-
Sodium fluoride (NaF) and sodium arsenite (NaAsO2) were purchased from Nanjing Shengqinghe Chemical Co. Ltd. and Shanghai Kanglang Biotechnology Co. Ltd., respectively.
-
Sixty SD rats (6–7 weeks old, 40 female rats weighing 160 ± 10 g, and 20 male rats weighing 200 ± 10 g) were purchased from Beijing SiPeiFu Biotechnology Co. The rats were housed at SPF level in a controlled environment (temperature, 24 ± 1 °C; relative humidity, 50%) with unrestricted access to food and water. All experiments were performed in accordance with the guidelines of the Animal Experimentation Center and approved by the Ethics Committee of Shanxi Medical University.
-
After one week of adaptation in separate cages for male and female rats, the rats were randomly divided into control (Con, drinking water only), arsenic (As, 70 mg/L NaAsO2), fluoride (F, 100 mg/L NaF), and arsenic-fluoride group (AsF, 70 mg/L NaAsO2 and 100 mg/L NaF). Rats were exposed to free water drinking according to the grouping described above, from first day of mating (female: male = 2:1) until the end of the gestation period. Subsequently, offspring were exposed to arsenic and/or fluoride through parental lactation during the 21-day lactation period. Afterwards, the weaned rats were continued to be exposed to the chemicals in similar manner until postnatal day (PND) 90.
-
Feces and urine were collected from six rats per group on the day of PND 90 before euthanasia. The urine was cryogenically centrifuged, and the supernatant was removed and stored at ‒80 °C. Urinary arsenic and fluoride levels were measured as described in later context. Meanwhile, the feces were collected under the principle of asepsis, rapidly frozen in a liquid nitrogen tank for 30 s, and stored at a ‒80 °C. The fecal samples were used for 16S rRNA sequencing to detect gut microbiomes and LC-MC to detect gut nontargeted metabolomics. Finally, the hippocampal region of the brain tissue was collected after intraperitoneal injection of 20% Uratan in anesthetized littermates, and pathological changes in the CA1 region of the hippocampus were observed by hematoxylin and eosin (H&E) staining.
-
The Morris water maze test, consisting of a spatial navigation training and a spatial exploration task, was used to evaluate their spatial learning and memory abilities. The water maze testing tank was 150 cm in diameter and 50 cm in depth. The circular water maze test chamber divided the pool into equal quadrants I, II, III, and IV. Black ink was added to the water before the experiment to prevent the camera from being able to track the white rats. The white rats were transferred to darkroom for at least 30 min before the experiment to adapt to the darkroom environment to reduce stress. The Morris water maze test was divided into two phases: spatial navigation training (days 1–5) and spatial exploration task (day 6). In the first phase, the rats were placed in the water from the appropriate position facing the wall of the pool each time and allowed to swim for up to 120 s. The escape latency was recorded for rats that found the platform and stayed on it for at least 5 s; otherwise, it was recorded for 120 s, and the rats were finally guided to find the platform and stay there for 5 s. The experiment was performed four times per day per rat for five consecutive days. In the second phase, the rats’ metrics (time spent in the target quadrant, swimming distance in the target quadrant, and number of crossing the platform) were recorded using a behavioral video tracking system (Hayward) after removal of the platform for subsequent analysis. Consistency of the experimental conditions was maintained throughout the experiments.
-
Following preconditioning of cardiac perfusion, hippocampal CA1 regions of rat brain tissues were collected and fixed in 4% paraformaldehyde for 24 h at room temperature. After continuous ethanol dehydration (70%, 80%, 90%, and 100% alcohol), it was embedded in paraffin and sectioned, followed by rehydration, H&E staining, and sealing. Lastly, the sections were observed under an inverted microscope and photographed for neuronal and nuclear analysis.
-
The urinary arsenic and fluorine levels were measured to assess the in vivo metabolism of arsenic and fluorine. First, urine was mixed with total ionic strength adjustment buffer (TISAB) in equal volumes, and the urinary fluorine content was measured using a fluoride ion meter (Shanghai Xianfan Instruments Co., Ltd.) with reference to the fluorine standard curve. While urine arsenic content was measured by digestion at low- temperature after centrifugation and dilution. Then, thiourea-vitamin C solution was added, and the volume was fixed using 5% dilute hydrochloric acid. Finally, urinary arsenic content was determined with an ASF-8530 atomic fluorescence photometer (Beijing Haiguang Instrument Company) using the arsenic standard curve as a reference.
-
Total genomic DNA extraction, PCR amplification, and library construction for fecal samples were performed by Shanghai Obio Technology Co. DNA was extracted using the MagPure Soil DNA LQ Kit. Next, primer 343F (5’-TACGGRAGGCAGCAG-3’) and 798R (5’-AGGGTATCTAATCCT-3’), targeting the high variability region V3V4 were selected for PCR amplification.
The original image data file obtained by high-throughput sequencing was transformed into raw data using base-calling analysis. Raw data processing flow: quality filtering, sequence splicing, base sequence removal, chimera removal to obtain valid tags for later operational taxonomic units (OTU) delineation. The average length of valid tags ranged from 408.87 to 420.07 bp, and the number of OTUs in each sample ranged from 1,838 to 2,514. Subsequently, the OTU classification of the quality sequence valid tags was performed according to 97% similarity using Vsearch2.4.2 software (Oslo, Norway) to obtain the representative sequences. Following OTU cluster analysis, the species were annotated and the relative abundance was calculated at the phylum, class, order, family, and genus levels.
The alpha and beta diversities were analyzed reflecting microbial community differences within and between samples. Principal coordinate analysis (PCoA) using the unweighted UniFrac distance metric can simplify complex multidimensional data into intuitive two-dimensional data to identify similarities or differences between communities. Finally, linear discriminant analysis effect size (LEfSe) was used to reveal the composition of different species in two or more groups of biomes. The results of LEfSe analysis are presented as differential species contribution to the magnitude of differences analysis and differential species annotation analysis, respectively.
-
Fecal metabolomics analysis was performed using a QE high-resolution mass spectrometer (Thermo Fisher World Technology Corporation) and Nexera ultra-performance liquid chromatography (UPLC) (Shimadzu Corporation). The gut metabolites were extracted as follows: First, 20 μL of the internal standard, 600 μL of methanol, and water were added to 60 mg of the sample. After grinding, sonication (10 min), resting (‒20 °C, 30 min), and low-temperature centrifugation (4 °C), 300 μL of supernatant was obtained and evaporated dry. The dried extract was redissolved in 400 μL of methanol/water (v:v = 1:4), vortexed for 30 s, sonicated (2 min), and centrifuged. Finally, 50 μL of the supernatant was aspirated for onboard LC-MS analysis.
Metabolites were quantified with UPLC. Chromatographic separation was carried out on a column (ACQUITY UPLC HSST3, 100 mm × 2.1 mm, 1.8 um) at 45 °C. The mobile phase consisted of water and acetonitrile and separation was achieved using a gradient (Supplementary Table S1, available in www.besjournal.com). The flow rate was 0.35 mL/min, and the injection volume was 2 μL. The mass range was m/z 125 to 1,000. The resolution was set at 70,000 for full MS scans and 17,500 for HCD MS/MS scans. The mass spectrometer operated as follows: spray voltage, 3,500 V (+) and 3,000 V (−); capillary temperature, 350 °C.
Table S1. Elution gradient of mobile phase
Time A% B% 0.01 95 5 2 95 5 4 70 30 8 50 50 10 20 80 14 0 100 15 0 100 15.1 95 5 16 95 5 Note. A-water (0.1% formic acid), B-acetonitrile (0.1% formic acid). Raw data were collected using UNIFI software (version 1.8.1) and analyzed qualitatively using the metabolomics processing software Progenesis QI (version 2.3) (Nonlinear Dynamics, Newcastle, UK). The positive and negative ion data (30-point scale) were combined into a data matrix table for subsequent analysis. Data analysis included univariate (Student’s t-test and fold-change analysis) and multivariate analyses. Multivariate statistical analyses included principal component analysis (PCA), supervised partial least squares analysis (PLS-DA), and orthogonal partial least-squares analysis (OPLS-DA). In this study, Variable important in projection (VIP) > 1 and P < 0.05 were adopted to screen for differential metabolites. Finally, screening of differential metabolites was enriched to a complete set of metabolic pathways using KEGG enrichment analysis.
-
Data analysis was conducted using SPSS24.0 software (IBM, USA). All values are presented as the mean ± standard error of the mean (SEM). Statistical analysis of multiple comparisons was performed by analysis of variance (ANOVA) or Wilcoxon’s test. After ANOVA, the least significant difference (LSD) test was used between the two groups. Escape latency was analyzed using repeated measures ANOVA. Correlations between the microbiome, metabolome, and learning memory metrics were analyzed using the Spearman correlation test. The interaction between As and F was analyzed by single variable statistical method of general linear model. The determination of arsenic and fluorine contents was calculated from the regression equation of the standard curve. Statistical significance was set at P < 0.05.
doi: 10.3967/bes2023.028
Mechanism of Learning and Memory Impairment in Rats Exposed to Arsenic and/or Fluoride Based on Microbiome and Metabolome
-
Abstract:
Objective Arsenic (As) and fluoride (F) are two of the most common elements contaminating groundwater resources. A growing number of studies have found that As and F can cause neurotoxicity in infants and children, leading to cognitive, learning, and memory impairments. However, early biomarkers of learning and memory impairment induced by As and/or F remain unclear. In the present study, the mechanisms by which As and/or F cause learning memory impairment are explored at the multi-omics level (microbiome and metabolome). Methods We stablished an SD rats model exposed to arsenic and/or fluoride from intrauterine to adult period. Results Arsenic and/fluoride exposed groups showed reduced neurobehavioral performance and lesions in the hippocampal CA1 region. 16S rRNA gene sequencing revealed that As and/or F exposure significantly altered the composition and diversity of the gut microbiome, featuring the Lachnospiraceae_NK4A136_group, Ruminococcus_1, Prevotellaceae_NK3B31_group, [Eubacterium]_xylanophilum_group. Metabolome analysis showed that As and/or F-induced learning and memory impairment may be related to tryptophan, lipoic acid, glutamate, gamma-aminobutyric acidergic (GABAergic) synapse, and arachidonic acid (AA) metabolism. The gut microbiota, metabolites, and learning memory indicators were significantly correlated. Conclusion Learning memory impairment triggered by As and/or F exposure may be mediated by different gut microbes and their associated metabolites. -
Key words:
- Arsenic /
- Fluoride /
- Learning and memory impairment /
- Microbiome /
- Metabolome
注释:1) ACKNOWLEDGEMENTS: -
Figure 1. Internal exposure and interaction of As and/or F. Urinary arsenic content (A). Urinary fluoride content (B). Interaction of urinary arsenic (C). Interaction of urinary fluoride (D). The values are expressed as the mean ± SEM; n = 6; aP < 0.05 vs. Con group; bP < 0.05 vs. As group; cP < 0.05 vs. F group.
Figure 2. Effects of As and/or F exposure on spatial learning and memory and pathological changes. Escape latency (A). Number of crossing platform (B). Swimming distance in target quadrant (C). Time spent in target quadrant (D). Interaction of time spent in target quadrant (E). Histopathological changes of hippocampal CA1 area after H&E staining (scale bar, 50 µm) (F). Histopathological changes of hippocampal CA1 area after HE staining (scale bar, 20 µm) (G). The red arrow markers indicate nerve cells with pathological changes. The values are expressed as the mean ± SEM; n = 6; aP < 0.05 vs. Con group; bP < 0.05 vs. As group; cP < 0.05 vs. F group.
Figure 3. As and/or F exposure alters microbiome community structure and diversity. Histogram of relative abundance of the top 15 bacterial phyla (A). Histogram of the relative abundance of the top 15 bacterial genera (B). Community structure Krona plots were used to visualize the annotation results performed on the species, with the size of the sectors representing the relative proportions of the different OTU annotation results (C). Alpha diversity of bacterial communities (D). Microbiome patterns were distinguished between control and exposed groups by unsupervised principal coordinate analysis (PCoA) based on unweighted Unifrac distance (E). *P < 0.05 vs. Con group
Figure 4. Differential microbial species identified by linear discriminant analysis effect size (LEfSe) analysis. Heatmap of the relative abundances carried out by differential microbial species between groups at the genus level (Use the scale parameter to improve the comparability of data) (A). Example maps of differential species annotated branches constructed based on LEfSe analysis between groups (B). Differential microbial species meeting the linear discriminant analysis (LDA) score significance threshold > 3.6 (C). Red indicates high relative abundance of species; blue indicates low relative abundance of species.
Figure 5. LC/MS untargeted metabolome analysis. PLS-DA diagram based on all groups (A). Heat map of differential metabolite expression abundance based on all groups (Use the scale parameter to improve the comparability of data) (B). Path bubble diagram of based on group As and Con (C), F and Con (D), and AsF and Con (E), P < 0.05. The abscissa is the enrichment factor (number of significantly different metabolites/total metabolites in the pathway). Red indicates high metabolite expression abundance;blue indicates low metabolite expression abundance. The ordinate is the name of metabolic pathway. The color scale (red to green) indicates a decreasing P-value. The larger the point, the more metabolites enriched on the metabolite.
Figure 6. Heat map of correlation matrix for differential gut microbiome and differential metabolites. Tryptophan, lipoic acid, GABA, and AA pathways based on group As and Con (A). Tryptophan and lipoic acid pathways based on group F and Con (B). GABA pathway based on combined AsF and Con (C). Moderate correlations (0.5 ≤ |r| < 0.7) are highlighted as yellow-outline squares; strong correlations (|r| ≥ 0.7) are highlighted as green-outline squares. *, **, and *** P < 0.05, 0.01, and 0.001, respectively, versus the Con group.
Figure 7. Correlation between microbiome composition at genus level and Morris water maze behavioral indicators. Heat map of correlation matrix generated based on all groups (A). Heat map of correlation matrix generated based on control group and AsF combination exposure group (B). Moderate correlations (0.5 ≤ |r| < 0.7) are highlighted as yellow-outline squares; strong correlations (|r| ≥ 0.7) are highlighted as green-outline squares. *, **, and *** P < 0.05, 0.01, and 0.001, respectively, versus the Con group.
Figure 8. Correlations between metabolomic alterations in the KEGG pathway and Morris water maze behavioral indicators. Heat map of correlation matrix generated based on all groups (A). Heat map of correlation matrix generated based on control group and AsF combination exposure group (B). Moderate correlations (0.5 ≤ |r| < 0.7) are highlighted as yellow-outline squares; strong correlations (|r| ≥ 0.7) are highlighted as green-outline squares. *, **, and *** P < 0.05, 0.01, and 0.001, respectively, versus the C group.
S5. Correlation between metabolomic alterations and behavioral indicators. Heat map of correlation matrix generated based on control group and As exposure group (A). Heat map of correlation matrix generated based on control group and F exposure group (B). Moderate correlations (0.5 ≤ |r| < 0.7) are highlighted as yellow-outline squares; strong correlations (|r| ≥ 0.7) are highlighted as green-outline squares. *, **, and *** P-values < 0.05, 0.01, and 0.001, respectively, versus the C group.
S1. Elution gradient of mobile phase
Time A% B% 0.01 95 5 2 95 5 4 70 30 8 50 50 10 20 80 14 0 100 15 0 100 15.1 95 5 16 95 5 Note. A-water (0.1% formic acid), B-acetonitrile (0.1% formic acid). -
[1] WHO. Guidelines for drinking-water quality. 4th ed. World Health Organization. 2017. [2] Liang CM, Wu XY, Huang K, et al. Domain- and sex-specific effects of prenatal exposure to low levels of arsenic on children's development at 6 months of age: findings from the Ma'anshan birth cohort study in China. Environ Int, 2020; 135, 105112. doi: 10.1016/j.envint.2019.105112 [3] Tyler CR, Allan AM. The effects of arsenic exposure on neurological and cognitive dysfunction in human and rodent studies: a review. Curr Environ Health Rep, 2014; 1, 132−47. doi: 10.1007/s40572-014-0012-1 [4] Green R, Lanphear B, Hornung R, et al. Association between maternal fluoride exposure during pregnancy and IQ scores in offspring in Canada. JAMA Pediatr, 2019; 173, 940−8. doi: 10.1001/jamapediatrics.2019.1729 [5] Qiu YL, Chen XS, Yan XY, et al. Gut microbiota perturbations and neurodevelopmental impacts in offspring rats concurrently exposure to inorganic arsenic and fluoride. Environ Int, 2020; 140, 105763. doi: 10.1016/j.envint.2020.105763 [6] Li XJ, Brejnrod AD, Ernst M, et al. Heavy metal exposure causes changes in the metabolic health-associated gut microbiome and metabolites. Environ Int, 2019; 126, 454−67. doi: 10.1016/j.envint.2019.02.048 [7] Leclercq S, Mian FM, Stanisz AM, et al. Low-dose penicillin in early life induces long-term changes in murine gut microbiota, brain cytokines and behavior. Nat Commun, 2017; 8, 15062. doi: 10.1038/ncomms15062 [8] Cheng D, Li H, Zhou JP, et al. Chlorogenic acid relieves lead-induced cognitive impairments and hepato-renal damage via regulating the dysbiosis of the gut microbiota in mice. Food Funct, 2019; 10, 681−90. doi: 10.1039/C8FO01755G [9] Liu J, Wang HW, Lin L, et al. Intestinal barrier damage involved in intestinal microflora changes in fluoride-induced mice. Chemosphere, 2019; 234, 409−18. doi: 10.1016/j.chemosphere.2019.06.080 [10] Sherwin E, Bordenstein SR, Quinn JL, et al. Microbiota and the social brain. Science, 2019; 366, eaar2016. doi: 10.1126/science.aar2016 [11] Gonzales J, Marchix J, Aymeric L, et al. Fecal supernatant from adult with autism spectrum disorder alters digestive functions, intestinal epithelial barrier, and enteric nervous system. Microorganisms, 2021; 9, 1723. doi: 10.3390/microorganisms9081723 [12] Altimiras F, Garcia JA, Palacios-García I, et al. Altered gut microbiota in a fragile X syndrome mouse model. Front Neurosci, 2021; 15, 653120. doi: 10.3389/fnins.2021.653120 [13] Shahrizaila N, Lehmann HC, Kuwabara S. Guillain-Barré syndrome. Lancet, 2021; 397, 1214−28. doi: 10.1016/S0140-6736(21)00517-1 [14] Kesika P, Suganthy N, Sivamaruthi BS, et al. Role of gut-brain axis, gut microbial composition, and probiotic intervention in Alzheimer's disease. Life Sci, 2021; 264, 118627. doi: 10.1016/j.lfs.2020.118627 [15] Lu K, Abo RP, Schlieper KA, et al. Arsenic exposure perturbs the gut microbiome and its metabolic profile in mice: an integrated metagenomics and metabolomics analysis. Environ Health Perspect, 2014; 122, 284−91. doi: 10.1289/ehp.1307429 [16] Jia CN, Wei YP, Lan Y, et al. Comprehensive analysis of the metabolomic characteristics on the health lesions induced by chronic arsenic exposure: a metabolomics study. Int J Hyg Environ Health, 2019; 222, 434−45. doi: 10.1016/j.ijheh.2018.12.010 [17] Yue BJ, Zhang XH, Li WP, et al. Fluoride exposure altered metabolomic profile in rat serum. Chemosphere, 2020; 258, 127387. doi: 10.1016/j.chemosphere.2020.127387 [18] Sharon G, Garg N, Debelius J, et al. Specialized metabolites from the microbiome in health and disease. Cell Metab, 2014; 20, 719−30. doi: 10.1016/j.cmet.2014.10.016 [19] Pershing ML, Bortz DM, Pocivavsek A, et al. Elevated levels of kynurenic acid during gestation produce neurochemical, morphological, and cognitive deficits in adulthood: implications for schizophrenia. Neuropharmacology, 2015; 90, 33−41. doi: 10.1016/j.neuropharm.2014.10.017 [20] Zhang YH, Wang DW, Xu SF, et al. α-Lipoic acid improves abnormal behavior by mitigation of oxidative stress, inflammation, ferroptosis, and tauopathy in P301S Tau transgenic mice. Redox Biol, 2018; 14, 535−48. doi: 10.1016/j.redox.2017.11.001 [21] Wang DF, Sun XY, Yan JJ, et al. Alterations of eicosanoids and related mediators in patients with schizophrenia. J Psychiatr Res, 2018; 102, 168−78. doi: 10.1016/j.jpsychires.2018.04.002 [22] Zhang YJ, Jiang XJ, Zhang J, et al. Heterozygous disruption of beclin 1 mitigates arsenite-induced neurobehavioral deficits via reshaping gut microbiota-brain axis. J Hazard Mater, 2020; 398, 122748. doi: 10.1016/j.jhazmat.2020.122748 [23] Xin JG, Wang HS, Sun N, et al. Probiotic alleviate fluoride-induced memory impairment by reconstructing gut microbiota in mice. Ecotoxicol Environ Saf, 2021; 215, 112108. doi: 10.1016/j.ecoenv.2021.112108 [24] Idowu OS, Duckworth RM, Valentine RA, et al. Biomarkers for the assessment of exposure to fluoride in adults. Caries Res, 2021; 55, 292−300. doi: 10.1159/000516091 [25] Hughes MF. Biomarkers of exposure: a case study with inorganic arsenic. Environ Health Perspect, 2006; 114, 1790−6. doi: 10.1289/ehp.9058 [26] Zhu YP, Xi SH, Li MY, et al. Fluoride and arsenic exposure affects spatial memory and activates the ERK/CREB signaling pathway in offspring rats. NeuroToxicology, 2017; 59, 56−64. doi: 10.1016/j.neuro.2017.01.006 [27] Wang SX, Wang ZH, Cheng XT, et al. Arsenic and fluoride exposure in drinking water: children's IQ and growth in Shanyin county, Shanxi province, China. Environ Health Perspect, 2007; 115, 643−7. doi: 10.1289/ehp.9270 [28] Du XY, Zhang J, Zhang X, et al. Persistence and reversibility of arsenic-induced gut microbiome and metabolome shifts in male rats after 30-days recovery duration. Sci Total Environ, 2021; 776, 145972. doi: 10.1016/j.scitotenv.2021.145972 [29] Chen FB, Luo Y, Li CJ, et al. Sub-chronic low-dose arsenic in rice exposure induces gut microbiome perturbations in mice. Ecotoxicol Environ Saf, 2021; 227, 112934. doi: 10.1016/j.ecoenv.2021.112934 [30] Li D, Yang Y, Li YX, et al. Changes induced by chronic exposure to high arsenic concentrations in the intestine and its microenvironment. Toxicology, 2021; 456, 152767. doi: 10.1016/j.tox.2021.152767 [31] Wang JT, Hu W, Yang HL, et al. Arsenic concentrations, diversity and co-occurrence patterns of bacterial and fungal communities in the feces of mice under sub-chronic arsenic exposure through food. Environ Int, 2020; 138, 105600. doi: 10.1016/j.envint.2020.105600 [32] Chen GJ, Hu PC, Xu ZC, et al. The beneficial or detrimental fluoride to gut microbiota depends on its dosages. Ecotoxicol Environ Saf, 2021; 209, 111732. doi: 10.1016/j.ecoenv.2020.111732 [33] Saji N, Niida S, Murotani K, et al. Analysis of the relationship between the gut microbiome and dementia: a cross-sectional study conducted in Japan. Sci Rep, 2019; 9, 1008. doi: 10.1038/s41598-018-38218-7 [34] Rong WW, Han KF, Zhao ZH, et al. The protective effect of Xanthoceras sorbifolia Bunge husks on cognitive disorder based on metabolomics and gut microbiota analysis. J Ethnopharmacol, 2021; 279, 113094. doi: 10.1016/j.jep.2020.113094 [35] Chen DL, Qi LK, Guo YR, et al. CircNF1-419 improves the gut microbiome structure and function in AD-like mice. Aging, 2020; 12, 260−87. doi: 10.18632/aging.102614 [36] Asnicar F, Berry SE, Valdes AM, et al. Microbiome connections with host metabolism and habitual diet from 1, 098 deeply phenotyped individuals. Nat Med, 2021; 27, 321−32. doi: 10.1038/s41591-020-01183-8 [37] Chai YY, Luo JY, Bao YH. Effects of Polygonatum sibiricum saponin on hyperglycemia, gut microbiota composition and metabolic profiles in type 2 diabetes mice. Biomed Pharmacother, 2021; 143, 112155. doi: 10.1016/j.biopha.2021.112155 [38] Chakraborti B, Verma D, Guhathakurta S, et al. Gender-specific effect of 5-HT and 5-HIAA on threshold level of behavioral symptoms and sex-bias in prevalence of autism spectrum disorder. Front Neurosci, 2019; 13, 1375. [39] Takahashi K, Mizukami H, Osonoi S, et al. Inhibitory effects of xanthine oxidase inhibitor, topiroxostat, on development of neuropathy in db/db mice. Neurobiol Dis, 2021; 155, 105392. doi: 10.1016/j.nbd.2021.105392 [40] Nelson-Mora J, Escobar ML, Rodríguez-Durán L, et al. Gestational exposure to inorganic arsenic (iAs3+) alters glutamate disposition in the mouse hippocampus and ionotropic glutamate receptor expression leading to memory impairment. Arch Toxicol, 2018; 92, 1037−48. doi: 10.1007/s00204-017-2111-x [41] Mo TT, Dai H, Du H, et al. Gas chromatography-mass spectrometry based metabolomics profile of hippocampus and cerebellum in mice after chronic arsenic exposure. Environ Toxicol, 2019; 34, 103−11. doi: 10.1002/tox.22662 [42] Ning X, Li B, Ku TT, et al. Comprehensive hippocampal metabolite responses to PM2.5 in young mice. Ecotoxicol Environ Saf, 2018; 165, 36−43. doi: 10.1016/j.ecoenv.2018.08.080 [43] Zhang RY, Tu JB, Ran RT, et al. Using the metabolome to understand the mechanisms linking chronic arsenic exposure to microglia activation, and learning and memory impairment. Neurotox Res, 2021; 39, 720−39. doi: 10.1007/s12640-020-00286-x [44] Jordan W, Dobrowolny H, Bahn S, et al. Oxidative stress in drug-naïve first episode patients with schizophrenia and major depression: effects of disease acuity and potential confounders. Eur Arch Psychiatry Clin Neurosci, 2018; 268, 129−43. doi: 10.1007/s00406-016-0749-7 [45] Minhas PS, Latif-Hernandez A, McReynolds MR, et al. Restoring metabolism of myeloid cells reverses cognitive decline in ageing. Nature, 2021; 590, 122−8. doi: 10.1038/s41586-020-03160-0 [46] Dos Santos SM, Romeiro CFR, Rodrigues CA, et al. Mitochondrial dysfunction and alpha-lipoic acid: beneficial or harmful in Alzheimer's disease? Oxid Med Cell Longev, 2019; 2019, 8409329. [47] Nurchi VM, Djordjevic AB, Crisponi G, et al. Arsenic toxicity: molecular targets and therapeutic agents. Biomolecules, 2020; 10, 235. doi: 10.3390/biom10020235 -
22169Supplementary Materials.pdf