Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters

Publication year range
1.
FASEB J ; 38(1): e23346, 2024 01.
Article in English | MEDLINE | ID: mdl-38095297

ABSTRACT

Folate deficiency contribute to neural tube defects (NTDs) which could be rescued by folate supplementation. However, the underlying mechanisms are still not fully understood. Besides, there is considerable controversy concerning the forms of folate used for supplementation. To address this controversy, we prepared culture medium with different forms of folate, folic acid (FA), and 5-methyltetrahydrofolate (5mTHF), at concentrations of 5 µM, 500 nM, 50 nM, and folate free, respectively. Mouse embryonic fibroblasts (MEFs) were treated with different folates continuously for three passages, and cell proliferation and F-actin were monitored. We determined that compared to 5mTHF, FA showed stronger effects on promoting cell proliferation and F-actin formation. We also found that FOLR1 protein level was positively regulated by folate concentration and the non-canonical Wnt/planar cell polarity (PCP) pathway signaling was significantly enriched among different folate conditions in RNA-sequencing analyses. We demonstrated for the first time that FOLR1 could promote the transcription of Vangl2, one of PCP core genes. The transcription of Vangl2 was down-regulated under folate-deficient condition, which resulted in a decrease in PCP activity and F-actin formation. In summary, we identified a distinct advantage of FA in cell proliferation and F-actin formation over 5mTHF, as well as demonstrating that FOLR1 could promote transcription of Vangl2 and provide a new mechanism by which folate deficiency can contribute to the etiology of NTDs.


Subject(s)
Folic Acid Deficiency , Neural Tube Defects , Animals , Mice , Folic Acid/metabolism , Actins/metabolism , Folate Receptor 1/genetics , Folate Receptor 1/metabolism , Cell Polarity/genetics , Fibroblasts/metabolism , Wnt Signaling Pathway , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Folic Acid Deficiency/metabolism
2.
BMC Biol ; 22(1): 146, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956599

ABSTRACT

BACKGROUND: Metabolic associated fatty liver disease (MAFLD), a prevalent liver disorder affecting one-third of the global population, encompasses a spectrum ranging from fatty liver to severe hepatic steatosis. Both genetic and lifestyle factors, particularly diet and nutrition, contribute to its etiology. Folate deficiency, a frequently encountered type of malnutrition, has been associated with the pathogenesis of MAFLD and shown to impact lipid deposition. However, the underlying mechanisms of this relationship remain incompletely understood. We investigated the impact of disturbed folate-mediated one-carbon metabolism (OCM) on hepatic lipid metabolism both in vitro using human hepatoma cells and in vivo using transgenic fluorescent zebrafish displaying extent-, stage-, and duration-controllable folate deficiency upon induction. RESULTS: Disturbed folate-mediated one-carbon metabolism, either by inducing folate deficiency or adding anti-folate drug, compromises autophagy and causes lipid accumulation in liver cells. Disturbed folate status down-regulates cathepsin L, a key enzyme involved in autophagy, through inhibiting mTOR signaling. Interfered mitochondrial biology, including mitochondria relocation and increased fusion-fission dynamics, also occurs in folate-deficient hepatocytes. Folate supplementation effectively mitigated the impaired autophagy and lipid accumulation caused by the inhibition of cathepsin L activity, even when the inhibition was not directly related to folate deficiency. CONCLUSIONS: Disruption of folate-mediated OCM diminishes cathepsin L expression and impedes autophagy via mTOR signaling, leading to lipid accumulation within hepatocytes. These findings underscore the crucial role of folate in modulating autophagic processes and regulating lipid metabolism in the liver.


Subject(s)
Autophagy , Folic Acid , Hepatocytes , Homeostasis , Lipid Metabolism , Zebrafish , Autophagy/physiology , Folic Acid/metabolism , Humans , Hepatocytes/metabolism , Animals , Folic Acid Deficiency/metabolism
3.
Mutagenesis ; 39(3): 196-204, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38417824

ABSTRACT

The developmental origins of health and disease hypothesis suggest early-life environment impacts health outcomes throughout the life course. In particular, epigenetic marks, including DNA methylation, are thought to be key mechanisms through which environmental exposures programme later-life health. Adequate maternal folate status before and during pregnancy is essential in the protection against neural tube defects, but data are emerging that suggest early-life folate exposures may also influence neurocognitive outcomes in childhood and, potentially, thereafter. Since folate is key to the supply of methyl donors for DNA methylation, we hypothesize that DNA methylation may be a mediating mechanism through which maternal folate influences neurocognitive outcomes. Using bisulphite sequencing, we measured DNA methylation of five genes (Art3, Rsp16, Tspo, Wnt16, and Pcdhb6) in the brain tissue of adult offspring of dams who were depleted of folate (n = 5, 0.4 mg folic acid/kg diet) during pregnancy (~19-21 days) and lactation (mean 22 days) compared with controls (n = 6, 2 mg folic acid/kg diet). Genes were selected as methylation of their promoters had previously been found to be altered by maternal folate intake in mice and humans across the life course, and because they have potential associations with neurocognitive outcomes. Maternal folate depletion was significantly associated with Art3 gene hypomethylation in subcortical brain tissue of adult mice at 28 weeks of age (mean decrease 6.2%, P = .03). For the other genes, no statistically significant differences were found between folate depleted and control groups. Given its association with neurocognitive outcomes, we suggest Art3 warrants further study in the context of lifecourse brain health. We have uncovered a potential biomarker that, once validated in accessible biospecimens and human context, may be useful to track the impact of early-life folate exposure on later-life neurocognitive health, and potentially be used to develop and monitor the effects of interventions.


Subject(s)
Brain , DNA Methylation , Folic Acid , Prenatal Exposure Delayed Effects , Animals , DNA Methylation/drug effects , Female , Brain/metabolism , Brain/drug effects , Pregnancy , Mice , Prenatal Exposure Delayed Effects/genetics , Folic Acid Deficiency/genetics , Epigenesis, Genetic , Male
4.
Eur J Nutr ; 63(2): 563-572, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38129362

ABSTRACT

PURPOSE: To our knowledge, there are very few studies evaluating if the levels of folate modify the risk of cervical intraepithelial neoplasia grade 2 and higher (CIN2+ and CIN3+) associated with the levels of HPV genome methylation, two cofactors related to single carbon metabolism and independently associated with cervical cancer in previous studies. We conducted a case-control study nested in a three-arm randomized clinical pragmatic trial (ASCUS-COL trial) to evaluate the risk of CIN3+ associated with methylation levels according to serum folate concentrations. METHODS: Cases (n = 155) were women with histologically confirmed CIN2+ (113 CIN2, 38 CIN3, and 4 SCC) and controls were age and follow-up time at diagnosis-matched women with histologically confirmed ≤ CIN1 (n = 155), selected from the 1122 hrHPV + women of this trial. The concentrations of serum folate were determined by the radioimmunoassay SimulTRAC-SNB-VitaminB12/Folate-RIAKit and the methylation levels by the S5 classifier. Stepwise logistic regression models were used to estimate the association between folate or methylation levels and CIN2+ or CIN3+. The joint effect of folate levels and methylation on the risk of CIN3+ was estimated using combinations of categorical stratifications. RESULTS: Folate levels were significantly lower in women with CIN3+ than in other diagnostic groups (p = 0.019). The risk of CIN3+ was eight times higher (OR 8.9, 95% CI 3.4-24.9) in women with folate deficiency and high methylation levels than in women with normal folate and high methylation levels (OR 1.4, 95% CI 0.4-4.6). CONCLUSION: High methylation and deficient folate independently increased the risk of CIN3+ while deficient folate combined with high methylation was associated with a substantially elevated risk of CIN3+.


Subject(s)
Atypical Squamous Cells of the Cervix , Folic Acid Deficiency , Uterine Cervical Dysplasia , Female , Humans , Case-Control Studies , DNA Methylation , Folic Acid , Folic Acid Deficiency/genetics , Uterine Cervical Dysplasia/diagnosis , Uterine Cervical Dysplasia/genetics , Uterine Cervical Dysplasia/pathology
5.
Appl Microbiol Biotechnol ; 108(1): 40, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38175236

ABSTRACT

Folic acid deficiency is common worldwide and is linked to an imbalance in gut microbiota. However, based on model animals used to study the utilization of folic acid by gut microbes, there are challenges of reproducibility and individual differences. In this study, an in vitro fecal slurry culture model of folic acid deficiency was established to investigate the effects of supplementation with 5-methyltetrahydrofolate (MTHF) and non-reduced folic acid (FA) on the modulation of gut microbiota. 16S rRNA sequencing results revealed that both FA (29.7%) and MTHF (27.9%) supplementation significantly reduced the relative abundance of Bacteroidetes compared with control case (34.3%). MTHF supplementation significantly improved the relative abundance of Firmicutes by 4.49%. Notably, compared with the control case, FA and MTHF supplementation promoted an increase in fecal levels of Lactobacillus, Bifidobacterium, and Pediococcus. Short-chain fatty acid (SCFA) analysis showed that folic acid supplementation decreased acetate levels and increased fermentative production of isobutyric acid. The in vitro fecal slurry culture model developed in this study can be utilized as a model of folic acid deficiency in humans to study the gut microbiota and demonstrate that exogenous folic acid affects the composition of the gut microbiota and the level of SCFAs. KEY POINTS: • Establishment of folic acid deficiency in an in vitro culture model. • Folic acid supplementation regulates intestinal microbes and SCFAs. • Connections between microbes and SCFAs after adding folic acid are built.


Subject(s)
Folic Acid Deficiency , Gastrointestinal Microbiome , Animals , Humans , Folic Acid , Fermentation , RNA, Ribosomal, 16S/genetics , Reproducibility of Results , Fatty Acids, Volatile
6.
Int J Biometeorol ; 68(7): 1343-1356, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38607561

ABSTRACT

Previous studies have suggested that exposure to air pollutants may be associated with specific blood indicators or anemia in certain populations. However, there is insufficient epidemiological data and prospective evidence to evaluate the relationship between environmental air pollution and specific types of anemia. We conducted a large-scale prospective cohort study based on the UK Biobank. Annual average concentrations of NO2, PM2.5, PM2.5-10, and PM10 were obtained from the ESCAPE study using the Land Use Regression (LUR) model. The association between atmospheric pollutants and different types of anemia was investigated using the Cox proportional hazards model. Furthermore, restricted cubic splines were used to explore exposure-response relationships for positive associations, followed by stratification and effect modification analyses by gender and age. After adjusting for demographic characteristics, 3-4 of the four types of air pollution were significantly associated with an increased risk of iron deficiency, vitamin B12 deficiency and folate deficiency anemia, while there was no significant association with other defined types of anemia. After full adjustment, we estimated that the hazard ratios (HRs) of iron deficiency anemia associated with each 10 µg/m3 increase in NO2, PM2.5, and PM10 were 1.04 (95%CI: 1.02, 1.07), 2.00 (95%CI: 1.71, 2.33), and 1.10 (95%CI: 1.02, 1.20) respectively. The HRs of folate deficiency anemia with each 10 µg/m3 increase in NO2, PM2.5, PM2.5-10, and PM10 were 1.25 (95%CI: 1.12, 1.40), 4.61 (95%CI: 2.03, 10.47), 2.81 (95%CI: 1.11, 7.08), and 1.99 (95%CI: 1.25, 3.15) respectively. For vitamin B12 deficiency anemia, no significant association with atmospheric pollution was found. Additionally, we estimated almost linear exposure-response curves between air pollution and anemia, and interaction analyses suggested that gender and age did not modify the association between air pollution and anemia. Our research provided reliable evidence for the association between long-term exposure to PM10, PM2.5, PM2.5-10, NO2, and several types of anemia. NO2, PM2.5, and PM10 significantly increased the risk of iron deficiency anemia and folate deficiency anemia. Additionally, we found that the smaller the PM diameter, the higher the risk, and folate deficiency anemia was more susceptible to air pollution than iron deficiency anemia. No association was observed between the four types of air pollution and hemolytic anemia, aplastic anemia, and other types of anemia. Although the mechanisms are not well understood, we emphasize the need to limit the levels of PM and NO2 in the environment to reduce the potential impact of air pollution on folate and iron deficiency anemia.


Subject(s)
Air Pollutants , Anemia , Particulate Matter , Humans , Male , Female , Air Pollutants/analysis , Air Pollutants/adverse effects , Middle Aged , United Kingdom/epidemiology , Particulate Matter/analysis , Particulate Matter/adverse effects , Anemia/epidemiology , Anemia/blood , Prospective Studies , Aged , Adult , Nitrogen Dioxide/analysis , Nitrogen Dioxide/adverse effects , Proportional Hazards Models , Biological Specimen Banks , Risk Assessment , Air Pollution/adverse effects , Air Pollution/analysis , Environmental Exposure/adverse effects , Environmental Exposure/analysis , Folic Acid Deficiency/epidemiology , Folic Acid Deficiency/blood , UK Biobank
7.
J Assoc Physicians India ; 72(1): 113, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38736088

ABSTRACT

We found the article on "The Digital Technology in Clinical Medicine: From Calculators to ChatGPT" interesting.1 According to Kulkarni et al., humanity has witnesses four important social system changes, starting with the primitive huntersgatherers and progressing to horticultural, agricultural, industrial, and the current fifth, which is based on digital information technology and has altered the way we present, recognize, and utilize different factors of production. In clinical medicine, digital technology has advanced significantly since the days of computations. According to Kulkarni et al., we have to benefit from these advancements as we all improve the lives of our patients while being cautious not to overturn the doctor-patient relationship. If technology, clinical expertise, and humanistic values are properly balanced, Kulkarni et al. concluded that the future is quite glorious.1 Regulatory organizations are pushing for improvements through clinical trials as a result of recognition of the expanding influence of digital technology in healthcare delivery. The "World Health Organizations Guidelines for Digital Interventions" and the "Food and Drug Administration's Digital Health Center of Excellence" are only two of the projects that are currently being highlighted in the study as efforts to analyze and implement digital health services.


Subject(s)
Folic Acid Deficiency , Pancytopenia , Humans , Pancytopenia/etiology , Pancytopenia/diagnosis , Folic Acid/therapeutic use
8.
Pract Neurol ; 24(1): 56-59, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38135499

ABSTRACT

A 25-year-old woman with childhood-onset refractory epilepsy and developmental delay experienced a gradually progressive marked deterioration in mobility and seizure control, with language regression. Investigation identified a homozygous deletion within the contactin-associated protein-like 2 gene (CNTNAP2), underlying her early presentation, but also cerebral folate deficiency that most likely contributed to her later deterioration. Following antiseizure medication adjustment and treatment with folinic acid, she stabilised with improved seizure control and limited improvement in language and motor function; she has remained neurologically stable for more than a decade. That the previously observed neurological decline was halted by folinic acid replacement supports this being due to cerebral folate deficiency. Metabolic conditions are less well recognised in adults and can be under-diagnosed. They are potentially treatable and should be considered even in the presence of another cause, particularly when the presentation is not fully compatible.


Subject(s)
Epilepsy , Folate Receptor 1/deficiency , Folic Acid Deficiency , Neuroaxonal Dystrophies , Adult , Female , Humans , Child , Leucovorin/genetics , Leucovorin/therapeutic use , Folic Acid Deficiency/diagnosis , Folic Acid Deficiency/drug therapy , Folic Acid Deficiency/genetics , Homozygote , Sequence Deletion , Seizures
9.
Brain Res ; 1822: 148639, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37858854

ABSTRACT

Folate is essential for the normal growth and development of the fetus. Folic acid supplementation during the fetal period affects postnatal brain development and reduces the incidence of mental disorders in animal and human studies. However, the association between folate deficiency (FD) during pregnancy and developmental disorders in children remains poorly understood. In this study, we investigated whether prenatal FD is associated with neurodevelopmental disorders in offspring. ICR mice were fed a control diet (2 mg folic acid/kg diet) or a folate-deficient diet (0.3 mg folic acid/kg diet) from embryonic day 1 until parturition. We evaluated locomotor activity, anxiety, grooming, sociability and learning memory in male offspring at 7-10 weeks of age. No differences were found in locomotor activity or anxiety in the open field test, nor in grooming time in the self-grooming test. However, sociability, spatial memory, and novel object recognition were impaired in the FD mice compared with control offspring. Furthermore, we measured protein expression levels of the NMDA and AMPA receptors, as well as PSD-95 and the GABA-synthesizing enzymes GAD65/67 in the frontal cortex and hippocampus. In FD mice, expression levels of AMPA receptor 1 and PSD-95 in both regions were reduced compared with control mice. Moreover, NMDA receptor subunit 2B and GAD65/67 were significantly downregulated in the frontal cortex of prenatal FD mice compared with the controls. Collectively, these findings suggest that prenatal FD causes behavioral deficits together with a reduction in synaptic protein levels in the frontal cortex and hippocampus.


Subject(s)
Folic Acid Deficiency , Prenatal Exposure Delayed Effects , Humans , Pregnancy , Female , Child , Animals , Male , Mice , Folic Acid/metabolism , Mice, Inbred ICR , Folic Acid Deficiency/complications , Folic Acid Deficiency/metabolism , Diet , Brain/metabolism , Prenatal Exposure Delayed Effects/metabolism
10.
Reprod Toxicol ; 125: 108576, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38479591

ABSTRACT

Folate is a micronutrient essential for DNA synthesis, cell division, fetal growth and development. Folate deficiency leads to genomic instability. Inadequate intake of folate during conception may lead to neural tube defects (NTDs) in the offspring. Folate influences the DNA methylation, histone methylation and homocysteine mediated gene methylation. DNA methylation influences the expression of microRNAs (miRNAs). Folate deficiency may be associated with miRNAs misregulation leading to NTDs. Mitochondrial epigenetics and folate metabolism has proved to be involved in embryogenesis and neural tube development. Folate related genetic variants also cause the occurrence of NTDs. Unmetabolized excessive folate may affect health adversely. Hence estimation of folate levels in the blood plays an important role in high-risk cases.


Subject(s)
Folic Acid Deficiency , MicroRNAs , Neural Tube Defects , Humans , Folic Acid , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Folic Acid Deficiency/complications , Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Epigenesis, Genetic , DNA Methylation , MicroRNAs/genetics , Neural Tube/metabolism
11.
Sci Rep ; 14(1): 3951, 2024 02 17.
Article in English | MEDLINE | ID: mdl-38365858

ABSTRACT

We investigated the clinical implications of the mean corpuscular volume (MCV) in patients with major trauma. This single-center retrospective review included 2021 trauma patients admitted to the intensive care unit between January 2016 and June 2020. We included 1218 patients aged [Formula: see text] 18 years with an injury severity score [Formula: see text] 16 in the final analysis. The clinical and laboratory variables were compared between macrocytosis (defined as MCV [Formula: see text] 100 fL) and non-macrocytosis groups. Cox regression analysis was performed to calculate the hazard ratios (HRs) of variables for 30-day mortality, with adjustment for other potential confounding factors. The initial mean value of MCV was 102.7 fL in the macrocytosis group (n = 199) and 93.7 fL in the non-macrocytosis group (n = 1019). The macrocytosis group showed a significantly higher proportion of initial hypotension, transfusion within 4 and 24 h, and 30-day mortality than the non-macrocytosis group. Age ([Formula: see text] 65 years), hypotension (systolic blood pressure [Formula: see text] 90 mmHg), transfusion (within 4 h), anemia (Hb < 12 g/day in women, < 13 g/day in men), and macrocytosis were significantly associated with 30-day mortality (adjusted HR = 1.4; 95% confidence interval 1.01-1.94; p = 0.046) in major trauma patients. Thus, initial macrocytosis independently predicted 30-day mortality in patients with major trauma at a Level I trauma center.


Subject(s)
Anemia, Macrocytic , Anemia , Folic Acid Deficiency , Hypotension , Male , Humans , Female , Aged , Erythrocyte Indices , Retrospective Studies , Prognosis
12.
J Vis Exp ; (208)2024 06 28.
Article in English | MEDLINE | ID: mdl-39007568

ABSTRACT

Maternal nutrition during pregnancy and lactation plays an important role in the neurodevelopment of offspring. One-carbon (1C) metabolism, which centers around folic acid and choline, as well as other B vitamins, plays a key role during the closure of the neural tube of the developing fetus. However, the impact of these maternal nutritional deficiencies during pregnancy on offspring health outcomes after birth remains relatively undefined. Furthermore, maternal dietary deficiencies in folic acid or choline may impact other health outcomes in offspring - making this a valuable model. This protocol aims to outline the procedure for inducing a deficiency in 1C metabolism in female mice through dietary modifications. Females are placed on diets at weaning, up to 2 months of age, for 4-6 weeks prior to mating and remain on diet throughout pregnancy and lactation. Offspring from these females can be evaluated for health outcomes. Females can be used multiple times to generate offspring, and tissues from females can be collected to measure for 1C metabolite measurements. This protocol provides an overview of how to induce maternal dietary deficiencies in folic acid or choline to study offspring health outcomes.


Subject(s)
Choline Deficiency , Diet , Folic Acid Deficiency , Maternal Nutritional Physiological Phenomena , Folic Acid/metabolism , Choline/metabolism , Female , Animals , Mice , Folic Acid Deficiency/pathology , Choline Deficiency/pathology
13.
Mol Nutr Food Res ; 68(5): e2300355, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38327171

ABSTRACT

SCOPE: Disturbances in one-carbon metabolism contribute to nonalcoholic fatty liver disease (NAFLD) which encompasses steatosis, steatohepatitis, fibrosis, and cirrhosis. The goal is to examine impact of folate deficiency and the Mthfr677C >T variant on NAFLD. METHODS AND RESULTS: This study uses the new Mthfr677C >T mouse model for the human MTHFR677C >T variant. Mthfr677CC and Mthfr677TT mice were fed control diet (CD) or folate-deficient (FD) diets for 4 months. FD and Mthfr677TT alter choline/methyl metabolites in liver and/or plasma (decreased S-adenosylmethionine (SAM):S-adenosylhomocysteine (SAH) ratio, methyltetrahydrofolate, and betaine; increased homocysteine [Hcy]). FD, with contribution from Mthfr677TT, provokes fibrosis in males. Studies of normal livers reveal alterations in plasma markers and gene expression that suggest an underlying predisposition to fibrosis induced by FD and/or Mthfr677TT in males. These changes are absent or reverse in females, consistent with the sex disparity of fibrosis. Sex-based differences in methylation potential, betaine, sphingomyelin, and trimethylamine-N-oxide (TMAO) levels may prevent fibrogenesis in females. In contrast, Mthfr677TT alters choline metabolism, dysregulates expression of lipid metabolism genes, and promotes steatosis in females. CONCLUSION: This study suggests that folate deficiency predisposes males to fibrosis, which is exacerbated by Mthfr677TT, whereas Mthfr677TT predisposes females to steatosis, and reveal novel contributory mechanisms for these NAFLD-related disorders.


Subject(s)
Folic Acid Deficiency , Methylenetetrahydrofolate Reductase (NADPH2) , Non-alcoholic Fatty Liver Disease , Animals , Female , Humans , Male , Mice , Betaine , Choline/metabolism , Folic Acid , Folic Acid Deficiency/metabolism , Genotype , Homocysteine , Liver Cirrhosis/etiology , Non-alcoholic Fatty Liver Disease/etiology , S-Adenosylmethionine
14.
Science ; 384(6695): 584-590, 2024 05 03.
Article in English | MEDLINE | ID: mdl-38696583

ABSTRACT

Meningomyelocele is one of the most severe forms of neural tube defects (NTDs) and the most frequent structural birth defect of the central nervous system. We assembled the Spina Bifida Sequencing Consortium to identify causes. Exome and genome sequencing of 715 parent-offspring trios identified six patients with chromosomal 22q11.2 deletions, suggesting a 23-fold increased risk compared with the general population. Furthermore, analysis of a separate 22q11.2 deletion cohort suggested a 12- to 15-fold increased NTD risk of meningomyelocele. The loss of Crkl, one of several neural tube-expressed genes within the minimal deletion interval, was sufficient to replicate NTDs in mice, where both penetrance and expressivity were exacerbated by maternal folate deficiency. Thus, the common 22q11.2 deletion confers substantial meningomyelocele risk, which is partially alleviated by folate supplementation.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 22 , Meningomyelocele , Animals , Female , Humans , Male , Mice , Chromosomes, Human, Pair 22/genetics , DiGeorge Syndrome/genetics , Exome Sequencing , Folic Acid/administration & dosage , Folic Acid Deficiency/complications , Folic Acid Deficiency/genetics , Meningomyelocele/epidemiology , Meningomyelocele/genetics , Penetrance , Spinal Dysraphism/genetics , Risk , Adaptor Proteins, Signal Transducing/genetics
15.
Nutrients ; 16(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892606

ABSTRACT

The prevalence of anaemia in India remains high in children, especially those in rural areas, and in women of childbearing age, and its impairment of neurological development can have serious lifelong effects. It is concerning that the most recent official data (2019-21) indicate an increased prevalence compared with 2015-16. There is also considerable variability in childhood anaemia between Indian states with socioeconomic factors, such as wealth and education contributing to the risk of anaemia among adolescent women and their children. Dietary iron deficiency is often regarded as the main contributor to anaemia but increasing evidence accumulated from the authors' ongoing literature database coupled with recent literature research suggests that it has a multifactorial aetiology, some of which is not related to nutrition. This narrative review focused on these multifactorial issues, notably the contribution of vitamin B12/folate deficiency, which also has a high prevalence in India. It was also noted that the dietary intake of bioavailable iron remains an important contributor for reducing anaemia, and the role of millets as an improved iron source compared to traditional staple cereals is briefly discussed. The overall conclusion is that anaemia has a multifactorial aetiology requiring multifactorial assessment that must include assessment of vitamin B12 status.


Subject(s)
Anemia , Folic Acid Deficiency , Vitamin B 12 Deficiency , Humans , India/epidemiology , Prevalence , Vitamin B 12 Deficiency/epidemiology , Female , Anemia/epidemiology , Anemia/etiology , Folic Acid Deficiency/epidemiology , Adolescent , Child , Anemia, Iron-Deficiency/epidemiology , Anemia, Iron-Deficiency/etiology , Male , Iron, Dietary/administration & dosage , Nutritional Status , Adult , Risk Factors , Diet/adverse effects , Vitamin B 12/administration & dosage , Vitamin B 12/blood
16.
J Glob Health ; 14: 04120, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38991209

ABSTRACT

Background: Preterm birth (PTB) and its complications are important public health problems. Its aetiology is multifactorial and involves both modifiable and non-modifiable factors. Among the modifiable risk factors, micronutrient deficiencies, including maternal folate deficiency, are increasingly being studied in PTB. In this study, we estimated the prevalence of folate deficiency during pregnancy and examined its association with PTB among rural Bangladeshi women. Methods: We conducted a nested case-control study using data from a population-based cohort of 3000 pregnant women who were enrolled between 8 and 19 weeks of gestation following ultrasound confirmation of gestational age. Sociodemographic, epidemiologic, clinical, and pregnancy outcomes data were collected through home visits, while blood samples were collected at enrolment and 24-28 weeks of gestation during pregnancy. We included all women who delivered preterm (defined as live births <37 weeks of gestation) as cases (n = 235) and a random sample of women having a term birth as controls (n = 658). The main exposure was folate concentrations in maternal serum during 24-28 weeks of pregnancy. We categorised women into folate deficient (<3 ng/mL) and not deficient (≥3 ng/mL). We then performed multivariable logistic regression analysis to examine the association between maternal folate levels and PTB, adjusting for relevant covariates. Results: Thirty-eight per cent of the enrolled pregnant women were folate deficient. Maternal serum folate deficiency was significantly associated with PTB (adjusted OR (aOR) = 1.73; 95% confidence interval (CI) = 1.27-2.36). The risk of PTB was also higher among women who were of short stature (aOR = 1.83; 95% CI = 1.27-2.63), primiparous (aOR = 1.60; 95% CI = 1.15-2.22), and had exposure to passive smoking (aOR = 1.54; 95% CI = 1.02-2.31). Conclusions: The prevalence of folate deficiency was high among pregnant women in rural Bangladesh, and folate deficiency was significantly associated with an increased risk of preterm birth.


Subject(s)
Folic Acid Deficiency , Premature Birth , Humans , Female , Pregnancy , Case-Control Studies , Premature Birth/epidemiology , Adult , Folic Acid Deficiency/epidemiology , Bangladesh/epidemiology , Risk Factors , Young Adult , Pregnancy Complications/epidemiology , Prevalence , Folic Acid/blood , Rural Population/statistics & numerical data
17.
Birth Defects Res ; 116(8): e2390, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39162364

ABSTRACT

BACKGROUND: Folate and vitamin B12 deficiencies in pregnant women are associated with increased risk for adverse maternal and infant health outcomes, including neural tube defects (NTDs). METHODS: A population-based cross-sectional survey was conducted in two rural areas in Ambala District, Haryana, India in 2017 to assess baseline folate and vitamin B12 status among women of reproductive age (WRA) and predict the prevalence of NTDs. We calculated the prevalence of folate and vitamin B12 deficiency and insufficiency by demographic characteristics among 775 non-pregnant, non-lactating WRA (18-49 years). Using red blood cell (RBC) folate distributions and an established Bayesian model, we predicted NTD prevalence. All analyses were conducted using SAS-callable SUDAAN Version 11.0.4 to account for complex survey design. RESULTS: Among WRA, 10.1% (95% CI: 7.9, 12.7) and 9.3% (95% CI: 7.4, 11.6) had serum (<7 nmol/L) and RBC folate (<305 nmol/L) deficiency, respectively. The prevalence of RBC folate insufficiency (<748 nmol/L) was 78.3% (95% CI: 75.0, 81.3) and the predicted NTD prevalence was 21.0 (95% uncertainly interval: 16.9, 25.9) per 10,000 live births. Prevalences of vitamin B12 deficiency (<200 pg/mL) and marginal deficiency (≥200 pg/mL and ≤300 pg/mL) were 57.7% (95% CI: 53.9, 61.4) and 23.5% (95% CI: 20.4, 26.9), respectively. CONCLUSIONS: The magnitude of folate insufficiency and vitamin B12 deficiency in this Northern Indian population is a substantial public health concern. The findings from the survey help establish the baseline against which results from future post-fortification surveys can be compared.


Subject(s)
Folic Acid Deficiency , Folic Acid , Neural Tube Defects , Rural Population , Vitamin B 12 Deficiency , Vitamin B 12 , Humans , Female , Neural Tube Defects/epidemiology , Neural Tube Defects/etiology , India/epidemiology , Adult , Folic Acid/blood , Vitamin B 12/blood , Prevalence , Cross-Sectional Studies , Pregnancy , Vitamin B 12 Deficiency/epidemiology , Folic Acid Deficiency/epidemiology , Folic Acid Deficiency/blood , Adolescent , Young Adult , Middle Aged , Bayes Theorem
18.
J Osteopath Med ; 124(5): 213-218, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38329818

ABSTRACT

CONTEXT: Folate deficiency is often observed in patients with inflammatory diseases, raising questions about its role in knee osteoarthritis (OA) progression. OBJECTIVES: This study aimed to assess the association of folate deficiency with the clinical and radiological severity of knee OA. METHODS: A prospective cross-sectional study was conducted from January 1, 2019 to January 1, 2020. Primary knee OA patients referred to orthopedic clinics in Zabol, Iran were included. Radiographic severity was gauged utilizing the Kellgren-Lawrence (KL) classification. For clinical severity, patients completed the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) questionnaire. IBM SPSS v.27 facilitated the statistical analysis. RESULTS: Forty-nine knee OA patients, averaging 67.45±13.44 years in age, were analyzed. Spearman correlation analysis revealed a negative correlation between folate levels and both WOMAC and KL scores. The correlation was stronger between folate and KL score (Spearman correlation coefficient: -0.75) than between folate and WOMAC total score (Spearman correlation coefficient: -0.46). Additionally, a significantly higher KL score was observed in patients with folate deficiency (p=0.004). CONCLUSIONS: Our study highlights a significant correlation between folate deficiency and increased severity of OA, which is evident in radiological and clinical assessments. These findings suggest that folate plays a key role in OA pathogenesis and could be a modifiable factor in its management.


Subject(s)
Folic Acid Deficiency , Osteoarthritis, Knee , Radiography , Severity of Illness Index , Humans , Osteoarthritis, Knee/diagnostic imaging , Male , Female , Cross-Sectional Studies , Prospective Studies , Aged , Middle Aged , Aged, 80 and over , Iran/epidemiology , Folic Acid/blood , Surveys and Questionnaires
19.
Life Sci Alliance ; 7(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39043420

ABSTRACT

Folate is a vitamin required for cell growth and is present in fortified foods in the form of folic acid to prevent congenital abnormalities. The impact of low-folate status on life-long health is poorly understood. We found that limiting folate levels with the folate antagonist methotrexate increased the lifespan of yeast and worms. We then restricted folate intake in aged mice and measured various health metrics, metabolites, and gene expression signatures. Limiting folate intake decreased anabolic biosynthetic processes in mice and enhanced metabolic plasticity. Despite reduced serum folate levels in mice with limited folic acid intake, these animals maintained their weight and adiposity late in life, and we did not observe adverse health outcomes. These results argue that the effectiveness of folate dietary interventions may vary depending on an individual's age and sex. A higher folate intake is advantageous during the early stages of life to support cell divisions needed for proper development. However, a lower folate intake later in life may result in healthier aging.


Subject(s)
Folic Acid , Longevity , Animals , Folic Acid/administration & dosage , Folic Acid/metabolism , Mice , Male , Female , Aging/metabolism , Diet/methods , Mice, Inbred C57BL , Methotrexate/pharmacology , Folic Acid Deficiency/metabolism , Caenorhabditis elegans , Saccharomyces cerevisiae/metabolism
20.
J Am Nutr Assoc ; 43(5): 452-463, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38289269

ABSTRACT

OBJECTIVE: This study evaluated the effect of an altered ratio of maternal RBC folate (MRF) to serum vitamin B12 (MB12) on pregnancy and newborn outcomes. METHODS: Blood samples were collected from pregnant women and the umbilical cord at the time of delivery. Estimations of RBC folate and serum vitamin B12 from maternal and cord blood samples and total homocysteine (HCY) were performed. Maternal and newborn anthropometric parameters like placental weight (PW), head circumference (HC), chest circumference (CC), and body weight (BW) were measured in offsprings after birth. We stratified the pregnant women into six groups (a) vitamin B12 normal and folic acid normal (BNFN)-control group, (b) vitamin B12 normal and folic acid elevated (BNFE), (c) vitamin B12 normal and folic acid deficient (BNFD), (d) vitamin B12 deficient and folic acid normal (BDFN), (e) vitamin B12 deficient and folic acid elevated (BDFE) and (f) vitamin B12 deficient and folic acid deficient (BDFD) based on their levels of RBC folate (MRF) and vitamin B12 (MB12). The expression of the one-carbon metabolism genes (methionine synthase (MS), glycine N-methyltransferase (GNMT), and cystathionine ß-synthase (CBS) was also studied in placental tissue by using real-time PCR. RESULTS: Cord blood RBC folate was significantly reduced in groups BDFE and BDFD as compared to the control group (BNFN). The cord blood vitamin B12 levels were also reduced in the BDFE group as compared to the BDFD. All the newborn parameters viz. PW, HC, CC, and BW, were reduced in the altered MRF/MB12 ratio (low & high vs. normal ratio). Total HCY was significantly elevated in the groups with (BDFE & BDFN) an imbalance of maternal RBC folate and serum vitamin B12 as compared to the control group. Downregulation of one-carbon metabolism genes like MS (p < 0.001), GNMT (p < 0.05), and CBS (p < 0.01) in placental tissue was observed in the high MRF/MB12 ratio group as compared to the normal ratio group. A strong positive correlation was also observed between MRF, MB12, and newborn parameters. CONCLUSIONS: The altered ratio of folate to vitamin B12 in the maternal blood is associated with adverse growth and development of the newborn.


Subject(s)
Birth Weight , Fetal Blood , Folic Acid , Vitamin B 12 , Humans , Female , Vitamin B 12/blood , Pregnancy , Folic Acid/blood , Infant, Newborn , Adult , Fetal Blood/metabolism , Fetal Blood/chemistry , Vitamin B 12 Deficiency/blood , Vitamin B 12 Deficiency/metabolism , Homocysteine/blood , Erythrocytes/metabolism , Folic Acid Deficiency/blood , Young Adult , Head/anatomy & histology , Placenta/metabolism , Maternal Nutritional Physiological Phenomena
SELECTION OF CITATIONS
SEARCH DETAIL