1
|
Wang M, Shi L, Tong Z, Liu Y, Bai Y, Yang X, Wang Y, Gong Z, Zhang Q, Zhang X. The impact of folic acid deficiency on ischemic stroke: Role of inflammation and long noncoding RNA H19. J Nutr Biochem 2025; 142:109916. [PMID: 40187372 DOI: 10.1016/j.jnutbio.2025.109916] [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: 11/19/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
It has been validated that folic acid deficiency (FD) is associated with an increased risk of stroke and a worse prognosis. However, the specific mechanisms by which FD exerts its detrimental effects on ischemic stroke (IS) have not been fully understood. The results of this case-control study indicated that patients with IS had a decreased serum folate level, along with up-regulated long non-coding RNA H19 (lncRNA H19) and enhanced inflammatory responses. Meanwhile, it was corroborated that the serum folate level was negatively correlated with H19 expression and the systemic immune-inflammation index (SII). Similarly, FD was demonstrated to exacerbate neurological injury in the middle cerebral artery occlusion/reperfusion (MCAO/R) rats by up-regulating the expression of inflammatory cytokines and H19 in both peripheral blood and brain tissue. Notably, the alterations in the expression of these factors in peripheral blood were consistent with those observed in brain tissue. Additionally, in a co-culture of N2a neurons and BV2 microglia, FD promoted the transition of BV2 cells towards a pro-inflammatory state by up-regulating the expression of H19, which aggravated neuronal injury. Moreover, blocking H19 in BV2 cells mitigated inflammation and partially reversed the injury in N2a cells exacerbated by FD after the treatment with oxygen-glucose deprivation and reperfusion (OGD/R). These findings provide a more in-depth insight into the regulatory role of H19-mediated systemic inflammatory responses in the context of FD, suggesting the potential clinical utility of folic acid in managing ischemic brain injury.
Collapse
Affiliation(s)
- Meng Wang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health; Center for International Collaborative Research on Environment; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, Tianjin, China
| | - Linran Shi
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Business Office, Tongzhou District Centre for Disease Prevention and Control, Tongzhou District, Beijing, China
| | - Zonghang Tong
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health; Center for International Collaborative Research on Environment; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, Tianjin, China
| | - Yinyue Liu
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health; Center for International Collaborative Research on Environment; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, Tianjin, China
| | - Yuxuan Bai
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xueli Yang
- Tianjin Key Laboratory of Environment, Nutrition and Public Health; Center for International Collaborative Research on Environment; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, Tianjin, China; Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yanhong Wang
- Affiliated Hospital of Changzhi Institute of TCM, Changzhi City, Shanxi Province, China
| | - Zhongying Gong
- Department of Neurology, Tianjin First Center Hospital, Tianjin, China
| | - Qiang Zhang
- Tianjin Key Laboratory of Environment, Nutrition and Public Health; Center for International Collaborative Research on Environment; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, Tianjin, China; Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xumei Zhang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health; Center for International Collaborative Research on Environment; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
2
|
Oner M, Chen MC, Cheng PT, Li YH, Cheng YC, Celik A, Soong SW, Hsu LW, Lin DY, Hossain Prince GMS, Dhar T, Cheng HC, Tang PC, Lin H. Impact of metformin on neocortical development during pregnancy: Involvement of ERK and p35/CDK5 pathways. CHEMOSPHERE 2024; 358:142124. [PMID: 38677614 DOI: 10.1016/j.chemosphere.2024.142124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 04/29/2024]
Abstract
Metformin, the most commonly prescribed drug for the treatment of diabetes, is increasingly used during pregnancy to address various disorders such as diabetes, obesity, preeclampsia, and metabolic diseases. However, its impact on neocortex development remains unclear. Here, we investigated the direct effects of metformin on neocortex development, focusing on ERK and p35/CDK5 regulation. Using a pregnant rat model, we found that metformin treatment during pregnancy induces small for gestational age (SGA) and reduces relative cortical thickness in embryos and neonates. Additionally, we discovered that metformin inhibits neural progenitor cell proliferation in the sub-ventricular zone (SVZ)/ventricular zone (VZ) of the developing neocortex, a process possibly mediated by ERK inactivation. Furthermore, metformin induces neuronal apoptosis in the SVZ/VZ area of the developing neocortex. Moreover, metformin retards neuronal migration, cortical lamination, and differentiation, potentially through p35/CDK5 inhibition in the developing neocortex. Remarkably, compensating for p35 through in utero electroporation partially rescues metformin-impaired neuronal migration and development. In summary, our study reveals that metformin disrupts neocortex development by inhibiting neuronal progenitor proliferation, neuronal migration, cortical layering, and cortical neuron maturation, likely via ERK and p35/CDK5 inhibition. Consequently, our findings advocate for caution in metformin usage during pregnancy, given its potential adverse effects on fetal brain development.
Collapse
Affiliation(s)
- Muhammet Oner
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Mei-Chih Chen
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Pang-Ting Cheng
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Yu-Hsuan Li
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Yu-Chiao Cheng
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Ayse Celik
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Shiuan-Woei Soong
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Li-Wen Hsu
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Din-You Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | | | - Trayee Dhar
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Hsu-Chen Cheng
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Pin-Chi Tang
- Department of Animal Science, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan.
| |
Collapse
|
3
|
Kubant R, Cho CE, Pannia E, Hammoud R, Yang NV, Simonian R, Anderson GH. Methyl donor micronutrients, hypothalamic development and programming for metabolic disease. Neurosci Biobehav Rev 2024; 157:105512. [PMID: 38128771 DOI: 10.1016/j.neubiorev.2023.105512] [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: 05/18/2023] [Revised: 11/14/2023] [Accepted: 12/17/2023] [Indexed: 12/23/2023]
Abstract
Nutriture in utero is essential for fetal brain development through the regulation of neural stem cell proliferation, differentiation, and apoptosis, and has a long-lasting impact on risk of disease in offspring. This review examines the role of maternal methyl donor micronutrients in neuronal development and programming of physiological functions of the hypothalamus, with a focus on later-life metabolic outcomes. Although evidence is mainly derived from preclinical studies, recent research shows that methyl donor micronutrients (e.g., folic acid and choline) are critical for neuronal development of energy homeostatic pathways and the programming of characteristics of the metabolic syndrome in mothers and their children. Both folic acid and choline are active in one-carbon metabolism with their impact on epigenetic modification of gene expression. We conclude that an imbalance of folic acid and choline intake during gestation disrupts DNA methylation patterns affecting mechanisms of hypothalamic development, and thus elevates metabolic disease risk. Further investigation, including studies to determine translatability to humans, is required.
Collapse
Affiliation(s)
- Ruslan Kubant
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Clara E Cho
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Emanuela Pannia
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Rola Hammoud
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Neil Victor Yang
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Rebecca Simonian
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - G Harvey Anderson
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Lyu R, Wu J, He Y, You Q, Qian Y, Jiang N, Cai Y, Chen D, Wu Z. Folate supports IL-25-induced tuft cell expansion following enteroviral infections. FASEB J 2024; 38:e23430. [PMID: 38243751 DOI: 10.1096/fj.202301928r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/11/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024]
Abstract
Intestinal tuft cells, a kind of epithelial immune cells, rapidly expand in response to pathogenic infections, which is associated with infection-induced interleukin 25 (IL-25) upregulation. However, the metabolic mechanism of IL-25-induced tuft cell expansion is largely unknown. Folate metabolism provides essential purine and methyl substrates for cell proliferation and differentiation. Thus, we aim to investigate the roles of folate metabolism playing in IL-25-induced tuft cell expansion by enteroviral infection and recombinant murine IL-25 (rmIL-25) protein-stimulated mouse models. At present, enteroviruses, such as EV71, CVA16, CVB3, and CVB4, upregulated IL-25 expression and induced tuft cell expansion in the intestinal tissues of mice. However, EV71 did not induce intestinal tuft cell expansion in IL-25-/- mice. Interestingly, compared to the mock group, folate was enriched in the intestinal tissues of both the EV71-infected group and the rmIL-25 protein-stimulated group. Moreover, folate metabolism supported IL-25-induced tuft cell expansion since both folate-depletion and anti-folate MTX-treated mice had a disrupted tuft cell expansion in response to rmIL-25 protein stimulation. In summary, our data suggested that folate metabolism supported intestinal tuft cell expansion in response to enterovirus-induced IL-25 expression, which provided a new insight into the mechanisms of tuft cell expansion from the perspective of folate metabolism.
Collapse
Affiliation(s)
- Ruining Lyu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Jing Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Yating He
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Qiao You
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Yajie Qian
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Na Jiang
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Yurong Cai
- School of Life Science, Ningxia University, Yinchuan, China
| | - Deyan Chen
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Zhiwei Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
- School of Life Science, Ningxia University, Yinchuan, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
5
|
Fardous AM, Heydari AR. Uncovering the Hidden Dangers and Molecular Mechanisms of Excess Folate: A Narrative Review. Nutrients 2023; 15:4699. [PMID: 37960352 PMCID: PMC10648405 DOI: 10.3390/nu15214699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
This review delves into the intricate relationship between excess folate (vitamin B9) intake, especially its synthetic form, namely, folic acid, and its implications on health and disease. While folate plays a pivotal role in the one-carbon cycle, which is essential for DNA synthesis, repair, and methylation, concerns arise about its excessive intake. The literature underscores potential deleterious effects, such as an increased risk of carcinogenesis; disruption in DNA methylation; and impacts on embryogenesis, pregnancy outcomes, neurodevelopment, and disease risk. Notably, these consequences stretch beyond the immediate effects, potentially influencing future generations through epigenetic reprogramming. The molecular mechanisms underlying these effects were examined, including altered one-carbon metabolism, the accumulation of unmetabolized folic acid, vitamin-B12-dependent mechanisms, altered methylation patterns, and interactions with critical receptors and signaling pathways. Furthermore, differences in the effects and mechanisms mediated by folic acid compared with natural folate are highlighted. Given the widespread folic acid supplementation, it is imperative to further research its optimal intake levels and the molecular pathways impacted by its excessive intake, ensuring the health and well-being of the global population.
Collapse
Affiliation(s)
- Ali M. Fardous
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA;
| | - Ahmad R. Heydari
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA;
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
6
|
Tan Z, Li W, Cheng X, Zhu Q, Zhang X. Non-Coding RNAs in the Regulation of Hippocampal Neurogenesis and Potential Treatment Targets for Related Disorders. Biomolecules 2022; 13:biom13010018. [PMID: 36671403 PMCID: PMC9855933 DOI: 10.3390/biom13010018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Non-coding RNAs (ncRNAs), including miRNAs, lncRNAs, circRNAs, and piRNAs, do not encode proteins. Nonetheless, they have critical roles in a variety of cellular activities-such as development, neurogenesis, degeneration, and the response to injury to the nervous system-via protein translation, RNA splicing, gene activation, silencing, modifications, and editing; thus, they may serve as potential targets for disease treatment. The activity of adult neural stem cells (NSCs) in the subgranular zone of the hippocampal dentate gyrus critically influences hippocampal function, including learning, memory, and emotion. ncRNAs have been shown to be involved in the regulation of hippocampal neurogenesis, including proliferation, differentiation, and migration of NSCs and synapse formation. The interaction among ncRNAs is complex and diverse and has become a major topic within the life science. This review outlines advances in research on the roles of ncRNAs in modulating NSC bioactivity in the hippocampus and discusses their potential applications in the treatment of illnesses affecting the hippocampus.
Collapse
Affiliation(s)
- Zhengye Tan
- Department of Anatomy, Institute of Neurobiology, Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Wen Li
- Department of Anatomy, Institute of Neurobiology, Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Xiang Cheng
- Department of Anatomy, Institute of Neurobiology, Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Qing Zhu
- School of Pharmacy, Nantong University, Nantong 226001, China
- Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong 226001, China
| | - Xinhua Zhang
- Department of Anatomy, Institute of Neurobiology, Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
- Central Lab, Yancheng Third People’s Hospital, The Sixth Affiliated Hospital of Nantong University, Yancheng 224001, China
- Correspondence:
| |
Collapse
|
7
|
Xu X, Johnson Z, Wang A, Padget RL, Smyth JW, Xie H. Folate regulates RNA m 5C modification and translation in neural stem cells. BMC Biol 2022; 20:261. [PMID: 36424632 PMCID: PMC9686110 DOI: 10.1186/s12915-022-01467-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Folate is an essential B-group vitamin and a key methyl donor with important biological functions including DNA methylation regulation. Normal neurodevelopment and physiology are sensitive to the cellular folate levels. Either deficiency or excess of folate may lead to neurological disorders. Recently, folate has been linked to tRNA cytosine-5 methylation (m5C) and translation in mammalian mitochondria. However, the influence of folate intake on neuronal mRNA m5C modification and translation remains largely unknown. Here, we provide transcriptome-wide landscapes of m5C modification in poly(A)-enriched RNAs together with mRNA transcription and translation profiles for mouse neural stem cells (NSCs) cultured in three different concentrations of folate. RESULTS NSCs cultured in three different concentrations of folate showed distinct mRNA methylation profiles. Despite uncovering only a few differentially expressed genes, hundreds of differentially translated genes were identified in NSCs with folate deficiency or supplementation. The differentially translated genes induced by low folate are associated with cytoplasmic translation and mitochondrial function, while the differentially translated genes induced by high folate are associated with increased neural stem cell proliferation. Interestingly, compared to total mRNAs, polysome mRNAs contained high levels of m5C. Furthermore, an integrative analysis indicated a transcript-specific relationship between RNA m5C methylation and mRNA translation efficiency. CONCLUSIONS Altogether, our study reports a transcriptome-wide influence of folate on mRNA m5C methylation and translation in NSCs and reveals a potential link between mRNA m5C methylation and mRNA translation.
Collapse
Affiliation(s)
- Xiguang Xu
- Epigenomics and Computational Biology Lab, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, 24061, USA
- Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, VA, 24061, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Zachary Johnson
- Epigenomics and Computational Biology Lab, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, 24061, USA
- Genetics, Bioinformatics and Computational Biology Program, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Amanda Wang
- Epigenomics and Computational Biology Lab, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Rachel L Padget
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, 24061, USA
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, 24016, USA
| | - James W Smyth
- Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, VA, 24061, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, 24061, USA
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
| | - Hehuang Xie
- Epigenomics and Computational Biology Lab, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, 24061, USA.
- Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, VA, 24061, USA.
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA.
- Genetics, Bioinformatics and Computational Biology Program, Virginia Tech, Blacksburg, VA, 24061, USA.
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
8
|
Irvine N, England-Mason G, Field CJ, Dewey D, Aghajafari F. Prenatal Folate and Choline Levels and Brain and Cognitive Development in Children: A Critical Narrative Review. Nutrients 2022; 14:nu14020364. [PMID: 35057545 PMCID: PMC8778665 DOI: 10.3390/nu14020364] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 01/13/2023] Open
Abstract
Women’s nutritional status during pregnancy can have long-term effects on children’s brains and cognitive development. Folate and choline are methyl-donor nutrients and are important for closure of the neural tube during fetal development. They have also been associated with brain and cognitive development in children. Animal studies have observed that prenatal folate and choline supplementation is associated with better cognitive outcomes in offspring and that these nutrients may have interactive effects on brain development. Although some human studies have reported associations between maternal folate and choline levels and child cognitive outcomes, results are not consistent, and no human studies have investigated the potential interactive effects of folate and choline. This lack of consistency could be due to differences in the methods used to assess folate and choline levels, the gestational trimester at which they were measured, and lack of consideration of potential confounding variables. This narrative review discusses and critically reviews current research examining the associations between maternal levels of folate and choline during pregnancy and brain and cognitive development in children. Directions for future research that will increase our understanding of the effects of these nutrients on children’s neurodevelopment are discussed.
Collapse
Affiliation(s)
- Nathalie Irvine
- O’Brien Centre for the Bachelor of Health Sciences, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada;
| | - Gillian England-Mason
- Owerko Centre, Alberta Children’s Hospital Research Institute, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada; (G.E.-M.); (D.D.)
- Department of Pediatrics, Cumming School of Medicine, Alberta Children’s Hospital, 28 Oki Drive NW, Calgary, AB T3B 6A8, Canada
| | - Catherine J. Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 4-126C Li Ka Shing Centre for Research, 11203-87th Ave NW, Edmonton, AB T6G 2H5, Canada;
| | - Deborah Dewey
- Owerko Centre, Alberta Children’s Hospital Research Institute, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada; (G.E.-M.); (D.D.)
- Department of Pediatrics, Cumming School of Medicine, Alberta Children’s Hospital, 28 Oki Drive NW, Calgary, AB T3B 6A8, Canada
- Hotchkiss Brain Institute, Health Research Innovation Centre, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, 3D10, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada
| | - Fariba Aghajafari
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, 3D10, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada
- Department of Family Medicine, Cumming School of Medicine, University of Calgary, G012, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
- Correspondence:
| |
Collapse
|
9
|
Selhub J, Miller JW, Troen AM, Mason JB, Jacques PF. Perspective: The High-Folate-Low-Vitamin B-12 Interaction Is a Novel Cause of Vitamin B-12 Depletion with a Specific Etiology-A Hypothesis. Adv Nutr 2021; 13:16-33. [PMID: 34634124 PMCID: PMC8803489 DOI: 10.1093/advances/nmab106] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/26/2021] [Accepted: 09/02/2021] [Indexed: 11/12/2022] Open
Abstract
Vitamin B-12 is a water-soluble vitamin that plays important roles in intermediary metabolism. Vitamin B-12 deficiency has many identifiable causes, including autoimmune and other gastrointestinal malabsorption disorders, dietary deficiency, and congenital defects in genes that are involved in vitamin B-12 trafficking and functions. Another putative cause of vitamin B-12 deficiency is the high-folate-low vitamin B-12 interaction, first suspected as the cause for observed relapse and exacerbation of the neurological symptoms in patients with pernicious anemia who were prescribed high oral doses of folic acid. We propose that this interaction is real and represents a novel cause of vitamin B-12 depletion with specific etiology. We hypothesize that excessive intake of folic acid depletes serum holotranscobalamin (holoTC), thereby decreasing active vitamin B-12 in the circulation and limiting its availability for tissues. This effect is specific for holoTC and does not affect holohaptocorrin, the inert form of serum vitamin B-12. Depletion of holoTC by folic acid in individuals with already low vitamin B-12 status further compromises the availability of vitamin B-12 coenzymes to their respective enzymes, and consequently a more pronounced state of biochemical deficiency. This hypothesis is drawn from evidence of observational and intervention studies of vitamin B-12-deficient patients and epidemiological cohorts. The evidence also suggests that, in a depleted state, vitamin B-12 is diverted to the hematopoietic system or the kidney. This most likely reflects a selective response of tissues expressing folate receptors with high affinity for unmetabolized folic acid (UMFA; e.g., hematopoietic progenitors and renal tubules) compared with those tissues (e.g., liver) that only express the reduced folate carrier, which is universally expressed but has poor affinity for UMFA. The biochemical and physiological mechanisms underlying this interaction require elucidation to clarify its potential public health significance.
Collapse
Affiliation(s)
| | | | - Aron M Troen
- School of Nutritional Sciences and Institute of Biochemistry Food Science and Nutrition, The Robert H Smith Faculty of Agriculture Food and Environment, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Joel B Mason
- Tufts–USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| | - Paul F Jacques
- Tufts–USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| |
Collapse
|
10
|
Sun J, Zhou X, Wu J, Xiao R, Chen Y, Lu Y, Lang H. Ligustilide enhances hippocampal neural stem cells activation to restore cognitive function in the context of postoperative cognitive dysfunction. Eur J Neurosci 2021; 54:5000-5015. [PMID: 34192824 DOI: 10.1111/ejn.15363] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 05/16/2021] [Accepted: 06/16/2021] [Indexed: 12/27/2022]
Abstract
Ligustilide exerts potential neuroprotective effects against various cerebral ischaemic insults and neurodegenerative disorders. However, the function and mechanisms of LIG-mediated hippocampal neural stem cells (H-NSCs) activation as well as cognitive recovery in the context of post-operative cognitive dysfunction (POCD) remain elusive and need to be explored. Mice were subjected to transient global cerebral ischaemia and reperfusion (tGCI/R) injury and treated with LIG (80 mg/kg) or vehicle for 1 month. Morris water maze test and western blot were employed to assess cognitive function. Nissl staining and immunofluorescence (IF) staining were used to detect H-NSCs proliferation and neurogenesis in hippocampus. Subsequently, primary H-NSCs were treated with LIG, and the level of H-NSCs proliferation and neuronal-differentiation was examined by IF staining for Edu and β-Tubulin III. The protein levels of ERK1/2, β-catenin, NICD, TLR4, Akt and FoxO1 were examined using western blotting. Finally, pretreatment with the ERK agonist SCH772984 was performed to observe the change in ERK expression. LIG treatment promoted H-NSCs proliferation and neurogenesis, increased the number of neurons in the hippocampal subfields, and ultimately reversed cognitive impairment in tGCI/R injury. Furthermore, LIG also promoted primary H-NSCs proliferation and neuronal-differentiation, as well as ERK1/2 phosphorylation. Pretreatment with SCH772984 effectively reversed the ability of LIG to induce ERK1/2 phosphorylation and promote H-NSCs proliferation and neuronal-differentiation. LIG can promote cognitive recovery after tGCI/R injury by activating ERK1/2 in H-NSCs to promote their proliferation and neurogenesis in the hippocampus. Therefore, LIG has potential for use in the prevention and/or treatment of POCD.
Collapse
Affiliation(s)
- Jing Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiang Zhou
- Department of Neurosurgery, The Fifth Affiliated Hospital of Nanchang University, Fuzhou, China
| | - Jusheng Wu
- Department of Anesthesiology, The Zhuji People's Hospital of Zhejiang Province, Zhuji, China
| | - Renjie Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yong Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yimei Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Haili Lang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
11
|
The association between dietary diversity and development among children under 24 months in rural Uganda: analysis of a cluster-randomised maternal education trial. Public Health Nutr 2021; 24:4286-4296. [PMID: 33706831 DOI: 10.1017/s136898002100077x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To assess the association between dietary diversity and development among children under 24 months in rural Uganda and to establish other factors that could be associated with development among these children. DESIGN A secondary data analysis of a cluster-randomised controlled maternal education trial (n 511) was conducted on a sub-sample of 385 children. We used adjusted ORs (AORs) to assess the associations of dietary diversity scores (DDS) and other baseline factors assessed at 6-8 months with child development domains (communication, fine motor, gross motor, personal-social and problem solving) at 20-24 months of age. SETTING Rural areas in Kabale and Kisoro districts of south-western Uganda. PARTICIPANTS Children under 24 months. RESULTS After multivariable analysis, DDS at 6-8 months were positively associated with normal fine motor skills development at 20-24 months (AOR = 1·18; 95 % CI 1·01, 1·37; P = 0·02). No significant association was found between DDS and other development domains. Children who were not ill at 6-8 months had higher odds of developing normal communication (AOR = 1·73; 95 % CI 1·08, 2·77) and gross motor (AOR = 1·91; 95 % CI 1·09, 3·36) skills than sick children. Girls had lower odds of developing normal gross motor skills compared with boys (AOR = 0·58; 95 % CI 0·33, 0·98). Maternal/caregiver nutritional education intervention was positively associated with development of gross motor, fine motor and problem-solving skills (P-values < 0·05). CONCLUSIONS We found an association between child DDS at 6-8 months and improvement in fine motor skills development at 20-24 months. Child illness status, maternal/caregiver nutritional education intervention and sex were other significant baseline predictors of child development at 20-24 months.
Collapse
|
12
|
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
|
13
|
Yang H, Qin D, Xu S, He C, Sun J, Hua J, Peng S. Folic acid promotes proliferation and differentiation of porcine pancreatic stem cells into insulin-secreting cells through canonical Wnt and ERK signaling pathway. J Steroid Biochem Mol Biol 2021; 205:105772. [PMID: 33091596 DOI: 10.1016/j.jsbmb.2020.105772] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/29/2020] [Accepted: 10/04/2020] [Indexed: 11/16/2022]
Abstract
Porcine pancreatic stem cells (pPSCs) can be induced to differentiate into insulin-producing cells in vitro and thus serve as a major cells source for β-cell regeneration. However, this application is limited by the weak cell proliferation ability and low insulin induction efficiency. In this study, we explored the role of folic acid in the proliferation of pPSCs and the formation of insulin-secreting cells. We found that FA-treated pPSCs cells had a high EDU positive rate, and the proliferation marker molecules PCNA, CyclinD1 and c-Myc were up-regulated, while the expression of folate receptor α (FOLRα) was up-regulated. In further research, interference FOLRα or adding canonical Wnt signaling pathway or ERK signaling pathway inhibitors could significantly inhibit the effect of FA on pPSCs proliferation. Meanwhile, during the differentiation of pPSCs into insulin-secreting cells, we found that the maturation marker genes Insulin, NKX6.1, MafA, and NeuroD1 was upregulated in insulin-secreting cell masses differentiationed from pPSCs after FA treatment, and the functional molecules Insulin and C-peptide were increased, the ability to secrete insulin in response to high glucose was also increased. With the addition of Wnt and ERK signaling pathway inhibitors, the pro-differentiation effect of FA was weakened. In conclusion, FA promotes the proliferation of pPSCs by binding to folate receptor α (FOLRα) and increase the efficiency of directed differentiation of pPSCs into insulin-producing cells by regulating canonical Wnt and ERK signaling pathway. This study lays theoretical foundation for solving the bottleneck in the treatment of diabetes with stem cell transplantation in future.
Collapse
Affiliation(s)
- Hong Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Dezhe Qin
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Shuanshuan Xu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Chen He
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Jing Sun
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Sha Peng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi, 712100, PR China.
| |
Collapse
|
14
|
Maruvada P, Stover PJ, Mason JB, Bailey RL, Davis CD, Field MS, Finnell RH, Garza C, Green R, Gueant JL, Jacques PF, Klurfeld DM, Lamers Y, MacFarlane AJ, Miller JW, Molloy AM, O'Connor DL, Pfeiffer CM, Potischman NA, Rodricks JV, Rosenberg IH, Ross SA, Shane B, Selhub J, Stabler SP, Trasler J, Yamini S, Zappalà G. Knowledge gaps in understanding the metabolic and clinical effects of excess folates/folic acid: a summary, and perspectives, from an NIH workshop. Am J Clin Nutr 2020; 112:1390-1403. [PMID: 33022704 PMCID: PMC7657327 DOI: 10.1093/ajcn/nqaa259] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022] Open
Abstract
Folate, an essential nutrient found naturally in foods in a reduced form, is present in dietary supplements and fortified foods in an oxidized synthetic form (folic acid). There is widespread agreement that maintaining adequate folate status is critical to prevent diseases due to folate inadequacy (e.g., anemia, birth defects, and cancer). However, there are concerns of potential adverse effects of excess folic acid intake and/or elevated folate status, with the original concern focused on exacerbation of clinical effects of vitamin B-12 deficiency and its role in neurocognitive health. More recently, animal and observational studies have suggested potential adverse effects on cancer risk, birth outcomes, and other diseases. Observations indicating adverse effects from excess folic acid intake, elevated folate status, and unmetabolized folic acid (UMFA) remain inconclusive; the data do not provide the evidence needed to affect public health recommendations. Moreover, strong biological and mechanistic premises connecting elevated folic acid intake, UMFA, and/or high folate status to adverse health outcomes are lacking. However, the body of evidence on potential adverse health outcomes indicates the need for comprehensive research to clarify these issues and bridge knowledge gaps. Three key research questions encompass the additional research needed to establish whether high folic acid or total folate intake contributes to disease risk. 1) Does UMFA affect biological pathways leading to adverse health effects? 2) Does elevated folate status resulting from any form of folate intake affect vitamin B-12 function and its roles in sustaining health? 3) Does elevated folate intake, regardless of form, affect biological pathways leading to adverse health effects other than those linked to vitamin B-12 function? This article summarizes the proceedings of an August 2019 NIH expert workshop focused on addressing these research areas.
Collapse
Affiliation(s)
- Padma Maruvada
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Patrick J Stover
- Texas A&M University College of Agriculture and Life Sciences, Texas A&M University AgriLife, College Station, TX, USA
| | - Joel B Mason
- Jean Mayer USDA Human Nutrition Research Center on Aging, Friedman School of Nutrition Science and Policy, and School of Medicine, Tufts University, Boston, MA, USA
| | - Regan L Bailey
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| | - Cindy D Davis
- Office of Dietary Supplements, NIH, Bethesda, MD, USA
| | - Martha S Field
- Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, NY, USA
| | - Richard H Finnell
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Cutberto Garza
- Professor Emeritus, Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, NY, USA
| | - Ralph Green
- Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, Sacramento, CA, USA
| | - Jean-Louis Gueant
- University of Lorraine and University Regional Hospital Centre of Nancy, Nancy, France
| | - Paul F Jacques
- Tufts University Friedman School of Nutritional Science and Policy and the Jean Mayer USDA Human Nutrition Research Center, Boston, MA, USA
| | - David M Klurfeld
- Department of Nutrition, Food Safety, and Quality, USDA Agricultural Research Service, Beltsville, MD, USA
| | - Yvonne Lamers
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Joshua W Miller
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Anne M Molloy
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Deborah L O'Connor
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | - Irwin H Rosenberg
- Jean Mayer USDA Human Nutrition Research Center on Aging, Friedman School of Nutrition Science and Policy, and School of Medicine, Tufts University, Boston, MA, USA
| | | | - Barry Shane
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Jacob Selhub
- Tufts University Friedman School of Nutritional Science and Policy and the Jean Mayer USDA Human Nutrition Research Center, Boston, MA, USA
| | - Sally P Stabler
- Department of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Sedigheh Yamini
- Office of Nutrition and Food Labeling, Center for Food Safety and Applied Nutrition, US FDA, College Park, MD, USA
| | - Giovanna Zappalà
- Division of Geriatrics and Clinical Gerontology, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
Wang X, Li W, Li Z, Ma Y, Yan J, Wilson JX, Huang G. Maternal Folic Acid Supplementation During Pregnancy Promotes Neurogenesis and Synaptogenesis in Neonatal Rat Offspring. Cereb Cortex 2020; 29:3390-3397. [PMID: 30137237 DOI: 10.1093/cercor/bhy207] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/08/2018] [Indexed: 12/25/2022] Open
Abstract
Maternal folic acid supplementation during pregnancy is associated with improved cognitive performances in offspring. However, the effect of supplementation on offspring's neurogenesis and synaptogenesis is unknown, and whether supplementation should be continued throughout pregnancy is controversial. In present study, 3 groups of female rats were fed a folate-normal diet, folate-deficient diet, or folate-supplemented diet from 1 week before mating until the end of pregnancy. A fourth group fed folate-normal diet from 1 week before mating until mating, then fed folate-supplemented diet for 10 consecutive days, then fed folate-normal diet until the end of pregnancy. Offspring were sacrificed on postnatal day 0 for measurement of neurogenesis and synaptogenesis by immunofluorescence and western blot. Additionally neural stem cells (NSCs) were cultured from offspring's hippocampus for immunocytochemical measurement of their rates of proliferation and neuronal differentiation. The results demonstrated that maternal folic acid supplementation stimulated hippocampal neurogenesis by increasing proliferation and neuronal differentiation of NSCs, and also enhanced synaptogenesis in cerebral cortex of neonatal offspring. Hippocampal neurogenesis was stimulated more when supplementation was continued throughout pregnancy instead of being limited to the periconceptional period. In conclusion, maternal folic acid supplementation, especially if continued throughout pregnancy, improves neurogenesis and synaptogenesis in neonatal offspring.
Collapse
Affiliation(s)
- Xinyan Wang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Wen Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Zhenshu Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yue Ma
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jing Yan
- Department of Social Medicine and Health Administration, School of Public Health, Tianjin Medical University, Tianjin, China
| | - John X Wilson
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Guowei Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China
| |
Collapse
|
16
|
Yu Y, Wang J, Kaul SC, Wadhwa R, Miyako E. Folic Acid Receptor-Mediated Targeting Enhances the Cytotoxicity, Efficacy, and Selectivity of Withania somnifera Leaf Extract: In vitro and in vivo Evidence. Front Oncol 2019; 9:602. [PMID: 31334122 PMCID: PMC6621239 DOI: 10.3389/fonc.2019.00602] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/18/2019] [Indexed: 11/17/2022] Open
Abstract
Nanomedicine holds great potential for drug delivery to achieve more effective and safer cancer treatment. Earlier, we reported that the alcoholic extract of Withania somnifera leaves (i-Extract) has selective cancer cell killing activity. Herein, we developed a folate receptor-targeting i-Extract nanocomplex (FRi-ExNC) that suspends well in water and possesses enhanced selective anticancer activity in both in vitro and in vivo assays. Comparative analyses of folate receptor (FR)-positive and -negative cells revealed that FRi-ExNC caused a stronger decrease in Cyclin D/Cdk4 and anti-apoptotic protein Bcl-2, as well as a higher increase in the growth arrest regulating protein p21WAF1 and pro-apoptotic protein PARP-1, in FR-enriched cancer cells. Our results demonstrate that FRi-ExNC could be a natural source-based nanomedicine for targeted cancer therapy.
Collapse
Affiliation(s)
- Yue Yu
- Department of Materials and Chemistry, Nanomaterials Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Jia Wang
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), AIST, Tsukuba, Japan.,Graduate School of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan
| | - Sunil C Kaul
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), AIST, Tsukuba, Japan
| | - Renu Wadhwa
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), AIST, Tsukuba, Japan
| | - Eijiro Miyako
- Department of Materials and Chemistry, Nanomaterials Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| |
Collapse
|
17
|
Craenen K, Verslegers M, Baatout S, Abderrafi Benotmane M. An appraisal of folates as key factors in cognition and ageing-related diseases. Crit Rev Food Sci Nutr 2019; 60:722-739. [PMID: 30729795 DOI: 10.1080/10408398.2018.1549017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Folic acid (FA) is often consumed as a food supplement and can be found in fortified staple foods in various western countries. Even though FA supplementation during pregnancy is known to prevent severe congenital anomalies in the developing child (e.g., neural tube defects), much less is known about its influence on cognition and neurological functioning. In this review, we address the advances in this field and situate how folate intake during pregnancy, postnatal life, adulthood and in the elderly affects cognition. In addition, an association between folate status and ageing, dementia and other neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis is discussed. While its role in the incidence and severity of these diseases is becoming apparent, the underlying action of folates and related metabolites remains elusive. Finally, the potential of FA as a nutraceutical has been proposed, although the efficacy will highly depend on the interplay with other micronutrients, the disease stage and the duration of supplementation. Hence, the lack of consistent data urges for more animal studies and (pre)clinical trials in humans to ascertain a potential beneficial role for folates in the treatment or amelioration of cognitive decline and ageing-related disorders.
Collapse
Affiliation(s)
- Kai Craenen
- Radiobiology Unit, Belgian Nuclear Research Centre SCK•CEN, Mol, Belgium.,Biology Department, Research Group Neural Circuit Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Mieke Verslegers
- Radiobiology Unit, Belgian Nuclear Research Centre SCK•CEN, Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Belgian Nuclear Research Centre SCK•CEN, Mol, Belgium
| | | |
Collapse
|
18
|
Kim GB, Chen Y, Kang W, Guo J, Payne R, Li H, Wei Q, Baker J, Dong C, Zhang S, Wong PK, Rizk EB, Yan J, Yang J. The critical chemical and mechanical regulation of folic acid on neural engineering. Biomaterials 2018; 178:504-516. [PMID: 29657092 PMCID: PMC6328061 DOI: 10.1016/j.biomaterials.2018.03.059] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 03/24/2018] [Accepted: 03/31/2018] [Indexed: 12/29/2022]
Abstract
The mandate of folic acid supplementation in grained products has reduced the occurrence of neural tube defects by one third in the U.S since its introduction by the Food and Drug Administration in 1998. However, the advantages and possible mechanisms of action of using folic acid for peripheral nerve engineering and neurological diseases still remain largely elusive. Herein, folic acid is described as an inexpensive and multifunctional niche component that modulates behaviors in different cells in the nervous system. The multiple benefits of modulation include: 1) generating chemotactic responses on glial cells, 2) inducing neurotrophin release, and 3) stimulating neuronal differentiation of a PC-12 cell system. For the first time, folic acid is also shown to enhance cellular force generation and global methylation in the PC-12 cells, thereby enabling both biomechanical and biochemical pathways to regulate neuron differentiation. These findings are evaluated in vivo for clinical translation. Our results suggest that folic acid-nerve guidance conduits may offer significant benefits as a low-cost, off-the-shelf product for reaching the functional recovery seen with autografts in large sciatic nerve defects. Consequently, folic acid holds great potential as a critical and convenient therapeutic intervention for neural engineering, regenerative medicine, medical prosthetics, and drug delivery.
Collapse
Affiliation(s)
- Gloria B Kim
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Yongjie Chen
- Department of Orthopedic Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, PR China
| | - Weibo Kang
- Department of Orthopedic Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, PR China
| | - Jinshan Guo
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Russell Payne
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Hui Li
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Qiong Wei
- Department of Engineering Science and Mechanics, Materials Research Institute, The Pennsylvania State University, University Park, PA 16802, USA
| | - Julianne Baker
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Cheng Dong
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Sulin Zhang
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Pak Kin Wong
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Elias B Rizk
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Jiazhi Yan
- Department of Orthopedic Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, PR China.
| | - Jian Yang
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
19
|
Gong L, Cao L, Shen Z, Shao L, Gao S, Zhang C, Lu J, Li W. Materials for Neural Differentiation, Trans-Differentiation, and Modeling of Neurological Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1705684. [PMID: 29573284 DOI: 10.1002/adma.201705684] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/04/2017] [Indexed: 05/02/2023]
Abstract
Neuron regeneration from pluripotent stem cells (PSCs) differentiation or somatic cells trans-differentiation is a promising approach for cell replacement in neurodegenerative diseases and provides a powerful tool for investigating neural development, modeling neurological diseases, and uncovering the mechanisms that underlie diseases. Advancing the materials that are applied in neural differentiation and trans-differentiation promotes the safety, efficiency, and efficacy of neuron regeneration. In the neural differentiation process, matrix materials, either natural or synthetic, not only provide a structural and biochemical support for the monolayer or three-dimensional (3D) cultured cells but also assist in cell adhesion and cell-to-cell communication. They play important roles in directing the differentiation of PSCs into neural cells and modeling neurological diseases. For the trans-differentiation of neural cells, several materials have been used to make the conversion feasible for future therapy. Here, the most current applications of materials for neural differentiation for PSCs, neuronal trans-differentiation, and neurological disease modeling is summarized and discussed.
Collapse
Affiliation(s)
- Lulu Gong
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Lining Cao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhenmin Shen
- The VIP Department, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Li Shao
- The VIP Department, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Shaorong Gao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Chao Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jianfeng Lu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Weida Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| |
Collapse
|
20
|
Folic acid inhibits nasopharyngeal cancer cell proliferation and invasion via activation of FRα/ERK1/2/TSLC1 pathway. Biosci Rep 2017; 37:BSR20170772. [PMID: 29070520 PMCID: PMC5705775 DOI: 10.1042/bsr20170772] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 10/19/2017] [Accepted: 10/23/2017] [Indexed: 12/26/2022] Open
Abstract
Folic acid (FA), which is necessary for normal cell division of mammals, has been implicated to be involved in many tumors. Dietary FA intake has been reported to be associated with a lower risk of nasopharyngeal cancer (NPC). However, the molecular mechanisms of FA in NPC cells remain unclear. In the present study, we found that FA treatment dose dependently inhibited the proliferation, invasion and migration of NPC cells, via folate receptor α (FRα). We further found that FA, bound to FRα, induced the activation of MEK/ERK1/2, and increased the expressions of TSLC1 and E-cadherin. Moreover, blocking of ERK1/2 activation attenuated FA-mediated increase in TSLC1 expression. In addition, knockdown of TSLC1 abolished the FA-mediated inhibition of cell proliferation, invasion and migration, and suppressed the FA-mediated increase oinE-cadherin expression in NPC cells. Taken together, our data suggest that FA treatment inhibits NPC cell proliferation and invasion via activation of FRα/ERK1/2/ TSLC1 signaling pathway. Therefore, FA could be explored as a therapeutic drug for the treatment of NPC, and TSLC1 may act as a tumor suppressor in NPC.
Collapse
|
21
|
Chaudhari SN, Mukherjee M, Vagasi AS, Bi G, Rahman MM, Nguyen CQ, Paul L, Selhub J, Kipreos ET. Bacterial Folates Provide an Exogenous Signal for C. elegans Germline Stem Cell Proliferation. Dev Cell 2017; 38:33-46. [PMID: 27404357 DOI: 10.1016/j.devcel.2016.06.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 05/04/2016] [Accepted: 06/09/2016] [Indexed: 11/29/2022]
Abstract
Here we describe an in vitro primary culture system for Caenorhabditis elegans germline stem cells. This culture system was used to identify a bacterial folate as a positive regulator of germ cell proliferation. Folates are a family of B-complex vitamins that function in one-carbon metabolism to allow the de novo synthesis of amino acids and nucleosides. We show that germ cell proliferation is stimulated by the folate 10-formyl-tetrahydrofolate-Glun both in vitro and in animals. Other folates that can act as vitamins to rescue folate deficiency lack this germ cell stimulatory activity. The bacterial folate precursor dihydropteroate also promotes germ cell proliferation in vitro and in vivo, despite its inability to promote one-carbon metabolism. The folate receptor homolog FOLR-1 is required for the stimulation of germ cells by 10-formyl-tetrahydrofolate-Glun and dihydropteroate. This work defines a folate and folate-related compound as exogenous signals to modulate germ cell proliferation.
Collapse
Affiliation(s)
- Snehal N Chaudhari
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | | | - Alexandra S Vagasi
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Gaofeng Bi
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Mohammad M Rahman
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Christine Q Nguyen
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Ligi Paul
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Jacob Selhub
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Edward T Kipreos
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
22
|
Components of the folate metabolic pathway and ADHD core traits: an exploration in eastern Indian probands. J Hum Genet 2017; 62:687-695. [PMID: 28250422 DOI: 10.1038/jhg.2017.23] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 01/11/2023]
Abstract
We investigated role of the folate-homocysteine metabolic pathway in the etiology of attention-deficit hyperactivity disorder (ADHD) due to its importance in maintaining DNA integrity as well as neurotransmission. Functional gene variants in MTR (rs1805087), CBS (rs5742905), MTHFR (rs1801133 &rs1801131), MTHFD (rs2236225), RFC1 (rs1051266), plasma vitamin B12, folate and homocysteine were analyzed. rs1805087 'A' showed strong association with ADHD. Vitamin B12 deficiency of ADHD probands (P=0.01) correlated with rs1801133 'T' and rs1805087'GG'. Mild hyperhomocysteinemia (P=0.05) in the probands was associated with rs1805087 'AA'. Probands having rs1805087 'GG' and rs1051266 'G' was more inattentive. Hyperactivity-impulsivity score revealed association with rs5742905 'TT' and rs2236225 'CC', while rs1801133 'CC' showed association with inattentiveness and hyperactivity-impulsivity. rs1801131 exhibited strong synergistic interaction with rs1051266 and rs2236225. This indicated that the folate-homocysteine pathway gene variants may affect ADHD etiology through mild hyperhomocysteinemia and vitamin B12 deficiency, factors known to be associated with cognitive deficit.
Collapse
|
23
|
Tavakol S, Mousavi SMM, Tavakol B, Hoveizi E, Ai J, Sorkhabadi SMR. Mechano-Transduction Signals Derived from Self-Assembling Peptide Nanofibers Containing Long Motif of Laminin Influence Neurogenesis in In-Vitro and In-Vivo. Mol Neurobiol 2016; 54:2483-2496. [DOI: 10.1007/s12035-016-9836-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/04/2016] [Indexed: 01/01/2023]
|
24
|
Tavakol S, Saber R, Hoveizi E, Tavakol B, Aligholi H, Ai J, Rezayat SM. Self-Assembling Peptide Nanofiber Containing Long Motif of Laminin Induces Neural Differentiation, Tubulin Polymerization, and Neurogenesis: In Vitro, Ex Vivo, and In Vivo Studies. Mol Neurobiol 2015; 53:5288-99. [DOI: 10.1007/s12035-015-9448-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 09/16/2015] [Indexed: 01/03/2023]
|
25
|
Hansen MF, Greibe E, Skovbjerg S, Rohde S, Kristensen ACM, Jensen TR, Stentoft C, Kjær KH, Kronborg CS, Martensen PM. Folic acid mediates activation of the pro-oncogene STAT3 via the Folate Receptor alpha. Cell Signal 2015; 27:1356-68. [PMID: 25841994 DOI: 10.1016/j.cellsig.2015.03.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 03/14/2015] [Accepted: 03/29/2015] [Indexed: 01/01/2023]
Abstract
The signal transducer and activator of transcription 3 (STAT3) is a well-described pro-oncogene found constitutively activated in several cancer types. Folates are B vitamins that, when taken up by cells through the Reduced Folate Carrier (RFC), are essential for normal cell growth and replication. Many cancer cells overexpress a glycophosphatidylinositol (GPI)-anchored Folate Receptor α (FRα). The function of FRα in cancer cells is still poorly described, and it has been suggested that transport of folate is not its primary function in these cells. We show here that folic acid and folinic acid can activate STAT3 through FRα in a Janus Kinase (JAK)-dependent manner, and we demonstrate that gp130 functions as a transducing receptor for this signalling. Moreover, folic acid can promote dose dependent cell proliferation in FRα-positive HeLa cells, but not in FRα-negative HEK293 cells. After folic acid treatment of HeLa cells, up-regulation of the STAT3 responsive genes Cyclin A2 and Vascular Endothelial Growth Factor (VEGF) were verified by qRT-PCR. The identification of this FRα-STAT3 signal transduction pathway activated by folic and folinic acid contributes to the understanding of the involvement of folic acid in preventing neural tube defects as well as in tumour growth. Previously, the role of folates in these diseases has been attributed to their roles as one-carbon unit donors following endocytosis into the cell. Our finding that folic acid can activate STAT3 via FRα adds complexity to the established roles of B9 vitamins in cancer and neural tube defects.
Collapse
Affiliation(s)
- Mariann F Hansen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Eva Greibe
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Signe Skovbjerg
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Sarah Rohde
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Anders C M Kristensen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Trine R Jensen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Charlotte Stentoft
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Karina H Kjær
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Camilla S Kronborg
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Pia M Martensen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|
26
|
Suppressed expression of mitogen-activated protein kinases in hyperthermia induced defective neural tube. Neurosci Lett 2015; 594:6-11. [PMID: 25818329 DOI: 10.1016/j.neulet.2015.03.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/05/2015] [Accepted: 03/23/2015] [Indexed: 11/23/2022]
Abstract
Neural tube defects (NTDs) are common congenital malformations. Mitogen-activated protein kinases (MAPKs) pathway is involved in many physiological processes. HMGB1 has been showed closely associated with neurulation and NTDs induced by hyperthermia and could activate MAPKs pathway. Since hyperthermia caused increased activation of MAPKs in many systems, the present study aims to investigate whether HMGB1 contributes to hyperthermia induced NTDs through MAPKs pathway. The mRNA levels of MAPKs and HMGB1 between embryonic day 8.5 and 10 (E8.5-10) in hyperthermia induced defective neural tube were detected by real-time quantitative polymerase chain reaction (qPCR). By immunofluorescence and western blotting, the expressions of HMGB1 and phosphorylated MAPKs (ERK1/2, JNK and p38) in neural tubes after hyperthermia were studied. The mRNA levels of MAPKs and HMGB1, as well as the expressions of HMGB1 along with phosphorylated JNK, p38 and ERK, were downregulated in NTDs groups induced by hyperthermia compared with control. The findings suggested that HMGB1 may contribute to hyperthermia induced NTDs formation through decreased cell proliferation due to inhibited phosphorylated ERK1/2 MAPK.
Collapse
|
27
|
Rodriguez JM, Miranda D, Bunout D, Ronco AM, de la Maza MP, Hirsch S. Folates Induce Colorectal Carcinoma HT29 Cell Line Proliferation Through Notch1 Signaling. Nutr Cancer 2015; 67:706-11. [PMID: 25802929 DOI: 10.1080/01635581.2015.1011285] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Folic acid (FA) consumption at high levels has been associated with colon cancer risk. Several mechanisms have been proposed to explain this association. The Notch signal pathway has been implicated in the regulation of cellular proliferation. Our aim was to demonstrate that high concentrations of FA or its reduced form, 5-methyltetrahydrofolic acid (5-MTHF), increase colorectal carcinoma HT29 cell proliferation through an increase of Notch1 activation and to prove if the inhibition of Notch1 activation by gamma secretase inhibitor, reduce the effect of folic acid. HT29 cells were cultured in high (400 nM), low (20 nM), or 0 nM FA or 5-MTHF concentrations during 96 h with or without DAPT (gamma secretase inhibitor). Cell proliferation was determined by the methylthiazole tetrazolium method, and Notch1-intracellular domain (NICD) was analyzed by flow cytometry. HT29 cells exposed to 400 nM FA or 5-MTHF showed higher proliferation rate than those exposed to 20 nM of FA or 5-MTHF (P < 0.01) during 96 h. NICD expression increased at higher FA or 5-MTHF concentrations compared with lower concentrations (P < 0.01). This effect on proliferation was partially reversible when we blocked Notch1 activation with the inhibitor of γ-secretase (P < 0.05).These data suggest that high concentration of FA and 5-MTHF induce HT29 cell proliferation activating Notch1 pathway.
Collapse
Affiliation(s)
- Juan M Rodriguez
- a Institute of Nutrition and Food Technology , University of Chile , Santiago , Chile
| | | | | | | | | | | |
Collapse
|
28
|
JIN LEI, ZHAO JIAN, JING WENSEN, YAN SHIJU, WANG XIN, XIAO CHUN, MA BAOAN. Role of miR-146a in human chondrocyte apoptosis in response to mechanical pressure injury in vitro. Int J Mol Med 2014; 34:451-63. [PMID: 24939082 PMCID: PMC4094584 DOI: 10.3892/ijmm.2014.1808] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 06/11/2014] [Indexed: 02/04/2023] Open
Abstract
MicroRNA (miR)-146a is known to be overexpressed in osteoarthritis (OA). However, the role of miR-146a in OA has not yet been fully elucidated. In the present study, we applied mechanical pressure of 10 MPa to human chondrocytes for 60 min in order to investigate the expression of miR-146a and apoptosis following the mechanical pressure injury. Normal human chondrocytes were transfected with an miR-146a mimic or an inhibitor to regulate miR-146a expression. Potential target genes of miR-146a were predicted using bioinformatics. Moreover, luciferase reporter assay confirmed that Smad4 was a direct target of miR-146a. The expression levels of miR-146a, Smad4 and vascular endothelial growth factor (VEGF) were quantified by quantitative reverse transcription PCR and/or western blot analysis. The effects of miR-146a on apoptosis were detected by Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) flow cytometry. The results indicated that mechanical pressure affected chondrocyte viability and induced the early apoptosis of chondrocytes. Mechanical pressure injury increased the expression levels of miR-146a and VEGF and decreased the levels of Smad4 in the chondrocytes. In the human chondrocytes, the upregulation of miR-146a induced apoptosis, upregulated VEGF expression and downregulated Smad4 expression. In addition, the knockdown of miR-146a reduced cell apoptosis, upregulated Smad4 expression and downregulated VEGF expression. Smad4 was identified as a direct target of miR-146a by harboring a miR‑146a binding sequence in the 3'-untranslated region (3'-UTR) of its mRNA. Furthermore, the upregulation of VEGF induced by miR‑146a was mediated by Smad4 in the chondrocytes subjected to mechanical pressure injury. These results demonstrated that miR-146a was overexpressed in our chondrocyte model of experimentally induced human mechanical injury, accompanied by the upregulation of VEGF and the downregulation of Smad4 in vitro. Moreover, our data suggest that miR-146a is involved in human chondrocyte apoptosis in response to mechanical injury, and may contribute to the mechanical injury of chondrocytes, as well as to the pathogenesis of OA by increasing the levels of VEGF and damaging the transforming growth factor (TGF)-β signaling pathway through the targeted inhibition of Smad4 in cartilage.
Collapse
Affiliation(s)
| | | | | | - SHIJU YAN
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710038, P.R. China
| | - XIN WANG
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710038, P.R. China
| | - CHUN XIAO
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710038, P.R. China
| | - BAOAN MA
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710038, P.R. China
| |
Collapse
|
29
|
Saha T, Dutta S, Rajamma U, Sinha S, Mukhopadhyay K. A pilot study on the contribution of folate gene variants in the cognitive function of ADHD probands. Neurochem Res 2014; 39:2058-67. [PMID: 25079255 DOI: 10.1007/s11064-014-1393-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 07/15/2014] [Accepted: 07/17/2014] [Indexed: 11/28/2022]
Abstract
Genetic abnormalities in components important for the folate cycle confer risk for various disorders since adequate folate turnover is necessary for normal methylation, gene expression and chromosome structure. However, the system has rarely been studied in children diagnosed with attention deficit hyperactivity disorder (ADHD). We hypothesized that ADHD related cognitive deficit could be attributed to abnormalities in the folate cycle and explored functional single nucleotide polymorphisms in methylenetetrahydrofolate dehydrogenase (rs2236225), reduced folate carrier (rs1051266), and methylenetetrahydrofolate reductase (rs1801131 and rs1801133) in families with ADHD probands (N = 185) and ethnically matched controls (N = 216) recruited following the DSM-IV. After obtaining informed written consent for participation, peripheral blood was collected for genomic DNA isolation and PCR-based analysis of target sites. Data obtained was analyzed by UNPHASED. Interaction between sites was analyzed by the multi dimensionality reduction (MDR) program. Genotypic frequencies of the Indian population were strikingly different from other ethnic groups. rs1801133 "T" allele showed biased transmission in female probands (p < 0.05). Significant difference in genotypic frequencies for female probands was also noticed. rs1801131 and rs1801133 showed an association with low intelligence quotient (IQ). MDR analysis exhibited independent effects and contribution of these sites to IQ, thus indicating a role of these genes in ADHD related cognitive deficit.
Collapse
Affiliation(s)
- T Saha
- Manovikas Biomedical Research and Diagnostic Centre, 482, Madudah, Plot I-24, Sec.-J, E.M. Bypass, Kolkata, 700107, India
| | | | | | | | | |
Collapse
|
30
|
Yan H, Zhang X, Luo S, Liu H, Wang X, Gao Y, Wilson JX, Huang G. Effects of homocysteine on ERK signaling and cell proliferation in fetal neural stem cells in vitro. Cell Biochem Biophys 2013; 66:131-7. [PMID: 23109178 DOI: 10.1007/s12013-012-9461-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The aim of the present study was to determine if the excitatory amino acid homocysteine (Hcy) alters ERK signaling and cell proliferation in fetal neural stem cells (NSCs) in vitro. NSCs were isolated from fetal rats and grown in serum-free suspension medium. The cells were identified as NSCs by their expression of immunoreactive Sox2. NSCs were assigned to one of four treatment groups: vehicle control, low-dose Hcy group (Hcy-L, medium contained 30 μmol/L Hcy), middle-dose Hcy group (Hcy-M, 100 μmol/L Hcy) and high-dose Hcy group (Hcy-H, 300 μmol/L Hcy). Cell proliferation was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Protein expression levels of ERK1/2 and phosphorylated ERK1/2 were detected by Western blot. The effects of Hcy on NSC death, including apoptosis, were assessed by using flow cytometry and trypan blue exclusion. The results showed that NSCs grew as neurospheres in the serum-free medium. Hcy decreased ERK1/2 protein phosphorylation and NSC proliferation, but it did not induce cell death or apoptosis within the concentration from 30 to 300 μmol/L. The above results are consistent with the hypothesis that Hcy decreases fetal NSC proliferation by inhibiting ERK signaling.
Collapse
Affiliation(s)
- Hai Yan
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Self-assembling nanofibers improve cognitive impairment in a transgenic mice model of Alzheimer's disease. Neurosci Lett 2013; 556:63-8. [DOI: 10.1016/j.neulet.2013.09.063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 09/11/2013] [Accepted: 09/26/2013] [Indexed: 11/17/2022]
|
32
|
Pang W, Leng X, Lu H, Yang H, Song N, Tan L, Jiang Y, Guo C. Depletion of intracellular zinc induces apoptosis of cultured hippocampal neurons through suppression of ERK signaling pathway and activation of caspase-3. Neurosci Lett 2013; 552:140-5. [PMID: 23954826 DOI: 10.1016/j.neulet.2013.07.057] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 07/15/2013] [Accepted: 07/31/2013] [Indexed: 11/19/2022]
Abstract
Although Zinc depletion induces apoptosis in different cells and tissues, exact mechanism of this action of zinc depletion is not completely understood. In our previous study, the results suggested that the significant down-regulation of MEK/ERK signaling pathway was observed in zinc deficiency neurons. Here, we investigate whether, in hippocampal neurons, this increased rate of apoptosis induced by zinc depletion is the result of hypophosphorylation of ERK pathway. In this study, we found that NGF, ERK agonist, prevented neurons against TPEN-induced apoptosis, whereas TPEN-induced apoptosis was potentiated by U0126, inhibitors of ERK. Moreover, TPEN-induced caspase-3 activity was further increased by the pretreatment with U0126, but it was further decreased by the pretreatment with NGF. However, pretreatment of the cells with U0126 or NGF had no effect on the changes of Bcl-2 and Bax protein expression induced by zinc depletion. Thus, the results indicate that TPEN induces apoptosis of hippocampal neurons through inhibition of ERK and, in turn, activation of caspase-3.
Collapse
Affiliation(s)
- Wei Pang
- Department of Nutrition, Institute of Hygiene & Environmental Medicine, Academy of Military Medical Science, Tianjin, China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Guéant JL, Namour F, Guéant-Rodriguez RM, Daval JL. Folate and fetal programming: a play in epigenomics? Trends Endocrinol Metab 2013; 24:279-89. [PMID: 23474063 DOI: 10.1016/j.tem.2013.01.010] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 01/27/2013] [Accepted: 01/30/2013] [Indexed: 01/12/2023]
Abstract
Folate plays a key role in the interactions between nutrition, fetal programming, and epigenomics. Maternal folate status influences DNA methylation, inheritance of the agouti phenotype, expression of imprinting genes, and the effects of mycotoxin FB1 on heterochromatin assembly in rodent offspring. Deficiency in folate and other methyl donors increases birth defects and produces visceral manifestations of fetal programming, including liver and heart steatosis, through imbalanced methylation and acetylation of PGC1-α and decreased SIRT1 expression, and produces persistent cognitive and learning disabilities through impaired plasticity and hippocampal atrophy. Maternal folate supplementation also produces long-term epigenomic effects in offspring, some beneficial and others negative. Deciphering these mechanisms will help understanding the discordances between experimental models and population studies of folate deficiency and supplementation.
Collapse
Affiliation(s)
- Jean-Louis Guéant
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 954, Department of Nutrition-Genetics-Environmental Risk Exposure, University of Lorraine and University Hospital of Nancy, Vandoeuvre-lès-Nancy, France.
| | | | | | | |
Collapse
|
34
|
Arigony ALV, de Oliveira IM, Machado M, Bordin DL, Bergter L, Prá D, Pêgas Henriques JA. The influence of micronutrients in cell culture: a reflection on viability and genomic stability. BIOMED RESEARCH INTERNATIONAL 2013; 2013:597282. [PMID: 23781504 PMCID: PMC3678455 DOI: 10.1155/2013/597282] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 04/23/2013] [Accepted: 05/03/2013] [Indexed: 12/31/2022]
Abstract
Micronutrients, including minerals and vitamins, are indispensable to DNA metabolic pathways and thus are as important for life as macronutrients. Without the proper nutrients, genomic instability compromises homeostasis, leading to chronic diseases and certain types of cancer. Cell-culture media try to mimic the in vivo environment, providing in vitro models used to infer cells' responses to different stimuli. This review summarizes and discusses studies of cell-culture supplementation with micronutrients that can increase cell viability and genomic stability, with a particular focus on previous in vitro experiments. In these studies, the cell-culture media include certain vitamins and minerals at concentrations not equal to the physiological levels. In many common culture media, the sole source of micronutrients is fetal bovine serum (FBS), which contributes to only 5-10% of the media composition. Minimal attention has been dedicated to FBS composition, micronutrients in cell cultures as a whole, or the influence of micronutrients on the viability and genetics of cultured cells. Further studies better evaluating micronutrients' roles at a molecular level and influence on the genomic stability of cells are still needed.
Collapse
Affiliation(s)
- Ana Lúcia Vargas Arigony
- Laboratório de Reparação de DNA em Eucariotos, Departamento de Biofísica/Centro de Biotecnologia, UFRGS, Avenida Bento Gonçalves 9500, Prédio 43422, Setor IV, Campus do Vale, 91501-970 Porto Alegre, RS, Brazil
| | - Iuri Marques de Oliveira
- Laboratório de Reparação de DNA em Eucariotos, Departamento de Biofísica/Centro de Biotecnologia, UFRGS, Avenida Bento Gonçalves 9500, Prédio 43422, Setor IV, Campus do Vale, 91501-970 Porto Alegre, RS, Brazil
| | - Miriana Machado
- Laboratório de Reparação de DNA em Eucariotos, Departamento de Biofísica/Centro de Biotecnologia, UFRGS, Avenida Bento Gonçalves 9500, Prédio 43422, Setor IV, Campus do Vale, 91501-970 Porto Alegre, RS, Brazil
- Instituto de Educação para Pesquisa, Desenvolvimento e Inovação Tecnológica—ROYAL, Unidade GENOTOX—ROYAL, Centro de Biotecnologia, UFRGS, Avenida Bento Gonçalves 9500, Prédio 43421, Setor IV, Campus do Vale, 91501-970 Porto Alegre, RS, Brazil
| | - Diana Lilian Bordin
- Laboratório de Reparação de DNA em Eucariotos, Departamento de Biofísica/Centro de Biotecnologia, UFRGS, Avenida Bento Gonçalves 9500, Prédio 43422, Setor IV, Campus do Vale, 91501-970 Porto Alegre, RS, Brazil
| | - Lothar Bergter
- Instituto de Educação para Pesquisa, Desenvolvimento e Inovação Tecnológica—ROYAL, Unidade GENOTOX—ROYAL, Centro de Biotecnologia, UFRGS, Avenida Bento Gonçalves 9500, Prédio 43421, Setor IV, Campus do Vale, 91501-970 Porto Alegre, RS, Brazil
| | - Daniel Prá
- Laboratório de Reparação de DNA em Eucariotos, Departamento de Biofísica/Centro de Biotecnologia, UFRGS, Avenida Bento Gonçalves 9500, Prédio 43422, Setor IV, Campus do Vale, 91501-970 Porto Alegre, RS, Brazil
- PPG em Promoção da Saúde, Universidade de Santa Cruz do Sul (UNISC), Avenida Independência 2293, 96815-900 Santa Cruz do Sul, RS, Brazil
| | - João Antonio Pêgas Henriques
- Laboratório de Reparação de DNA em Eucariotos, Departamento de Biofísica/Centro de Biotecnologia, UFRGS, Avenida Bento Gonçalves 9500, Prédio 43422, Setor IV, Campus do Vale, 91501-970 Porto Alegre, RS, Brazil
- Instituto de Educação para Pesquisa, Desenvolvimento e Inovação Tecnológica—ROYAL, Unidade GENOTOX—ROYAL, Centro de Biotecnologia, UFRGS, Avenida Bento Gonçalves 9500, Prédio 43421, Setor IV, Campus do Vale, 91501-970 Porto Alegre, RS, Brazil
- Instituto de Biotecnologia, Departamento de Ciências Biomédicas, Universidade de Caxias do Sul (UCS), Rua Francisco Getúlio Vargas 1130, 95070-560 Caxias do Sul, RS, Brazil
| |
Collapse
|
35
|
Nyaradi A, Li J, Hickling S, Foster J, Oddy WH. The role of nutrition in children's neurocognitive development, from pregnancy through childhood. Front Hum Neurosci 2013; 7:97. [PMID: 23532379 PMCID: PMC3607807 DOI: 10.3389/fnhum.2013.00097] [Citation(s) in RCA: 281] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 03/07/2013] [Indexed: 12/20/2022] Open
Abstract
This review examines the current evidence for a possible connection between nutritional intake (including micronutrients and whole diet) and neurocognitive development in childhood. Earlier studies which have investigated the association between nutrition and cognitive development have focused on individual micronutrients, including omega-3 fatty acids, vitamin B12, folic acid, choline, iron, iodine, and zinc, and single aspects of diet. The research evidence from observational studies suggests that micronutrients may play an important role in the cognitive development of children. However, the results of intervention trials utilizing single micronutrients are inconclusive. More generally, there is evidence that malnutrition can impair cognitive development, whilst breastfeeding appears to be beneficial for cognition. Eating breakfast is also beneficial for cognition. In contrast, there is currently inconclusive evidence regarding the association between obesity and cognition. Since individuals consume combinations of foods, more recently researchers have become interested in the cognitive impact of diet as a composite measure. Only a few studies to date have investigated the associations between dietary patterns and cognitive development. In future research, more well designed intervention trials are needed, with special consideration given to the interactive effects of nutrients.
Collapse
Affiliation(s)
- Anett Nyaradi
- Centre for Child Health Research, Telethon Institute for Child Health Research, The University of Western AustraliaPerth, WA, Australia
- School of Population Health, The University of Western AustraliaPerth, WA, Australia
| | - Jianghong Li
- Centre for Child Health Research, Telethon Institute for Child Health Research, The University of Western AustraliaPerth, WA, Australia
- Centre for Population Health Research, Curtin Health Innovation Research Institute, Curtin UniversityPerth, WA, Australia
- Social Science Research CenterBerlin, Germany
| | - Siobhan Hickling
- Centre for Child Health Research, Telethon Institute for Child Health Research, The University of Western AustraliaPerth, WA, Australia
- School of Population Health, The University of Western AustraliaPerth, WA, Australia
| | - Jonathan Foster
- Centre for Child Health Research, Telethon Institute for Child Health Research, The University of Western AustraliaPerth, WA, Australia
- School of Psychology and Speech Pathology, Curtin UniversityPerth, WA, Australia
- Neurosciences Unit, Health Department of Western AustraliaPerth, WA, Australia
- School of Paediatrics and Child Health, The University of Western AustraliaPerth, WA, Australia
| | - Wendy H. Oddy
- Centre for Child Health Research, Telethon Institute for Child Health Research, The University of Western AustraliaPerth, WA, Australia
| |
Collapse
|
36
|
Li W, Yu M, Luo S, Liu H, Gao Y, Wilson JX, Huang G. DNA methyltransferase mediates dose-dependent stimulation of neural stem cell proliferation by folate. J Nutr Biochem 2013; 24:1295-301. [PMID: 23332600 DOI: 10.1016/j.jnutbio.2012.11.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Revised: 10/31/2012] [Accepted: 11/09/2012] [Indexed: 11/30/2022]
Abstract
The proliferative response of neural stem cells (NSCs) to folate may play a critical role in the development, function and repair of the central nervous system. It is important to determine the dose-dependent effects of folate in NSC cultures that are potential sources of transplantable cells for therapies for neurodegenerative diseases. To determine the optimal concentration and mechanism of action of folate for stimulation of NSC proliferation in vitro, NSCs were exposed to folic acid or 5-methyltetrahydrofolate (5-MTHF) (0-200 μmol/L) for 24, 48 or 72 h. Immunocytochemistry and methyl thiazolyl tetrazolium assay showed that the optimal concentration of folic acid for NSC proliferation was 20-40 μmol/L. Stimulation of NSC proliferation by folic acid was associated with DNA methyltransferase (DNMT) activation and was attenuated by the DNMT inhibitor zebularine, which implies that folate dose-dependently stimulates NSC proliferation through a DNMT-dependent mechanism. Based on these new findings and previously published evidence, we have identified a mechanism by which folate stimulates NSC growth.
Collapse
Affiliation(s)
- Wen Li
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Folic Acid Acts Through DNA Methyltransferases to Induce the Differentiation of Neural Stem Cells into Neurons. Cell Biochem Biophys 2013; 66:559-66. [DOI: 10.1007/s12013-012-9503-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
38
|
Yuan Q, Zhao S, Liu S, Zhang Y, Fu J, Wang F, Liu Q, Ling EA, Hao A. Folic acid supplementation changes the fate of neural progenitors in mouse embryos of hyperglycemic and diabetic pregnancy. J Nutr Biochem 2012; 24:1202-12. [PMID: 23260036 DOI: 10.1016/j.jnutbio.2012.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 09/03/2012] [Accepted: 09/17/2012] [Indexed: 11/28/2022]
Abstract
Folic acid has been shown to decrease the incidence of neural tube defects (NTDs) in normal and hyperglycemic conditions, but the influence of folic acid on the development of central nervous system is not fully understood. Here, we aimed to explore the effects of folic acid, especially high dose of folic acid, on the characteristics of neural progenitors in embryos of hyperglycemic and diabetic mouse. Hyperglycemic and diabetic pregnant mice were given 3 mg/kg or 15 mg/kg folic acid from embryonic day 0.5 (E0.5) and were euthanased on E11.5, E13.5 or E18.5. The incidence of NTDs at E13.5 was counted. The proliferation, apoptosis and differentiation of neural progenitors and neuronal migration were determined using BrdU incorporation assay, TUNEL assay, immunofluorescence, Western blot and real-time reverse transcriptase polymerase chain reaction. Both normal and high doses of folic acid decreased the incidence of NTDs, promoted proliferation and reduced apoptosis of neuroepithelial cells in embryos of hyperglycemic and diabetic mice. Importantly, folic acid, especially at high dose, might affect the premature differentiation of neural progenitors in embryos of hyperglycemic and diabetic pregnancy. This may be attributed to changes of messenger RNA expression levels of some basic-helix-loop-helix transcription factors. In addition, folic acid might be involved in regulating neuronal migration in embryos of hyperglycemic and diabetic pregnancy. These findings suggest that periconceptional supplementation of folic acid, especially at high dose, may be a double-edged sword because it may result in undesirable outcomes affecting both the neuronal and glial differentiation in hyperglycemic and diabetic pregnancy.
Collapse
Affiliation(s)
- Qiuhuan Yuan
- Key Laboratory of the Ministry of Education for Experimental Teratology, Department of Histology and Embryology, Shandong University School of Medicine, Jinan 250012, China
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Guo J, Xie H, Wang J, Zhao H, Wang F, Liu C, Wang L, Lu X, Bao Y, Zou J, Wang G, Niu B, Zhang T. The maternal folate hydrolase gene polymorphism is associated with neural tube defects in a high-risk Chinese population. GENES AND NUTRITION 2012; 8:191-7. [PMID: 22918695 DOI: 10.1007/s12263-012-0309-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 07/24/2012] [Indexed: 11/28/2022]
Abstract
Folate hydrolase 1 (FOLH1) gene encodes intestinal folate hydrolase, which regulates intestinal absorption of dietary folate. Previous studies on the association between polymorphisms rs202676 and rs61886492 and the risk of neural tube defects (NTDs) were inconclusive. A case-control study of women with NTD-affected pregnancies (n = 160) and controls (n = 320) was conducted in the Chinese population of Lvliang, a high-risk area for NTDs. We genotyped the polymorphic sites rs202676 and rs61886492 and assessed maternal plasma folate and total homocysteine (tHcy). Our results showed that in case group, plasma folate concentrations were 18 % lower compared with those of control group (8.32 vs. 6.79 nmol/L, p = 0.033) and tHcy concentrations were 17 % higher (10.47 vs. 12.65 μmol/L, p = 0.047). Almost all samples had the rs61886492 GG genotype (99.78 %). The result showed that the frequency of GG genotype in rs202676 was significantly higher in group with multiple NTDs than in controls (p = 0.030, OR = 2.157, 95 % CI, 1.06-4.38). The multiple-NTD group showed higher maternal plasma concentrations of tHcy (10.47 vs. 13.96 μmol/L, p = 0.024). The GG genotype of rs202676 had a lower maternal folate and higher tHcy concentrations than other genotypes with no significant differences. The result of structural prediction indicated that this variation might change the spatial structure of the protein. These results suggested that the maternal polymorphism rs202676 was a potential risk factor for multiple NTDs in this Chinese population. The allele G might affect maternal plasma folate and tHcy concentration.
Collapse
Affiliation(s)
- Jin Guo
- Capital Institute of Pediatrics, Beijing, 100020, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ro SH, Liu D, Yeo H, Paik JH. FoxOs in neural stem cell fate decision. Arch Biochem Biophys 2012; 534:55-63. [PMID: 22902436 DOI: 10.1016/j.abb.2012.07.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 07/30/2012] [Accepted: 07/31/2012] [Indexed: 02/07/2023]
Abstract
Neural stem cells (NSCs) persist over the lifespan of mammals to give rise to committed progenitors and their differentiated cells in order to maintain the brain homeostasis. To this end, NSCs must be able to self-renew and otherwise maintain their quiescence. Suppression of aberrant proliferation or undesired differentiation is crucial to preclude either malignant growth or precocious depletion of NSCs. The PI3K-Akt-FoxO signaling pathway plays a central role in the regulation of multiple stem cells including one in the mammalian brain. In particular, members of FoxO family transcription factors are highly expressed in these stem cells. As an important downstream effector of growth, differentiation, and stress stimuli, mammalian FoxO transcription factor family controls cellular proliferation, oxidative stress response, homeostasis, and eventual maintenance of long-term repopulating potential. The review will focus on the current understanding of FoxO function in NSCs as well as discuss their biological activities that contribute to determining neural stem cell fate.
Collapse
Affiliation(s)
- Seung-Hyun Ro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | |
Collapse
|
41
|
Xie H, Guo J, Wang J, Wang F, Zhao H, Liu C, Wang L, Lu X, Wu L, Bao Y, Zou J, Zhang T, Niu B. Glutamate carboxypeptidase II gene polymorphisms and neural tube defects in a high-risk Chinese population. Metab Brain Dis 2012; 27:59-65. [PMID: 22124883 DOI: 10.1007/s11011-011-9272-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 11/08/2011] [Indexed: 11/27/2022]
Abstract
Glutamate carboxypeptidase II (GCPII) catalyzes the hydrolysis of N-acetylaspartylglutamate into N-acetylaspartate and glutamate in the brain. Animal experiments suggested that GCPII plays an essential role in early embryonic development. Previous studies provided conflicting results on the effect of the GCPII rs61886492 C>T (or 1561C>T) polymorphism on NTDs. In the Lvliang area of Shanxi province, where the incidence of NTDs is the highest in China, a case-control study was conducted to investigate possible association between the GCPII rs61886492 and rs202676 polymorphisms and NTD risk. Results indicated all the case and control samples displayed the rs61886492 GG genotype. Although no significant differences in rs202676 genotype or allele frequencies were found between the NTD and control groups, the combined AG+GG genotype group was significantly associated with anencephaly (p = 0.03, OR = 2.11, 95% CI, 1.11-4.01), but not with spina bifida or encephalocele. Overall, the rs202676 A>G polymorphism is a potential risk factor for anencephaly. The results of this study suggest that phenotypic heterogeneity may exist among NTDs in this Chinese population.
Collapse
Affiliation(s)
- Hua Xie
- Capital Institute of Pediatrics, Beijing 100020, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ichi S, Nakazaki H, Boshnjaku V, Singh RM, Mania-Farnell B, Xi G, McLone DG, Tomita T, Mayanil CSK. Fetal Neural Tube Stem Cells from Pax3 Mutant Mice Proliferate, Differentiate, and Form Synaptic Connections When Stimulated with Folic Acid. Stem Cells Dev 2012; 21:321-30. [DOI: 10.1089/scd.2011.0100] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Shunsuke Ichi
- Developmental Biology Program, Division of Pediatric Neurosurgery, Children's Memorial Hospital and Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Neurosurgery, University of Tokyo, Tokyo, Japan
| | - Hiromichi Nakazaki
- Developmental Biology Program, Division of Pediatric Neurosurgery, Children's Memorial Hospital and Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Vanda Boshnjaku
- Developmental Biology Program, Division of Pediatric Neurosurgery, Children's Memorial Hospital and Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Ravneet Monny Singh
- Developmental Biology Program, Division of Pediatric Neurosurgery, Children's Memorial Hospital and Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Guifa Xi
- Developmental Biology Program, Division of Pediatric Neurosurgery, Children's Memorial Hospital and Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - David G. McLone
- Developmental Biology Program, Division of Pediatric Neurosurgery, Children's Memorial Hospital and Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Tadanori Tomita
- Developmental Biology Program, Division of Pediatric Neurosurgery, Children's Memorial Hospital and Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Chandra Shekhar K. Mayanil
- Developmental Biology Program, Division of Pediatric Neurosurgery, Children's Memorial Hospital and Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
43
|
Pang W, Lu H, Hu YD, Yang HP, Leng X, Jiang YG. Depletion of intracellular zinc induced apoptosis in cultured hippocampal neurons through Raf/MEK/ERK pathways. Nutr Neurosci 2012; 15:18-24. [PMID: 22305649 DOI: 10.1179/1476830511y.0000000031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
An experiment was performed to observe the changes in Raf-1 kinase/mitogen-activated protein kinase ERK (MEK)/extracellular signal-regulated kinase (ERK) signaling pathways in cultured hippocampal neurons and its correlation with neurons apoptosis induced by intracellular zinc depletion. Cultured hippocampal neurons were exposed to a cell membrane-permeant zinc chelator TPEN (2 µM), and to TPEN plus zinc sulfate (5 µM) for 24 h. Cultures were then processed to detect neuronal viability by the methyl thiazolyl tetrazolium assay, while apoptosis rate was simultaneously observed by the flow cytometric analysis. Caspase-3, Raf-1, pMEK, pERK1/2, and pCREB protein levels were examined by Western blot assays. The viability in TPEN-incubated neurons was notably decreased, apoptosis rate and expression of caspase-3 significantly increased compared to untreated controls. The significant down-regulation of Raf/MEK/ERK signaling pathway and expression of pCREB were decreased in TPEN-treated neurons. Co-addition of zinc almost completely reversed TPEN-induced alterations described. The results demonstrated zinc-modulated apoptosis and the expression of Raf/MEK/ERK at the protein level in hippocampal neurons. It is possible that zinc depletion-induced apoptosis in cultured hippocampal neurons may be relevant to the changes of Raf/MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Wei Pang
- Department of Nutrition, Institute of Hygiene & Environmental Medicine, Academy of Military Medical Science, Tianjin, China
| | | | | | | | | | | |
Collapse
|
44
|
Maldonado E, Murillo J, Barrio C, del Río A, Pérez-Miguelsanz J, López-Gordillo Y, Partearroyo T, Paradas I, Maestro C, Martínez-Sanz E, Varela-Moreiras G, Martínez-Álvarez C. Occurrence of cleft-palate and alteration of Tgf-β(3) expression and the mechanisms leading to palatal fusion in mice following dietary folic-acid deficiency. Cells Tissues Organs 2011; 194:406-20. [PMID: 21293104 DOI: 10.1159/000323213] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2010] [Indexed: 02/02/2023] Open
Abstract
Folic acid (FA) is essential for numerous bodily functions. Its decrease during pregnancy has been associated with an increased risk of congenital malformations in the progeny. The relationship between FA deficiency and the appearance of cleft palate (CP) is controversial, and little information exists on a possible effect of FA on palate development. We investigated the effect of a 2-8 weeks' induced FA deficiency in female mice on the development of CP in their progeny as well as the mechanisms leading to palatal fusion, i.e. cell proliferation, cell death, and palatal-shelf adhesion and fusion. We showed that an 8 weeks' maternal FA deficiency caused complete CP in the fetuses although a 2 weeks' maternal FA deficiency was enough to alter all the mechanisms analyzed. Since transforming growth factor-β(3) (TGF-β(3)) is crucial for palatal fusion and since most of the mechanisms impaired by FA deficiency were also observed in the palates of Tgf-β(3)null mutant mice, we investigated the presence of TGF-β(3) mRNA, its protein and phospho-SMAD2 in FA-deficient (FAD) mouse palates. Our results evidenced a large reduction in Tgf-β(3) expression in palates of embryos of dams fed an FAD diet for 8 weeks; Tgf-β(3) expression was less reduced in palates of embryos of dams fed an FAD diet for 2 weeks. Addition of TGF-β(3) to palatal-shelf cultures of embryos of dams fed an FAD diet for 2 weeks normalized all the altered mechanisms. Thus, an insufficient folate status may be a risk factor for the development of CP in mice, and exogenous TGF-β(3) compensates this deficit in vitro.
Collapse
Affiliation(s)
- Estela Maldonado
- Departamento de Anatomía y Embriología Humana I, Facultad de Odontología, Universidad Complutense, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|