Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.571
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 299(7): 104909, 2023 07.
Article in English | MEDLINE | ID: mdl-37307917

ABSTRACT

Sustainable TGF-ß1 signaling drives organ fibrogenesis. However, the cellular adaptation to maintain TGF-ß1 signaling remains unclear. In this study, we revealed that dietary folate restriction promoted the resolution of liver fibrosis in mice with nonalcoholic steatohepatitis. In activated hepatic stellate cells, folate shifted toward mitochondrial metabolism to sustain TGF-ß1 signaling. Mechanistically, nontargeted metabolomics screening identified that α-linolenic acid (ALA) is exhausted by mitochondrial folate metabolism in activated hepatic stellate cells. Knocking down serine hydroxymethyltransferase 2 increases the bioconversion of ALA to docosahexaenoic acid, which inhibits TGF-ß1 signaling. Finally, blocking mitochondrial folate metabolism promoted liver fibrosis resolution in nonalcoholic steatohepatitis mice. In conclusion, mitochondrial folate metabolism/ALA exhaustion/TGF-ßR1 reproduction is a feedforward signaling to sustain profibrotic TGF-ß1 signaling, and targeting mitochondrial folate metabolism is a promising strategy to enforce liver fibrosis resolution.


Subject(s)
Folic Acid , Liver Cirrhosis , Mitochondria , alpha-Linolenic Acid , Animals , Mice , alpha-Linolenic Acid/deficiency , alpha-Linolenic Acid/metabolism , Hepatic Stellate Cells/metabolism , Liver/cytology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/complications , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/metabolism , Transforming Growth Factor beta1/metabolism , Folic Acid/metabolism , Mitochondria/metabolism , Folic Acid Deficiency/complications , Folic Acid Deficiency/metabolism , Signal Transduction , Feedback, Physiological
2.
Neurosurg Rev ; 47(1): 724, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39365348

ABSTRACT

Neural tube defects (NTDs), such as spina bifida and anencephaly, are severe congenital anomalies affecting the development of the brain and spine. These conditions are often linked to folic acid deficiency during early pregnancy, a modifiable risk factor. While high-income countries have implemented mandatory folic acid fortification in staple foods, resulting in significant reductions in NTD prevalence, low- and middle-income countries (LMICs) continue to experience disproportionately high rates of these birth defects. Folic acid supplementation and fortification are proven interventions for preventing NTDs, but many LMICs face political, financial, and logistical barriers to implementing these programs. This paper highlights the importance of mandatory folic acid fortification as a cost-effective public health intervention and advocates for its expansion in LMICs. It reviews the successes of fortification programs in high-income countries, explores alternative food vehicles like rice for regions with different dietary staples, and discusses the potential of multi-nutrient fortification strategies. Additionally, this paper emphasizes the need for global collaboration, enhanced monitoring and evaluation, and public health education campaigns to ensure that women of reproductive age, especially in LMICs, receive adequate folic acid. By addressing these challenges, the global health community can significantly reduce the incidence of NTDs, improve maternal and child health, and promote health equity worldwide. The time to act is now, as the benefits of folic acid fortification far outweigh the costs of inaction.


Subject(s)
Folic Acid , Food, Fortified , Neural Tube Defects , Spinal Dysraphism , Humans , Folic Acid/therapeutic use , Folic Acid/administration & dosage , Spinal Dysraphism/prevention & control , Spinal Dysraphism/epidemiology , Neural Tube Defects/prevention & control , Neural Tube Defects/epidemiology , Female , Pregnancy , Folic Acid Deficiency/prevention & control , Folic Acid Deficiency/complications , Dietary Supplements
3.
J Nutr ; 153(3): 760-770, 2023 03.
Article in English | MEDLINE | ID: mdl-36792392

ABSTRACT

BACKGROUND: Evidence on the association of serum folate and homocysteine concentrations with risk of mortality in the general population is unclear. OBJECTIVES: This study aimed to examine the associations of serum folate and homocysteine concentrations with all-cause, CVD, and cancer mortality risk in Korean men and women aged ≥40 y. METHODS: In this population-based prospective cohort study, serum folate and homocysteine concentrations were measured in a subset of participants enrolled between 2005 and 2012. A total of 21,260 participants were linked to mortality data from the survey date to 31 December 2019. Cox proportional hazards models and restricted cubic splines were used to identify the associations of serum folate and homocysteine concentrations with mortality. RESULTS: During a median follow-up of 12.3 y, 2501, 549, and 842 deaths were attributed to all-cause, CVD, and cancer, respectively. The prevalence of folate deficiency and hyperhomocysteinemia were higher in men than in women. In men, a nonlinear inverse association was observed between serum folate concentrations and all-cause mortality. Men in the third quartile of serum folate concentrations exhibited a lower risk of all-cause mortality (HR: 0.85; 95% CI: 0.73, 0.99) than those in the lowest quartile. Serum homocysteine concentration was positively associated with all-cause and CVD mortality. Men and women in the highest compared with those in the lowest serum homocysteine quartile showed a higher risk of CVD mortality (HR: 1.60; 95% CI: 1.07, 2.39; and HR: 1.79; 95% CI: 1.11, 2.89, respectively). Hyperhomocysteinemia combined with folate deficiency was associated with increased all-cause, CVD, and cancer-related mortality rates. CONCLUSIONS: Higher serum homocysteine and lower serum folate concentrations were associated with an increased risk of all-cause, CVD, and cancer-related mortality in Korean adults. The finding of a nonlinear inverse relationship between serum folate concentration and mortality in men warrants further investigation.


Subject(s)
Cardiovascular Diseases , Folic Acid Deficiency , Hyperhomocysteinemia , Neoplasms , Male , Adult , Humans , Female , Prospective Studies , Folic Acid , Folic Acid Deficiency/complications , Republic of Korea/epidemiology , Homocysteine , Risk Factors
4.
JAMA ; 330(5): 454-459, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37526713

ABSTRACT

Importance: Neural tube defects are among the most common congenital malformations in the US, with an estimated 3000 pregnancies affected each year. Many of these neural tube defects are caused by low folate levels in the body. Objective: The US Preventive Services Task Force (USPSTF) commissioned a reaffirmation evidence update on the benefits and harms of folic acid supplementation. Population: Persons who are planning to or could become pregnant. Evidence Assessment: The USPSTF concludes that, for persons who are planning to or could become pregnant, there is high certainty that folic acid supplementation has a substantial net benefit to prevent neural tube defects in their offspring. Recommendation: The USPSTF recommends that all persons planning to or who could become pregnant take a daily supplement containing 0.4 to 0.8 mg (400 to 800 µg) of folic acid. (A recommendation).


Subject(s)
Dietary Supplements , Folic Acid Deficiency , Folic Acid , Neural Tube Defects , Pregnancy Complications , Female , Humans , Pregnancy , Advisory Committees , Folic Acid/administration & dosage , Folic Acid/therapeutic use , Mass Screening , Neural Tube Defects/etiology , Neural Tube Defects/prevention & control , Preventive Health Services , Folic Acid Deficiency/complications , Folic Acid Deficiency/diagnosis , Folic Acid Deficiency/drug therapy , Pregnancy Complications/etiology , Pregnancy Complications/prevention & control , Preconception Care/standards
5.
J Cell Mol Med ; 26(3): 776-788, 2022 02.
Article in English | MEDLINE | ID: mdl-34953021

ABSTRACT

Sperm DNA injury is one of the common causes of male infertility. Folic acid deficiency would increase the methylation level of the important genes, including those involved in DNA double-strand break (DSB) repair pathway. In the early stages, we analysed the correlation between seminal plasma folic acid concentration and semen parameters in 157 infertility patients and 91 sperm donor volunteers, and found that there was a significant negative correlation between seminal folic acid concentration and sperm DNA Fragmentation Index (DFI; r = -0.495, p < 0.01). Then through reduced representation bisulphite sequencing, global DNA methylation of sperm of patients in the low folic acid group and the high folic acid group was analysed, it was found that the methylation level in Rad54 promoter region increased in the folic acid deficiency group compared with the normal folic acid group. Meanwhile, the results of animal model and spermatocyte line (GC-2) also found that folic acid deficiency can increase the methylation level in Rad54 promoter region, increased sperm DFI in mice, increased the expression of γ-H2AX, that is, DNA injury marker protein, and increased sensitivity of GC-2 to external damage and stimulation. The study indicates that the expression of Rad54 is downregulated by folic acid deficiency via DNA methylation. This may be one of the mechanisms of sperm DNA damage caused by folate deficiency.


Subject(s)
Folic Acid Deficiency , Infertility, Male , Animals , DNA Damage , DNA Fragmentation , Folic Acid/metabolism , Folic Acid Deficiency/complications , Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Humans , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Mice , Semen/chemistry , Semen/metabolism , Sperm Count , Spermatozoa/metabolism
7.
Cereb Cortex ; 31(1): 635-649, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32995858

ABSTRACT

Folate is an essential micronutrient required for both cellular proliferation through de novo nucleotide synthesis and epigenetic regulation of gene expression through methylation. This dual requirement places a particular demand on folate availability during pregnancy when both rapid cell generation and programmed differentiation of maternal, extraembryonic, and embryonic/fetal tissues are required. Accordingly, prenatal neurodevelopment is particularly susceptible to folate deficiency, which can predispose to neural tube defects, or when effective transport into the brain is impaired, cerebral folate deficiency. Consequently, adequate folate consumption, in the form of folic acid (FA) fortification and supplement use, is widely recommended and has led to a substantial increase in the amount of FA intake during pregnancy in some populations. Here, we show that either maternal folate deficiency or FA excess in mice results in disruptions in folate metabolism of the offspring, suggesting diversion of the folate cycle from methylation to DNA synthesis. Paradoxically, either intervention causes comparable neurodevelopmental changes by delaying prenatal cerebral cortical neurogenesis in favor of late-born neurons. These cytoarchitectural and biochemical alterations are accompanied by behavioral abnormalities in FA test groups compared with controls. Our findings point to overlooked potential neurodevelopmental risks associated with excessively high levels of prenatal FA intake.


Subject(s)
Behavior, Animal/drug effects , Epigenesis, Genetic/drug effects , Folic Acid/pharmacology , Pregnancy/drug effects , Animals , DNA Methylation/drug effects , Dietary Supplements/adverse effects , Female , Folic Acid Deficiency/complications , Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Mice, Inbred C57BL
8.
Clin Lab ; 68(9)2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36125146

ABSTRACT

BACKGROUND: Deficiency of vitamin B(12) or folate causes megaloblastic anemia (MA). The disease presents with pancytopenia due to the excessive cellular apoptosis of hematopoietic progenitor. MA is characterized by the presence of high mean corpuscular volume in the blood routine test and hyperlobulation nuclei of the granulocytes in the peripheral blood smears, and megaloblasts in the bone marrow. METHODS: We report a rare case, in which megaloblastic anemia was masked by an unrecognized hemoglobinopathy and presented with normocytic anemia and atypical morphological features of bone marrow. RESULTS: The patient was finally diagnosed with coexistence of MA and a-thalassemia minor due to determination of folate deficiency and genetic mutation for a-thalassemia. CONCLUSIONS: The case focuses on the contribution of the peripheral circulating blood smear examination in the diagnosis of anemia.


Subject(s)
Anemia, Megaloblastic , Folic Acid Deficiency , Thalassemia , Anemia, Megaloblastic/complications , Anemia, Megaloblastic/diagnosis , Folic Acid , Folic Acid Deficiency/complications , Folic Acid Deficiency/diagnosis , Humans , Vitamins
9.
BMC Pregnancy Childbirth ; 22(1): 16, 2022 Jan 06.
Article in English | MEDLINE | ID: mdl-34986796

ABSTRACT

BACKGROUND: The Sustainable development goals, which focus strongly on equity, aim to end all forms of malnutrition by 2030. However, a significant cause of intergenerational transfer of malnutrition, anaemia in pregnancy, is still a challenge. It is especially so in the low- and middle-income settings where possible context-specific aetiologies leading to anaemia have been poorly explored. This study explores the prevalence of etiological factors significantly contributing to anaemia in pregnancy in Sri Lanka, a lower-middle-income country with a high prevalence of malnutrition albeit robust public health infrastructure. METHODS: All first-trimester pregnant women registered in the public maternal care programme in the Anuradhapura district from July to September 2019 were invited to participate in Rajarata Pregnancy Cohort (RaPCo). After a full blood count analysis, high-performance liquid chromatography, peripheral blood film examination, serum B12 and folate levels were performed in anaemic participants, guided by an algorithm based on the red cell indices in the full blood count. In addition, serum ferritin was tested in a random subsample of 213 participants. Anaemic women in this subsample underwent B12 and folate testing. RESULTS: Among 3127 participants, 14.4% (95%CI 13.2-15.7, n = 451) were anaemic. Haemoglobin ranged between 7.4 to 19.6 g/dl. 331(10.6%) had mild anaemia. Haemoglobin ≥13 g/dl was observed in 39(12.7%). Microcytic, normochromic-normocytic, hypochromic-normocytic and macrocytic anaemia was observed in 243(54%), 114(25.3%), 80(17.8%) and two (0.4%) of full blood counts in anaemic women, respectively. Microcytic anaemia with a red cell count ≥5 * 106 /µl demonstrated a 100% positive predictive value for minor haemoglobinopathies. Minor hemoglobinopathies were present in at least 23.3%(n = 105) of anaemic pregnant women. Prevalence of iron deficiency, B12 deficiency and Southeast Asian ovalocytosis among the anaemic was 41.9% (95%CI 26.4-59.2), 23.8% (95%CI 10.6-45.1) and 0.9% (95%CI 0.3-2.3%), respectively. Folate deficiency was not observed. CONCLUSION: Even though iron deficiency remains the primary cause, minor hemoglobinopathies, B 12 deficiency and other aetiologies substantially contribute to anaemia in pregnancy in this study population. Public health interventions, including screening for minor hemoglobinopathies and multiple micronutrient supplementation in pregnancy, should be considered in the national programme for areas where these problems have been identified.


Subject(s)
Anemia/classification , Anemia/epidemiology , Anemia/etiology , Pregnancy Complications, Hematologic/classification , Pregnancy Complications, Hematologic/epidemiology , Pregnancy Complications, Hematologic/etiology , Pregnancy Trimester, First , Adult , Anemia/blood , Cohort Studies , Erythrocyte Indices , Female , Ferritins/blood , Folic Acid Deficiency/complications , Hemoglobinopathies/complications , Hemoglobins/analysis , Humans , Iron Deficiencies/complications , Pregnancy , Pregnancy Complications, Hematologic/blood , Prevalence , Sri Lanka/epidemiology , Vitamin B 12 Deficiency/complications
10.
Int J Vitam Nutr Res ; 92(3-4): 169-181, 2022 Jul.
Article in English | MEDLINE | ID: mdl-31841076

ABSTRACT

Folate and vitamin B12(V-B12) deficiencies are associated with metabolic diseases that may impair memory function. We hypothesized that folate and V-B12 may differently alter mild cognitive impairment, glucose metabolism, and inflammation by modulating the gut microbiome in rats with Alzheimer's disease (AD)-like dementia. The hypothesis was examined in hippocampal amyloid-ß infused rats, and its mechanism was explored. Rats that received an amyloid-ß(25-35) infusion into the CA1 region of the hippocampus were fed either control(2.5 mg folate plus 25 µg V-B12/kg diet; AD-CON, n = 10), no folate(0 folate plus 25 µg V-B12/kg diet; AD-FA, n = 10), no V-B12(2.5 mg folate plus 0 µg V-B12/kg diet; AD-V-B12, n = 10), or no folate plus no V-B12(0 mg folate plus 0 µg V-B12/kg diet; AD-FAB12, n = 10) in high-fat diets for 8 weeks. AD-FA and AD-VB12 exacerbated bone mineral loss in the lumbar spine and femur whereas AD-FA lowered lean body mass in the hip compared to AD-CON(P < 0.05). Only AD-FAB12 exacerbated memory impairment by 1.3 and 1.4 folds, respectively, as measured by passive avoidance and water maze tests, compared to AD-CON(P < 0.01). Hippocampal insulin signaling and neuroinflammation were attenuated in AD-CON compared to Non-AD-CON. AD-FAB12 impaired the signaling (pAkt→pGSK-3ß) and serum TNF-α and IL-1ß levels the most among all groups. AD-CON decreased glucose tolerance by increasing insulin resistance compared to Non-AD-CON. AD-VB12 and AD-FAB12 increased insulin resistance by 1.2 and 1.3 folds, respectively, compared to the AD-CON. AD-CON and Non-AD-CON had a separate communities of gut microbiota. The relative counts of Bacteroidia were lower and those of Clostridia were higher in AD-CON than Non-AD-CON. AD-FA, but not V-B12, separated the gut microbiome community compared to AD-CON and AD-VB12(P = 0.009). In conclusion, folate and B-12 deficiencies impaired memory function by impairing hippocampal insulin signaling and gut microbiota in AD rats.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Folic Acid Deficiency , Gastrointestinal Microbiome , Vitamin B 12 Deficiency , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Amyloid beta-Peptides/administration & dosage , Animals , Diet, High-Fat , Folic Acid , Folic Acid Deficiency/complications , Insulin , Insulin Resistance , Memory , Rats , Rats, Sprague-Dawley , Vitamin B 12 , Vitamin B 12 Deficiency/complications , Vitamins
11.
Int J Mol Sci ; 23(13)2022 Jun 22.
Article in English | MEDLINE | ID: mdl-35805953

ABSTRACT

Early life stage folate status may influence neurodevelopment in offspring. The developmental origin of health and disease highlights the importance of the period of the first 1000 days (from conception to 2 years) of life. This study aimed to evaluate the effect of early life stage folic acid deficiency on de novo telomere synthesis, neurobehavioral development, and the cognitive function of offspring rats. The rats were divided into three diet treatment groups: folate-deficient, folate-normal, and folate-supplemented. They were fed the corresponding diet from 5 weeks of age to the end of the lactation period. After weaning, the offspring rats were still fed with the corresponding diet for up to 100 days. Neurobehavioral tests, folic acid and homocysteine (Hcy) levels, relative telomere length in brain tissue, and uracil incorporation in telomere in offspring were measured at different time points. The results showed that folic acid deficiency decreased the level of folic acid, increased the level of Hcy of brain tissue in offspring, increased the wrong incorporation of uracil into telomeres, and hindered de novo telomere synthesis. However, folic acid supplementation increased the level of folic acid, reduced the level of Hcy of brain tissue in offspring, reduced the wrong incorporation of uracil into telomeres, and protected de novo telomere synthesis of offspring, which was beneficial to the development of early sensory-motor function, spatial learning, and memory in adolescence and adulthood. In conclusion, early life stage folic acid deficiency had long-term inhibiting effects on neurodevelopment and cognitive function in offspring.


Subject(s)
Folic Acid Deficiency , Animals , Cognition , Dietary Supplements , Female , Folic Acid/metabolism , Folic Acid Deficiency/complications , Folic Acid Deficiency/metabolism , Rats , Telomere/metabolism , Uracil
12.
Vestn Oftalmol ; 138(4): 29-34, 2022.
Article in Russian | MEDLINE | ID: mdl-36004588

ABSTRACT

Folate metabolism disorders are known to have a potential involvement in the pathophysiology of mitochondrial diseases. Many researchers suggest that profound systemic folate deficiency may contribute to mitochondrial folate deficiency. Folic acid metabolism is closely related to vitamin B12 and homocysteine. Considering that hereditary optic neuropathies (HON) are mitochondrial diseases, it is important to study the folate status, the content of vitamin B12 and homocysteine in patients with this pathology. OBJECTIVE: To compare the content of folic acid, vitamin B12 and homocysteine in the blood serum of patients with Leber's hereditary optic neuropathy (LHON) and autosomal recessive optic neuropathy (ARON), optic neuropathy of other genesis, and the comparison group. MATERIAL AND METHODS: The study involved 58 patients with LHON and ARON, the control group of 49 patients with ischemic, inflammatory, traumatic and compressive optic neuropathies, and the comparison group of 20 healthy volunteers. RESULTS: A decrease in blood folic acid levels was revealed (4.0±1.6 ng/mL) in patients with HON compared to the control group (p=1.3·10-8) and the comparison group (p=1·10-17). The content of vitamin B12 in patients with HON was 380.8±168.1 pg/mL, which was significantly lower than in the comparison group (p=0.0001). The homocysteine content was 14.1±5.6 µmol/L in patients with HON, which was significantly higher than in the control group (p=0.0007) and the comparison group (p=0.000003). At the same time, an increase in homocysteine level of more than 10 µmol/L was revealed in 75% of patients with HON. Similar metabolic disorders were found in groups with various mutations in mitochondrial and nuclear DNA. CONCLUSION: Patients with HON showed marked decrease in the levels of folic acid and vitamin B12, as well as hyperhomocysteinemia. It is very important to identify the causes of metabolic disorders in order to determine the role of folate deficiency in the development of HON, as well as the possibility of its pharmacological treatment.


Subject(s)
Folic Acid Deficiency , Hyperhomocysteinemia , Optic Atrophy, Hereditary, Leber , Optic Nerve Diseases , Folic Acid , Folic Acid Deficiency/complications , Folic Acid Deficiency/diagnosis , Homocysteine , Humans , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/diagnosis , Optic Atrophy, Hereditary, Leber/diagnosis , Optic Nerve Diseases/diagnosis , Optic Nerve Diseases/etiology , Vitamins
13.
Andrologia ; 53(4): e14003, 2021 May.
Article in English | MEDLINE | ID: mdl-33550658

ABSTRACT

The aim of this study was to assess the relationship between serum folic acid (FA) levels and erectile dysfunction (ED) through a meta-analysis. A research was conducted in MEDLINE via PubMed, Cochrane Library, EMBASE and Web of Science up to 22 November 2020 to identify studies related to FA and ED. Two authors independently screened the literature, evaluated methodological quality and extracted the data. We used RevMan5.3 and STATA 14.0 for meta-analysis. A total of six studies including 1,842 participants were included, and the results showed that the FA levels in the non-ED group were significantly higher than those in the ED group (MD = 3.37, 95% CI 1.49-5.52, p = 0.004). Subgroup analysis indicated that with the increase in ED severity, the difference in FA levels between groups was more obvious (MD: 1.99 vs. 4.63 vs. 5.63). The differences in FA levels between groups seem more significant in the younger group (MD = 4.87, 95% CI 2.58-6.89, p < 0.001) than in the older group (MD = 3.15, 95% CI 2.21-4.08, p < 0.001). In conclusion, FA deficiency is closely related to ED, and the degree of FA deficiency may reflect the severity of ED. In addition, the association seems to be more pronounced in the younger group.


Subject(s)
Erectile Dysfunction , Folic Acid Deficiency , Folic Acid , Folic Acid Deficiency/complications , Humans , Male
14.
Int J Neurosci ; 131(5): 468-477, 2021 May.
Article in English | MEDLINE | ID: mdl-32241207

ABSTRACT

OBJECTIVE: Neural tube defects (NTDs) are one of the most common and serious birth defects in human beings caused by genetic and environmental factors. Folate insufficiency is involved in the occurrence of NTDs and folic acid supplementation can prevent NTDs occurrence, however, the underlying mechanism remains poorly understood. METHODS: We established cell and animal models of folic acid deficiency to detect the methylation modification and expression levels of genes by MassARRAY and real-time PCR, respectively. Results and conclusion: In the present study, we found firstly that in human folic acid-insufficient NTDs, the methylation level of imprinted gene Mest/Peg1 was decreased. By using a folic acid-deficient cell model, we demonstrated that Mest/Peg1 methylation was descended. Meanwhile, the mRNA level of Mest/Peg1 was up-regulated via hypomethylation modification under low folic acid conditions. Consistent with the results in cell models, Mest/Peg1 expression was elevated through hypomethylation regulation in folate-deficient animal models. Furthermore, the up-regulation of Mest/Peg1 inhibited the expression of Lrp6 gene, a crucial component of Wnt pathway. Similar results with Lrp6 down-regulation of fetal brain were verified in animal models under folic acid-deficient condition. Taken together, our findings indicated folic acid increased the expression of Mest/Peg1 via hypomethylation modification, and then inhibited Lrp6 expression, which may ultimately impact on the development of nervous system through the inactivation of Wnt pathway.


Subject(s)
Brain/metabolism , Folic Acid Deficiency/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Neural Tube Defects/metabolism , Proteins/metabolism , Wnt Signaling Pathway/genetics , Animals , Cells, Cultured , Disease Models, Animal , Female , Fetus , Folic Acid Deficiency/complications , Gene Expression Regulation , Humans , Methylation , Mice , Mice, Inbred C57BL , Neural Tube Defects/etiology
15.
J Formos Med Assoc ; 120(2): 819-826, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32888843

ABSTRACT

BACKGROUND/PURPOSE: Our previous study found the serum gastric parietal cell antibody (GPCA) positivity in 12.3% of burning mouth syndrome (BMS) patients. This study assessed whether GPCA-positive BMS (GPCA+BMS) patients had significantly higher frequencies of macrocytosis, anemia, hematinic deficiencies, and hyperhomocysteinemia than healthy control subjects or GPCA-negative BMS (GPCA-BMS) patients. METHODS: The mean corpuscular volume, blood hemoglobin (Hb), and serum iron, vitamin B12, folic acid, homocysteine, and GPCA levels were measured and compared between any two of three groups of 109 GPCA+BMS patients, 775 GPCA-BMS patients, and 442 healthy control subjects. RESULTS: We found that 109 GPCA+BMS patients had significantly higher frequencies of macrocytosis, blood Hb and serum iron and vitamin B12 deficiencies, and hyperhomocysteinemia than 442 healthy control subjects (all P-values < 0.001) and significantly higher frequencies of macrocytosis, blood Hb and serum vitamin B12 deficiencies, and hyperhomocysteinemia than 775 GPCA-BMS patients (all P-values < 0.01). Moreover, 775 GPCA-BMS patients had significantly higher frequencies of macrocytosis, blood Hb and serum iron, vitamin B12, and folic acid deficiencies, and hyperhomocysteinemia than 442 healthy control subjects (all P-values < 0.005). Pernicious anemia (45.5%) and normocytic anemia (24.2%) were the two most common types of anemia in 33 anemic GPCA+BMS patients. Moreover, normocytic anemia (61.3%), thalassemia trait-induced anemia (15.5%), and iron deficiency anemia (14.1%) were the three most common types of anemia in 142 anemic GPCA-BMS patients. CONCLUSION: GPCA+BMS patients have significantly higher frequencies of macrocytosis, blood Hb and serum vitamin B12 deficiencies, and hyperhomocysteinemia than healthy control subjects or GPCA-BMS patients.


Subject(s)
Anemia , Burning Mouth Syndrome , Hematinics , Hyperhomocysteinemia , Burning Mouth Syndrome/epidemiology , Folic Acid , Folic Acid Deficiency/complications , Folic Acid Deficiency/epidemiology , Glossitis , Hemoglobins/analysis , Humans , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/epidemiology , Iron , Parietal Cells, Gastric , Vitamin B 12 , Vitamin B 12 Deficiency/complications , Vitamin B 12 Deficiency/epidemiology
16.
J Nutr ; 150(Suppl 1): 2532S-2537S, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33000156

ABSTRACT

Homocysteine (Hcy) is methylated by methionine synthase to form methionine with methyl-cobalamin as a cofactor. The reaction demethylates 5-methyltetrahydrofolate to tetrahydrofolate, which is required for DNA and RNA synthesis. Deficiency of either of the cobalamin (Cbl) and/or folate cofactors results in elevated Hcy and megaloblastic anemia. Elevated Hcy is a sensitive biomarker of Cbl and/or folate status and more specific than serum vitamin assays. Elevated Hcy normalizes when the correct vitamin is given. Elevated Hcy is associated with alcohol use disorder and drugs that target folate or Cbl metabolism, and is a risk factor for thrombotic vascular disease. Elevated methionine and cystathionine are associated with liver disease. Elevated Hcy, cystathionine, and cysteine, but not methionine, are common in patients with chronic renal failure. Higher cysteine predicts obesity and future weight gain. Serum S-adenosylhomocysteine (AdoHcy) is elevated in Cbl deficiency and chronic renal failure. Drugs that require methylation for catabolism may deplete liver S-adenosylmethionine and raise AdoHcy and Hcy. Deficiency of Cbl or folate or perturbations of their metabolism cause major changes in sulfur amino acids.


Subject(s)
Amino Acids, Sulfur/metabolism , Folic Acid Deficiency/complications , Folic Acid/blood , Hyperhomocysteinemia/blood , Nutritional Status , Vitamin B 12 Deficiency/complications , Vitamin B 12/blood , Alcoholism/blood , Amino Acids, Sulfur/blood , Anemia, Megaloblastic/blood , Biomarkers/blood , Cardiovascular Diseases/blood , Folic Acid Deficiency/blood , Humans , Hyperhomocysteinemia/complications , Kidney Failure, Chronic/blood , Liver Diseases/blood , Obesity/blood , S-Adenosylhomocysteine/blood , Vitamin B 12 Deficiency/blood
17.
Crit Rev Food Sci Nutr ; 60(5): 722-739, 2020.
Article in English | MEDLINE | ID: mdl-30729795

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.


Subject(s)
Aging/metabolism , Cognition , Folic Acid/metabolism , Neurodegenerative Diseases/metabolism , Aging/drug effects , Cognition/drug effects , Dietary Supplements , Folic Acid/therapeutic use , Folic Acid Deficiency/complications , Folic Acid Deficiency/diet therapy , Folic Acid Deficiency/drug therapy , Folic Acid Deficiency/metabolism , Humans , Neurodegenerative Diseases/complications , Neurodegenerative Diseases/drug therapy
18.
J Pharmacol Sci ; 143(2): 97-105, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32173264

ABSTRACT

Folate deficiency has been suggested as a risk factor for depression in preclinical and clinical studies. Several hypotheses of mechanisms underlying folate deficiency-induced depressive symptoms have been proposed, but the detailed mechanisms are still unclear. In this study, we assessed whether post-weaning folate deficiency affect neurological and psychological function. The low folate diet-fed mice showed depression-like behavior in the forced swim test. In contrast, spontaneous locomotor activity, social behavior, coordinated motor skills, anxiety-like behavior and spatial memory did not differ between control and low folate diet-fed mice. In the dentate gyrus (DG) of the hippocampus, decreased number of newborn mature neurons and increased number of immature neurons were observed in low folate diet-fed mice. Staining with Golgi-Cox method revealed that dendritic complexity, spine density and the number of mature spines of neurons were markedly reduced in the DG of low folate diet-fed mice. Stress response of neurons indicated as c-Fos expression was also reduced in the DG of low folate diet-fed mice. These results suggest that reduction in the degree of maturation of newborn hippocampal neurons underlies folate deficiency-induced depressive symptoms.


Subject(s)
Dentate Gyrus/cytology , Dentate Gyrus/pathology , Depression/etiology , Depression/pathology , Folic Acid Deficiency/complications , Neurons/pathology , Weaning , Animals , Gene Expression , Male , Mice, Inbred Strains , Neurons/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism
19.
BMC Fam Pract ; 21(1): 13, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31964335

ABSTRACT

BACKGROUND: The study aimed to analyze anemia management in non-pregnant, and non-menopausal women aged from 18 to 50 years old, in a French primary care setting. METHODS: An observational descriptive prospective study was conducted between November 2018 and February 2019. Inclusion criteria were as followed: anemia diagnosed in women aged from 18 to 50, not pregnant and not menopausal. Quantitative and qualitative data were anonymized and collected through an electronic survey. Investigating general practitioners completed the questionnaire for each newly diagnosed woman. Mean values and medians were calculated for the quantitative data. Answers to the open questions were encoded manually and proportions of the different modalities have been calculated. RESULTS: Altogether, 43 women with anemia were ascertained. Moderate microcytic anemia, due to an iron deficiency in a context of menorrhagia, was the most observed anemia profile. The mean value of hemoglobin was 10.5 ± 1 g/dl. Among these women: 32 (74%) presented an iron deficiency, 17 (53%) had inappropriate intakes, and 9 (28%) reported menorrhagia. For 17 (40%) women, unnecessary or inappropriate exams were prescribed. The investigations did not allow to establish a differential diagnosis for 12 women (28%). Even for similar clinical situations, anemia management was variable. Among the women who presented iron deficiency, 15 (47%) were informed about an iron-rich diet and received a daily iron supplementation of ferrous sulfate between 80 mg and 160 mg. CONCLUSIONS: Our study highlights that, in the absence of specific national guidelines for anemia management in non-pregnant, non-menopausal women in primary care settings, French GPs undergo various clinical management strategies leading to a heterogeneous, sometimes inappropriate follow-up. Women with iron deficiency were prescribed higher daily iron supplementation than recommended, according to new evidence, suggesting a maximal daily dose of 50 mg of elementary iron in a context of Hepcidin up-regulation in the case of an iron overload. Additional longitudinal studies with a bigger sample size and randomized controlled trials are needed to confirm our results and to elaborate national guidelines.


Subject(s)
Anemia, Iron-Deficiency/therapy , Ferrous Compounds/therapeutic use , Hematinics/therapeutic use , Iron, Dietary/therapeutic use , Practice Patterns, Physicians' , Adolescent , Adult , Anemia/diagnosis , Anemia/metabolism , Anemia/therapy , Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/etiology , Anemia, Iron-Deficiency/metabolism , Diet Therapy , Disease Management , Erythrocyte Indices , Female , Ferritins/blood , Folic Acid Deficiency/complications , France , Guideline Adherence , Hemoglobins/metabolism , Humans , Menorrhagia/complications , Middle Aged , Practice Guidelines as Topic , Premenopause , Primary Health Care , Prospective Studies , Vitamin B 12 Deficiency/complications , Young Adult
20.
J Formos Med Assoc ; 119(6): 1109-1115, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31362840

ABSTRACT

BACKGROUND/PURPOSE: Normocytosis is defined as having the mean corpuscular volume (MCV) between 80 fL and 99.9 fL. This study evaluated whether 944 atrophic glossitis (AG) patients with normocytosis had significantly higher frequencies of anemia, hematinic deficiencies, hyperhomocysteinemia, and serum gastric parietal cell antibody (GPCA) positivity than 532 healthy control subjects or 1064 AG patients. METHODS: Complete blood count, serum iron, vitamin B12, folic acid, homocysteine, and GPCA levels in 944 AG patients with normocytosis, 1064 AG patients, and 532 healthy control subjects were measured and compared. RESULTS: We found that 12.4%, 14.5%, 2.3%, 2.0%, 9.0%, and 25.7% of 944 AG patients with normocytosis had blood hemoglobin (Hb), iron, vitamin B12, and folic acid deficiencies, hyperhomocysteinemia, and serum GPCA positivity, respectively. Furthermore, 944 AG patients with normocytosis had significantly higher frequencies of blood Hb, iron, vitamin B12, folic acid deficiencies, hyperhomocysteinemia, and serum GPCA positivity than 532 healthy control subjects (all P-values < 0.01). On the contrary, 944 AG patients with normocytosis had significantly lower frequencies of blood Hb and vitamin B12 deficiencies and hyperhomocysteinemia than overall 1064 AG patients (all P-values < 0.05). CONCLUSION: We conclude that there are significantly higher frequencies of anemia and serum iron, vitamin B12, and folic acid deficiencies, hyperhomocysteinemia, and serum GPCA positivity in AG patients with normocytosis than in healthy control subjects. On the contrary, AG patients with normocytosis have significantly lower frequencies of blood Hb and vitamin B12 deficiencies and hyperhomocysteinemia than overall AG patients.


Subject(s)
Folic Acid Deficiency , Glossitis , Hematinics , Hyperhomocysteinemia , Vitamin B 12 Deficiency , Case-Control Studies , Erythrocyte Indices , Folic Acid , Folic Acid Deficiency/complications , Glossitis/complications , Hemoglobins/analysis , Humans , Parietal Cells, Gastric , Vitamin B 12 , Vitamin B 12 Deficiency/complications
SELECTION OF CITATIONS
SEARCH DETAIL