PMCID: PMC8535925 DOI: 10.3390/genes12101634] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/01/2021] [Accepted: 10/12/2021] [Indexed: 01/13/2023] Open
Abstract
The maternal environment during the periconceptional period influences foetal growth and development, in part, via epigenetic mechanisms moderated by one-carbon metabolic pathways. During embryonic development, one-carbon metabolism is involved in brain development and neural programming. Derangements in one-carbon metabolism increase (i) the short-term risk of embryonic neural tube-related defects and (ii) long-term childhood behaviour, cognition, and autism spectrum disorders. Here we investigate the association between maternal one-carbon metabolism and foetal and neonatal brain growth and development. Database searching resulted in 26 articles eligible for inclusion. Maternal vitamin B6, vitamin B12, homocysteine, and choline were not associated with foetal and/or neonatal head growth. First-trimester maternal plasma folate within the normal range (>17 nmol/L) associated with increased foetal head size and head growth, and high erythrocyte folate (1538-1813 nmol/L) with increased cerebellar growth, whereas folate deficiency (<7 nmol/L) associated with a reduced foetal brain volume. Preconceptional folic acid supplement use and specific dietary patterns (associated with increased B vitamins and low homocysteine) increased foetal head size. Although early pregnancy maternal folate appears to be the most independent predictor of foetal brain growth, there is insufficient data to confirm the link between maternal folate and offspring risks for neurodevelopmental diseases.
Collapse
Affiliation(s)
- Eleonora Rubini
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, 3000 CA Rotterdam, The Netherlands; (E.R.); (I.M.M.B.); (S.S.); (M.R.)
| | - Inge M. M. Baijens
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, 3000 CA Rotterdam, The Netherlands; (E.R.); (I.M.M.B.); (S.S.); (M.R.)
| | - Alex Horánszky
- Department of Physiology and Animal Health, Institute of Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, H-2100 Gödöllő, Hungary; (A.H.); (A.D.)
- BioTalentum Ltd., H-2100 Gödöllő, Hungary;
| | - Sam Schoenmakers
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, 3000 CA Rotterdam, The Netherlands; (E.R.); (I.M.M.B.); (S.S.); (M.R.)
| | - Kevin D. Sinclair
- School of Biosciences, University of Nottingham, Nottingham LE12 5RD, UK;
| | | | - András Dinnyés
- Department of Physiology and Animal Health, Institute of Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, H-2100 Gödöllő, Hungary; (A.H.); (A.D.)
- BioTalentum Ltd., H-2100 Gödöllő, Hungary;
- HCEMM-USZ Stem Cell Research Group, Hungarian Centre of Excellence for Molecular Medicine, H-6723 Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Régine P. M. Steegers-Theunissen
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, 3000 CA Rotterdam, The Netherlands; (E.R.); (I.M.M.B.); (S.S.); (M.R.)
| | - Melek Rousian
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, 3000 CA Rotterdam, The Netherlands; (E.R.); (I.M.M.B.); (S.S.); (M.R.)
| |
Collapse
|
29
|
Powers BE, Velazquez R, Strawderman MS, Ginsberg SD, Mufson EJ, Strupp BJ. Maternal Choline Supplementation as a Potential Therapy for Down Syndrome: Assessment of Effects Throughout the Lifespan. Front Aging Neurosci 2021; 13:723046. [PMID: 34690739 PMCID: PMC8527982 DOI: 10.3389/fnagi.2021.723046] [Citation(s) in RCA: 10] [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: 06/09/2021] [Accepted: 08/20/2021] [Indexed: 12/23/2022] Open
Abstract
Maternal choline supplementation (MCS) has emerged as a promising therapy to lessen the cognitive and affective dysfunction associated with Down syndrome (DS). Choline is an essential nutrient, especially important during pregnancy due to its wide-ranging ontogenetic roles. Using the Ts65Dn mouse model of DS, our group has demonstrated that supplementing the maternal diet with additional choline (4-5 × standard levels) during pregnancy and lactation improves spatial cognition, attention, and emotion regulation in the adult offspring. The behavioral benefits were associated with a rescue of septohippocampal circuit atrophy. These results have been replicated across a series of independent studies, although the magnitude of the cognitive benefit has varied. We hypothesized that this was due, at least in part, to differences in the age of the subjects at the time of testing. Here, we present new data that compares the effects of MCS on the attentional function of adult Ts65Dn offspring, which began testing at two different ages (6 vs. 12 months of age). These data replicate and extend the results of our previous reports, showing a clear pattern indicating that MCS has beneficial effects in Ts65Dn offspring throughout life, but that the magnitude of the benefit (relative to non-supplemented offspring) diminishes with aging, possibly because of the onset of Alzheimer's disease-like neuropathology. In light of growing evidence that increased maternal choline intake during pregnancy is beneficial to the cognitive and affective functioning of all offspring (e.g., neurotypical and DS), the addition of this nutrient to a prenatal vitamin regimen would be predicted to have population-wide benefits and provide early intervention for fetuses with DS, notably including babies born to mothers unaware that they are carrying a fetus with DS.
Collapse
Affiliation(s)
- Brian E. Powers
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
- Edward Hines Jr. VA Hospital, Hines, IL, United States
| | - Ramon Velazquez
- Department of Psychology, Cornell University, Ithaca, NY, United States
- Arizona State University-Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Myla S. Strawderman
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
- Department Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- New York University Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| | - Elliott J. Mufson
- Departments of Translational Neuroscience and Neurology, Barrow Neurological Institute, St. Joseph's Medical Center, Phoenix, AZ, United States
| | - Barbara J. Strupp
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
- Department of Psychology, Cornell University, Ithaca, NY, United States
| |
Collapse
|
30
|
Coley EJL, Hsiao EY. Malnutrition and the microbiome as modifiers of early neurodevelopment. Trends Neurosci 2021; 44:753-764. [PMID: 34303552 DOI: 10.1016/j.tins.2021.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/23/2021] [Accepted: 06/11/2021] [Indexed: 01/16/2023]
Abstract
Malnutrition refers to a dearth, excess, or altered differential ratios of calories, macronutrients, or micronutrients. Malnutrition, particularly during early life, is a pressing global health and socioeconomic burden that is increasingly associated with neurodevelopmental impairments. Understanding how perinatal malnutrition influences brain development is crucial to uncovering fundamental mechanisms for establishing behavioral neurocircuits, with the potential to inform public policy and clinical interventions for neurodevelopmental conditions. Recent studies reveal that the gut microbiome can mediate dietary effects on host physiology and that the microbiome modulates the development and function of the nervous system. This review discusses evidence that perinatal malnutrition alters brain development and examines the maternal and neonatal microbiome as a potential contributing factor.
Collapse
Affiliation(s)
- Elena J L Coley
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Elaine Y Hsiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
31
|
Kowluru RA, Radhakrishnan R, Mohammad G. Regulation of Rac1 transcription by histone and DNA methylation in diabetic retinopathy. Sci Rep 2021; 11:14097. [PMID: 34238980 PMCID: PMC8266843 DOI: 10.1038/s41598-021-93420-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/17/2021] [Indexed: 12/16/2022] Open
Abstract
Cytosolic ROS, generated by NADPH oxidase 2 (Nox2) in diabetes, damage retinal mitochondria, which leads to the development of retinopathy. A small molecular weight G-protein essential for Nox2 activation, Rac1, is also transcriptionally activated via active DNA methylation-hydroxymethylation. DNA methylation is a dynamic process, and can also be regulated by histone modifications; diabetes alters retinal histone methylation machinery. Our aim is to investigate the role of histone methylation (H3K9me3) of Rac1 promoter in dynamic DNA methylation- transcriptional activation. Using human retinal endothelial cells in 20 mM D-glucose, H3K9me3 at Rac1 promoter was quantified by chromatin-Immunoprecipitation technique. Crosstalk between H3K9me3 and DNA methylation was examined in cells transfected with siRNA of histone trimethyl-transferase, Suv39H1, or Dnmt1, exposed to high glucose. Key parameters were confirmed in retinal microvessels from streptozotocin-induced diabetic mice, with intravitreally administered Suv39H1-siRNA or Dnmt1-siRNA. Compared to cells in normal glucose, high glucose increased H3K9me3 and Suv39H1 binding at Rac1 promoter, and Suv39H1-siRNA prevented glucose-induced increase 5 hydroxy methyl cytosine (5hmC) and Rac1 mRNA. Similarly, in diabetic mice, Suv39H1-siRNA attenuated increase in 5hmC and Rac1 mRNA. Thus, H3K9me3 at Rac1 promoter assists in active DNA methylation-hydroxymethylation, activating Rac1 transcription. Regulation of Suv39H1-H3K9 trimethylation could prevent further epigenetic modifications, and prevent diabetic retinopathy.
Collapse
Affiliation(s)
- Renu A Kowluru
- Ophthalmology, Visual and Anatomical Sciences, Kresge Eye Institute, Wayne State University, Detroit, MI, 48201, USA.
| | - Rakesh Radhakrishnan
- Ophthalmology, Visual and Anatomical Sciences, Kresge Eye Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Ghulam Mohammad
- Ophthalmology, Visual and Anatomical Sciences, Kresge Eye Institute, Wayne State University, Detroit, MI, 48201, USA
| |
Collapse
|
32
|
Rahul DR, Ponniah RJ. The Modularity of Dyslexia. Pediatr Neonatol 2021; 62:240-248. [PMID: 33775610 DOI: 10.1016/j.pedneo.2021.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/24/2020] [Accepted: 03/05/2021] [Indexed: 10/21/2022] Open
Abstract
There is a growing interest in understanding dyslexia and the mechanisms involved in reading difficulties. Inquiries into the morphological and physiological changes of the brain have contributed to our increased understanding of reading ability and dyslexia. Similarly, inquiries into brain chemistry and reading provide a neurometabolic framework of dyslexia in terms of poor reading and phonological measures. Also, studies of the genetic etiology of reading yield substantial evidence of genes and SNPs associated with dyslexia. However, little is known about the interface between these distinct areas of knowledge. Therefore, we offer an exhaustive perspective on dyslexia using the idea of modularity by assimilating the findings and implications from the brain morphological, neurophysiological, neurochemical, genetic, and educational insights into dyslexia. We contend that this endeavor will provide a beneficial foundation for aiming at the possibilities of a holistic intervention and informed solutions for reading difficulties.
Collapse
Affiliation(s)
- D R Rahul
- Department of Humanities and Social Sciences, National Institute of Technology, Tiruchirappalli, India
| | - R Joseph Ponniah
- Department of Humanities and Social Sciences, National Institute of Technology, Tiruchirappalli, India.
| |
Collapse
|
33
|
Schieffler DA, Matta SE. Evidence to Support the Use of S-Adenosylmethionine for Treatment of Post-Concussive Sequelae in the Military. Mil Med 2021; 187:e1182-e1192. [PMID: 33900393 DOI: 10.1093/milmed/usab130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/26/2021] [Accepted: 03/26/2021] [Indexed: 11/12/2022] Open
Abstract
INTRODUCTION Since the year 2000, over 413,000 service members have sustained traumatic brain injury (TBI) and may present with post-concussive sequelae including headaches, fatigue, irritability, cognitive problems, depression, insomnia, and chronic pain. Although the focus of the article is on military TBI, the usefulness of S-adenosylmethionine (SAMe) would extend to both civilian and military populations. This narrative review examines the preclinical and clinical literature of SAMe's metabolism and alterations seen in disease states such as depressive disorders, pain disorders, fatigue, cognition, dementia, use in pregnancy and peripartum, children, adolescents, and adults, to the elderly with and without dementia, stroke, and neurodegeneration, in order to highlight its potential benefit in post-concussive sequelae after TBI. MATERIALS AND METHODS A MEDLINE/PubMed and Cochrane Database search was conducted between May 3, 2018 and July 30, 2019 by combining search terms for SAMe with terms for relevant disease states including depression, brain injury, dementia, Alzheimer's disease, Parkinson's disease, cognition, fatigue, and pain. This search retrieved a total of 676 references. 439 were excluded for being over a 10-year publication date, except where clinically relevant. After additional removal of repeated articles, the number of articles were totaled 197. An additional 59 articles were excluded: 10 not in English, 4 duplicates, 4 not original investigations, and 41 outside the scope of this article. The remaining 138 articles were used in this review and included 25 clinical studies, 46 preclinical studies, 63 reviews, and 4 case reports. RESULTS This narrative review examined the preclinical and clinical literature of SAMe's metabolism and alterations seen in MDD, pain disorders, fatigue, cognition and memory, dementia, and other disorders to highlight the potential benefit of SAMe in post-concussive sequelae in mTBI. The literature showed potential for improvement, safety, and tolerability in these symptom clusters commonly seen in military mild TBI (mTBI). CONCLUSION There is evidence of a potential benefit of SAMe as an intervention to help with symptoms across the range of post-concussive sequelae and syndromes commonly seen in military mTBI. Since the discovery of SAMe in 1952, this pleiotropic molecule has shown the significance of its involvement in several metabolic cascades in such disparate systems as epigenetics, bioenergetics, DNA methylation, neurotransmitter systems, and potential usefulness in military TBI. Significant limitations include disparate presentations seen in patients with mild TBI, those with post-concussive syndrome, as well as those with comorbid depression and posttraumatic stress disorder. Also, over-the-counter medications are not regulated and SAMe products may vary widely in price and quality. Given the potential for mania in patients with bipolar disorder, evaluation and recommendations should be made by a physician able to evaluate the underlying bipolar diathesis. Furthermore, this narrative review serves as the rationale for future open-label and double-blind placebo-controlled trials in military mTBI and SAMe.
Collapse
Affiliation(s)
| | - Sofia E Matta
- Naval Hospital Camp Pendleton, Oceanside, CA 92055, USA
| |
Collapse
|
34
|
Functional Expression of Choline Transporters in Human Neural Stem Cells and Its Link to Cell Proliferation, Cell Viability, and Neurite Outgrowth. Cells 2021; 10:cells10020453. [PMID: 33672580 PMCID: PMC7924032 DOI: 10.3390/cells10020453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 01/11/2023] Open
Abstract
Choline and choline metabolites are essential for all cellular functions. They have also been reported to be crucial for neural development. In this work, we studied the functional characteristics of the choline uptake system in human neural stem cells (hNSCs). Additionally, we investigated the effect of extracellular choline uptake inhibition on the cellular activities in hNSCs. We found that the mRNAs and proteins of choline transporter-like protein 1 (CTL1) and CTL2 were expressed at high levels. Immunostaining showed that CTL1 and CTL2 were localized in the cell membrane and partly in the mitochondria, respectively. The uptake of extracellular choline was saturable and performed by a single uptake mechanism, which was Na+-independent and pH-dependent. We conclude that CTL1 is responsible for extracellular choline uptake, and CTL2 may uptake choline in the mitochondria and be involved in DNA methylation via choline oxidation. Extracellular choline uptake inhibition caused intracellular choline deficiency in hNSCs, which suppressed cell proliferation, cell viability, and neurite outgrowth. Our findings contribute to the understanding of the role of choline in neural development as well as the pathogenesis of various neurological diseases caused by choline deficiency or choline uptake impairment.
Collapse
|
35
|
Liu X. Effects of Methyl Donors on L-Tryptophan Fermentation. Bioengineered 2021:21655979.2021.1882821. [PMID: 33522354 DOI: 10.1080/21655979.2021.1882821] [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: 12/03/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 10/22/2022] Open
Abstract
Methyl donors, a class of compounds that supply methyl groups to methyl acceptors, play important roles in the function, growth, and proliferation of cells; however, the methyl donor content in cells is not sufficient to meet their normal needs. In L-tryptophan production with E. coli, the growth and acid-producing ability of E. coli cells are weak due to the presence of exogenous plasmids that inhibit the growth of E. coli, and reduce the efficiency of exogenous gene expression. Therefore, the effect of methyl donors on L-tryptophan production was investigated. Among the methyl donors tested, choline chloride showed the most significant effect in promoting fermentation, followed by methionine. The optimum addition method involved the addition of 1.5 g/L methionine to the culture medium, combined with continuous feeding with a glucose solution containing 1 g/L choline chloride. The final tryptophan titer reached 53.5 g/L; the highest biomass of bacteria reached 51.8 g/L; and the main by-product, acetic acid, was reduced to 2.23 g/L, which had a significant impact on the fermentation results.
Collapse
Affiliation(s)
- Xiaocui Liu
- Department of Life Science of Shanxi Datong University, Datong Shanxi 037009, China
| |
Collapse
|
36
|
The Mechanisms of Improving IVF Outcomes of Liu-Wei-Di-Huang Pill Acting on DOR Patients. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:5183017. [PMID: 33178317 PMCID: PMC7648682 DOI: 10.1155/2020/5183017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 07/26/2020] [Accepted: 10/19/2020] [Indexed: 11/17/2022]
Abstract
Diminished ovarian reserve (DOR) is the weakening of ovarian oocyte production and quality. It will further become premature ovarian failure without timely cure. However, disease pathology and diagnostic markers are still incompletely understood. Liu-Wei-Di-Huang (LWDH) pill, a traditional Chinese medicine formula, is commonly used in the treatment of DOR in China. To explore the mechanism of the effect of LWDH on in vitro fertilization (IVF) outcomes in patients with DOR, a pseudotargeted metabolomics study combined with multivariate data processing strategy was carried out. A liquid chromatography tandem mass spectrometry-based metabolomics approach was applied to characterize metabolic biomarker candidates. Multiple pattern recognition was used to determine groups and confirm important variables. A total of 21 potential biomarkers were characterized, and related metabolic pathways were identified. The study displayed that the established pseudotargeted metabolomics strategy is a powerful approach for investigating the mechanism of DOR and LWDH. In addition, the approach may highlight biomarkers and metabolic pathways and can capture subtle metabolite changes from headache, which may lead to an improved mechanism understanding of DOR diseases and LWDH treatment.
Collapse
|
37
|
Interplay between Metabolism, Nutrition and Epigenetics in Shaping Brain DNA Methylation, Neural Function and Behavior. Genes (Basel) 2020; 11:genes11070742. [PMID: 32635190 PMCID: PMC7397264 DOI: 10.3390/genes11070742] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/25/2020] [Accepted: 07/02/2020] [Indexed: 12/14/2022] Open
Abstract
Gene expression in the brain is dramatically regulated by a variety of stimuli. While the role of neural activity has been extensively studied, less is known about the effects of metabolism and nutrition on transcriptional control mechanisms in the brain. Extracellular signals are integrated at the chromatin level through dynamic modifications of epigenetic marks, which in turn fine-tune gene transcription. In the last twenty years, it has become clear that epigenetics plays a crucial role in modulating central nervous system functions and finally behavior. Here, we will focus on the effect of metabolic signals in shaping brain DNA methylation, both during development and adulthood. We will provide an overview of maternal nutrition effects on brain methylation and behavior in offspring. In addition, the impact of different diet challenges on cytosine methylation dynamics in the adult brain will be discussed. Finally, the possible role played by the metabolic status in modulating DNA hydroxymethylation, which is particularly abundant in neural tissue, will be considered.
Collapse
|
38
|
Derbyshire E, Obeid R. Choline, Neurological Development and Brain Function: A Systematic Review Focusing on the First 1000 Days. Nutrients 2020; 12:E1731. [PMID: 32531929 PMCID: PMC7352907 DOI: 10.3390/nu12061731] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/22/2022] Open
Abstract
The foundations of neurodevelopment across an individual's lifespan are established in the first 1000 days of life (2 years). During this period an adequate supply of nutrients are essential for proper neurodevelopment and lifelong brain function. Of these, evidence for choline has been building but has not been widely collated using systematic approaches. Therefore, a systematic review was performed to identify the animal and human studies looking at inter-relationships between choline, neurological development, and brain function during the first 1000 days of life. The database PubMed was used, and reference lists were searched. In total, 813 publications were subject to the title/abstract review, and 38 animal and 16 human studies were included after evaluation. Findings suggest that supplementing the maternal or child's diet with choline over the first 1000 days of life could subsequently: (1) support normal brain development (animal and human evidence), (2) protect against neural and metabolic insults, particularly when the fetus is exposed to alcohol (animal and human evidence), and (3) improve neural and cognitive functioning (animal evidence). Overall, most offspring would benefit from increased choline supply during the first 1000 days of life, particularly in relation to helping facilitate normal brain development. Health policies and guidelines should consider re-evaluation to help communicate and impart potential choline benefits through diet and/or supplementation approaches across this critical life stage.
Collapse
Affiliation(s)
| | - Rima Obeid
- Department of Clinical Chemistry, University Hospital of the Saarland, Building 57, 66424 Homburg, Germany;
| |
Collapse
|
39
|
Rosenfeld CS, Hekman JP, Johnson JL, Lyu Z, Ortega MT, Joshi T, Mao J, Vladimirova AV, Gulevich RG, Kharlamova AV, Acland GM, Hecht EE, Wang X, Clark AG, Trut LN, Behura SK, Kukekova AV. Hypothalamic transcriptome of tame and aggressive silver foxes (Vulpes vulpes) identifies gene expression differences shared across brain regions. GENES BRAIN AND BEHAVIOR 2019; 19:e12614. [PMID: 31605445 DOI: 10.1111/gbb.12614] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 12/15/2022]
Abstract
The underlying neurological events accompanying dog domestication remain elusive. To reconstruct the domestication process in an experimental setting, silver foxes (Vulpes vulpes) have been deliberately bred for tame vs aggressive behaviors for more than 50 generations at the Institute for Cytology and Genetics in Novosibirsk, Russia. The hypothalamus is an essential part of the hypothalamic-pituitary-adrenal axis and regulates the fight-or-flight response, and thus, we hypothesized that selective breeding for tameness/aggressiveness has shaped the hypothalamic transcriptomic profile. RNA-seq analysis identified 70 differentially expressed genes (DEGs). Seven of these genes, DKKL1, FBLN7, NPL, PRIMPOL, PTGRN, SHCBP1L and SKIV2L, showed the same direction expression differences in the hypothalamus, basal forebrain and prefrontal cortex. The genes differentially expressed across the three tissues are involved in cell division, differentiation, adhesion and carbohydrate processing, suggesting an association of these processes with selective breeding. Additionally, 159 transcripts from the hypothalamus demonstrated differences in the abundance of alternative spliced forms between the tame and aggressive foxes. Weighted gene coexpression network analyses also suggested that gene modules in hypothalamus were significantly associated with tame vs aggressive behavior. Pathways associated with these modules include signal transduction, interleukin signaling, cytokine-cytokine receptor interaction and peptide ligand-binding receptors (eg, G-protein coupled receptor [GPCR] ligand binding). Current studies show the selection for tameness vs aggressiveness in foxes is associated with unique hypothalamic gene profiles partly shared with other brain regions and highlight DEGs involved in biological processes such as development, differentiation and immunological responses. The role of these processes in fox and dog domestication remains to be determined.
Collapse
Affiliation(s)
- Cheryl S Rosenfeld
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri.,Biomedical Sciences, University of Missouri, Columbia, Missouri.,Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, Missouri.,MU Informatics Institute, University of Missouri, Columbia, Missouri
| | - Jessica P Hekman
- Department of Animal Sciences, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois, Urbana, Illinois.,The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Jennifer L Johnson
- Department of Animal Sciences, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois, Urbana, Illinois
| | - Zhen Lyu
- Department of Computer Science, University of Missouri, Columbia, Missouri
| | - Madison T Ortega
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri.,Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Trupti Joshi
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri.,MU Informatics Institute, University of Missouri, Columbia, Missouri.,Department of Computer Science, University of Missouri, Columbia, Missouri.,Department of Health Management and Informatics, University of Missouri, Columbia, Missouri
| | - Jiude Mao
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri.,Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Anastasiya V Vladimirova
- The Laboratory of Evolutionary Genetics, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Rimma G Gulevich
- The Laboratory of Evolutionary Genetics, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Anastasiya V Kharlamova
- The Laboratory of Evolutionary Genetics, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Gregory M Acland
- Baker Institute for Animal Health, Cornell University, College of Veterinary Medicine, Ithaca, New York
| | - Erin E Hecht
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts
| | - Xu Wang
- Department of Pathobiology, Auburn University, College of Veterinary Medicine, Auburn, Alabama
| | - Andrew G Clark
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York
| | - Lyudmila N Trut
- The Laboratory of Evolutionary Genetics, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Susanta K Behura
- MU Informatics Institute, University of Missouri, Columbia, Missouri.,Division of Animal Sciences, University of Missouri, Columbia, Missouri
| | - Anna V Kukekova
- Department of Animal Sciences, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois, Urbana, Illinois
| |
Collapse
|
40
|
Bekdash RA. Neuroprotective Effects of Choline and Other Methyl Donors. Nutrients 2019; 11:nu11122995. [PMID: 31817768 PMCID: PMC6950346 DOI: 10.3390/nu11122995] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/20/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022] Open
Abstract
Recent evidence suggests that physical and mental health are influenced by an intricate interaction between genes and environment. Environmental factors have been shown to modulate neuronal gene expression and function by epigenetic mechanisms. Exposure to these factors including nutrients during sensitive periods of life could program brain development and have long-lasting effects on mental health. Studies have shown that early nutritional intervention that includes methyl-donors improves cognitive functions throughout life. Choline is a micronutrient and a methyl donor that is required for normal brain growth and development. It plays a pivotal role in maintaining structural and functional integrity of cellular membranes. It also regulates cholinergic signaling in the brain via the synthesis of acetylcholine. Via its metabolites, it participates in pathways that regulate methylation of genes related to memory and cognitive functions at different stages of development. Choline-related functions have been dysregulated in some neurodegenerative diseases suggesting choline role in influencing mental health across the lifespan.
Collapse
Affiliation(s)
- Rola A Bekdash
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| |
Collapse
|
41
|
Choline: Exploring the Growing Science on Its Benefits for Moms and Babies. Nutrients 2019; 11:nu11081823. [PMID: 31394787 PMCID: PMC6722688 DOI: 10.3390/nu11081823] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/01/2019] [Accepted: 08/05/2019] [Indexed: 12/23/2022] Open
Abstract
The importance of ensuring adequate choline intakes during pregnancy is increasingly recognized. Choline is critical for a number of physiological processes during the prenatal period with roles in membrane biosynthesis and tissue expansion, neurotransmission and brain development, and methyl group donation and gene expression. Studies in animals and humans have shown that supplementing the maternal diet with additional choline improves several pregnancy outcomes and protects against certain neural and metabolic insults. Most pregnant women in the U.S. are not achieving choline intake recommendations of 450 mg/day and would likely benefit from boosting their choline intakes through dietary and/or supplemental approaches.
Collapse
|
42
|
Trujillo-Gonzalez I, Friday WB, Munson CA, Bachleda A, Weiss ER, Alam NM, Sha W, Zeisel SH, Surzenko N. Low availability of choline in utero disrupts development and function of the retina. FASEB J 2019; 33:9194-9209. [PMID: 31091977 PMCID: PMC6662989 DOI: 10.1096/fj.201900444r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022]
Abstract
Adequate supply of choline, an essential nutrient, is necessary to support proper brain development. Whether prenatal choline availability plays a role in development of the visual system is currently unknown. In this study, we addressed the role of in utero choline supply for the development and later function of the retina in a mouse model. We lowered choline availability in the maternal diet during pregnancy and assessed proliferative and differentiation properties of retinal progenitor cells (RPCs) in the developing prenatal retina, as well as visual function in adult offspring. We report that low choline availability during retinogenesis leads to persistent retinal cytoarchitectural defects, ranging from focal lesions with displacement of retinal neurons into subretinal space to severe hypocellularity and ultrastructural defects in photoreceptor organization. We further show that low choline availability impairs timely differentiation of retinal neuronal cells, such that the densities of early-born retinal ganglion cells, amacrine and horizontal cells, as well as cone photoreceptor precursors, are reduced in low choline embryonic d 17.5 retinas. Maintenance of higher proportions of RPCs that fail to exit the cell cycle underlies aberrant neuronal differentiation in low choline embryos. Increased RPC cell cycle length, and associated reduction in neurofibromin 2/Merlin protein, an upstream regulator of the Hippo signaling pathway, at least in part, explain aberrant neurogenesis in low choline retinas. Furthermore, we find that animals exposed to low choline diet in utero exhibit a significant degree of intraindividual variation in vision, characterized by marked functional discrepancy between the 2 eyes in individual animals. Together, our findings demonstrate, for the first time, that choline availability plays an essential role in the regulation of temporal progression of retinogenesis and provide evidence for the importance of adequate supply of choline for proper development of the visual system.-Trujillo-Gonzalez, I., Friday, W. B., Munson, C. A., Bachleda, A., Weiss, E. R., Alam, N. M., Sha, W., Zeisel, S. H., Surzenko, N. Low availability of choline in utero disrupts development and function of the retina.
Collapse
Affiliation(s)
- Isis Trujillo-Gonzalez
- Nutrition Research Institute, University of North Carolina–Chapel Hill, Kannapolis, North Carolina, USA
| | - Walter B. Friday
- Nutrition Research Institute, University of North Carolina–Chapel Hill, Kannapolis, North Carolina, USA
| | - Carolyn A. Munson
- Nutrition Research Institute, University of North Carolina–Chapel Hill, Kannapolis, North Carolina, USA
| | - Amelia Bachleda
- Department of Cell Biology and Physiology, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ellen R. Weiss
- Department of Cell Biology and Physiology, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nazia M. Alam
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, USA
- Center for Visual Restoration, Burke Neurological Institute, White Plains, New York, USA
| | - Wei Sha
- Bioinformatics Services Division, University of North Carolina–Charlotte, Kannapolis, North Carolina, USA
| | - Steven H. Zeisel
- Nutrition Research Institute, University of North Carolina–Chapel Hill, Kannapolis, North Carolina, USA
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, USA
| | - Natalia Surzenko
- Nutrition Research Institute, University of North Carolina–Chapel Hill, Kannapolis, North Carolina, USA
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
43
|
Abstract
Appraising success in meeting the world's nutritional needs has largely focused on infant mortality and anthropometric measurements with an emphasis on the first 1,000 days (conception to approximately age 2 years). This ignores the unique nutritional needs of the human brain. Although the intrauterine environment and the early postnatal years are important, equally critical periods follow during which the brain's intricate wiring is established for a lifetime of experience-driven remodeling. At the peak of this process during childhood, the human brain may account for 50% of the body's basal nutritional requirement. Thus, the consequences of proper nutritional management of the brain play out over a lifetime. Our motivation in preparing this review was to move the human brain into a more central position in the planning of nutritional programs. Here we review the macro- and micronutrient requirements of the human brain and how they are delivered, from conception to adulthood.
Collapse
Affiliation(s)
- Manu S. Goyal
- Mallinckrodt Institute of Radiology and Department of Neurology, Washington University School of Medicine, Washington University, St. Louis, Missouri 63130, USA
| | - Lora L. Iannotti
- Brown School, Institute for Public Health, Washington University, St. Louis, Missouri 63130, USA
| | - Marcus E. Raichle
- Mallinckrodt Institute of Radiology and Department of Neurology, Washington University School of Medicine, Washington University, St. Louis, Missouri 63130, USA
| |
Collapse
|
44
|
Hippocampus-dependent memory and allele-specific gene expression in adult offspring of alcohol-consuming dams after neonatal treatment with thyroxin or metformin. Mol Psychiatry 2018; 23:1643-1651. [PMID: 28727687 PMCID: PMC5775940 DOI: 10.1038/mp.2017.129] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 02/06/2023]
Abstract
Fetal alcohol spectrum disorder (FASD), the result of fetal alcohol exposure (FAE), affects 2-11% of children worldwide, with no effective treatments. Hippocampus-based learning and memory deficits are key symptoms of FASD. Our previous studies show hypothyroxinemia and hyperglycemia of the alcohol-consuming pregnant rat, which likely affects fetal neurodevelopment. We administered vehicle, thyroxine (T4) or metformin to neonatal rats post FAE and rats were tested in the hippocampus-dependent contextual fear-conditioning paradigm in adulthood. Both T4 and metformin alleviated contextual fear memory deficit induced by FAE, and reversed the hippocampal expression changes in the thyroid hormone-inactivating enzyme, deiodinase-III (Dio3) and insulin-like growth factor 2 (Igf2), genes that are known to modulate memory processes. Neonatal T4 restored maternal allelic expressions of the imprinted Dio3 and Igf2 in the adult male hippocampus, while metformin restored FAE-caused changes in Igf2 expression only. The decreased hippocampal expression of DNA methyltransferase 1 (Dnmt1) that maintains the imprinting of Dio3 and Igf2 during development was normalized by both treatments. Administering Dnmt1 inhibitor to control neonates resulted in FAE-like deficits in fear memory and hippocampal allele-specific expression of Igf2, which were reversed by metformin. We propose that neonatal administration of T4 and metformin post FAE affect memory via elevating Dnmt1 and consequently normalizing hippocampal Dio3 and Igf2 expressions in the adult offspring. The present results indicate that T4 and metformin, administered during the neonatal period that is equivalent to the third trimester of human pregnancy, are potential treatments for FASD and conceivably for other neurodevelopmental disorders with cognitive deficits.
Collapse
|
45
|
Degroote S, Hunting D, Takser L. Periconceptional folate deficiency leads to autism-like traits in Wistar rat offspring. Neurotoxicol Teratol 2018; 66:132-138. [PMID: 29305196 DOI: 10.1016/j.ntt.2017.12.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/13/2017] [Accepted: 12/26/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND Folates in their role as key one carbon donors, are essential for two major pathways: the synthesis of DNA and RNA precursors and DNA methylation. A growing body of evidence from epidemiological studies indicates a possible association between nutritional and functional deficiency in folates and Autism Spectrum Disorders (ASD). However, there are no available behavioral animal studies on periconceptional one‑carbon donor deficiency during gestation and the autistic phenotype. OBJECTIVE The objective of this study was to determine if the periconceptional alteration of one‑carbon metabolism induced with a folate deficient diet would affect the behaviour of rat offspring. METHODS Female Wistar rats were divided in two groups: control (basal diet, in compliance with standards of regular laboratory diets), or exposed during one month before breeding until Gestational Day 15 to a modified diet with no added folic acid (0.2mg/kg of food), reduced choline (750mg/kg of food), and added 1% SST (a non-absorbable antibiotic used to inhibit folate synthesis by gut bacteria). We administered behavioral tests to offspring, i.e., open field (P20), social interactions (P25), marble burying (P30), elevated plus maze (P35), and prepulse inhibition of the acoustic startle reflex (sensorimotor gating) (P45). Blood homocysteine levels were used to confirm the deficit in one‑carbon donors. RESULTS Compared to controls, offspring with the periconceptional deficit in folate showed: (i) congenital body malformations; (ii) reduced social interactions, with a ~30% decrease in social sniffing behavior; (iii) reduced exploration of the open arm by 50% in the elevated plus maze test, indicating increased anxiety; (iv) a ~160% increased number of marbles buried, indicating repetitive behaviors; and (v) altered sensorimotor gating, with a global 50% decrease in startle inhibition. CONCLUSION Maternal periconceptional deficit in folate provokes alterations in the behavior of offspring relevant to the autistic-like phenotype.
Collapse
Affiliation(s)
- Stéphanie Degroote
- Département de Pédiatrie, Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, Québec, Canada
| | - Darel Hunting
- Département de médecine nucléaire et radiobiologie, Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, Québec, Canada
| | - Larissa Takser
- Département de Pédiatrie, Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, Québec, Canada.
| |
Collapse
|
46
|
Iannotti LL, Lutter CK, Waters WF, Gallegos Riofrío CA, Malo C, Reinhart G, Palacios A, Karp C, Chapnick M, Cox K, Aguirre S, Narvaez L, López F, Sidhu R, Kell P, Jiang X, Fujiwara H, Ory DS, Young R, Stewart CP. Eggs early in complementary feeding increase choline pathway biomarkers and DHA: a randomized controlled trial in Ecuador. Am J Clin Nutr 2017; 106:1482-1489. [PMID: 29092879 PMCID: PMC5698841 DOI: 10.3945/ajcn.117.160515] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 10/03/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Choline status has been associated with stunting among young children. Findings from this study showed that an egg intervention improved linear growth by a length-for-age z score of 0.63.Objective: We aimed to test the efficacy of eggs introduced early in complementary feeding on plasma concentrations of biomarkers in choline pathways, vitamins B-12 and A, and essential fatty acids.Design: A randomized controlled trial, the Lulun ("egg" in Kichwa) Project, was conducted in a rural indigenous population of Ecuador. Infants aged 6-9 mo were randomly assigned to treatment (1 egg/d for 6 mo; n = 80) and control (no intervention; n = 83) groups. Socioeconomic data, anthropometric measures, and blood samples were collected at baseline and endline. Household visits were made weekly for morbidity surveillance. We tested vitamin B-12 plasma concentrations by using chemiluminescent competitive immunoassay and plasma concentrations of choline, betaine, dimethylglycine, retinol, essential fatty acids, methionine, dimethylamine (DMA), trimethylamine, and trimethylamine-N-oxide (TMAO) with the use of liquid chromatography-tandem mass spectrometry.Results: Socioeconomic factors and biomarker concentrations were comparable at baseline. Of infants, 11.4% were vitamin B-12 deficient and 31.7% marginally deficient at baseline. In adjusted generalized linear regression modeling, the egg intervention increased plasma concentrations compared with control by the following effect sizes: choline, 0.35 (95% CI: 0.12, 0.57); betaine, 0.29 (95% CI: 0.01, 0.58); methionine, 0.31 (95% CI: 0.03, 0.60); docosahexaenoic acid, 0.43 (95% CI: 0.13, 0.73); DMA, 0.37 (95% CI: 0.37, 0.69); and TMAO, 0.33 (95% CI: 0.08, 0.58). No significant group differences were found for vitamin B-12, retinol, linoleic acid (LA), α-linolenic acid (ALA), or ratios of betaine to choline and LA to ALA.Conclusion: The findings supported our hypothesis that early introduction of eggs significantly improved choline and other markers in its methyl group metabolism pathway. This trial was registered at clinicaltrials.gov as NCT02446873.
Collapse
Affiliation(s)
| | - Chessa K Lutter
- School of Public Health, University of Maryland, College Park, MD;,RTI International, Research Triangle Park, NC
| | - William F Waters
- Institute for Research in Health and Nutrition, Universidad San Francisco de Quito, Quito, Ecuador
| | - Carlos Andres Gallegos Riofrío
- Brown School, Institute for Public Health, and,Institute for Research in Health and Nutrition, Universidad San Francisco de Quito, Quito, Ecuador
| | - Carla Malo
- Institute for Research in Health and Nutrition, Universidad San Francisco de Quito, Quito, Ecuador
| | - Gregory Reinhart
- The Mathile Institute for the Advancement of Human Nutrition, Dayton, OH
| | - Ana Palacios
- The Mathile Institute for the Advancement of Human Nutrition, Dayton, OH;,Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX
| | - Celia Karp
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | | | | | | | - Luis Narvaez
- NETLAB Laboratorios Especializados, Quito, Ecuador; and
| | | | - Rohini Sidhu
- Diabetic Cardiovascular Disease Center, Washington University in St. Louis, St. Louis, MO
| | - Pamela Kell
- Diabetic Cardiovascular Disease Center, Washington University in St. Louis, St. Louis, MO
| | - Xuntian Jiang
- Diabetic Cardiovascular Disease Center, Washington University in St. Louis, St. Louis, MO
| | - Hideji Fujiwara
- Diabetic Cardiovascular Disease Center, Washington University in St. Louis, St. Louis, MO
| | - Daniel S Ory
- Diabetic Cardiovascular Disease Center, Washington University in St. Louis, St. Louis, MO
| | - Rebecca Young
- Department of Nutrition, University of California, Davis, Davis, CA
| | | |
Collapse
|
47
|
Neuroprotective Actions of Dietary Choline. Nutrients 2017; 9:nu9080815. [PMID: 28788094 PMCID: PMC5579609 DOI: 10.3390/nu9080815] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/21/2017] [Accepted: 07/25/2017] [Indexed: 12/22/2022] Open
Abstract
Choline is an essential nutrient for humans. It is a precursor of membrane phospholipids (e.g., phosphatidylcholine (PC)), the neurotransmitter acetylcholine, and via betaine, the methyl group donor S-adenosylmethionine. High choline intake during gestation and early postnatal development in rat and mouse models improves cognitive function in adulthood, prevents age-related memory decline, and protects the brain from the neuropathological changes associated with Alzheimer’s disease (AD), and neurological damage associated with epilepsy, fetal alcohol syndrome, and inherited conditions such as Down and Rett syndromes. These effects of choline are correlated with modifications in histone and DNA methylation in brain, and with alterations in the expression of genes that encode proteins important for learning and memory processing, suggesting a possible epigenomic mechanism of action. Dietary choline intake in the adult may also influence cognitive function via an effect on PC containing eicosapentaenoic and docosahexaenoic acids; polyunsaturated species of PC whose levels are reduced in brains from AD patients, and is associated with higher memory performance, and resistance to cognitive decline.
Collapse
|
48
|
Metabolic profiling of stages of healthy pregnancy in Hu sheep using nuclear magnetic resonance (NMR). Theriogenology 2017; 92:121-128. [DOI: 10.1016/j.theriogenology.2017.01.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 01/12/2017] [Accepted: 01/15/2017] [Indexed: 01/03/2023]
|
49
|
Bahous RH, Jadavji NM, Deng L, Cosín-Tomás M, Lu J, Malysheva O, Leung KY, Ho MK, Pallàs M, Kaliman P, Greene ND, Bedell BJ, Caudill MA, Rozen R. High dietary folate in pregnant mice leads to pseudo-MTHFR deficiency and altered methyl metabolism, with embryonic growth delay and short-term memory impairment in offspring. Hum Mol Genet 2017; 26:888-900. [PMID: 28069796 PMCID: PMC5409086 DOI: 10.1093/hmg/ddx004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/06/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022] Open
Abstract
Methylenetetrahydrofolate reductase (MTHFR) generates methyltetrahydrofolate for methylation reactions. Severe MTHFR deficiency results in homocystinuria and neurologic impairment. Mild MTHFR deficiency (677C > T polymorphism) increases risk for complex traits, including neuropsychiatric disorders. Although low dietary folate impacts brain development, recent concerns have focused on high folate intake following food fortification and increased vitamin use. Our goal was to determine whether high dietary folate during pregnancy affects brain development in murine offspring. Female mice were placed on control diet (CD) or folic acid-supplemented diet (FASD) throughout mating, pregnancy and lactation. Three-week-old male pups were evaluated for motor and cognitive function. Tissues from E17.5 embryos, pups and dams were collected for choline/methyl metabolite measurements, immunoblotting or gene expression of relevant enzymes. Brains were examined for morphology of hippocampus and cortex. Pups of FASD mothers displayed short-term memory impairment, decreased hippocampal size and decreased thickness of the dentate gyrus. MTHFR protein levels were reduced in FASD pup livers, with lower concentrations of phosphocholine and glycerophosphocholine in liver and hippocampus, respectively. FASD pup brains showed evidence of altered acetylcholine availability and Dnmt3a mRNA was reduced in cortex and hippocampus. E17.5 embryos and placentas from FASD dams were smaller. MTHFR protein and mRNA were reduced in embryonic liver, with lower concentrations of choline, betaine and phosphocholine. Embryonic brain displayed altered development of cortical layers. In summary, high folate intake during pregnancy leads to pseudo-MTHFR deficiency, disturbed choline/methyl metabolism, embryonic growth delay and memory impairment in offspring. These findings highlight the unintended negative consequences of supplemental folic acid.
Collapse
Affiliation(s)
- Renata H. Bahous
- Departments of Human Genetics and Pediatrics, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Nafisa M. Jadavji
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Liyuan Deng
- Departments of Human Genetics and Pediatrics, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Marta Cosín-Tomás
- Pharmacology Unit, Faculty of Pharmacy, Institut de Neurociència Universitat de Barcelona (IBUB), Nucli Universitari de Pedralbes, Barcelona, Spain
| | - Jessica Lu
- Departments of Human Genetics and Pediatrics, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Olga Malysheva
- Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY, USA
| | - Kit-Yi Leung
- Developmental Biology and Cancer Programme, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Ming-Kai Ho
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Mercè Pallàs
- Pharmacology Unit, Faculty of Pharmacy, Institut de Neurociència Universitat de Barcelona (IBUB), Nucli Universitari de Pedralbes, Barcelona, Spain
| | - Perla Kaliman
- Institute of Biomedical Investigation of Barcelona, Spanish National Research Council, Barcelona, Spain
- Center for Mind and Brain, University of California Davis, Davis, CA, USA
| | - Nicholas D.E. Greene
- Developmental Biology and Cancer Programme, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Barry J. Bedell
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Marie A. Caudill
- Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY, USA
| | - Rima Rozen
- Departments of Human Genetics and Pediatrics, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| |
Collapse
|
50
|
Mellott TJ, Huleatt OM, Shade BN, Pender SM, Liu YB, Slack BE, Blusztajn JK. Perinatal Choline Supplementation Reduces Amyloidosis and Increases Choline Acetyltransferase Expression in the Hippocampus of the APPswePS1dE9 Alzheimer's Disease Model Mice. PLoS One 2017; 12:e0170450. [PMID: 28103298 PMCID: PMC5245895 DOI: 10.1371/journal.pone.0170450] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 01/05/2017] [Indexed: 12/27/2022] Open
Abstract
Prevention of Alzheimer's disease (AD) is a major goal of biomedical sciences. In previous studies we showed that high intake of the essential nutrient, choline, during gestation prevented age-related memory decline in a rat model. In this study we investigated the effects of a similar treatment on AD-related phenotypes in a mouse model of AD. We crossed wild type (WT) female mice with hemizygous APPswe/PS1dE9 (APP.PS1) AD model male mice and maintained the pregnant and lactating dams on a control AIN76A diet containing 1.1 g/kg of choline or a choline-supplemented (5 g/kg) diet. After weaning all offspring consumed the control diet. As compared to APP.PS1 mice reared on the control diet, the hippocampus of the perinatally choline-supplemented APP.PS1 mice exhibited: 1) altered levels of amyloid precursor protein (APP) metabolites-specifically elevated amounts of β-C-terminal fragment (β-CTF) and reduced levels of solubilized amyloid Aβ40 and Aβ42 peptides; 2) reduced number and total area of amyloid plaques; 3) preserved levels of choline acetyltransferase protein (CHAT) and insulin-like growth factor II (IGF2) and 4) absence of astrogliosis. The data suggest that dietary supplementation of choline during fetal development and early postnatal life may constitute a preventive strategy for AD.
Collapse
Affiliation(s)
- Tiffany J. Mellott
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| | - Olivia M. Huleatt
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Bethany N. Shade
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Sarah M. Pender
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Yi B. Liu
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Barbara E. Slack
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jan K. Blusztajn
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|