Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Eur J Nutr ; 63(1): 291-302, 2024 Feb.
Article En | MEDLINE | ID: mdl-37870657

PURPOSE: Oxidative stress has been reported to cause telomere attrition, which triggers cell apoptosis. Apoptosis of neurocytes may play an essential role in the pathogenesis of neurodegenerative diseases. This study hypothesized that folic acid (FA) supplementation decreased neurocyte apoptosis by alleviating oxidative stress-induced telomere attrition in 25-month-old Sprague Dawley (SD) rats. METHODS: Three-month-old male SD rats were randomly divided into four diet groups by different concentrations of folic acid in equal numbers, with intervention for 22 months. Folate, homocysteine (Hcy), reactive oxygen species (ROS) levels, antioxidant activities, and telomere length in the brain tissues were tested at 11, 18, and 22 months of intervention, and 8-hydroxy-deoxyguanosine (8-OHdG) levels, neurocyte apoptosis and telomere length in the cerebral cortex and hippocampal regions were tested during the 22-month intervention. An automated chemiluminescence system, auto-chemistry analyzer, Q-FISH, qPCR, and TUNEL assay were used in this study. RESULTS: The rats had lower folate concentrations and higher Hcy, ROS, and 8-OHdG concentrations in brain tissue with aging. However, FA supplementation increased folate concentrations and antioxidant activities while decreasing Hcy, ROS, and 8-OHdG levels in rat brain tissue after 11, 18, and 22 months of intervention. Furthermore, FA supplementation alleviated telomere length shortening and inhibited neurocyte apoptosis during the 22-month intervention. CONCLUSION: FA supplementation alleviated oxidative stress-induced telomere attrition and inhibited apoptosis of neurocytes in 25-month-old rats.


Antioxidants , Folic Acid , Rats , Male , Animals , Folic Acid/pharmacology , Antioxidants/pharmacology , Reactive Oxygen Species , Rats, Sprague-Dawley , Oxidative Stress , Apoptosis , 8-Hydroxy-2'-Deoxyguanosine , Telomere
2.
Int J Mol Sci ; 24(19)2023 Sep 25.
Article En | MEDLINE | ID: mdl-37833955

Research demonstrated that folate deficiency in either the mother or father could impact the biological functions of the offspring's of neural cells. Folate deficiency can also impair the methionine cycle, thus contributing to the conversion of S-adenosylmethionine (SAM) to S-adenosylhomocysteine (SAH), which could potentially cause damage to the central nervous system. The study focused on the effect of parental folate deficiency on neural cell apoptosis in offspring neonatal rats and whether it is mediated by the levels of SAM and SAH in brains. The experimental design was conducted by feeding female and male Sprague Dawley (SD) rats with either folate-deficient or folate-normal diets, sacrificing the offspring within 24 h and isolating their brain tissue. Rats were divided into four groups: the maternal-folate-deficient and paternal-folate-deficient (D-D) group; the maternal-folate-deficient and paternal-folate-normal (D-N) group; the maternal-folate-normal and paternal-folate-deficient (N-D) group; and the maternal-folate-normal and paternal-folate-normal (N-N) group. There was down-regulation of B-cell lymphoma 2 (Bcl-2) expression, up-regulation of Bcl-2-associated X protein (Bax) and Caspase-3 expression of neural cells, and pathological changes in the brain ultrastructure, as well as decreased SAM levels, increased SAH levels, and a decreased SAM/SAH ratio in the rat fetal brain via parental folate deficiency. In conclusion, parental folate deficiency could induce the apoptosis of neural cells in neonatal offspring rats, while biparental folate deficiency had the greatest effect on offspring, and the unilateral effect was greater in mothers than in fathers. This process may be mediated by the levels of SAM and SAH in the rat fetal brain.


Folic Acid Deficiency , Rats , Animals , Male , Female , Animals, Newborn , bcl-2-Associated X Protein/genetics , Caspase 3 , Rats, Sprague-Dawley , Folic Acid Deficiency/metabolism , Folic Acid , Apoptosis/physiology , S-Adenosylmethionine/metabolism
3.
J Nutr Biochem ; 122: 109455, 2023 12.
Article En | MEDLINE | ID: mdl-37788724

Maternal folate status during pregnancy is associated with the neurodevelopment of offspring; however, study results on the association between paternal folate status and offspring neurodevelopment are inconsistent. This study aimed to explore whether parental folic acid deficiency affects the neurobehavioral development of offspring by affecting the differentiation of neural stem cells (NSCs) into neurons. In the present study, the offspring were divided into four groups: parental folic acid deficient group (D-D), maternal folic acid deficient and paternal folic acid normal group (D-N), maternal folic acid normal and paternal folic acid deficient group (N-D), and parental folic acid normal group (N-N). For in vivo study, neurobehavioral indexes, and neuron-specific nuclear protein (NeuN) and glial fibrillary acidic protein (GFAP) expression in the brain hippocampus and cerebral cortex of offspring were measured at different time points. For in vitro study, NSCs were cultured from the hippocampus and striatum, and neuronal and astrocytic differentiation were measured. The results demonstrated that parental folic acid deficiency decreased the brain folate level in offspring, delayed early sensory-motor reflex development, impaired spatial learning and memory ability in adolescence and adulthood, decreased differentiation of NSCs into neurons and increased differentiation of NSCs into astrocytes in vivo and in vitro. These impacts on the neurodevelopment of offspring were most pronounced in D-D group, followed by D-N group and N-D group. In conclusion, parental folic acid deficiency inhibits the neurobehavioral development of offspring, possibly by inhibiting the differentiation of NSCs into neurons.


Folic Acid Deficiency , Neural Stem Cells , Pregnancy , Female , Rats , Animals , Neural Stem Cells/physiology , Neurons/metabolism , Folic Acid/pharmacology , Folic Acid/metabolism , Cell Differentiation
4.
Nutrients ; 15(19)2023 Oct 01.
Article En | MEDLINE | ID: mdl-37836528

The deterioration of brain glucose metabolism predates the clinical onset of Alzheimer's disease (AD). Medium-chain triglycerides (MCTs) and docosahexaenoic acid (DHA) positively improve brain glucose metabolism and decrease the expression of AD-related proteins. However, the effects of the combined intervention are unclear. The present study explored the effects of the supplementation of MCTs combined with DHA in improving brain glucose metabolism and decreasing AD-related protein expression levels in APP/PS1 mice. The mice were assigned into four dietary treatment groups: the control group, MCTs group, DHA group, and MCTs + DHA group. The corresponding diet of the respective groups was fed to mice from the age of 3 to 11 months. The results showed that the supplementation of MCTs combined with DHA could increase serum octanoic acid (C8:0), decanoic acid (C10:0), DHA, and ß-hydroxybutyrate (ß-HB) levels; improve glucose metabolism; and reduce nerve cell apoptosis in the brain. Moreover, it also aided with decreasing the expression levels of amyloid beta protein (Aß), amyloid precursor protein (APP), ß-site APP cleaving enzyme-1 (BACE1), and presenilin-1 (PS1) in the brain. Furthermore, the supplementation of MCTs + DHA was significantly more beneficial than that of MCTs or DHA alone. In conclusion, the supplementation of MCTs combined with DHA could improve energy metabolism in the brain of APP/PS1 mice, thus decreasing nerve cell apoptosis and inhibiting the expression of Aß.


Alzheimer Disease , Amyloid beta-Peptides , Mice , Animals , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid Precursor Protein Secretases/metabolism , Docosahexaenoic Acids/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Mice, Transgenic , Aspartic Acid Endopeptidases/metabolism , Disease Models, Animal , Alzheimer Disease/drug therapy , Brain/metabolism , Dietary Supplements , Triglycerides/metabolism
5.
Nutrients ; 15(13)2023 Jun 22.
Article En | MEDLINE | ID: mdl-37447170

The effect of maternal folate status on the fetal central nervous system (CNS) is well recognized, while evidence is emerging that such an association also exists between fathers and offspring. The biological functions of telomeres and telomerase are also related to neural cell proliferation and apoptosis. The study aimed to investigate the effect of parental folate deficiency on the proliferation and apoptosis of neural stem cells (NSCs) in neonatal offspring and the role of telomeres in this effect. In this study, rats were divided into four groups: maternal folate-deficient and paternal folate-deficient diet (D-D) group; maternal folate-deficient and paternal folate-normal diet (D-N) group; maternal folate-normal and paternal folate-deficient diet (N-D) group; and the maternal folate-normal and paternal folate-normal diet (N-N) group. The offspring were sacrificed at postnatal day 0 (PND0), and NSCs were cultured from the hippocampus and striatum tissues of offspring for future assay. The results revealed that parental folate deficiency decreased folate levels, increased homocysteine (Hcy) levels of the offspring's brain tissue, inhibited proliferation, increased apoptosis, shortened telomere length, and aggravated telomere attrition of offspring NSCs in vivo and in vitro. In vitro experiments further showed that offspring NSCs telomerase activity was inhibited due to parental folate deficiency. In conclusion, parental folate deficiency inhibited the proliferation and increased apoptosis of offspring NSCs, maternal folate deficiency had more adverse effects than paternal, and the mechanisms may involve the telomere attrition of NSCs.


Folic Acid Deficiency , Neural Stem Cells , Telomerase , Animals , Rats , Telomere Shortening , Telomere , Folic Acid/pharmacology , Apoptosis , Cell Proliferation
6.
JAMA Netw Open ; 6(7): e2324031, 2023 07 03.
Article En | MEDLINE | ID: mdl-37462970

Importance: Apolipoprotein E polymorphism ε4 (APOE ε4) and methylenetetrahydrofolate reductase (MTHFR) TT genotype are genetic risk factors of mild cognitive impairment (MCI), but whether this risk can be changed by modifiable lifestyle factors is unknown. Objective: To explore whether unhealthy lifestyle (unhealthy dietary intake, current smoking, nonlimited alcohol consumption, and irregular physical activities) is associated with a higher risk of age-related MCI considering genetic risk. Design, Setting, and Participants: This population-based cohort study used data from Tianjin Elderly Nutrition and Cognition (TENC) study participants, recruited from March 1, 2018, through June 30, 2021, and followed up until November 30, 2022. Participants were Chinese adults aged 60 years or older who completed the neuropsychological assessments, general physical examinations, and a personal interview. Exposures: Healthy lifestyle was defined according to the Chinese Dietary Guidelines 2022, including healthy diet, regular physical activity, limited alcohol consumption, and no current smoking, categorized into healthy and unhealthy lifestyles according to weighted standardized lifestyle score. Genetic risk was defined by MTHFR TT genotype and APOE ε4, categorized into low and high genetic risk according to weighted standardized genetic risk score. Main Outcomes and Measures: The main outcome was newly diagnosed MCI as identified using a modified version of Petersen criteria. Hazard ratios (HRs) and 95% CIs were estimated using Cox proportional hazard regression models. Results: A total of 4665 participants were included (mean [SD] age, 67.9 [4.9] years; 2546 female [54.6%] and 2119 male [45.4%]); 653 participants with new-onset MCI (mean [SD] age, 68.4 [5.4] years; 267 female [40.9%] and 386 male [59.1%]) were identified after a median follow-up of 3.11 years (range, 0.82-4.61 years). Individuals with a low genetic risk and an unhealthy lifestyle (HR, 3.01; 95% CI, 2.38-3.79), a high genetic risk and a healthy lifestyle (HR, 2.65; 95% CI, 2.03-3.44), and a high genetic risk and an unhealthy lifestyle (HR, 3.58; 95% CI, 2.73-4.69) had a higher risk of MCI compared with participants with a low genetic risk and a healthy lifestyle. There was a synergistic interaction between lifestyle categories and genetic risk (ß = 3.58; 95% CI, 2.73-4.69). Conclusions and Relevance: In this cohort study of TENC participants, the findings show that unhealthy lifestyle and high genetic risk were significantly associated with a higher risk of MCI among Chinese older adults. Unhealthy lifestyle factors were associated with a higher risk of MCI regardless of genetic risk, and lifestyle and genetic risk had synergistic interactions. These findings could contribute to the development of dietary guidelines and the prevention of early-stage dementia.


Apolipoprotein E4 , Cognitive Dysfunction , Methylenetetrahydrofolate Reductase (NADPH2) , Aged , Female , Humans , Male , Apolipoprotein E4/genetics , Cognitive Dysfunction/epidemiology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/prevention & control , Cohort Studies , East Asian People , Life Style , Risk Factors , Middle Aged , Methylenetetrahydrofolate Reductase (NADPH2)/genetics
7.
Acad Radiol ; 30 Suppl 1: S92-S103, 2023 09.
Article En | MEDLINE | ID: mdl-37173236

RATIONALE AND OBJECTIVES: To compare the efficacy and complications of ultrasound-guided percutaneous radiofrequency ablation of hepatocellular carcinoma (HCC) in the hepatocaval confluence with those of HCC in the non-hepatocaval confluence and to explore the risk factors that lead to radiofrequency ablation failure and patient local tumor progression (LTP). MATERIALS AND METHODS: From January 2017 to January 2022, 86 patients with HCC in the hepatocaval confluence who had radiofrequency ablation were included. A 1:1 propensity-matched group of patients with HCC in the non-hepatocaval confluence with comparable clinical baseline traits, such as tumor diameter and tumor number, served as the control group. The two groups' complications, primary efficacy rate (PER), technical success rate (TSR), and prognosis were estimated. RESULTS: After PSM, no significant difference of TSR (91.7% vs 95.8%, p = 0.491) and PER (95.8% vs 97.2%, p = 1.000) and 1-, 3-, and 5-year LTP rate (12.5% vs 9.9%, 28.2% vs 27.7%, 40.8% vs 43.8%, p = 0.959) and 1-, 3-, and 5-year DFS rate (87.5% vs 87.5%, 62.3% vs 54.2%, 18.1% vs 22.6%, p = 0.437) and 1-, 3-, and 5-year OS rate (94.3% vs 95.7%, 72.7% vs 69.6%, 20.9% vs 33.6%, p = 0.904) was detected between the two groups. The tumor-to-IVC distance was an independent risk factor for radiofrequency ablation failure in HCC patients in the hepatocaval confluence (OR = 0.611, p = 0.022). Besides, tumor diameter was an independent risk factor for predicting LTP in patients with HCC in the hepatocaval confluence (HR = 2.209, p = 0.046). CONCLUSION: HCC in the hepatocaval confluence can be effectively treated with radiofrequency ablation. To maximize treatment efficacy, the tumor-to-IVC distance and tumor diameter should be assessed before the operation begins.


Carcinoma, Hepatocellular , Catheter Ablation , Liver Neoplasms , Radiofrequency Ablation , Humans , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/surgery , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/surgery , Liver Neoplasms/pathology , Propensity Score , Treatment Outcome , Retrospective Studies
8.
Front Pharmacol ; 14: 1183619, 2023.
Article En | MEDLINE | ID: mdl-37251319

Surgery remains the preferred treatment option for early-stage gallbladder cancer (GBC). According to the anatomical position of the primary tumor, accurate preoperative stage and strict control of surgical indications, appropriate surgical strategies are selected to achieve the optimal surgical effect. However, most patients have already been at the locally advanced stage or the tumor has metastasized at the initial diagnosis. The postoperative recurrence rate and 5-year survival rate remain unsatisfactory even after radical resection for gallbladder cancer. Hence, there is an urgent need for more treatment options, such as neoadjuvant therapy, postoperative adjuvant therapy and first-line and second-line treatments of local progression and metastasis, in the whole-course treatment management of gallbladder cancer patients. In recent years, the application of molecular targeted drugs and immunotherapy has brought greater hope and broader prospects for the treatment of gallbladder cancer, but their effects in improving the prognosis of patients still lack sufficient evidence-based medicine evidence, so many problems should be addressed by further research. Based on the latest progress in gallbladder cancer research, this review systematically analyzes the treatment trends of gallbladder cancer.

9.
J Nutr Biochem ; 117: 109328, 2023 07.
Article En | MEDLINE | ID: mdl-36958416

The brain has high energy demand making it sensitive to changes in energy fuel supply. Aging shrinks brain volume, decreases glucose uptake availability of the brain, and finally, causes cognitive dysfunction. Folic acid supplementation delayed cognitive decline and neurodegeneration. However, whether folic acid affects brain energy metabolism and structural changes is unclear. The study aimed to determine if long-term dietary folic acid supplementation could alleviate age-related cognitive decline by attenuating hippocampus atrophy and promoting brain glucose uptake in Sprague-Dawley (SD) rats. According to folic acid levels in diet, 3-months old male SD rats were randomly divided into four intervention groups for 22 months in equal numbers: folic acid-deficient diet (FA-D) group, folic acid-normal diet (FA-N) group, low folic acid-supplemented diet (FA-L) group, and high folic acid-supplemented diet (FA-H) group. The results showed that serum folate concentrations decreased and serum homocysteine (Hcy) concentrations increased with age, and dietary folic acid supplementation increased serum folate concentrations and decreased Hcy concentrations at 11, 18, and 22 months of intervention. Dietary folic acid supplementation attenuated aging-induced hippocampus atrophy, which was showed by higher fractional anisotropy and lower mean diffusivity in the hippocampus, increased brain 18F-Fluorodeoxyglucose (18F-FDG) uptake, then stimulated neuronal survival, and alleviated age-related cognitive decline in SD rats. In conclusion, long-term dietary folic acid supplementation alleviated age-related cognitive decline by attenuating hippocampus atrophy and promoting brain glucose uptake in SD rats.


Cognitive Dysfunction , Diet , Rats , Animals , Male , Rats, Sprague-Dawley , Folic Acid/metabolism , Dietary Supplements , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/metabolism , Aging , Hippocampus/metabolism , Glucose/metabolism
10.
Appl Physiol Nutr Metab ; 48(5): 393-402, 2023 May 01.
Article En | MEDLINE | ID: mdl-36809211

Folic acid (FA) could improve cognitive performance and attenuate brain cell injury in the aging brain; FA supplementation is also associated with inhibiting neural stem cell (NSC) apoptosis. However, its role in age-associated telomere attrition remains unclear. We hypothesized that FA supplementation attenuates age-associated apoptosis of NSCs in mice via alleviating telomere attrition in senescence-accelerated mouse prone 8 (SAMP8). In this study, 4-month-old male SAMP8 mice were assigned equal numbers to four different diet groups (n = 15). Fifteen age-matched senescence-accelerated mouse resistant 1 mice, fed with the FA-normal diet, were used as the standard aging control group. After FA treatment for 6 months, all mice were sacrificed. NSC apoptosis, proliferation, oxidative damage, and telomere length were evaluated by immunofluorescence and Q-fluorescent in situ hybridization. The results showed that FA supplementation inhibited age-associated NSC apoptosis and prevented telomere attrition in the cerebral cortex of SAMP8 mice. Importantly, this effect might be explained by the decreased levels of oxidative damage. In conclusion, we demonstrate it may be one of the mechanisms by which FA inhibits age-associated NSC apoptosis by alleviating telomere length shortening.


Folic Acid , Neural Stem Cells , Mice , Male , Animals , Folic Acid/pharmacology , In Situ Hybridization, Fluorescence , Aging , Apoptosis , Telomere
11.
Immun Ageing ; 20(1): 1, 2023 Jan 05.
Article En | MEDLINE | ID: mdl-36604719

BACKGROUND: Diet and chronic inflammation might play a major role in the pathogenesis of mild cognitive impairment (MCI). In addition, peripheral blood leukocyte telomere length (LTL) and mitochondrial DNA copy number (mtDNAcn) might mediate the relationship between inflammation and MCI risk. The purpose of the present study is to evaluate whether inflammatory potential of diet assessed by dietary inflammatory index (DII), chronic inflammation, peripheral blood LTL, and mtDNAcn were associated with the risk of MCI. RESULTS: A population-based cohort study was conducted with a total of 2944 participants. During a median follow-up of 2 years, 438 (14.90%) individuals were new-onset MCI. After adjustment, a higher score of DII (hazard ratio [HR]: 1.056, 95% CI: 1.005, 1.109), a higher log systemic immune inflammation index (SII) (HR: 1.333, 95% CI: 1.089, 1.633) and log system inflammation response index (SIRI) (HR: 1.487, 95% CI: 1.024, 2.161) predicted elevated risk of MCI. An increased mtDNAcn (HR: 0.843, 95% CI: 0.712, 0.997), but not LTL, predicted a decreased risk of MCI. Negative associations of log SII with LTL (ß:-0.359, 95% CI: -0.445, -0.273) and mtDNAcn (ß:-0.048, 95% CI: -0.090, -0.006) were found. Additionally, negative associations of log SIRI with LTL (ß: -0.035, 95% CI: -0.052, -0.017) and mtDNAcn (ß:-0.136, 95% CI: -0.216, -0.056) were also found. Path analysis suggested that SIRI, LTL, and mtDNAcn, in series, have mediation roles in the association between DII score and MCI risk. CONCLUSIONS: Higher DII, SII, and SIRI might predict a greater risk of MCI, while a longer LTL and an increased mtDNAcn were linked to a reduced risk of MCI among the older population. LTL and mtDNAcn could play mediation roles in the association between DII and MCI risk.

12.
Curr Alzheimer Res ; 2022 10 07.
Article En | MEDLINE | ID: mdl-36214304

BACKGROUND: Recent findings suggest that both dietary protein intake and hand grip strength (HGS) were associated with cognitive function, however, few studies have been devoted specifically to the mediation effect of HGS on the association of dietary protein with cognitive function. OBJECTIVES: To confirm the hypothesis that HGS mediated the association of dietary protein intake with cognitive function in the elderly, which was modified by triglyceride level and methylenetetrahydrofolate reductase (MTHFR) gene status. METHODS: This cross-sectional study included 3,268 participants. Dietary protein intake, HGS, and cognitive function were collected by food frequency questionnaires (FFQ), grip measurements and mini mental state examination (MMSE), respectively. In this mediation analysis, dietary protein intake was entered as independent variable, HGS was entered as mediator, and cognitive function was entered as dependent variable. RESULTS: HGS significantly mediated the associations of dietary protein (ß = 0.0013, 95% CI: 0.0007, 0.0022), animal protein (ß = 0.0024, 95% CI: 0.0012, 0.0037), and plant protein intake (ß = 0.0011, 95% CI: 0.0001, 0.0023) with cognitive function in total participants, with the mediated proportion of 16.19%, 12.45% and 20.57%, respectively. Furthermore, significant mediation effects of HGS on the associations of dietary protein, animal protein, and plant protein intake with MMSE score were found in the elderly without hypertriglyceridemia or in MTHFR C677T CC/CT carriers. CONCLUSION: This study suggested that HGS mediated the association of dietary protein intake with cognitive function, and this mediation effect was modified by triglyceride level and MTHFR C677T gene status.

13.
J Alzheimers Dis ; 90(1): 389-404, 2022.
Article En | MEDLINE | ID: mdl-36120779

BACKGROUND: The high cost, limited availability, and perceived invasiveness of amyloid PET and cerebrospinal fluid biomarkers limit their use for the diagnosis of Alzheimer's disease. OBJECTIVE: The present study aimed to assess the associations of mild cognitive impairment (MCI) with circulating amyloid-ß (Aß), methionine circulating metabolites (MCMs), and their downstream products, and to develop a nomogram based on these easily accessible blood indexes for the individualized prediction of MCI risk in older adults. METHODS: In this nested case-control study, we recruited 74 MCI patients and, for each, 3 matched controls (n = 222) within the context of the Tianjin Elderly Nutrition and Cognition (TENC) cohort, a population-based prospective study in China. Concentrations of Aß, MCMs, and their circulating downstream factors (i.e., leukocyte telomere length and inflammatory cytokines) were evaluated in fasting blood sample using standard procedures. We constructed a nomogram for MCI harnessed multivariable logistic models incorporating variables selected in the Lasso regression. RESULTS: Among the many biomarkers examined, the final prediction nomogram retained only 3 factors: Aß42/Aß40 ratio, Hcy, and SAM/SAH ratio. The model achieved favorable discrimination, with a C-statistic of 0.75 (95% confidence interval 0.69-0.81) in internal validation after adjustment of optimism. The calibration accuracy was satisfactory; the Brier score of the model was 0.161 in internal validation after adjustment of optimism. CONCLUSION: his study presents an individualized prediction nomogram incorporating only three blood biomarkers (i.e., Aß42/Aß40 ratio, Hcy, and SAM/SAH ratio), which can be conveniently utilized to facilitate early identification and the development of high-risk prevention strategies for MCI in older adults.


Alzheimer Disease , Cognitive Dysfunction , Humans , Aged , Case-Control Studies , Prospective Studies , Methionine , Amyloid beta-Peptides/cerebrospinal fluid , Alzheimer Disease/diagnosis , Alzheimer Disease/epidemiology , Biomarkers/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid
14.
Int J Mol Sci ; 23(13)2022 Jun 22.
Article En | MEDLINE | ID: mdl-35805953

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.


Folic Acid Deficiency , Animals , Cognition , Dietary Supplements , Female , Folic Acid/metabolism , Folic Acid Deficiency/complications , Folic Acid Deficiency/metabolism , Rats , Telomere/metabolism , Uracil
15.
Eur J Neurol ; 29(10): 2913-2924, 2022 10.
Article En | MEDLINE | ID: mdl-35735052

BACKGROUND: The longitudinal association between serum folate concentrations and the risk of cognitive impairment remains unclear in populations with low folate levels. We examined the association between serum folate concentrations and mild cognitive impairment (MCI) in older adults in China, where mandatory fortification of foods with folic acid has not been implemented. We further explored if homocysteine (Hcy) and leukocyte telomere length (LTL) mediate the association between serum folate and MCI. METHODS: We performed a longitudinal analysis of 3974 participants aged ≥60 years from the Tianjin Elderly Nutrition and Cognition (TENC) cohort study. The associations between serum folate level and the risk of cognitive impairment overall and stratified by apolipoprotein E (APOE) ε4 genotypes were evaluated using multivariable Cox proportional hazards models. The mediating effects of Hcy and LTL on the folate-MCI association were explored via a path analysis approach. RESULTS: Within a 3-year follow-up, we documented 560 incident MCI cases. After multivariable adjustment, higher serum folate concentrations were associated with lower incidence of MCI, with hazard ratios (95% confidence interval) across quartiles of folate (from lowest to highest concentrations) of 1.00 (reference), 0.66 (0.52, 0.83), 0.57 (0.45, 0.73), 0.66 (0.52, 0.84), respectively (p for trend <0.001). In mediation analyses, the status of serum folate deficiency and MCI were correlated via two intermediary pathways, Hcy and Hcy-telomere (p < 0.05). CONCLUSIONS: Lower folate concentrations, independently of APOE genotype, were associated with increased risk of MCI among elderly Chinese people, a population with relatively low folate intake. Our data were compatible with the mediation hypothesis that the association between folate status and MCI was mediated by Hcy and LTL.


Cognitive Dysfunction , Folic Acid , Aged , Apolipoprotein E4 , China/epidemiology , Cognitive Dysfunction/epidemiology , Cohort Studies , Homocysteine , Humans , Prospective Studies , Vitamin B 12
16.
Cereb Cortex ; 32(2): 286-297, 2022 01 10.
Article En | MEDLINE | ID: mdl-34223882

Folic acid (FA) has been reported to inhibit astrocyte apoptosis and improve aging-induced disorders; however, its role in telomere attrition remains unclear. In present study, 4-month-old senescence-accelerated mouse prone 8 (SAMP8) mice were assigned to four treatment groups for the in vivo experiment: FA-deficient diet (FA-D) group, FA-normal diet (FA-N) group, low FA-supplemented diet (FA-L) group, and high FA-supplemented diet (FA-H) group. These mice were euthanized when 10 months old. There was also a young SAMP8 (4 months old) control group (Con-Y) fed with FA-normal diet. In in vitro study, primary cultures of astrocytes from hippocampus and cerebral cortex were incubated for five generations with various concentrations of FA (0-40 µM) and were assigned to five groups: FA 0 µM (generation 5), FA 10 µM (generation 5), FA 20 µM (generation 5), FA 40 µM (generation 5), and FA 10 µM (generation 1). The results showed that FA supplementation inhibited aging-induced astrocytosis, astrocyte apoptosis, neurodegeneration, and prevented telomere attrition in hippocampus and cortex of SAMP8 mice. FA supplementation also decreased apoptosis and telomere attrition, and increased telomerase activity, in primary cultures of astrocytes. These results showed that it may be one of the mechanisms that FA inhibiting aging-induced apoptosis of astrocyte by alleviating telomere attrition.


Astrocytes , Folic Acid , Aging , Animals , Apoptosis , Folic Acid/pharmacology , Mice , Telomere
17.
Mol Neurobiol ; 59(1): 590-602, 2022 Jan.
Article En | MEDLINE | ID: mdl-34741234

DNA oxidative damage can cause telomere attrition or dysfunction that triggers cell senescence and apoptosis. The hypothesis of this study is that folic acid decreases apoptosis in neural stem cells (NSCs) by preventing oxidative stress-induced telomere attrition. Primary cultures of NSCs were incubated for 9 days with various concentrations of folic acid (0-40 µM) and then incubated for 24 h with a combination of folic acid and an oxidant (100-µM hydrogen peroxide, H2O2), antioxidant (10-mM N-acetyl-L-cysteine, NAC), or vehicle. Intracellular folate concentration, apoptosis rate, cell proliferative capacity, telomere length, telomeric DNA oxidative damage, telomerase activity, intracellular reactive oxygen species (ROS) levels, cellular oxidative damage, and intracellular antioxidant enzyme activities were determined. The results showed that folic acid deficiency in NSCs decreased intracellular folate concentration, cell proliferation, telomere length, and telomerase activity but increased apoptosis, telomeric DNA oxidative damage, and intracellular ROS levels. In contrast, folic acid supplementation dose-dependently increased intracellular folate concentration, cell proliferative capacity, telomere length, and telomerase activity but decreased apoptosis, telomeric DNA oxidative damage, and intracellular ROS levels. Exposure to H2O2 aggravated telomere attrition and oxidative damage, whereas NAC alleviated the latter. High doses of folic acid prevented telomere attrition and telomeric DNA oxidative damage by H2O2. In conclusion, inhibition of telomeric DNA oxidative damage and telomere attrition in NSCs may be potential mechanisms of inhibiting NSC apoptosis by folic acid.


Antioxidants/pharmacology , Apoptosis/drug effects , Folic Acid/pharmacology , Neural Stem Cells/drug effects , Oxidative Stress/drug effects , Telomere/drug effects , Animals , Cell Proliferation/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hydrogen Peroxide/pharmacology , Neural Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Telomere/metabolism
18.
J Nutr Biochem ; 97: 108796, 2021 11.
Article En | MEDLINE | ID: mdl-34102282

Disturbed deoxythymidine triphosphate biosynthesis due to the inhibition of thymidylate synthase (TS) can lead to uracil accumulation in DNA, eventually, lead to neurocytes apoptosis and cognitive decline. Folic acid supplementation delayed cognitive decline and neurodegeneration in senescence-accelerated mouse prone 8 (SAMP8). Whether folic acid, one of nutrition factor, the effect on the expression of TS is unknown. The study aimed to determine if folic acid supplementation could alleviate age-related cognitive decline and apoptosis of neurocytes by increasing TS expression in SAMP8 mice. According to folic acid concentration in diet, four-month-old male SAMP8 mice were randomly divided into three different diet groups by baseline body weight in equal numbers. Moreover, to evaluate the role of TS, a TS inhibitor was injected intraperitoneal. Cognitive test, apoptosis rates of neurocytes, expression of TS, relative uracil level in telomere, and telomere length in brain tissue were detected. The results showed that folic acid supplementation decreased deoxyuridine monophosphate accumulation, uracil misincorporation in telomere, alleviated telomere length shorting, increased expression of TS, then decreased apoptosis rates of neurocytes, and alleviated cognitive performance in SAMP8 mice. Moreover, at the same concentration of folic acid, TS inhibitor raltitrexed increased deoxyuridine monophosphate accumulation, uracil misincorporation in telomere, and exacerbated telomere length shorting, decreased expression of TS, then increased apoptosis rates of neurocytes, and decreased cognitive performance in SAMP8 mice. In conclusion, folic acid supplementation alleviated age-related cognitive decline and inhibited apoptosis of neurocytes by increasing TS expression in SAMP8 mice.


Aging , Brain/metabolism , Cognitive Dysfunction/diet therapy , Dietary Supplements , Folic Acid/administration & dosage , Neurons/physiology , Thymine Nucleotides/biosynthesis , Animals , Apoptosis , Folic Acid/blood , Folic Acid/metabolism , Male , Memory , Mice , Morris Water Maze Test , Quinazolines/pharmacology , Telomere Shortening , Thiophenes/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Uracil/metabolism
19.
Int J Mol Sci ; 21(1)2019 Dec 20.
Article En | MEDLINE | ID: mdl-31861819

Astrocytes are the most widely distributed cells in the brain, and astrocyte apoptosis may play an important role in the pathogenesis of neurodegenerative diseases. Folate is required for the normal development of the nervous system, but its effect on astrocyte apoptosis is unclear. In this study, we hypothesized that folic acid (the therapeutic form of folate) decreases astrocyte apoptosis by preventing oxidative stress-induced telomere attrition. Primary cultures of astrocytes were incubated for 12 days with various concentrations of folic acid (0-40 µmol/L), then cell proliferation, apoptosis, intracellular folate concentration, intracellular homocysteine (Hcy) concentration, intracellular reactive oxygen species (ROS) levels, telomeric DNA oxidative damage, and telomere length were determined. The results showed that folic acid deficiency decreased intracellular folate, cell proliferation, and telomere length, whereas it increased Hcy concentration, ROS levels, telomeric DNA oxidative damage, and apoptosis. In contrast, folic acid dose-dependently increased intracellular folate, cell proliferation, and telomere length but it decreased Hcy concentration, ROS levels, telomeric DNA oxidative damage, and apoptosis. In conclusion, folic acid inhibited apoptosis in astrocytes. The underlying mechanism for this protective effect may be that folic acid decreased oxidative stress and thereby prevented telomeric DNA oxidative damage and telomere attrition.


Apoptosis/drug effects , Astrocytes/drug effects , Folic Acid/pharmacology , Oxidative Stress/drug effects , Vitamin B Complex/pharmacology , Animals , Antioxidants/pharmacology , Astrocytes/cytology , Astrocytes/metabolism , Cells, Cultured , Rats, Sprague-Dawley , Telomere/drug effects , Telomere/metabolism
20.
Aging (Albany NY) ; 11(22): 10356-10373, 2019 11 22.
Article En | MEDLINE | ID: mdl-31757935

The occurrence of telomere attrition in brain may cause senescence and death of neurons, leading to cognitive decline. Folic acid (FA) has been reported to improve cognitive performance in mild cognitive impairment; however, its association with telomere remains unclear. The study aimed to investigate if alleviation of telomere attrition by FA supplementation could act as a potential mechanism to delay age-related cognitive decline in senescence-accelerated mouse prone 8 (SAMP8). Aged SAMP8 mice were assigned to four treatment groups: FAdeficient diet (FA-D) group, FA-normal diet (FA-N) group, low FA-supplemented diet (FA-L) group and high FAsupplemented diet (FA-H) group. There was also an age-matched senescence-accelerated mouse resistant 1 (SAMR1) control group (Con-R), and a young SAMP8 control group (Con-Y). The results demonstrated that FA supplementation delayed age-related cognitive decline and neurodegeneration in SAMP8 mice. Importantly, this effect could be attributed to the alleviated telomere attrition, which might be interpreted by the decreased levels of reactive oxygen species. Additionally, improved telomere integrity stimulated mitochondrial function via telomere-p53-mithondria pathway, consequently delayed neuronal degeneration. In conclusion, we demonstrate that FA supplementation delays age-related neurodegeneration and cognitive decline in SAMP8 mice, in which alleviated telomere attrition could serve as one influential factor in the process.


Aging/drug effects , Cognitive Dysfunction , Dietary Supplements , Folic Acid/pharmacology , Telomere Shortening/drug effects , Animals , Brain/drug effects , Brain/pathology , Male , Mice , Nerve Degeneration/pathology
...