1
|
Tyagi SC. A High-Fat Diet Induces Epigenetic 1-Carbon Metabolism, Homocystinuria, and Renal-Dependent HFpEF. Nutrients 2025; 17:216. [PMID: 39861346 PMCID: PMC11767380 DOI: 10.3390/nu17020216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Chronic gut dysbiosis due to a high-fat diet (HFD) instigates cardiac remodeling and heart failure with preserved ejection fraction (HFpEF), in particular, kidney/volume-dependent HFpEF. Studies report that although mitochondrial ATP citrate lyase (ACLY) supports cardiac function, it decreases more in human HFpEF than HFrEF. Interestingly, ACLY synthesizes lipids and creates hyperlipidemia. Epigenetically, ACLY acetylates histone. The mechanism(s) are largely unknown. METHODS/RESULTS One hypothesis is that an HFD induces epigenetic folate 1-carbon metabolism (FOCM) and homocystinuria. This abrogates dipping in sleep-time blood pressure and causes hypertension and morning heart attacks. We observed that probiotics/lactobacillus utilize fat/lipids post-biotically, increasing mitochondrial bioenergetics and attenuating HFpEF. We suggest novel and paradigm-shift epigenetic mitochondrial sulfur trans-sulfuration pathways that selectively target HFD-induced HFpEF. Previous studies from our laboratory, using a single-cell analysis, revealed an increase in the transporter (SLC25A) of s-adenosine-methionine (SAM) during elevated levels of homocysteine (Hcy, i.e., homocystinuria, HHcy), a consequence of impaired epigenetic recycling of Hcy back to methionine due to an increase in the FOCM methylation of H3K4, K9, H4K20, and gene writer (DNMT) and decrease in eraser (TET/FTO). Hcy is transported to mitochondria by SLC7A for clearance via sulfur metabolomic trans-sulfuration by 3-mercaptopyruvate sulfur transferase (3MST). CONCLUSIONS We conclude that gut dysbiosis due to HFD disrupts rhythmic epigenetic memory via FOCM and increases in DNMT1 and creates homocystinuria, leading to a decrease in mitochondrial trans-sulfuration and bioenergetics. The treatment with lactobacillus metabolites fat/lipids post-biotically and bi-directionally produces folic acid and lactone-ketone body that mitigates the HFD-induced mitochondrial remodeling and HFpEF.
Collapse
Affiliation(s)
- Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
2
|
Saferin N, Haseeb I, Taha AM, Beecroft SE, Pillai S, Neifer AE, Lakkuru R, Kistler BP, Nawor CN, Malik I, Hasan D, Carlson JA, Zade KK, Dressel SP, Carney EM, Shah R, Gautam S, Vergis J, Neifer KL, Johnson ZV, Gustison ML, Hall FS, Burkett JP. Folate prevents the autism-related phenotype caused by developmental pyrethroid exposure in prairie voles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625285. [PMID: 39651146 PMCID: PMC11623627 DOI: 10.1101/2024.11.25.625285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Neurodevelopmental disorders (NDDs) have dramatically increased in prevalence to an alarming one in six children, and yet both causes and preventions remain elusive. Recent human epidemiology and animal studies have implicated developmental exposure to pyrethroid pesticides, one of the most common classes of pesticides in the US, as an environmental risk factor for autism and neurodevelopmental disorders. Our previous research has shown that low-dose chronic developmental pyrethroid exposure (DPE) changes folate metabolites in the adult mouse brain. We hypothesize that DPE acts directly on molecular targets in the folate metabolism pathway, and that high-dose maternal folate supplementation can prevent or reduce the biobehavioral effects of DPE. We exposed pregnant prairie vole dams chronically to vehicle or low-dose deltamethrin (3 mg/kg/3 days) with or without high-dose folate supplementation (methylfolate, 5 mg/kg/3 days). The resulting DPE offspring showed broad deficits in five behavioral domains relevant to neurodevelopmental disorders (including the social domain); increased plasma folate concentrations; and increased neural expression of SHMT1, a folate cycle enzyme. Maternal folate supplementation prevented most of the behavioral phenotypes (except for repetitive behaviors) and caused potentially compensatory changes in neural expression of FOLR1 and MTHFR, two folate-related proteins. We conclude that DPE causes neurodevelopmental disorder-relevant behavioral deficits; DPE directly alters aspects of folate metabolism; and preventative interventions targeting folate metabolism are effective in reducing, but not eliminating, the behavioral effects of DPE.
Collapse
Affiliation(s)
- Nilanjana Saferin
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ibrahim Haseeb
- College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH 43606
| | - Adam M. Taha
- Department of Pharmacology and Experimental Therapeutics, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, OH, USA
| | - Sarah E. Beecroft
- Department of Pharmacology and Experimental Therapeutics, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, OH, USA
| | - Sangeetha Pillai
- College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH 43606
| | - Asha E. Neifer
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Rudhasri Lakkuru
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Brian P. Kistler
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Charlotte N. Nawor
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Isa Malik
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Dena Hasan
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Jonathan A. Carlson
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Kareem K. Zade
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Sydnee P. Dressel
- Department of Pharmacology and Experimental Therapeutics, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, OH, USA
| | - Eileen M. Carney
- Department of Pharmacology and Experimental Therapeutics, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, OH, USA
| | - Radha Shah
- College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH 43606
| | - Shudhant Gautam
- College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH 43606
| | - John Vergis
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Kari L. Neifer
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Zachary V. Johnson
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA; Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Morgan L. Gustison
- Department of Psychology, The University of Western Ontario, London, ON, Canada (current); Department of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| | - F. Scott Hall
- Department of Pharmacology and Experimental Therapeutics, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, OH, USA
| | - James P. Burkett
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
3
|
Tyagi SC. Lactobacillus Eats Amyloid Plaque and Post-Biotically Attenuates Senescence Due to Repeat Expansion Disorder and Alzheimer's Disease. Antioxidants (Basel) 2024; 13:1225. [PMID: 39456478 PMCID: PMC11506100 DOI: 10.3390/antiox13101225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Patients with Alzheimer's disease and related dementia (ADRD) are faced with a formidable challenge of focal amyloid deposits and cerebral amyloid angiopathy (CAA). The treatment of amyloid deposits in ADRD by targeting only oxidative stress, inflammation and hyperlipidemia has not yielded significant positive clinical outcomes. The chronic high-fat diet (HFD), or gut dysbiosis, is one of the major contributors of ADRD in part by disrupted transport, epigenetic DNMT1 and the folate 1-carbon metabolism (FOCM) cycle, i.e., rhythmic methylation/de-methylation on DNA, an active part of epigenetic memory during genes turning off and on by the gene writer (DNMT1) and eraser (TET2/FTO) and the transsulfuration pathway by mitochondrial 3-mercaptopyruvate sulfur transferase (3MST)-producing H2S. The repeat CAG expansion and m6A disorder causes senescence and AD. We aim to target the paradigm-shift pathway of the gut-brain microbiome axis that selectively inhibits amyloid deposits and increases mitochondrial transsulfuration and H2S. We have observed an increase in DNMT1 and decreased FTO levels in the cortex of the brain of AD mice. Interestingly, we also observed that probiotic lactobacillus-producing post-biotic folate and lactone/ketone effectively prevented FOCM-associated gut dysbiosis and amyloid deposits. The s-adenosine-methionine (SAM) transporter (SLC25A) was increased by hyperhomocysteinemia (HHcy). Thus, we hypothesize that chronic gut dysbiosis induces SLC25A, the gene writer, and HHcy, and decreases the gene eraser, leading to a decrease in SLC7A and mitochondrial transsulfuration H2S production and bioenergetics. Lactobacillus engulfs lipids/cholesterol and a tri-directional post-biotic, folic acid (an antioxidant and inhibitor of beta amyloid deposits; reduces Hcy levels), and the lactate ketone body (fuel for mitochondria) producer increases SLC7A and H2S (an antioxidant, potent vasodilator and neurotransmitter gas) production and inhibits amyloid deposits. Therefore, it is important to discuss whether lactobacillus downregulates SLC25A and DNMT1 and upregulates TET2/FTO, inhibiting β-amyloid deposits by lowering homocysteine. It is also important to discuss whether lactobacillus upregulates SLC7A and inhibits β-amyloid deposits by increasing the mitochondrial transsulfuration of H2S production.
Collapse
Affiliation(s)
- Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
4
|
Razavinia F, Ebrahimiyan A, Faal Siahkal S, Ghazinezhad N, Abedi P. Vitamins B 9 and B 12 in children with attention deficit hyperactivity disorder (ADHD). INT J VITAM NUTR RES 2024; 94:476-484. [PMID: 38904980 DOI: 10.1024/0300-9831/a000809] [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] [Indexed: 06/22/2024]
Abstract
Background: Attention deficit hyperactivity disorder (ADHD) is a common childhood neurodevelopmental disorder that begins before age 12. Given the role of B group vitamins in cell metabolism, synthesis of nucleotides, and neurotransmitters, the present study systematically investigated the plasma levels of vitamins B9 and B12 in children with ADHD. Methods: We searched electronic databases including Web of Science, MEDLINE, EMBASE, Scopus, Iran MEDEX, Cochran database, and SID from conception to June 2023. Full-text case-control or cross-sectional studies were included in this study. Participants in the case group were children with ADHD aged 6-12 years. Review Manager Software (RevMan 5.4) was used for statistical analyses. Standardized mean differences (SMD) with 95% CIs were used to determine the differences between the two groups. Results: Six studies were included in the present meta-analysis. They included 982 children, of whom, 204 were girls and 744 were boys. The mean age of the children was 8.86±2.03 years. The level of vitamin B9 was significantly different between children with and without ADHD [SMD -0.80, 95% CI (-1.55, -0.04)]. Vitamin B12 was significantly lower in children with ADHD [SMD -0.29, 95% CI (-0.42, -0.16)]. However, due to high heterogeneity (I2 = 93%), sensitivity analysis was used, I2 fell to 21%, and significant difference was observed between the two groups [SMD -0.19, 95% CI (-0.34, -0.04)]. Conclusion: The results of this systematic review showed that the level of vitamins B9 and B12 in children with ADHD was significantly lower than that in healthy children.
Collapse
Affiliation(s)
- Fatemeh Razavinia
- Midwifery Department, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Atefeh Ebrahimiyan
- Midwifery Department, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shahla Faal Siahkal
- Department of Midwifery, Marand Branch, Islamic Azad University, Marand, Iran
| | - Neda Ghazinezhad
- Faculty of Medical Sciences, Department of Reproductive Health and Midwifery, Tarbiat Modares University, Tehran, Iran
| | - Parvin Abedi
- Midwifery Department, Menopause Andropause Research Center, Ahvaz Jundisahpur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
5
|
Sfakianoudis K, Zikopoulos A, Grigoriadis S, Seretis N, Maziotis E, Anifandis G, Xystra P, Kostoulas C, Giougli U, Pantos K, Simopoulou M, Georgiou I. The Role of One-Carbon Metabolism and Methyl Donors in Medically Assisted Reproduction: A Narrative Review of the Literature. Int J Mol Sci 2024; 25:4977. [PMID: 38732193 PMCID: PMC11084717 DOI: 10.3390/ijms25094977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
One-carbon (1-C) metabolic deficiency impairs homeostasis, driving disease development, including infertility. It is of importance to summarize the current evidence regarding the clinical utility of 1-C metabolism-related biomolecules and methyl donors, namely, folate, betaine, choline, vitamin B12, homocysteine (Hcy), and zinc, as potential biomarkers, dietary supplements, and culture media supplements in the context of medically assisted reproduction (MAR). A narrative review of the literature was conducted in the PubMed/Medline database. Diet, ageing, and the endocrine milieu of individuals affect both 1-C metabolism and fertility status. In vitro fertilization (IVF) techniques, and culture conditions in particular, have a direct impact on 1-C metabolic activity in gametes and embryos. Critical analysis indicated that zinc supplementation in cryopreservation media may be a promising approach to reducing oxidative damage, while female serum homocysteine levels may be employed as a possible biomarker for predicting IVF outcomes. Nonetheless, the level of evidence is low, and future studies are needed to verify these data. One-carbon metabolism-related processes, including redox defense and epigenetic regulation, may be compromised in IVF-derived embryos. The study of 1-C metabolism may lead the way towards improving MAR efficiency and safety and ensuring the lifelong health of MAR infants.
Collapse
Affiliation(s)
- Konstantinos Sfakianoudis
- Centre for Human Reproduction, Genesis Athens Clinic, 14-16, Papanikoli, 15232 Athens, Greece; (K.S.); (K.P.)
| | - Athanasios Zikopoulos
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
- Obstetrics and Gynecology, Royal Cornwall Hospital, Treliske, Truro TR1 3LJ, UK
| | - Sokratis Grigoriadis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (E.M.); (P.X.)
| | - Nikolaos Seretis
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
| | - Evangelos Maziotis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (E.M.); (P.X.)
| | - George Anifandis
- Department of Obstetrics and Gynecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41222 Larisa, Greece;
| | - Paraskevi Xystra
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (E.M.); (P.X.)
| | - Charilaos Kostoulas
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
| | - Urania Giougli
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
| | - Konstantinos Pantos
- Centre for Human Reproduction, Genesis Athens Clinic, 14-16, Papanikoli, 15232 Athens, Greece; (K.S.); (K.P.)
| | - Mara Simopoulou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.G.); (E.M.); (P.X.)
| | - Ioannis Georgiou
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.); (C.K.); (U.G.); (I.G.)
| |
Collapse
|
6
|
Socha MW, Flis W, Wartęga M. Epigenetic Genome Modifications during Pregnancy: The Impact of Essential Nutritional Supplements on DNA Methylation. Nutrients 2024; 16:678. [PMID: 38474806 PMCID: PMC10934520 DOI: 10.3390/nu16050678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Pregnancy is an extremely stressful period in a pregnant woman's life. Currently, women's awareness of the proper course of pregnancy and its possible complications is constantly growing. Therefore, a significant percentage of women increasingly reach for various dietary supplements during gestation. Some of the most popular substances included in multi-ingredient supplements are folic acid and choline. Those substances are associated with positive effects on fetal intrauterine development and fewer possible pregnancy-associated complications. Recently, more and more attention has been paid to the impacts of specific environmental factors, such as diet, stress, physical activity, etc., on epigenetic modifications, understood as changes occurring in gene expression without the direct alteration of DNA sequences. Substances such as folic acid and choline may participate in epigenetic modifications by acting via a one-carbon cycle, leading to the methyl-group donor formation. Those nutrients may indirectly impact genome phenotype by influencing the process of DNA methylation. This review article presents the current state of knowledge on the use of folic acid and choline supplementation during pregnancy, taking into account their impacts on the maternal-fetal unit and possible pregnancy outcomes, and determining possible mechanisms of action, with particular emphasis on their possible impacts on epigenetic modifications.
Collapse
Affiliation(s)
- Maciej W. Socha
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland;
- Department of Obstetrics and Gynecology, St. Adalbert’s Hospital in Gdańsk, Copernicus Healthcare Entity, Jana Pawła II 50, 80-462 Gdańsk, Poland
| | - Wojciech Flis
- Department of Perinatology, Gynecology and Gynecologic Oncology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland;
- Department of Obstetrics and Gynecology, St. Adalbert’s Hospital in Gdańsk, Copernicus Healthcare Entity, Jana Pawła II 50, 80-462 Gdańsk, Poland
| | - Mateusz Wartęga
- Department of Pathophysiology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland;
| |
Collapse
|
7
|
Westmark PR, Lyon G, Gutierrez A, Boeck B, Van Hammond O, Ripp N, Pagan-Torres NA, Brower J, Held PK, Scarlett C, Westmark CJ. Effects of Soy Protein Isolate on Fragile X Phenotypes in Mice. Nutrients 2024; 16:284. [PMID: 38257177 PMCID: PMC10819477 DOI: 10.3390/nu16020284] [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: 11/28/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Obesity is a pediatric epidemic that is more prevalent in children with developmental disabilities. We hypothesize that soy protein-based diets increase weight gain and alter neurobehavioral outcomes. Our objective herein was to test matched casein- and soy protein-based purified ingredient diets in a mouse model of fragile X syndrome, Fmr1KO mice. The experimental methods included assessment of growth; 24-7 activity levels; motor coordination; learning and memory; blood-based amino acid, phytoestrogen and glucose levels; and organ weights. The primary outcome measure was body weight. We find increased body weight in male Fmr1KO from postnatal day 6 (P6) to P224, male wild type (WT) from P32-P39, female Fmr1KO from P6-P18 and P168-P224, and female Fmr1HET from P9-P18 as a function of soy. Activity at the beginning of the light and dark cycles increased in female Fmr1HET and Fmr1KO mice fed soy. We did not find significant differences in rotarod or passive avoidance behavior as a function of genotype or diet. Several blood-based amino acids and phytoestrogens were significantly altered in response to soy. Liver weight was increased in WT and adipose tissue in Fmr1KO mice fed soy. Activity levels at the beginning of the light cycle and testes weight were greater in Fmr1KO versus WT males irrespective of diet. DEXA analysis at 8-months-old indicated increased fat mass and total body area in Fmr1KO females and lean mass and bone mineral density in Fmr1KO males fed soy. Overall, dietary consumption of soy protein isolate by C57BL/6J mice caused increased growth, which could be attributed to increased lean mass in males and fat mass in females. There were sex-specific differences with more pronounced effects in Fmr1KO versus WT and in males versus females.
Collapse
Affiliation(s)
- Pamela R. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA;
| | - Greg Lyon
- Undergraduate Research Scholars Program, University of Wisconsin, Madison, WI 53706, USA; (G.L.); (O.V.H.)
| | - Alejandra Gutierrez
- Molecular Environmental Toxicology Master’s Program, University of Wisconsin, Madison, WI 53706, USA;
| | - Brynne Boeck
- Neurology Undergraduate Research, University of Wisconsin, Madison, WI 53706, USA; (B.B.); (N.R.)
| | - Olivia Van Hammond
- Undergraduate Research Scholars Program, University of Wisconsin, Madison, WI 53706, USA; (G.L.); (O.V.H.)
| | - Nathan Ripp
- Neurology Undergraduate Research, University of Wisconsin, Madison, WI 53706, USA; (B.B.); (N.R.)
| | - Nicole Arianne Pagan-Torres
- Molecular Environmental Toxicology Summer Research Opportunities Program, University of Wisconsin, Madison, WI 53706, USA;
| | - James Brower
- Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, WI 53706, USA; (J.B.); (P.K.H.)
| | - Patrice K. Held
- Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, WI 53706, USA; (J.B.); (P.K.H.)
| | - Cameron Scarlett
- School of Pharmacy, University of Wisconsin, Madison, WI 53706, USA;
| | - Cara J. Westmark
- Department of Neurology and Molecular Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
8
|
Yoshikawa M, Suemaru K. Prenatal folate deficiency impairs sociability and memory/recognition in mice offspring. Brain Res 2024; 1822:148639. [PMID: 37858854 DOI: 10.1016/j.brainres.2023.148639] [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: 07/10/2023] [Revised: 09/30/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
Folate is essential for the normal growth and development of the fetus. Folic acid supplementation during the fetal period affects postnatal brain development and reduces the incidence of mental disorders in animal and human studies. However, the association between folate deficiency (FD) during pregnancy and developmental disorders in children remains poorly understood. In this study, we investigated whether prenatal FD is associated with neurodevelopmental disorders in offspring. ICR mice were fed a control diet (2 mg folic acid/kg diet) or a folate-deficient diet (0.3 mg folic acid/kg diet) from embryonic day 1 until parturition. We evaluated locomotor activity, anxiety, grooming, sociability and learning memory in male offspring at 7-10 weeks of age. No differences were found in locomotor activity or anxiety in the open field test, nor in grooming time in the self-grooming test. However, sociability, spatial memory, and novel object recognition were impaired in the FD mice compared with control offspring. Furthermore, we measured protein expression levels of the NMDA and AMPA receptors, as well as PSD-95 and the GABA-synthesizing enzymes GAD65/67 in the frontal cortex and hippocampus. In FD mice, expression levels of AMPA receptor 1 and PSD-95 in both regions were reduced compared with control mice. Moreover, NMDA receptor subunit 2B and GAD65/67 were significantly downregulated in the frontal cortex of prenatal FD mice compared with the controls. Collectively, these findings suggest that prenatal FD causes behavioral deficits together with a reduction in synaptic protein levels in the frontal cortex and hippocampus.
Collapse
Affiliation(s)
- Misato Yoshikawa
- School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama 703-8516, Japan.
| | - Katsuya Suemaru
- School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama 703-8516, Japan.
| |
Collapse
|
9
|
Gevezova M, Sbirkov Y, Sarafian V, Plaimas K, Suratanee A, Maes M. Autistic spectrum disorder (ASD) - Gene, molecular and pathway signatures linking systemic inflammation, mitochondrial dysfunction, transsynaptic signalling, and neurodevelopment. Brain Behav Immun Health 2023; 30:100646. [PMID: 37334258 PMCID: PMC10275703 DOI: 10.1016/j.bbih.2023.100646] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 06/03/2023] [Indexed: 06/20/2023] Open
Abstract
Background Despite advances in autism spectrum disorder (ASD) research and the vast genomic, transcriptomic, and proteomic data available, there are still controversies regarding the pathways and molecular signatures underlying the neurodevelopmental disorders leading to ASD. Purpose To delineate these underpinning signatures, we examined the two largest gene expression meta-analysis datasets obtained from the brain and peripheral blood mononuclear cells (PBMCs) of 1355 ASD patients and 1110 controls. Methods We performed network, enrichment, and annotation analyses using the differentially expressed genes, transcripts, and proteins identified in ASD patients. Results Transcription factor network analyses in up- and down-regulated genes in brain tissue and PBMCs in ASD showed eight main transcription factors, namely: BCL3, CEBPB, IRF1, IRF8, KAT2A, NELFE, RELA, and TRIM28. The upregulated gene networks in PBMCs of ASD patients are strongly associated with activated immune-inflammatory pathways, including interferon-α signaling, and cellular responses to DNA repair. Enrichment analyses of the upregulated CNS gene networks indicate involvement of immune-inflammatory pathways, cytokine production, Toll-Like Receptor signalling, with a major involvement of the PI3K-Akt pathway. Analyses of the downregulated CNS genes suggest electron transport chain dysfunctions at multiple levels. Network topological analyses revealed that the consequent aberrations in axonogenesis, neurogenesis, synaptic transmission, and regulation of transsynaptic signalling affect neurodevelopment with subsequent impairments in social behaviours and neurocognition. The results suggest a defense response against viral infection. Conclusions Peripheral activation of immune-inflammatory pathways, most likely induced by viral infections, may result in CNS neuroinflammation and mitochondrial dysfunction, leading to abnormalities in transsynaptic transmission, and brain neurodevelopment.
Collapse
Affiliation(s)
- Maria Gevezova
- Department of Medical Biology, Medical University of Plovdiv, Bulgaria
- Research Institute at MU-Plovdiv, Bulgaria
| | - Yordan Sbirkov
- Department of Medical Biology, Medical University of Plovdiv, Bulgaria
- Research Institute at MU-Plovdiv, Bulgaria
| | - Victoria Sarafian
- Department of Medical Biology, Medical University of Plovdiv, Bulgaria
- Research Institute at MU-Plovdiv, Bulgaria
| | - Kitiporn Plaimas
- Advanced Virtual and Intelligent Computing (AVIC) Center, Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Apichat Suratanee
- Department of Mathematics, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
| | - Michael Maes
- Research Institute at MU-Plovdiv, Bulgaria
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
- Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, South Korea
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
10
|
Ssewanyana D, Knight JA, Matthews SG, Wong J, Khani NA, Lye J, Murphy KE, Foshay K, Okeke J, Lye SJ, Hung RJ. Maternal prenatal psychological distress and vitamin intake with children's neurocognitive development. Pediatr Res 2022; 92:1450-1457. [PMID: 35288638 DOI: 10.1038/s41390-022-02003-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/06/2022] [Accepted: 02/06/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Maternal prenatal psychological distress (PPD) is increasingly linked to sub-optimal child neurodevelopment. Daily intake of prenatal vitamin during pre-conception and early pregnancy may ameliorate the effects of PPD on cognition in the offspring. METHODS PPD was assessed in early (12-16 weeks) and late (28-32 weeks) gestation in the Ontario Birth Study. Prenatal vitamin supplement intake information was collected in early gestation. Child cognition at 4 years was assessed using the NIH Toolbox. Poisson regression was used to investigate associations between PPD and/or prenatal vitamin intake and child cognition. RESULTS Four hundred and eighteen mother-child dyads were assessed. Moderate-severe PPD experienced during early gestation was associated with reduced cognition (adjusted incidence rate ratio (IRRadj) = 3.71, 95% confidence interval (CI): 1.57-8.77, P = 0.003). Daily intake of prenatal vitamins was not associated with cognition (IRRadj = 1.34, 95% CI: 0.73-2.46, P = 0.34). Upon stratification, the experience of mild-severe PPD with daily intake of prenatal vitamins was associated with higher incident rates of suboptimal cognition compared to children of women with daily prenatal vitamin intake without any episode of PPD (IRRadj = 2.88, 95% CI: 1.1-7.4). CONCLUSIONS Moderate-severe PPD in early pregnancy is associated with poor cognition in children and daily intake of prenatal vitamin did not ameliorate this association. IMPACT Our findings expand on existing literature by highlighting that exposure to prenatal psychological distress (PPD), in moderate-to-severe form, in the early stages of pregnancy, can have detrimental effects on the offspring's cognitive development at 4 years. Overall, prenatal vitamin intake did not ameliorate the effects of PPD. Early screening and treatment of prenatal maternal mental illness is crucial.
Collapse
Affiliation(s)
- Derrick Ssewanyana
- Alliance for Human Development, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada.,Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Julia A Knight
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada.,Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Stephen G Matthews
- Alliance for Human Development, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynaecology, Sinai Health System, Toronto, ON, Canada
| | - Jody Wong
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Nadya Adel Khani
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Jennifer Lye
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Kellie E Murphy
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynaecology, Sinai Health System, Toronto, ON, Canada
| | - Kim Foshay
- Department of Obstetrics and Gynaecology, Sinai Health System, Toronto, ON, Canada
| | - Justin Okeke
- Department of Obstetrics and Gynaecology, Sinai Health System, Toronto, ON, Canada
| | - Stephen J Lye
- Alliance for Human Development, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynaecology, Sinai Health System, Toronto, ON, Canada
| | - Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada. .,Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
Vitamin B12 as a Cholinergic System Modulator and Blood Brain Barrier Integrity Restorer in Alzheimer's Disease. Eur J Pharm Sci 2022; 174:106201. [PMID: 35523375 DOI: 10.1016/j.ejps.2022.106201] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 12/22/2022]
Abstract
So far, the cholinergic hypothesis of Alzheimer's disease (AD) remains the fundamental explanation for the complex etiopathology of AD. However, therapeutics raising synaptic acetylcholine (Ach) or having cholinergic receptors agonistic activity had shown limited clinical efficacy, possibly, due to lacking capability to aggregate cholinergic receptors within the degenerated cholinergic neurons. Vitamin-B12 (B12) is an epigenetic modifier. It has a specific CNS transport system via the cubam receptors. The later enclose a cholinergic aggregator; agrin protein, suggesting that B12 administration may cause cholinergic receptors aggregation. Further, B12 involvement in homocysteine (Hcy) metabolism may restore blood brain barrier (BBB) integrity disrupted by elevated Hcy levels in AD. Here in, using a pharmacological model of cholinergic amnesia, three different B12 doses were compared to the standard of care; donepezil (DON) regarding cholinergic system modulation, and Hcy metabolic pathways. Further, AD-associated cerebro-vascular pathology was assessed by morphometric analyses of cerebro-vasculature morphology and ultrastructure using scanning and transmission electron-microscopes, respectively. Consequent effect on key AD-hallmarks and behavioral cognitive tests was also examined. The highest B12-tested dose (B12-HD) showed the greatest hippocampal cholinergic modulation with dose-dependent preferential upregulation of one cholinergic receptor over the other. Altered Hcy metabolism was proved to be a consequence of cholinergic disruption that was variably reversed by different B12 doses. In spite of equipotent effect of DON and B12-HD therapies in decreasing β-amyloid synthesis, B12-HD-treated group revealed the greatest restoration of BBB integrity indicating superior capability of β-amyloid clearance. Therefore, B12-HD therapy may represent a promising AD-modifying agent with extra-ability over conventional cholinergic modulators to aggregate cholinergic receptors.
Collapse
|
12
|
Yang X, Sun W, Wu Q, Lin H, Lu Z, Shen X, Chen Y, Zhou Y, Huang L, Wu F, Liu F, Chu D. Excess Folic Acid Supplementation before and during Pregnancy and Lactation Alters Behaviors and Brain Gene Expression in Female Mouse Offspring. Nutrients 2021; 14:nu14010066. [PMID: 35010941 PMCID: PMC8746785 DOI: 10.3390/nu14010066] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Use of folic acid (FA) during early pregnancy protects against birth defects. However, excess FA has shown gender-specific neurodevelopmental toxicity. Previously, we fed the mice with 2.5 times the recommended amount of FA one week prior to mating and during the pregnancy and lactation periods, and detected the activated expression of Fos and related genes in the brains of weaning male offspring, as well as behavioral abnormalities in the adults. Here, we studied whether female offspring were affected by the same dosage of FA. An open field test, three-chamber social approach and social novelty test, an elevated plus-maze, rotarod test and the Morris water maze task were used to evaluate their behaviors. RNA sequencing was performed to identify differentially expressed genes in the brains. Quantitative real time-PCR (qRT-PCR) and Western blots were applied to verify the changes in gene expression. We found increased anxiety and impaired exploratory behavior, motor coordination and spatial memory in FA-exposed females. The brain transcriptome revealed 36 up-regulated and 79 down-regulated genes in their brains at weaning. The increase of Tlr1; Sult1a1; Tph2; Acacb; Etnppl; Angptl4 and Apold1, as well as a decrease of Ppara mRNA were confirmed by qRT-PCR. Among these genes; the mRNA levels of Etnppl; Angptl4andApold1 were increased in the both FA-exposed female and male brains. The elevation of Sult1a1 protein was confirmed by Western blots. Our data suggest that excess FA alteres brain gene expression and behaviors in female offspring, of which certain genes show apparent gender specificity.
Collapse
Affiliation(s)
- Xingyue Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China; (H.L.); (F.W.)
| | - Wenyan Sun
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (W.S.); (Z.L.); (Y.C.); (Y.Z.)
| | - Qian Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
| | - Hongyan Lin
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China; (H.L.); (F.W.)
| | - Zhixing Lu
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (W.S.); (Z.L.); (Y.C.); (Y.Z.)
| | - Xin Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
| | - Yongqi Chen
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (W.S.); (Z.L.); (Y.C.); (Y.Z.)
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (W.S.); (Z.L.); (Y.C.); (Y.Z.)
| | - Li Huang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
| | - Feng Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China; (H.L.); (F.W.)
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
- Correspondence: (F.L.); (D.C.)
| | - Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
- Correspondence: (F.L.); (D.C.)
| |
Collapse
|
13
|
Wu Q, Zhou D, Wu R, Shi R, Shen X, Jin N, Gu J, Gu JH, Liu F, Chu D. Excess folic acid supplementation before and during pregnancy and lactation activates β-catenin in the brain of male mouse offspring. Brain Res Bull 2021; 178:133-143. [PMID: 34808323 DOI: 10.1016/j.brainresbull.2021.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/03/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
Folic acid (FA) supplementation in early pregnancy is recommended to protect against birth defects. But excess FA has exhibited neurodevelopmental toxicity. We previously reported that the mice treated with 2.5-fold the dietary requirement of FA one week before mating and throughout pregnancy and lactation displayed abnormal behaviors in the offspring. Here we found the levels of non-phosphorylated β-catenin (active) were increased in the brains of weaning and adult FA-exposed offspring. Meanwhile, demethylation of protein phosphatase 2 A catalytic subunit (PP2Ac), which suppresses its enzyme activity in regulatory subunit dependent manner, was significantly inhibited. Among the upstream regulators of β-catenin, PI3K/Akt/GSK-3β but not Wnt signaling was stimulated in FA-exposed brains only at weaning. In mouse neuroblastoma N2a cells, knockdown of PP2Ac or leucine carboxyl methyltransferase-1 (LCMT-1), or overexpression of PP2Ac methylation-deficient mutant decreased β-catenin dephosphorylation. These results suggest that excess FA may activate β-catenin via suppressing PP2Ac demethylation, providing a novel mechanism for the influence of FA on neurodevelopment.
Collapse
Affiliation(s)
- Qian Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Dingwei Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Ruozhen Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Ruirui Shi
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong University, 226018 Nantong, China
| | - Xin Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Nana Jin
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China
| | - Jianlan Gu
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, 226001 Nantong, China
| | - Jin-Hua Gu
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong University, 226018 Nantong, China.
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, United States.
| | - Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, 226001 Nantong, China.
| |
Collapse
|
14
|
Early-Life Iron Deficiency Anemia Programs the Hippocampal Epigenomic Landscape. Nutrients 2021; 13:nu13113857. [PMID: 34836113 PMCID: PMC8623089 DOI: 10.3390/nu13113857] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 01/04/2023] Open
Abstract
Iron deficiency (ID) anemia is the foremost micronutrient deficiency worldwide, affecting around 40% of pregnant women and young children. ID during the prenatal and early postnatal periods has a pronounced effect on neurodevelopment, resulting in long-term effects such as cognitive impairment and increased risk for neuropsychiatric disorders. Treatment of ID has been complicated as it does not always resolve the long-lasting neurodevelopmental deficits. In animal models, developmental ID results in abnormal hippocampal structure and function associated with dysregulation of genes involved in neurotransmission and synaptic plasticity. Dysregulation of these genes is a likely proximate cause of the life-long deficits that follow developmental ID. However, a direct functional link between iron and gene dysregulation has yet to be elucidated. Iron-dependent epigenetic modifications are one mechanism by which ID could alter gene expression across the lifespan. The jumonji and AT-rich interaction domain-containing (JARID) protein and the Ten-Eleven Translocation (TET) proteins are two families of iron-dependent epigenetic modifiers that play critical roles during neural development by establishing proper gene regulation during critical periods of brain development. Therefore, JARIDs and TETs can contribute to the iron-mediated epigenetic mechanisms by which early-life ID directly causes stable changes in gene regulation across the life span.
Collapse
|
15
|
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
|
16
|
Parental Reports on Early Autism Behaviors in Their Children with Fragile X Syndrome as a Function of Infant Feeding. Nutrients 2021; 13:nu13082888. [PMID: 34445048 PMCID: PMC8401950 DOI: 10.3390/nu13082888] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022] Open
Abstract
This study evaluates the prevalence of autistic behaviors in fragile X syndrome as a function of infant diet. Retrospective survey data from the Fragile X Syndrome Nutrition Study, which included data on infant feeding and caregiver-reported developmental milestones for 190 children with fragile X syndrome enrolled in the Fragile X Online Registry with Accessible Database (FORWARD), were analyzed. Exploratory, sex-specific associations were found linking the use of soy-based infant formula with worse autistic behaviors related to language in females and self-injurious behavior in males. These findings prompt prospective evaluation of the effects of soy-based infant formula on disease comorbidities in fragile X syndrome, a rare disorder for which newborn screening could be implemented if there was an intervention. Gastrointestinal problems were the most common reason cited for switching to soy-based infant formula. Thus, these findings also support the study of early gastrointestinal problems in fragile X syndrome, which may underly the development and severity of disease comorbidities. In conjunction with comorbidity data from the previous analyses of the Fragile X Syndrome Nutrition Study, the findings indicate that premutation fragile X mothers should be encouraged to breastfeed.
Collapse
|
17
|
Mondal A, Mukherjee S, Dar W, Singh S, Pati S. Role of glucose 6-phosphate dehydrogenase (G6PD) deficiency and its association to Autism Spectrum Disorders. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166185. [PMID: 34087423 DOI: 10.1016/j.bbadis.2021.166185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/27/2022]
Abstract
Autism Spectrum Disorder (ASD) is a common group of neurodevelopmental disorders which causes significant alterations in social and communication skills along with repetitive behavior and limited interests. The physiological understanding of ASD is ambiguous. Several reports suggested that environmental, genetic and epigenetic changes, neuroinflammation, mitochondrial dysfunction and metabolic alterations orchestrate the pathological outcomes of ASD. A recent report from Saudi Arabia found a mutation in X-chromosomal housekeeping glucose 6-phosphate dehydrogenase (G6PD) gene in two male ASD patients. Although, the involvement of G6PD-deficiency in the pathogenesis of ASD is poorly understood. Several reports suggested that G6PD deficiency impedes cellular detoxification of reactive oxygen species (ROS), which may result in neuronal damage and neuroinflammation. A deficiency of G6PD in newborn children may play a fundamental role in the pathogenesis of ASD. In this review, we will discuss the implications of G6PD deficiency in pathogenesis, male biasness and theranostics in ASD patients.
Collapse
Affiliation(s)
- Abir Mondal
- Neurobiology and Disease Modelling Laboratory, Host-Pathogen Interactions & Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida 201314, India
| | - Soumyadeep Mukherjee
- Neurobiology and Disease Modelling Laboratory, Host-Pathogen Interactions & Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida 201314, India
| | - Waseem Dar
- Neurobiology and Disease Modelling Laboratory, Host-Pathogen Interactions & Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida 201314, India
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India.
| | - Soumya Pati
- Neurobiology and Disease Modelling Laboratory, Host-Pathogen Interactions & Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida 201314, India.
| |
Collapse
|
18
|
Abstract
Pollutant agents are exponentially increasing in modern society since industrialization processes and technology are being developed worldwide. Impact of pollution on public health is well known but little has been described on the association between environmental pollutants and mental health. A literature search on PubMed and EMBASE has been conducted and 134 articles published on the issue of pollution and mental health have been included, cited, reviewed, and summarized. Emerging evidences have been collected on association between major environmental pollutants (air pollutants, heavy metals, ionizing radiation [IR], organophosphate pesticides, light pollution, noise pollution, environmental catastrophes) and various mental health disorders including anxiety, mood, and psychotic syndromes. Underlying pathogenesis includes direct and indirect effects of these agents on brain, respectively, due to their biological effect on human Central Nervous System or related to some levels of stress generated by the exposure to the pollutant agents over the time. Most of emerging evidences are still nonconclusive. Further studies should clarify how industrial production, the exploitation of certain resources, the proximity to waste and energy residues, noise, and the change in lifestyles are connected with psychological distress and mental health problems for the affected populations.
Collapse
|
19
|
Di Y, Li Z, Li J, Cheng Q, Zheng Q, Zhai C, Kang M, Wei C, Lan J, Fan J, Ren W, Tian Y. Maternal folic acid supplementation prevents autistic behaviors in a rat model induced by prenatal exposure to valproic acid. Food Funct 2021; 12:4544-4555. [DOI: 10.1039/d0fo02926b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Maternal FA supplementation at 4 mg kg−1 rescued the development delay, anxiety and core autism-like behaviors, and restored the abnormal synaptic spine morphology and synaptic protein expression in mPFC in the male offspring prenatally exposed to VPA.
Collapse
|
20
|
Zhu Y, Mordaunt CE, Durbin‐Johnson BP, Caudill MA, Malysheva OV, Miller JW, Green R, James SJ, Melnyk SB, Fallin MD, Hertz‐Picciotto I, Schmidt RJ, LaSalle JM. Expression Changes in Epigenetic Gene Pathways Associated With One-Carbon Nutritional Metabolites in Maternal Blood From Pregnancies Resulting in Autism and Non-Typical Neurodevelopment. Autism Res 2021; 14:11-28. [PMID: 33159718 PMCID: PMC7894157 DOI: 10.1002/aur.2428] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/16/2020] [Accepted: 10/21/2020] [Indexed: 12/15/2022]
Abstract
The prenatal period is a critical window for the development of autism spectrum disorder (ASD). The relationship between prenatal nutrients and gestational gene expression in mothers of children later diagnosed with ASD or non-typical development (Non-TD) is poorly understood. Maternal blood collected prospectively during pregnancy provides insights into the effects of nutrition, particularly one-carbon metabolites, on gene pathways and neurodevelopment. Genome-wide transcriptomes were measured with microarrays in 300 maternal blood samples in Markers of Autism Risk in Babies-Learning Early Signs. Sixteen different one-carbon metabolites, including folic acid, betaine, 5'-methyltretrahydrofolate (5-MeTHF), and dimethylglycine (DMG) were measured. Differential expression analysis and weighted gene correlation network analysis (WGCNA) were used to compare gene expression between children later diagnosed as typical development (TD), Non-TD and ASD, and to one-carbon metabolites. Using differential gene expression analysis, six transcripts (TGR-AS1, SQSTM1, HLA-C, and RFESD) were associated with child outcomes (ASD, Non-TD, and TD) with genome-wide significance. Genes nominally differentially expressed between ASD and TD significantly overlapped with seven high confidence ASD genes. WGCNA identified co-expressed gene modules significantly correlated with 5-MeTHF, folic acid, DMG, and betaine. A module enriched in DNA methylation functions showed a suggestive protective association with folic acid/5-MeTHF concentrations and ASD risk. Maternal plasma betaine and DMG concentrations were associated with a block of co-expressed genes enriched for adaptive immune, histone modification, and RNA processing functions. These results suggest that the prenatal maternal blood transcriptome is a sensitive indicator of gestational one-carbon metabolite status and changes relevant to children's later neurodevelopmental outcomes. LAY SUMMARY: Pregnancy is a time when maternal nutrition could interact with genetic risk for autism spectrum disorder. Blood samples collected during pregnancy from mothers who had a prior child with autism were examined for gene expression and nutrient metabolites, then compared to the diagnosis of the child at age three. Expression differences in gene pathways related to the immune system and gene regulation were observed for pregnancies of children with autism and non-typical neurodevelopment and were associated with maternal nutrients.
Collapse
Affiliation(s)
- Yihui Zhu
- Department of Medical Microbiology and Immunology, Genome Center, and Perinatal Origins of Disparities CenterUniversity of CaliforniaDavisCaliforniaUSA
- MIND Institute, School of MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Charles E. Mordaunt
- Department of Medical Microbiology and Immunology, Genome Center, and Perinatal Origins of Disparities CenterUniversity of CaliforniaDavisCaliforniaUSA
- MIND Institute, School of MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | | | - Marie A. Caudill
- Division of Nutritional SciencesCornell UniversityIthacaNew YorkUSA
| | | | - Joshua W. Miller
- Department of Nutritional SciencesRutgers UniversityNew BrunswickNew JerseyUSA
| | - Ralph Green
- Department of Pathology and Laboratory MedicineUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - S. Jill James
- Department of Pediatrics, University of Arkansas for Medical SciencesArkansas Children's Research InstituteLittle RockArkansasUSA
| | - Stepan B. Melnyk
- Department of Pediatrics, University of Arkansas for Medical SciencesArkansas Children's Research InstituteLittle RockArkansasUSA
| | - M. Daniele Fallin
- Department of Mental Health, Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Irva Hertz‐Picciotto
- MIND Institute, School of MedicineUniversity of CaliforniaDavisCaliforniaUSA
- Department of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Rebecca J. Schmidt
- MIND Institute, School of MedicineUniversity of CaliforniaDavisCaliforniaUSA
- Department of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Janine M. LaSalle
- Department of Medical Microbiology and Immunology, Genome Center, and Perinatal Origins of Disparities CenterUniversity of CaliforniaDavisCaliforniaUSA
- MIND Institute, School of MedicineUniversity of CaliforniaDavisCaliforniaUSA
| |
Collapse
|
21
|
Orjuela MA, Mejia-Rodriguez F, Quezada AD, Sanchez-Pimienta TG, Shamah-Levy T, Romero-Rendón J, Bhatt-Carreño S, Ponce-Castañeda MV, Castro MA, Paul L, Villalpando S. Fortification of bakery and corn masa-based foods in Mexico and dietary intake of folic acid and folate in Mexican national survey data. Am J Clin Nutr 2019; 110:1434-1448. [PMID: 31529037 PMCID: PMC6885481 DOI: 10.1093/ajcn/nqz224] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 08/14/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND In Mexico, wheat and corn flour fortification with folic acid (FA) was implemented in 2001 and mandated in 2008, but without direct enforcement. Current Mexican nutrient-content tables do not account for FA contained in bakery bread and corn masa-based foods, which are dietary staples in Mexico. OBJECTIVE The objective of this study was to examine the impact of FA fortification of dietary staples on the proportion of the population consuming below the Estimated Average Requirement (EAR) for folate or above the Tolerable Upper Intake Level (UL) for FA. METHODS We measured FA and folate content in dietary staples (bakery bread and tortillas) using microbial assays and MS, and we recalculated FA intake from 24-h recall dietary intake data collected in the 2012 Mexican National Health and Nutrition Survey (Encuesta Nacional de Salud y Nutrición) utilizing estimates from our food measurements, using nutrient concentrations from tortillas to approximate nutrient content of other corn masa-derived foods. The revised FA intake estimates were used to examine population-level intake of FA and dietary folate equivalent (DFE) accounting for geographic differences in FA content with statistical models. RESULTS FA content in dietary staples was variable, whereas use of FA-fortified flour in corn masa tortillas increased with population size in place of residence. Accounting for dietary staples' FA fortification increased population estimates for FA and DFE intake, resulting in a lower proportion with intake below the EAR and a higher proportion with intake above the UL. Despite accounting for FA-fortified staple foods, 9-33% of women of childbearing age still have intake below the EAR, whereas up to 12% of younger children have intake above the UL. CONCLUSIONS Unregulated FA fortification of dietary staples leads to unpredictable total folate intake without adequately impacting the intended target. Our findings suggest that monitoring, evaluation, and enforcement of mandatory fortification policies are needed. Without these, alternate strategies may be needed in order to reach women of childbearing age while avoiding overexposing children.
Collapse
Affiliation(s)
- Manuela A Orjuela
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA,Address correspondence to MAO (e-mail: )
| | - Fabiola Mejia-Rodriguez
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Mexico
| | - Amado D Quezada
- Center for Evaluation and Surveys Research, National Institute of Public Health, Cuernavaca, Mexico
| | - Tania G Sanchez-Pimienta
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Mexico
| | - Teresa Shamah-Levy
- Center for Evaluation and Surveys Research, National Institute of Public Health, Cuernavaca, Mexico
| | | | - Silvia Bhatt-Carreño
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | | | - Maria A Castro
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Ligi Paul
- Jean Mayer USDA Laboratories for Human Nutrition, Tufts University, Boston, MA, USA
| | - Salvador Villalpando
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Mexico
| |
Collapse
|
22
|
Zhang Q, Wu H, Zou M, Li L, Li Q, Sun C, Xia W, Cao Y, Wu L. Folic acid improves abnormal behavior via mitigation of oxidative stress, inflammation, and ferroptosis in the BTBR T+ tf/J mouse model of autism. J Nutr Biochem 2019; 71:98-109. [DOI: 10.1016/j.jnutbio.2019.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/27/2019] [Accepted: 05/09/2019] [Indexed: 01/09/2023]
|
23
|
Mircher C, Sacco S, Bouis C, Gallard J, Pichot A, Le Galloudec E, Cieuta C, Marey I, Greiner-Mahler O, Dorison N, Gambarini A, Stora S, Durand S, Polak M, Baruchel A, Schlumberger E, Dewailly J, Azar-Kolakez A, Guéant-Rodriguez RM, Guéant JL, Borderie D, Bonnefont-Rousselot D, Blondiaux E, Ravel A, Sturtz FG. Thyroid hormone and folinic acid in young children with Down syndrome: the phase 3 ACTHYF trial. Genet Med 2019; 22:44-52. [PMID: 31281181 DOI: 10.1038/s41436-019-0597-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/18/2019] [Indexed: 11/09/2022] Open
Abstract
PURPOSE To determine whether folinic acid (FA) and thyroxine, in combination or alone, benefit psychomotor development in young patients with Down syndrome (DS). METHODS The Assessment of Systematic Treatment With Folinic Acid and Thyroid Hormone on Psychomotor Development of Down Syndrome Young Children (ACTHYF) was a single-center, randomized, double-blind, placebo-controlled phase 3 trial in DS infants aged 6-18 months. Patients were randomly assigned to one of four treatments: placebo, folinic acid (FA), L-thyroxine, or FA+L-thyroxine, administered for 12 months. Randomization was done by age and sex. The primary endpoint was adjusted change from baseline in Griffiths Mental Development Scale global development quotient (GDQ) after 12 months. RESULTS Of 175 patients randomized, 143 completed the study. The modified intention-to-treat (mITT) population included all randomized patients who did not prematurely discontinue due to elevated baseline thyroid stimulating hormone (TSH). Baseline characteristics in the mITT were well balanced between groups, with reliable developmental assessment outcomes. Adjusted mean change in GDQ in the mITT showed similar decreases in all groups (placebo: -5.10 [95% confidence interval (CI) -7.84 to -2.37]; FA: -4.69 [95% CI -7.73 to -1.64]; L-thyroxine: -3.89 [95% CI -6.94 to -0.83]; FA+L-thyroxine: -3.86 [95% CI -6.67 to -1.06]), with no significant difference for any active treatment group versus placebo. CONCLUSION This trial does not support the hypotheses that thyroxine and/or folinic acid improve development of young children with DS or are synergistic. This trial is registered with ClinicalTrials.gov number, NCT01576705.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Michel Polak
- Endocrinologie gynécologie diabétologie pédiatriques, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Université Paris Descartes, INSERM U1016, Institut IMAGINE, Paris, France
| | - André Baruchel
- Pediatric Hematology-Immunology Department, University Hospital Robert Debré, Assistance Publique-Hôpitaux de Paris. Paris Diderot University, EA 3518; Institute of Hematology, Sorbonne Paris-Cité, Paris, France
| | - Emilie Schlumberger
- Reference Center for Language and Learning Disorders, Raymond Poincaré Hospital, Assistance Publique-Hôpitaux de Paris, Garches, France
| | | | - Ahlam Azar-Kolakez
- Endocrinology-Diabetology Department, Reference Center for Endocrine Growth and Developmental Disease, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Rosa-Maria Guéant-Rodriguez
- Research Unit (Inserm) UMRS 1256 N-GERE (Nutrition-Genetics-Environmental Risks), University de Lorraine, Faculty of Medicine, Nancy, France.,Department of Endocrinology, Diabetology and Nutrition, University Hospital of Nancy, Nancy, France
| | - Jean-Louis Guéant
- Research Unit (Inserm) UMRS 1256 N-GERE (Nutrition-Genetics-Environmental Risks), University de Lorraine, Faculty of Medicine, Nancy, France.,Department of Endocrinology, Diabetology and Nutrition, University Hospital of Nancy, Nancy, France
| | - Didier Borderie
- Biochemistry and Molecular Biology Laboratory, Cochin University Hospital, Paris, France
| | - Dominique Bonnefont-Rousselot
- Metabolic Biochemistry Department, Pitié-Salpêtrière-Charles Foix University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biochemistry Department, Faculty of Pharmacy, CNRS UMR 8258 - INSERM U1022, Paris Descartes University, Paris, France
| | | | | | - Franck G Sturtz
- Institut Jérôme Lejeune, Paris, France.,Biochemistry and Molecular Biology Department, CHU Limoges, Limoges, France.,Univ. Limoges, EA 6309, Limoges, France
| |
Collapse
|
24
|
Zou M, Sun C, Liang S, Sun Y, Li D, Li L, Fan L, Wu L, Xia W. Fisher discriminant analysis for classification of autism spectrum disorders based on folate-related metabolism markers. J Nutr Biochem 2019; 64:25-31. [DOI: 10.1016/j.jnutbio.2018.09.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/01/2018] [Accepted: 09/24/2018] [Indexed: 12/20/2022]
|
25
|
Raghavan R, Zuckerman B, Hong X, Wang G, Ji Y, Paige D, DiBari J, Zhang C, Fallin MD, Wang X. Fetal and Infancy Growth Pattern, Cord and Early Childhood Plasma Leptin, and Development of Autism Spectrum Disorder in the Boston Birth Cohort. Autism Res 2018; 11:1416-1431. [PMID: 30248249 PMCID: PMC6320256 DOI: 10.1002/aur.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/15/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Leptin is a proinflammatory cytokine that plays an important role in energy homeostasis. Emerging evidence suggests that leptin levels are altered in children with autism spectrum disorder (ASD); however, this has not been studied prospectively. Rapid growth during infancy and early childhood has been implicated in ASD, but the evidence is inconsistent. As leptin is involved in growth and is a potential risk factor for ASD, we explored the associations between (a) cord, early childhood leptin and ASD; and (b) birth weight for gestational age, early childhood weight gain, and ASD. We also assessed the mediating role of leptin in the relationship between weight gain during infancy and ASD. This study was conducted in a sample of 822 subjects from the Boston Birth Cohort. ASD was defined from diagnostic codes in electronic medical records. Extremely rapid weight gain during infancy was associated with a greater ASD risk and this persisted after adjusting for potential confounders (aOR: 3.11; 95% CI: 1.37, 7.07). Similarly, children that had higher plasma leptin levels, prior to ASD diagnosis, had an increased ASD risk in both unadjusted and adjusted models (aOR: 7.87; 95% CI: 2.06, 30.04). Further, early childhood leptin indirectly mediated the relationship between rapid weight gain and ASD. No associations were found between birth weight for gestational age, cord leptin and risk of ASD. Our findings provide a basis to further explore whether the combination of early life growth pattern and a biomarker such as leptin can predict ASD earlier. Autism Res 2018, 11: 1416-1431. © 2018 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: Is early life growth and a biomarker leptin related to ASD risk? To answer this question, we followed 822 children from birth and found that those who gained weight very quickly in infancy, had higher leptin levels in early childhood, had a greater chance of later ASD diagnosis. More research is needed to see if infant's weight gain pattern along with a biomarker (such as leptin) can be used to identify children with ASD sooner.
Collapse
Affiliation(s)
- Ramkripa Raghavan
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
| | - Barry Zuckerman
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, 850 Harrison Ave, 3 Floor, Suite 324L, Boston, MA 02118
| | - Xiumei Hong
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
| | - Guoying Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
| | - Yuelong Ji
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
| | - David Paige
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
| | - Jessica DiBari
- Office of Epidemiology and Research, Maternal & Child Health Bureau, Health Resources & Services Administration, 5600 Fishers Lane, 18N120, Rockville, MD 20857
| | - Cuilin Zhang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
- Division of Intramural Population and Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710 B, Rockledge Dr., Bethesda, MD 20817
| | - M. Daniele Fallin
- Wendy Klag Center for Autism and Developmental Disabilities & Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, 624 N. Broadway, HH 850, Baltimore, MD 21205
| | - Xiaobin Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
- Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205
| |
Collapse
|
26
|
Pjetri E, Zeisel SH. Deletion of one allele of Mthfd1 ( methylenetetrahydrofolate dehydrogenase 1 ) impairs learning in mice. Behav Brain Res 2017; 332:71-74. [DOI: 10.1016/j.bbr.2017.05.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/16/2017] [Accepted: 05/22/2017] [Indexed: 11/28/2022]
|
27
|
Wei H, Liang F, Meng G, Nie Z, Zhou R, Cheng W, Wu X, Feng Y, Wang Y. Redox/methylation mediated abnormal DNA methylation as regulators of ambient fine particulate matter-induced neurodevelopment related impairment in human neuronal cells. Sci Rep 2016; 6:33402. [PMID: 27624276 PMCID: PMC5022064 DOI: 10.1038/srep33402] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/24/2016] [Indexed: 12/22/2022] Open
Abstract
Fine particulate matter (PM2.5) has been implicated as a risk factor for neurodevelopmental disorders including autism in children. However, the underlying biological mechanism remains unclear. DNA methylation is suggested to be a fundamental mechanism for the neuronal responses to environmental cues. We prepared whole particle of PM2.5 (PM2.5), water-soluble extracts (Pw), organic extracts (Po) and carbon core component (Pc) and characterized their chemical constitutes. We found that PM2.5 induced significant redox imbalance, decreased the levels of intercellular methyl donor S-adenosylmethionine and caused global DNA hypomethylation. Furthermore, PM2.5 exposure triggered gene-specific promoter DNA hypo- or hypermethylation and abnormal mRNA expression of autism candidate genes. PM2.5-induced DNA hypermethylation in promoter regions of synapse related genes were associated with the decreases in their mRNA and protein expression. The inhibiting effects of antioxidative reagents, a methylation-supporting agent and a DNA methyltransferase inhibitor demonstrated the involvement of redox/methylation mechanism in PM2.5-induced abnormal DNA methylation patterns and synaptic protein expression. The biological effects above generally followed a sequence of PM2.5 ≥ Pwo > Po > Pw > Pc. Our results implicated a novel epigenetic mechanism for the neurodevelopmental toxicity of particulate air pollution, and that eliminating the chemical components could mitigate the neurotoxicity of PM2.5.
Collapse
Affiliation(s)
- Hongying Wei
- Shanghai Jiao Tong University School of Public Health; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai 200025, China
| | - Fan Liang
- Shanghai Jiao Tong University School of Public Health; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai 200025, China
| | - Ge Meng
- Shanghai Jiao Tong University School of Public Health; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai 200025, China
| | - Zhiqing Nie
- Shanghai Jiao Tong University School of Environmental Science and Engineering, Shanghai, 200240, China
| | - Ren Zhou
- Shanghai Jiao Tong University School of Public Health; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai 200025, China
| | - Wei Cheng
- Shanghai Jiao Tong University School of Public Health; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai 200025, China
| | - Xiaomeng Wu
- Shanghai Jiao Tong University School of Public Health; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai 200025, China
| | - Yan Feng
- Shanghai Jiao Tong University School of Public Health; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai 200025, China
| | - Yan Wang
- Shanghai Jiao Tong University School of Public Health; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai 200025, China.,Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
28
|
Sun C, Zou M, Zhao D, Xia W, Wu L. Efficacy of Folic Acid Supplementation in Autistic Children Participating in Structured Teaching: An Open-Label Trial. Nutrients 2016; 8:nu8060337. [PMID: 27338456 PMCID: PMC4924178 DOI: 10.3390/nu8060337] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/29/2016] [Accepted: 05/31/2016] [Indexed: 11/23/2022] Open
Abstract
Autism spectrum disorders (ASD) are recognized as a major public health issue. Here, we evaluated the effects of folic acid intervention on methylation cycles and oxidative stress in autistic children enrolled in structured teaching. Sixty-six autistic children enrolled in this open-label trial and participated in three months of structured teaching. Forty-four children were treated with 400 μg folic acid (two times/daily) for a period of three months during their structured teaching (intervention group), while the remaining 22 children were not given any supplement for the duration of the study (control group). The Autism Treatment Evaluation Checklist (ATEC) and Psychoeducational Profile-third edition (PEP-3) were measured at the beginning and end of the treatment period. Folic acid, homocysteine, and glutathione metabolism in plasma were measured before and after treatment in 29 autistic children randomly selected from the intervention group and were compared with 29 age-matched unaffected children (typical developmental group). The results illustrated folic acid intervention improved autism symptoms towards sociability, cognitive verbal/preverbal, receptive language, and affective expression and communication. Furthermore, this treatment also improved the concentrations of folic acid, homocysteine, and normalized glutathione redox metabolism. Folic acid supplementation may have a certain role in the treatment of children with autism.
Collapse
Affiliation(s)
- Caihong Sun
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, China.
| | - Mingyang Zou
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, China.
| | - Dong Zhao
- Zhejiang Provincial Center For Disease Prevention and Control, Hangzhou 310009, China.
| | - Wei Xia
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, China.
| | - Lijie Wu
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
29
|
Tran PV, Kennedy BC, Pisansky MT, Won KJ, Gewirtz JC, Simmons RA, Georgieff MK. Prenatal Choline Supplementation Diminishes Early-Life Iron Deficiency-Induced Reprogramming of Molecular Networks Associated with Behavioral Abnormalities in the Adult Rat Hippocampus. J Nutr 2016; 146:484-93. [PMID: 26865644 PMCID: PMC4763487 DOI: 10.3945/jn.115.227561] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 12/10/2015] [Accepted: 01/05/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Early-life iron deficiency is a common nutrient deficiency worldwide. Maternal iron deficiency increases the risk of schizophrenia and autism in the offspring. Postnatal iron deficiency in young children results in cognitive and socioemotional abnormalities in adulthood despite iron treatment. The rat model of diet-induced fetal-neonatal iron deficiency recapitulates the observed neurobehavioral deficits. OBJECTIVES We sought to establish molecular underpinnings for the persistent psychopathologic effects of early-life iron deficiency by determining whether it permanently reprograms the hippocampal transcriptome. We also assessed the effects of maternal dietary choline supplementation on the offspring's hippocampal transcriptome to identify pathways through which choline mitigates the emergence of long-term cognitive deficits. METHODS Male rat pups were made iron deficient (ID) by providing pregnant and nursing dams an ID diet (4 g Fe/kg) from gestational day (G) 2 through postnatal day (PND) 7 and an iron-sufficient (IS) diet (200 g Fe/kg) thereafter. Control pups were provided IS diet throughout. Choline (5 g/kg) was given to half the pregnant dams in each group from G11 to G18. PND65 hippocampal transcriptomes were assayed by next generation sequencing (NGS) and analyzed with the use of knowledge-based Ingenuity Pathway Analysis. Real-time polymerase chain reaction was performed to validate a subset of altered genes. RESULTS Formerly ID rats had altered hippocampal expression of 619 from >10,000 gene loci sequenced by NGS, many of which map onto molecular networks implicated in psychological disorders, including anxiety, autism, and schizophrenia. There were significant interactions between iron status and prenatal choline treatment in influencing gene expression. Choline supplementation reduced the effects of iron deficiency, including those on gene networks associated with autism and schizophrenia. CONCLUSIONS Fetal-neonatal iron deficiency reprograms molecular networks associated with the pathogenesis of neurologic and psychological disorders in adult rats. The positive response to prenatal choline represents a potential adjunctive therapeutic supplement to the high-risk group.
Collapse
Affiliation(s)
| | | | | | - Kyoung-Jae Won
- Institute for Diabetes, Obesity and Metabolism, Department of Genetics, and
| | - Jonathan C Gewirtz
- Graduate Program in Neuroscience, and Department of Psychology, University of Minnesota, Minneapolis, MN
| | - Rebecca A Simmons
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
30
|
Keil KP, Lein PJ. DNA methylation: a mechanism linking environmental chemical exposures to risk of autism spectrum disorders? ENVIRONMENTAL EPIGENETICS 2016; 2:dvv012. [PMID: 27158529 PMCID: PMC4856164 DOI: 10.1093/eep/dvv012] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
There is now compelling evidence that gene by environment interactions are important in the etiology of autism spectrum disorders (ASDs). However, the mechanisms by which environmental factors interact with genetic susceptibilities to confer individual risk for ASD remain a significant knowledge gap in the field. The epigenome, and in particular DNA methylation, is a critical gene expression regulatory mechanism in normal and pathogenic brain development. DNA methylation can be influenced by environmental factors such as diet, hormones, stress, drugs, or exposure to environmental chemicals, suggesting that environmental factors may contribute to adverse neurodevelopmental outcomes of relevance to ASD via effects on DNA methylation in the developing brain. In this review, we describe epidemiological and experimental evidence implicating altered DNA methylation as a potential mechanism by which environmental chemicals confer risk for ASD, using polychlorinated biphenyls (PCBs), lead, and bisphenol A (BPA) as examples. Understanding how environmental chemical exposures influence DNA methylation and how these epigenetic changes modulate the risk and/or severity of ASD will not only provide mechanistic insight regarding gene-environment interactions of relevance to ASD but may also suggest potential intervention strategies for these and potentially other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Kimberly P. Keil
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
- *Correspondence address. Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA. Tel:
(530) 752-1970
; Fax:
(530) 752-7690
; E-mail:
| |
Collapse
|
31
|
Rathod R, Kale A, Joshi S. Novel insights into the effect of vitamin B₁₂ and omega-3 fatty acids on brain function. J Biomed Sci 2016; 23:17. [PMID: 26809263 PMCID: PMC4727338 DOI: 10.1186/s12929-016-0241-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 01/20/2016] [Indexed: 01/16/2023] Open
Abstract
The prevalence of psychiatric disorders which are characterized by cognitive decline is increasing at an alarming rate and account for a significant proportion of the global disease burden. Evidences from human and animal studies indicate that neurocognitive development is influenced by various environmental factors including nutrition. It has been established that nutrition affects the brain throughout life. However, the mechanisms through which nutrition modulates mental health are still not well understood. It has been suggested that the deficiencies of both vitamin B12 and omega-3 fatty acids can have adverse effects on cognition and synaptic plasticity. Studies indicate a need for supplementation of vitamin B12 and omega-3 fatty acids to reduce the risk of cognitive decline, although the results of intervention trials using these nutrients in isolation are inconclusive. In the present article, we provide an overview of vitamin B12 and omega-3 fatty acids, the possible mechanisms and the evidences through which vitamin B12 and omega-3 fatty acids modulate mental health and cognition. Understanding the role of vitamin B12 and omega-3 fatty acids on brain functioning may provide important clues to prevent early cognitive deficits and later neurobehavioral disorders.
Collapse
Affiliation(s)
- Richa Rathod
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth Deemed University, Pune Satara Road, Pune, 411043, India
| | - Anvita Kale
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth Deemed University, Pune Satara Road, Pune, 411043, India
| | - Sadhana Joshi
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth Deemed University, Pune Satara Road, Pune, 411043, India.
| |
Collapse
|
32
|
Maternal folate status as a risk factor for autism spectrum disorders: a review of existing evidence. Br J Nutr 2015; 114:663-72. [PMID: 26243379 DOI: 10.1017/s0007114515002470] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Emerging evidence from epidemiological studies supports the notion that maternal folate status regulated by dietary and genetic factors early in pregnancy may influence the risk of autism spectrum disorders (ASD). In this review, we provide an overview of what is known about the role of folate in the aetiology of neurodevelopmental disorders; summarise relevant biological, genetic and epigenetic mechanisms; and synthesise the evidence from human observational studies and randomised controlled trials that have examined the relationship between maternal folate and ASD or related traits. Much of the existing literature on this topic is subject to limitations such as potential confounding by healthy behaviours and other dietary factors, and exposure assessed within limited exposure windows. As the existing evidence is inconclusive, further research remains to be conducted in order to verify this hypothesis. Complete assessment of maternal functional folate status through the pre- and peri-conceptional periods requires biological measurement of folate, vitamin B12 and homocysteine and genetic variants involved in one-carbon metabolism and epigenetic mechanisms. In addition to more complete assessment of maternal functional folate status, careful consideration of potential confounding is warranted.
Collapse
|
33
|
Abstract
Nutrition plays a fundamental role in supporting the structural and functional development of the human brain from conception, throughout early infancy and extending into later life. A growing body of evidence suggests that folate and the metabolically related B-vitamins are essential for brain health across all age groups, owing to their specific roles in C1 metabolism and particularly in the production of S-adenosylmethionine, a universal methyl donor essential for the production of neurotransmitters. Emerging, though not entirely consistent, evidence suggests that maternal folate status throughout pregnancy may influence neurodevelopment and behaviour of the offspring. Furthermore optimal B-vitamin status is associated with better cognitive health in ageing. Of note, a recent clinical trial provided evidence that supplementation with folic acid and related B-vitamins over a 2-year-period reduced global and regional brain atrophy, as measured by MRI scan in older adults. In terms of potential mechanisms, the effects of these B-vitamins on cognitive health may be independent or may be mediated by nutrient-nutrient and/or relevant gene-nutrient interactions. Furthermore, a new area of research suggests that the in utero environment influences health in later life. Folate, an important cofactor in C1 metabolism, is indirectly involved in DNA methylation, which in turn is considered to be one of the epigenetic mechanisms that may underlie fetal programming and brain development. The present review will explore the evidence that supports a role for folate and the related B-vitamins in brain health across the lifecycle, and potential mechanisms to explain such effects.
Collapse
|