You are on page 1of 15

The Journal of Nutrition 153 (2023) 3382–3396

journal homepage: https://jn.nutrition.org/

Genomics, Proteomics, and Metabolomics

Maternal-Periconceptional Vitamin B12 Deficiency in Wistar Rats


Leads to Sex-Specific Programming for Cardiometabolic Disease Risk in
the Next Generation
Praveen Singh 1, 3, #, Lovejeet Kaur 2, 4, #, Subhoshree Ghose 1, 3, Swati Varshney 1, 3,
Vislavath Jyothi 2, Sourav Ghosh 1, 3, Pujitha Kommineni 2, Shamsudheen KV 1, Vinod Scaria 1, 3,
Sridhar Sivasubbu 1, 3, Giriraj Ratan Chandak 2, 3, **, Shantanu Sengupta 1, 3, *
1
CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; 2 CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India;
3
Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India; 4 Translational Health Science and Technology Institute, Faridabad,
India

A B S T R A C T

Background: Maternal vitamin B12 deficiency plays a vital role in fetal programming, as corroborated by previous studies on murine
models and longitudinal human cohorts.
Objectives: This study assessed the effects of diet-induced maternal vitamin B12 deficiency on F1 offspring in terms of cardiometabolic
health and normalization of these effects by maternal-periconceptional vitamin B12 supplementation.
Methods: A diet-induced maternal vitamin B12 deficient Wistar rat model was generated in which female rats were either fed a control AIN-
76A diet (with 0.01 g/kg vitamin B12) or the same diet with vitamin B12 removed. Females from the vitamin B12-deficient group were
mated with males on the control diet. A subset of vitamin B12-deficient females was repleted with vitamin B12 on day 1 of conception. The
offspring in the F1 generation were assessed for changes in body composition, plasma biochemistry, and molecular changes in the liver. A
multiomics approach was used to obtain a mechanistic insight into the changes in the offspring liver.
Results: We showed that a 36% reduction in plasma vitamin B12 levels during pregnancy in F0 females can lead to continued vitamin B12
deficiency (60%–70% compared with control) in the F1 offspring and program them for cardiometabolic adversities. These adversities, such
as high triglycerides and low high-density lipoprotein cholesterol, were seen only among F1 males but not females. DNA methylome analysis
of the liver of F1 3-mo-old offspring highlights sexual dimorphism in the alteration of methylation status of genes critical to signaling
processes. Proteomics and targeted metabolomics analysis confirm that sex-specific alterations occur through modulations in PPAR signaling
and steroid hormone biosynthesis pathway. Repletion of deficient mothers with vitamin B12 at conception normalizes most of the molecular
and biochemical changes.
Conclusions: Maternal vitamin B12 deficiency has a programming effect on the next generation and increases the risk for cardiometabolic
syndrome in a sex-specific manner. Normalization of the molecular risk markers on vitamin B12 supplementation indicates a causal role.

Keywords: maternal vitamin B12 deficiency, intergenerational programming, repletion, sex-specific effects, cardiometabolic abnormalities

Abbreviations: C, control; DMR, differentially methylated region; FFM, fat-free mass; iTRAQ, isobaric tags for relative and absolute quantitation; LBM, lean body
mass; LC, liquid chromatography; MetS, metabolic syndrome; MeDIP-Seq, methylated DNA immunoprecipitation sequencing; MS, mass spectrometry; OCM, one-
carbon metabolic; PPAR, peroxisome proliferator-activated receptors; RC, repletion at conception; TC, total cholesterol; TG, triglyceride; TSS, transcription start site;
UTR, untranslated region; VR, vitamin-restricted.
* Corresponding author.
** Corresponding author. E-mail addresses: chandakgrc@ccmb.res.in (G.R. Chandak), shantanus@igib.res.in (S. Sengupta).
#
PS and LK contributed equally to this work.

https://doi.org/10.1016/j.tjnut.2023.08.032
Received 16 June 2023; Received in revised form 18 August 2023; Accepted 24 August 2023; Available online 1 September 2023
0022-3166/© 2023 American Society for Nutrition. Published by Elsevier Inc. All rights reserved.
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

Introduction offspring in the next generation. To ascertain causality, we


looked for the reversal of these effects by maternal-
In utero maternal nutritional status is known to influence the periconceptional vitamin B12 repletion. We observed that
offspring’s health. Under and/or overnutrition can predispose vitamin B12 deficiency leads to increased adiposity and altered
the fetus to the risk of developing noncommunicable diseases, plasma lipid profile in the F1 generation offspring. Multiomics
such as diabetes, hypertension, stroke, and coronary artery dis- approaches, including DNA methylation and proteomics studies
ease in adult life [1]. The Developmental Origins of Health and on the liver of 3-mo-old F1 Wistar rats revealed altered metabolic
Disease hypothesis has emerged based on epidemiologic obser- processes in male offspring but not in females, which were
vations that in utero malnutrition is associated with altered fetal supported by a targeted metabolomics analysis of the liver. Thus,
birth weight and future risk of the onset of metabolic syndrome our study provides evidence that maternal vitamin B12 defi-
(MetS) [2–4]. Quantity and quality of macronutrients, such as ciency dysregulates key metabolic pathways in a sex-specific
proteins, fat, and micronutrients, such as vitamins and minerals, manner and consequent future cardiometabolic risk.
play a critical role in fetal development [5–7]. Deficiencies of
vitamin B12 and/or vitamin B9 (folate) are known to influence Methods
fetal growth and differentiation through altered fetal program-
ming via regulating DNA methylation at several gene loci [8]. Animal experimentation and ethics statement
Both vitamin B12 and folate participate in the one-carbon The female vitamin B12 deficient model was developed at
metabolic (OCM) pathway in which folate acts as a methyl CSIR (Council of Scientific and Industrial Research) -Centre for
group carrier and participates in the biosynthesis of nucleotides Cellular and Molecular Biology, Hyderabad, India on Wistar rats
whereas vitamin B12 acts as a cofactor of methionine synthase, in accordance with the Committee for the Purpose of Control and
which catalyzes the conversion of homocysteine to methionine. Supervision of Experimental Animals guidelines of the Govern-
Epidemiologic studies demonstrate that low serum vitamin B12 ment of India. The experimental procedures were approved by
concentrations are common during pregnancy due to increased the “Institute’s Ethical Committee on Animal Experiments”
demand throughout the gestational period [9]; severe deficiency (CCMB/IAEC/27/2013).
can be associated with intrauterine growth restriction, neural
tube defects, preterm birth and low-birth weight, adiposity, and
other adverse pregnancy outcomes [10–12]. Generation of female Wistar rat model with vitamin
Our previous studies have shown that a 90% plasma vitamin B12 deficiency
B12 reduction in female Wistar rats is associated with insulin Female weanling Wistar (outbred) rats (Rattus norvegicus) (n ¼
resistance, adiposity, oxidative stress, and increased gluconeo- 18) were housed (n ¼ 2/cage) in propylene cages with wire mesh
genesis in male offspring [13]. Proteomics study performed on bottom (to avoid coprophagy) and maintained at 22  2 C and
the liver of offspring born to vitamin B12-deficient mothers 12-h light/dark cycle. The animals were divided into 2 groups and
revealed alteration of carbohydrate, amino acid, fatty acid, and consumed ad libitum for 3 mo either a casein-based control diet
lipid metabolism pathways [14]. Moreover, 90% reduction of (AIN-76A) (n ¼ 6) or the same diet with vitamin B12 restriction (0
maternal plasma vitamin B12 has been shown to alter the g/kg vitamin B12 in the diet; n ¼ 12). The diets were purchased
methylation status of candidate genes involved in the fatty acid from M/S Research Diets (diet composition: Supplementary
oxidation pathway and mitochondrial metabolism in the liver of Table 1A–C) and all animals had free access to deionized water.
F1 generation male offspring [15]. Interestingly, repletion of Feed intake and body weights were measured at 3 mo.
mothers with vitamin B12 at conception and parturition Biochemical estimations of vitamin B12, folate, homocysteine,
normalized the methylation signatures as well as the plasma and lipids in plasma were performed at the end of 3 mo. All
lipid levels [15]. However, this extreme deficiency model does control males were fed normal feed pellets from the time of
not appropriately mimic the vitamin B12 deficiency that is weaning, maintained under similar conditions, and housed in
routinely observed in the human population. Importantly, it has pairs until mating. After confirmation of vitamin B12 deficiency,
been shown previously that severe vitamin B12 deficiency could the F0 females were mated with control males (at 2 females:1
modulate the anthropometric, biochemical parameters, and in- male per cage). A subgroup of vitamin-restricted (VR) dams (n ¼
flammatory cytokines to a greater extent than moderate defi- 6) was repleted with control diet on day 1 of conception [repletion
ciency [12]. Therefore, it becomes pertinent to understand the at conception (RC)]. All 3 groups, Control (C), VR, and RC, were
phenotypes and underlying mechanisms associated with this allowed to deliver and were continued on their respective diets
range of vitamin B12 deficiency. Further, in our previous study, during lactation (Figure 1). The pups of F0 dams, i.e., F1 gener-
we analyzed intergenerational effects of vitamin B12 deficiency ation, were continued on their mother’s respective diets until age
only in male offspring; however, several studies have shown 1 y, and biochemical and body composition analysis was con-
differential bioavailability of vitamin B12 in males and females ducted at 3 and 12 mo. The rats were killed using standard pro-
[16–19]. Some of these differences have been attributed to the cedure following ethical principles by inhalation of carbon
levels of sex hormones, such as testosterone and estrogen, which dioxide. Extracted tissues were washed twice with normal saline
also affect the OCM pathway at multiple levels [20,21]. Thus, it (0.9% NaCl), aliquoted, and snap-frozen using liquid nitrogen
is also important to independently understand the intergenera- before storage in a 80 C freezer until further analysis.
tional effects of vitamin B12 deficiency in males and females.
In the present study, we generated a female Wistar rat model Body composition analysis
with 36% vitamin B12 deficiency in plasma (compared with Body composition was analyzed using total body electrical
control) and assessed its molecular and biologic effects on the conductivity at various time points as described previously [22].

3383
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

Figure 1. Schematic representation of the maternal vitamin B12 deficiency model of Wistar rat. RC, repleted with vitamin B12 at conception; VR,
vitamin B12 restricted.

Lean body mass (LBM) and fat-free mass (FFM) percentages were with an approximate size of 300–350 bp fragments. The quality
measured in a small animal body composition analysis system of the libraries was checked using Agilent 2100 Bioanalyzer and
(model SA 3000 multidetector EMSCAN). The difference be- the concentrations were estimated using Qubit ds DNA high
tween LBM and FFM was used to calculate the percentage of sensitivity assay in Qubit 3.0 fluorometer (Q33216). The li-
tissue-associated fat [13]. braries were loaded onto the flow cell, and sequencing was
performed in the Illumina HiSeq2500 instrument using version 4
Measurement of plasma biochemical parameters chemistry on single read mode for 50 cycles of amplification.
Plasma vitamin B12 and folate levels were measured using an
electrochemiluminescence immunoassay kit from Roche Di- Proteomic profiling of liver tissue from F1 pups
agnostics on a Cobas e411 automated platform using the man- Proteomic studies were performed on the liver of F1
ufacturer’s guidelines. Plasma homocysteine and cysteine offspring (both male and female) in 3 different groups (C, VR,
concentrations were measured by HPLC equipped with a fluo- and RC) with 3 biologic replicates of each experimental con-
rescence detector as described previously [23]. Lipid parameters dition. Samples were homogenized in lysis buffer [7M urea,
such as total cholesterol (TC), HDL cholesterol, LDL cholesterol, 2M thiourea, 4% 3-((3-cholamidopropyl) dimethylammonio)-
and triglyceride (TG) levels were measured using enzymatic 1-propanesulfonate, 25 mM ammonium bicarbonate, with
colorimetric assay kits from Roche Diagnostics on the COBAS complete protease inhibitor cocktail] and centrifuged at
INTEGRA 400 plus analyzer following the manufacturer’s 12,000  g for 30 min at 4 C. The supernatant was collected in
instructions. a fresh microcentrifuge tube. After quantification with Brad-
ford’s reagent, protein reduction, alkylation, tryptic digestion,
DNA methylome study and peptide labeling with isobaric tags for relative and abso-
Methylated DNA immunoprecipitation sequencing of F1 livers lute quantitation (iTRAQ) 4-plex reagent were performed as
To generate a genome-wide methylation profile, a methylated described previously [25]. The labeled peptides were frac-
DNA immunoprecipitation sequencing (MeDIP-Seq) based tionated into 8 fractions by strong cation exchange chroma-
approach was used. Genomic DNA was isolated from the liver of tography (5 μm, 300 Å bead from SCIEX) with increasing
3-mo-old Wistar rats from 3 groups (C, VR, and RC) of the F1 concentrations of ammonium formate (30–350 mM) buffer in
generation. MeDIP library construction and sequencing was 30% acetonitrile (vol:vol), pH 2.9.
performed as mentioned previously [15]. Briefly, genomic DNA The iTRAQ 4-plex labeled peptide fractions obtained after
was fragmented to an approximate size of 100–500 bp, purified strong cation exchange chromatography were separated on
followed by end repair, 30 end adenylation, and adapter ligation reverse phase LC using the Eksigent NanoLC-Ultra 2D plus sys-
using NEBNext DNA library preparation kit (E6040L) following tem in trap-elute fashion. Peptides were desalted on an Eksigent
the manufacturer’s instructions. Double-stranded DNA was de- trap (ChromXP C18-3 μm, 120 Å, 350 μm  0.5 mm) column for
natured at 95 C to single-stranded DNA and then 5-methylcyto- 40 min at a flow rate of 3 μL/min. They were resolved on an
sine-specific antibody (BI-MECY-0100) was used to precipitate Eksigent C18 column (ChromXP C18, 75 μm  15 cm) in a 110-
and enrich for methylated DNA fragments. The efficiency of min binary gradient with solvent A (0.1% formic acid) and sol-
enrichment was checked using PCR and qRT-PCR with primers vent B (99.9% acetonitrile and 0.1% formic acid) as the mobile
corresponding to unmethylated and methylated regions reported phase and a flow rate of 250 nL/min. The LC eluent was analyzed
in humans previously [24]. PCR amplification was performed using a NanoSpray III Source installed on the SCIEX TripleTOF
using NEBNext Q5 HotStart HiFi PCR master mix (M0543S) for 5600þ system. The instrument was operated in information-
12 cycles followed by agarose gel-based size selection of libraries dependent acquisition mode. The full mass spectrometry (MS)

3384
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

spectra were acquired in positive ion mode in m/z 350–1250 Da sufficient for whole genome coverage [33]. Depth of sequencing
with a 200 ms time-of-flight MS accumulation time, whereas the was checked using a systematic data reduction approach with
MS/MS product ion scan was performed in the mass range of MACS output [15,24]. The number of methylated peaks were
100–1800 Da with a 70 ms accumulation time and a total cycle comparable to a previously published rat liver methylome study
time of 1.93 s. Parent ions with a charge state from þ2 to þ5 [15]. Regions were considered to be differentially methylated
were considered for MS/MS acquisition. Optimized ion-source (DMR) if there was at least a 2-fold difference of methylation
parameters were used. Twenty-four candidate ions were moni- with a hypergeometric P value  5  105. The number of peak
tored per MS cycle, and the former target ion was excluded for 7 distributions was counted in different genomic regions (pro-
s. Information-dependent acquisition advanced “rolling collision moter, 50 untranslated region [UTR], exon, introns, and 30 UTR)
energy” was applied for subsequent MS and MS/MS scans. using in-house PERL script. For gene annotation of the DMRs, we
downloaded the Ref-Seq (rn6) gene coordinates from the UCSC
Targeted metabolite profiling of liver in the F1 genome browser, and regulatory region analysis was performed
offspring in the 10-kb region surrounding the transcription start site (TSS).
Metabolites were extracted from the liver of F1 generation KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway
offspring (both male and female) from control, VR, and RC analysis was performed using DAVID bioinformatics resources
groups with 3 biologic replicates in each experimental condition. [34,35]. Pathways were considered significant based on P value
Ten milligrams of freeze-dried liver tissue was used for the of 0.05 with minimum 3 genes in each category.
extraction of metabolites using the protocol defined previously
[26]. Metabolites were reconstituted in 125-μL methanol: Proteomics and metabolomics data analysis
acetonitrile: water (2:1:1), and polar and nonpolar metabolites All MS and MS/MS spectra generated were submitted for
were pooled in the ratio of 1:1 prior to LC-MS acquisition. Tar- database searching and quantitative analysis using ProteinPilot
geted metabolic profiling was performed for amino acids and version 4.0 (SCIEX). The Paragon algorithm was used in a
intermediates of glycolysis, tricarboxylic acid (TCA) cycle, and “Thorough ID” search mode against the Rattus norvegicus refer-
steroid hormone biosynthesis pathways (Supplementary Table 2 ence proteome from the UniProt database [36]. The following
shows the metabolites and transitions information). Samples parameters were included for search: trypsin as the digestion
were analyzed on a triple quadrupole hybrid ion trap mass enzyme with 2 missed cleavages, modifications by iodoaceta-
spectrometer (QTRAP 6500þ, SCIEX) coupled with a SCIEX mide as cysteine blocking reagent, iTRAQ 4-plex modification of
ExionLC UHPLC system, and data was acquired through Analyst the N-terminal of peptides and of the side chains of lysine. An
1.6.3 software in both negative and positive modes. Metabolites automatic decoy database search was also performed to calculate
were resolved on an Acquity UPLC BEH HILIC (1.7 μm, 2.1  100 the false discovery rate, and 1% global protein level false dis-
mm) column using mobile phases of 10 mM ammonium acetate covery rate was considered for protein identification. The
and 0.1% formic acid in water (buffer A) and 95% acetonitrile resulting data set was normalized using auto-bias correction to
with 10 mM ammonium acetate and 0.1% formic acid (buffer B) remove any experimental bias. A cutoff of 1.2-fold change was
with a flow rate of 0.3 mL/min. set for identifying differentially expressed proteins, as used in
previous studies [37,38]. Pathway enrichment analysis was
Data analysis performed in DAVID bioinformatics resources [34,35].
All biochemical variables are presented as mean  SEM. Metabolomics data was processed using MultiQuant software
Differences among the groups at specific ages were tested using v.3.0 (SCIEX), and data was exported to Microsoft Excel where
Student’s t test for 2 groups or analysis of variance (including a relative quantification and statistical analysis was performed.
test for the homogeneity of variance) for >2 groups, followed by Metabolites were considered significantly altered for a 1.5-fold
Bonferroni’s post hoc correction. Data sets were checked for any change and a P value of 0.05 in Student’s t test.
outliers and if found deviated, Mann–Whitney test for 2 groups
and Kruskal Wallis for >2 groups were used. P values of 0.05 Results
were considered significant. All heatmaps were generated using
Morpheus (https://software.broadinstitute.org/morpheus). Direct effects of plasma vitamin B12 deficiency on
Venn diagrams were plotted using Venny 2.1.0 (https:// F0 females
bioinfogp.cnb.csic.es/tools/venny/index.html). Plasma vitamin B12 levels were 36% lower in female rats fed
a vitamin B12-deficient diet for 3 mo than those fed with control
MeDIP-Seq data analysis diet. There were no significant changes in plasma folate and
The raw sequencing reads were processed through FastQC homocysteine levels (Table 1). Body weight of vitamin B12-
(https://www.bioinformatics.babraham.ac.uk/projects/fastqc) deficient female rats was lower than the control group
and Trimmomatic [27] to remove the adapters and low-quality (Table 1). The vitamin B12-deficient dams (F0-VR) and those
reads. The reads were mapped to the rat genome available in repleted at conception (F0-RC) did not show any significant
the UCSC genome browser (rn6 assembly) using Bowtie 2 difference in reproductive performance in terms of time to
[28–30]. Alignment percentages were calculated using both conception and litter size (data not shown).
aligned and unaligned reads. Further, Samtools [31] was used to
filter the uniquely mapped reads, and they were used for meth- Effects of maternal plasma vitamin B12 deficiency
ylated peak calling using MACS version 1.4.0 (Model-based on F1 offspring
Analysis of ChIP-Seq) [32] with default parameters. We gener- F1 generation offspring were followed at 3- and 12-mo,
ated ~50 to 200 million reads in each sample, which was equivalent to early and mid-adulthood in humans [39]. Body

3385
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

Table 1 completely correct to the level of controls in the RC groups


Biochemical parameters and body weight of the F0 females after 3 mo (Supplementary Table 3).
of vitamin B12-restricted diet1 In an attempt to account for the effect of sex, we conducted a
Parameters Control (n ¼ 6) Vitamin-restricted (n ¼ 6)2 sex-specific analysis. Sex-specific analysis in 3 mo-old offspring
revealed that male offspring born to vitamin B12-restricted
Body weight (g) 230.72  3.81 216.91  3.983
Vitamin B12 (pg/mL) 823.17  27.07 529.2  78.783 mothers had significantly higher body fat, lower FFM, and
Folate (ng/mL) 8.62  1.03 8.96  1.26 lower LBM percentages whereas females did not show any sig-
Homocysteine (μmol/L) 3.47  0.3 3.1  0.04 nificant differences in the body composition with respect to
1
Values are presented as mean  SEM. controls. Interestingly, although a significant vitamin B12 defi-
2
Biochemical parameters could not be measured in one animal due ciency was present in both male and female offspring at 3 mo,
to low blood volume; hence, the values represent mean value of 5 plasma homocysteine levels were significantly higher only in
animals. males. There were no significant changes in plasma folate and
3
P < 0.05 by Student’s t test. cysteine levels. Repletion with vitamin B12 at conception cor-
rected (albeit partially) the plasma vitamin B12 levels in both
composition analysis using total body electrical conductivity in sexes and homocysteine levels only in males. We also observed
3-mo-old offspring showed no overall difference between significantly higher TG levels and a trend of increasing TC and
vitamin B12-restricted and control group (Supplementary LDL cholesterol in VR males with respect to control whereas VR
Table 3). We also noted a ~71% reduction in plasma vitamin females had significantly lower LDL cholesterol and a trend of
B12 with significant increase in homocysteine levels in the lower TG and TC levels. Males from the RC group showed
3-mo-old pups born to VR mothers. However, no significant reversal of TGs to the control levels whereas RC females
differences were noted in plasma lipid parameters in the VR continued to have low TC and LDL cholesterol levels (Table 2).
group except an increasing trend for TG levels compared to pups Both male and female offspring in the VR group showed
born to control rats. Offspring from the VR group at 12 mo significantly lower levels of plasma vitamin B12 (65% and 55%
continued the same trend as 3 mo offspring with no significant compared with respective controls) and elevated homocysteine
differences in body composition. However, along with a ~60% levels at 12 mo. However, only males showed elevated cysteine
plasma vitamin B12 deficiency and elevated plasma homocys- levels and altered body composition and plasma lipid parame-
teine, the 12-mo-old pups in the VR groups had significantly ters. Males from the VR group were obese with higher body
higher levels of cysteine and TGs. These changes did not weight and body fat percentages and decreased LBM and FFM

Table 2
Body composition and plasma biochemical parameters in female and male offspring at 3-mo time points1
Parameters Age (mo) Male Female
Control (n ¼ 6) VR (n ¼ 6) RC (n ¼ 6) Control (n ¼ 6) VR (n ¼ 6) RC (n ¼ 6)
Body weight (g) 3 361.4  10.2 361.7  5.5 373.7  6.5 219.1  6.3 214.6  6.4 221.2  6.0
12 440.2  21.4 492  15.6 583.3  9.3 b*&,c#$ 281.7  12.4 271.7  10.3 308.2  11.8a#$
Body fat (%) 3 14.2  0.5 18.4  1.1b*$ 18  0.8 b*$ 15.0  0.9 12.9  1 17.0  0.6b#$
12 17.4  0.5 19.5  0.9 14.8  1.8 18.0  1.4 16.8  1.3 19.4  0.4
Lean body mass (%) 3 85.8  0.5 81.6  1.1b*$ 82.0  0.8b*$ 85.0  0.9 87.1  1.0 83.0  0.6b#$
12 82.6  0.5 80.5  0.8 85.2  1.8 82.0  1.4 83.2  1.3 80.6  0.4
Fat-free mass (%) 3 38.8  0.1 37.1  0.4b*$ 37.2  0.3c*$ 41.5  0.5 42.4  0.6 40.6  0.3a#$
12 45.8  0.3 43.7  0.8a*$ 46.9  1.4 47.4  1.3 48.6  1.2 45.8  0.4
Vitamin B12 (pg/mL) 3 731.8  31.7 190.2  16.7b*& 673.0  23.2b#& 678.9  18.0 217.9  29.3c*$ 660.2  45.2c#$
12 902.1  37.6 320.0  10.0b*& 770.6  44.6a*&,c#$ 727.8  18.7 330.3  24.0c*$ 669.4  33.3c#$
Folate (ng/mL) 3 31.7  1.8 32.4  2.2 33.0  2.0 21.5  0.7 22.5  1.4 21.3  1.4
12 21.6  1.4 23.3  1.2 20.0  1.2 23.5  1.0 22.2  1.0 21.7  0.6
Homocysteine (μmol/L) 3 6.3  0.7 8.9  0.5a*$ 7.0  0.4a#$ 5.0  0.3 6.5  1.6 5.1  0.56
12 5.4  0.4 9.0  0.7b*$ 7.9  0.3c*$ 5.2  0.4 7.9  0.8a*$ 5.8  0.4a#$
Cysteine (μmol/L) 3 135.0  10.7 154.8  11.5 155.8  8.6 162.9  7.7 158.7  19.5 173.4  18.7
12 94.7  7.2 132.3  14.7a*$ 99.8  1.8 96.2  5.8 103.9  9.8 126.0  7.3a*&
TG (mg/dL) 3 77.2  7.9 170.8  28.1a*$ 56.5  2.8a*$,b#$ 94.6  11.8 80.2  10.2 112.4  14.5
12 57.9  5.1 95.2  9.3b*$ 82.3  8.1a*$ 64.2  12.7 72.4  6.3 77.8  7.9
HDL-C (mg/dL) 3 60.8  4.3 69.7  5.0 55.3  1.8a#& 85.7  3.9 77.1  8.1 73.8  5.9
12 91.7  2.1 69.9  2.7c*$ 76.3  3.9b*$ 67.0  6.2 75.4  4.1 70.5  4.9
LDL-C (mg/dL) 3 6.7  0.6 9.9  1.5 6.1  0.3 15.8  2.4 8.8  1.2a*$ 7.8  0.82a*$
12 21.7  1.8 24.6  2.7 20.6  2.8 9.1  1.9 8.3  2.5 9.0  1.7
TC (mg/dL) 3 70.1  4.6 88.0  7.4 63.7  2.3a#$ 109.2  6.4 89.9  7.9 84.5  5.6a*$
12 124.9  11.1 145.4  12.8 129.2  12.9 108.9  6.0 104.3  5.2 109.4  9.6
1
Wistar rat offspring fed different diets: Control (C), Vitamin B12 restricted (VR), Repleted at Conception (RC). Values are presented as the mean
 SEM. Values in a row with different superscripts (a/b/c) are significantly different from others at P value (a < 0.05, b < 0.01, c < 0.001).
*
Difference from control group.
#
Difference from the vitamin-restricted group. The normality test was done using the Shapiro test. For homogenous distribution, t test was used
(represented as $), and for non-homogenous distribution, non-parametric Mann–Whitney test was done (represented as &). Abbreviations: HDL-C,
high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; TC, total cholesterol; TG, triglyceride.

3386
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

percentages. These male rats also showed significantly elevated 100-bp sliding window showed a characteristic V-shaped curve
plasma TG and low HDL cholesterol levels, indicating an indicating low methylation density at the TSS, which increased
increased risk for developing cardiovascular diseases. However, with increasing distance downstream of the TSS (Figure 2A).
altered body composition and plasma lipid parameters in VR This V-shaped pattern was consistent among all the groups of
males did not completely recover in the RC group and continued offspring indicating that the global methylation pattern around
to show earlier changes. However, females from the VR and RC the TSS was conserved across different groups and is in agree-
groups showed no difference in body composition and plasma ment with our previous study [15]. However, sex-specific anal-
lipid parameters compared with control females (Table 2). ysis revealed that vitamin B12-restricted male offspring had an
increased methylation density in the gene body region compared
Methylome sequencing reveals sex-specific with controls, which was partially restored following RC
differences in DNA methylation and associated (Figure 2A). No significant differences in global methylation
biologic pathways density were observed between control, vitamin B12-restricted,
RC rescued some of the phenotypic parameters, albeit and vitamin B12-repleted female offspring suggesting
partially, hinting toward epigenetic alteration due to vitamin sex-specific differences in programming of methylation alter-
B12 deficiency. We thus assessed the DNA methylation status in ations upon maternal vitamin B12 deficiency (Figure 2B).
the liver of 3-mo-old pups to ascertain DMRs in pups born to VR Further, both hypomethylated and hypermethylated regions [in
mothers and also to check if these alterations normalized in the all the genomic regions (promoter, 5’UTR, exon, 3’UTR and
repleted pups. The average methylation pattern in the 10-kb intron)] were higher in males than in females in VR pups,
region (5 kb upstream and downstream) around the TSS in a compared with control (Figure 2C, D). Interestingly, the majority

Figure 2. Average methylation pattern around the transcription start site (TSS) in control, vitamin B12 restricted, vitamin B12 repleted male
(A) and female (B) offspring in a 100-base pair (bp) sliding window. Distribution of hypermethylated (C) and hypomethylated (D) regions across
different genomic regions in the liver of F1 generation male and female offspring. Venn diagram showing the intersection of genes with
hypermethylated (E) and hypomethylated (F) promoter regions in vitamin-restricted male and female livers. UTR, untranslated region.

3387
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

of the DMRs in liver were sex-specific. We found a total of 4264 leading to metabolic changes. Therefore, we looked at the
and 703 unique hypermethylated and 290 and 127 hypo- methylation status of genes involved in energy metabolism. The
methylated DMRs in the promoter region of male and female promoter regions of genes that code for enzymes in the glycolysis
liver, respectively. This corroborates with our previous study pathway, glucose-6-phosphatase, catalytic subunit (G6pc), phos-
that showed a significant association of DNA hypermethylation phofructokinase (Pfkl), 6-phosphofructo-2-kinase/fructose-2,
with hyperhomocysteinemia due to vitamin B12 deficiency [15, 6-bisphosphatase (Pfklb3), glyceraldehyde 3-phosphate dehydro-
40]. Methylation levels at ~75% to 90% of DMRs were reversed genase (Gapdh), phosphoglycerate mutase (Pgam1), enolase
upon vitamin B12 RC, suggesting a causal inference (Supple- (Eno3), and pyruvate kinase (Pk), were hypermethylated in VR
mentary Table 4). male liver. Similarly, genes coding for enzymes in the TCA cycle
such as citrate synthase (Cs), aconitase (Aco1 and Aco2), isocitrate
dehydrogenase (Idh1), fumarate hydratase (Fh), and malate de-
Global methylome study indicates perturbation of hydrogenase (Mdh2) were hypermethylated. Promoters of genes
important metabolic pathways coding for phosphoenolpyruvate carboxykinase (Pck1) and pyru-
Biologic pathway enrichment analysis revealed that the DMR vate carboxylase (Pc) also showed hypermethylation in the VR
genes in the VR male liver were majorly enriched in signaling male liver, indicating lower gene expression.
pathways like Hippo signaling, FoxO signaling, MAPK (Mitogen- Genes related to fatty acid transport, desaturation, and elon-
activated protein kinase) signaling, AGE-RAGE (Advanced gation were hypermethylated in the male liver, indicating that
glycation end products (AGEs) and their receptor (Receptor for β-oxidation in the mitochondria could be affected. The trans-
Advanced Glycation Endproducts, RAGE)) signaling, Wnt porter genes (Cpt1a, Cpt1b, Slc27a1, and Slc27a4) that import
signaling, AMPK (AMP-activated protein kinase) signaling, and fatty acids in the mitochondria showed hypermethylation. We
PI3K-Akt signaling (Figure 3A). These signaling pathways are observed that the promoter of genes such as Elovl, Acsl4, Fads,
complex, dynamic, and highly interconnected and regulate Fabp1, Hmgcs, Acat2, Acaa1b, and Acox2 were hypermethylated
glucose, lipid metabolism, and insulin sensitivity along with in male liver, indicating downregulation. ACOX is the rate-
processes, such as cell proliferation, differentiation, and stress limiting enzyme in fatty acid β-oxidation. Srebf1 and Srebf2,
response [41–46]. DMR genes in VR female liver showed genes that code for sterol regulatory element binding proteins, a
enrichment in pathways like Rap1 signaling, phospholipase D family of transcription factors that regulate lipid homeostasis by
signaling, and Ras signaling (Figure 3B). Pathways in cancer controlling the expression of a range of enzymes required for
identified from DMR genes were enriched in both VR male and endogenous cholesterol, fatty acid, triacylglycerol, and phos-
female liver. pholipid synthesis, also showed hypermethylation in their pro-
The list of DMR genes shows hypermethylation in the promoter moter regions in VR male liver [50]. Interestingly, altered
regions of 3 peroxisome proliferator-activated receptors (PPARs), methylation status in the promoters of genes from glucose and
a family of nutrient sensing nuclear receptors (Ppara, Ppard, lipid metabolism were only found in VR male liver and not in
Pparg). PPARα (Ppara) is the PPAR member that is majorly female.
expressed in liver and controls energy metabolism (glucose and In the amino acid metabolism pathway, we observed that
lipid metabolism) [47–49]. Promoter hypermethylation in PPARα genes involved in transamination (Gpt and Agxt) were hyper-
suggests lower expression under vitamin B12 restriction in liver, methylated in the male liver, suggesting altered levels of

Figure 3. KEGG pathway enrichment analysis of differentially methylated genes in vitamin B12-restricted male (A) and female (B) liver. X-axis
denotes -log10 (adjusted P value) and the Y-axis denotes pathway names.

3388
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

glutamate, glutamine, alanine, and serine. Moreover, aldehyde and 110 downregulated) proteins were differentially expressed
dehydrogenases (Aldh4a1, Aldh5a1, and Aldh9a1) were also in male and female offspring, respectively. Interestingly, only
hypermethylated in the male liver. ~10% of the proteins that had altered expression were common
Rap1 signaling pathway was the most affected pathway in between male and female offspring, again suggesting sex-specific
female liver, and Rap1-deficient female liver is known to upre- proteome alterations (Figure 4B). The majority of the differen-
gulate genes involved in different metabolic pathways including tially expressed proteins (62% in males and 65% in females)
lipid, fatty acid, steroid, and cholesterol metabolism [51]. Thus, showed reversal of expression in pups born to vitamin B12-
alteration of multiple metabolic pathways, including branched deficient mothers who were repleted at conception. KEGG
chain amino acid degradation, PPAR signaling pathway, glyc- pathway analysis of differentially expressed proteins revealed
erolipids, and fatty acid metabolism, reflects alterations in lipid that metabolic pathways related to amino acid metabolism,
homeostasis. Glycolysis and gluconeogenesis as well as carbon metabolism, glutathione metabolism, and drug meta-
diabetes-related pathways have also been found to be deregu- bolism involving cytochrome P450 were enriched in vitamin
lated due to RAP1 deficiency [51]. We previously reported B12-restricted males (Figure 5A) whereas females showed al-
altered methylation and protein expression of PPAR signaling in terations in metabolic pathways such as oxidative phosphoryla-
the rat liver of male pups born to vitamin B12-deficient mothers tion and drug metabolism involving cytochrome P450 pathway
[14]. In the female liver, genes involved in cysteine and methi- (Figure 5B).
onine metabolism (Mthfr, Ahcyl, Sds, and Srm) were found to be Amino acid metabolism was the most significantly altered
hypermethylated. Estrogen receptor (Esrra and Esrrg in male and metabolic process in males upon vitamin B12 restriction, with
Esrrb in female), retinoid X receptor (Rxra in males and Rxrb in differential expression of multiple proteins in the amino acid
both sexes), and glucocorticoid receptor (Nr3c1) genes in males biosynthesis process (Table 3).
also showed hypermethylation in the promoter region in vitamin Carbon metabolism showed significant alterations upon
B12-restricted offspring liver. maternal vitamin B12 deficiency as proteins majorly involved in
glycolysis, TCA cycle, and OCM were altered in both male and
Proteomic profiling indicates sex-specific female liver, which was in line with the methylome data.
differences of differential proteins and biologic Expression of glycolytic and TCA cycle enzymes PFKL, ENO3, CS,
pathways and MDH in vitamin-restricted females showed elevated
DNA methylation analysis in this study showed alterations in expression with respect to controls whereas aldolase (ALDOA),
methylation status of promoter and gene body region of a PC, and PCK1 showed diminished expression in at least 2 of the 3
number of genes in a sex-specific fashion. To confirm if these replicates. In the male liver, pyruvate kinase enzyme level was
changes were also reflected at the protein level, proteomic low in restricted group whereas level of isocitrate dehydrogenase
profiling was performed in the liver tissue of F1 generation 3-mo- was high. Proteins involved in oxidative phosphorylation from
old male and female offspring using iTRAQ 4-plex tag based complex I (NADH dehydrogenase), complex III, complex IV, and
quantitative proteomics technique. A total of 1279 proteins in complex V (ATP synthase subunits) showed increased expression
males and 1134 proteins in females were quantified in the 3 upon vitamin restriction in female liver (Table 3).
biologic replicates, of which ~930 proteins were common be- Liver proteomic profiling of F1 tissue revealed that the
tween male and female liver. These proteins showed sex-specific enzyme betaine-homocysteine methyltransferase (BHMT),
alterations in liver upon vitamin B12 restriction (Figure 4A). which is involved in remethylation of homocysteine to methio-
Among the list of commonly quantified proteins, 239 (129 nine in liver, had elevated expression in the male offspring under
upregulated and 110 downregulated) and 305 (195 upregulated vitamin B12 restriction. Higher expression of enzymes involved

Figure 4. (A) Heatmap represents distinct protein expression in vitamin-restricted male and female groups with respect to the control group.
(B) Venn diagram showing the common and unique proteins identified between vitamin B12 restricted male and female offspring liver as
compared to controls.

3389
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

Figure 5. KEGG pathway enrichment analysis of differentially expressed proteins in the vitamin B12-restricted male (A) and female liver (B).

Table 3
Differentially expressed proteins in different metabolic pathways in the liver upon vitamin B12 restriction1
Metabolic pathways VR male liver VR female liver
Up Down Up Down
(>1.2-fold change) (<0.8-fold change) (>1.2-fold change) (<0.8-fold change)
Amino acid metabolism ASL1, ASS1, CPS1, CTH, PKM, SDSL ENO3, OTC CPS
ENO3, GLUL, GPT, GOT1,
IDH1, IDH2, OTC, PAH, SDS
Energy metabolism IDH PKM PFKL, ENO3, CS, MDH, ALDOA, PC, PCK1
(glycolysis, TCA cycle, NDUFA6, NDUFA4, NDUFS3,
oxidative phosphorylation) NDUFS8, NDUFA9,
NDUFA13, NDUFB6,
UQCRC2, COX5A, COX6B1,
COX6A1, COX7A2, ATP5A1,
ATP5H, ATP5O, ATP5I,
ATP5B, ATP5I
One-carbon metabolism BHMT, CTH
Glutathione metabolism GST, GPX, GPT2, GOT1, GCLC, GST, GLS2
GLUL, GLS2
Arginine biosynthesis and ASS1, ASL, OTC, CPS1, GLUL, OTC CPS1, GLS2
urea cycle GPT2, GOT1
Cytochrome P450-dependent UGT2B, FMO3, GSTA3, GSTA1, AKR7A3 UGT2B37, GSTA3, GSTK1, GSTM2, ADH1
xenobiotics and drug GSTK1, GSTM3, GSTT2 GSTP1, SULT2A1, FMO1,
metabolism CYP1A2, F1LTB8,
RGD1559459
Steroid hormone biosynthesis UGT2B, AKR1D1, CYP2D5, CYP1A2, CYP2C12, CYP2B3, HSD11B1
SULT1E1 UGT2B37, RGD1559459
1
Differentially expressed proteins in the liver under vitamin B12 restriction in males (VR male) and females (VR female), revealed by iTRAQ
analysis. Abbreviations: iTRAQ, isobaric tags for relative and absolute quantitation; TCA, tricarboxylic acid.

in the transsulfuration pathway, such as cystathionine gamma- male liver. Females also showed alterations in expression of
lyase indicated an increased flux of homocysteine toward GSTs, glutamate cysteine ligase, and GLS2. Proteins involved
glutathione biosynthesis and metabolism through increase in directly in arginine biosynthesis, and the urea cycle in general,
cysteine levels (as reflected in plasma). Proteins, such as gluta- showed increased expression upon vitamin restriction in males
thione S-transferase (GST) and glutathione peroxidases (GPX) (Table 3).
showed elevated levels under vitamin restriction in male liver. Cytochrome P450-dependent metabolic processes were
Glutamate pyruvate transaminase 2 (GPT2), glutamate- among the top dysregulated pathways in liver of the vitamin-
oxaloacetate transaminase 1 (GOT1), GLUL (glutamine synthe- restricted male and female offspring. Several proteins
tase), and glutaminase (GLS2) were other proteins involved in involved in cytochrome P450-dependent xenobiotics and drug
glutamate biosynthesis and glutamine/glutamate metabolism metabolism pathways show differential levels (Table 3).
that showed elevated expression upon vitamin restriction in Interestingly, some of these cytochrome p450 monooxygenases

3390
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

(CYPs) are also involved in the process of steroid hormone and phosphoenolpyruvate indicate lower glycolytic flux in VR
biosynthesis. Steroid hormone biosynthesis involves enzymes males. Altered TCA intermediates (α-ketoglutarate and oxaloac-
from 2 classes—CYPs and hydroxysteroid dehydrogenases etate in VR males and succinate and oxaloacetate in VR females)
(HSDs) along with some accessory enzymes. Proteins like may also result in alteration of amino acid metabolism and
UGT2B, AKR1D1, CYP2D5, and SULT1E1 were dysregulated in transamination reactions [as highlighted in pathway enrichment
males whereas females showed dysregulated expression of analysis using differentially expressed proteins and methylated
proteins encoded by CYP1A2, CYP2B3, CYP2C12, HSD11B1, genes (Figure 6A)] as they utilize TCA cycle intermediates like
UGT2B37, and RGD1559459. Changes in levels of these pro- α-ketoglutarate and oxaloacetate [52,53].
teins from the steroid hormone biosynthesis pathway might Alteration in amino acid levels was distinctly visible in VR
result in sex-specific outcomes. male liver as all the measured amino acids levels were altered in
males, whereas in females the extent of alteration was lower
Targeted metabolite profiling to understand the (Figure 6D). Glutamine and glutamate, involved in glutathione
effects of maternal vitamin B12 restriction biosynthesis, particularly showed sex-specific patterns as they
DNA methylation and proteomics studies in the liver of F1 were only altered in VR males and were unaffected in VR fe-
offspring born to VR females hinted toward the alteration of males, as also indicated in proteomics data. Glutathione
amino acid, glycolysis, energy metabolism, and steroid hormone biosynthesis was also impacted through an increased trans-
biosynthesis pathways, which could be due to PPARα dysregu- sulfuration process from the OCM pathway through different
lation. To check if the metabolites in these pathways show al- metabolic intermediates. Metabolites like S-adenosylmethionine
terations, we performed a targeted metabolomics study, majorly (SAM), 5,10-methylenetetrahydrofolate, 5-methyltetrahy-
focusing on these pathways. drofolate, cystathionine, and cysteine sulfinate are all elevated in
Glycolysis intermediates, fructose bisphosphate (FBP), 3- VR males (>1.5-fold change, P < 0.05) whereas only VR females
phosphoglycerate/2-phosphoglycerate (3-PG/2-PG), and phos- showed elevated SAM while other intermediates were largely
phoenolpyruvate were lower only in VR male liver. FBP is an unaltered. Proteomics data showed increased BHMT expression
important regulator of glycolytic flux, and low FBP, 3-PG/2-PG, only in VR males. Betaine levels were low specifically in VR male

Figure 6. Status of dysregulated proteins and biologic pathways in the vitamin B12-restricted group with respect to controls with their metabolite
status involved in (A) glucose metabolism, (B) one-carbon metabolism and glutathione biosynthesis, (C) arginine metabolism along with some
intermediates of steroid hormone biosynthesis, and (D) amino acid profile among vitamin-restricted and vitamin-repleted rat livers with respect to
control liver.

3391
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

liver indicating increased utilization in males for the generation with women (20%), and there was a decrease in insulin sensi-
of SAM through methionine (Figure 6B). tivity with age. Those with glucose intolerance showed lower
An altered arginine biosynthesis and urea cycle flux was also compensatory insulin secretion from childhood [61]. Another
found in VR males at the metabolite level, as high arginine, study in the Mysore Parthenon Cohort showed an association of
argininosuccinate, ornithine, citrulline, and aspartate were low maternal vitamin B12 status with greater cortisol response to
found only in VR males. This is consistent with high ASS1, ASL, stress in the offspring, and higher plasma total homocysteine
OTC, and CPS1 found at the protein level. In females, this concentration was significantly associated with a higher heart
pathway remained unimpacted at the metabolite level rate response to stress at 13.5 y [62]. Both studies provide evi-
(Figure 6C). dence that vitamin B12-deficient status during pregnancy leads
Differential steroid hormone regulation as indicated from to fetal programming for future risk of cardiometabolic
methylation and proteomic data was reflected at the metabolite disorders.
level, as several intermediates from steroid hormone biogenesis Global DNA methylome analysis of liver tissues from 3-mo-
like pregnenolone, hydroxypregnenolone, aldosterone, deoxy- old vitamin B12-restricted F1 male offspring show alteration of
corticosterone, and hydroxycorticosterone, were all elevated in signaling pathways like Hippo, FoxO, MAPK, Wnt, and AMPK,
VR males and relatively unaltered in VR females. Cortisol, a which regulate complex metabolic functions and maintain en-
glucocorticoid associated with stress, was also significantly ergy homeostasis by regulating glucose, lipid metabolism, and
increased only in VR male liver whereas VR female showed insulin sensitivity [41–46]. These signaling pathways are also
levels comparable with control female. involved in processes like cell proliferation, differentiation, and
stress response. VR F1 female offspring show altered Rap1,
Discussion phospholipase D, and Ras signaling in liver. Rap1 and Ras
signaling pathways affect processes like cell growth, prolifera-
Vitamin B12 is a key micronutrient essential for fetal growth, tion, differentiation, and cell survival [63]. Rap1 is a crucial
development, and various physiologic functions [54–57]. Defi- player in the process of tumor cell migration, invasion, and
ciency of vitamin B12 at critical stages of development is asso- metastasis [64,65]. Rap1-deficient mice show deregulation of
ciated with poor gestational outcome and adult onset of metabolic programs, including fatty acid, glucose metabolism,
cardiometabolic diseases [56,58–60]. The present study high- and PPARα signaling [51]. Phospholipase D signaling has been
lights the effects of dietary vitamin B12 deficiency in pregnant shown to be positive regulator of glucose metabolism and insulin
Wistar rats leading to distinct effects on the OCM pathway, body sensitivity [66]. Dysregulation of these signaling pathways
composition, and lipid parameters in the offspring in a highlights impaired energy homeostasis (glucose and lipid
sex-specific manner. Multiomics (DNA methylome, proteome, metabolism) and suggests progression toward a MetS phenotype
and metabolome) profiling of liver tissue of F1 generation Wistar with insulin resistance and fatty liver in males. Major hepatic
rat offspring combined with analysis of anthropometric and transcription factors involved in nutrient sensing and regulating
biochemical parameters provide us an insight into the molecular metabolism that are reported to be dysregulated in MetS-related
mechanisms of early developmental programming for risk of perturbations in both clinical and preclinical studies are perox-
cardiometabolic diseases in the future due to perturbation of in isome proliferator-activated receptor α (Ppra), cAMP responsive
utero vitamin B12 levels. Our present study shows that offspring element binding protein 3 like 3 (Creb3l3), and forkhead box O1
in the F1 generation continued on the same vitamin (Foxo1) [67]. Interestingly, these genes show promoter hyper-
B12-restricted diet as their mothers show a further reduction in methylation in VR male liver but not in VR female. Targets of
plasma vitamin B12 with elevated plasma homocysteine levels. these transcription factors also show perturbed methylation
Our study further provides evidence that repletion of pregnant status in the gene promoter region. Proteomics analysis in the VR
mothers with vitamin B12 at conception could partially restore male liver tissue also shows enrichment of metabolic pathways
plasma vitamin B12 levels and also attenuate the metabolic ab- from dysregulated proteins. Pathways related to biosynthesis
normalities in the repleted offspring through altered epigenetic and metabolism of amino acids, such as arginine metabolism,
programming. Sex-specific analysis of body composition and glutathione metabolism, carbon metabolism, cytochrome
plasma biochemical parameters are in line with an earlier report P450-related metabolic processes, pentose and glucuronate
[14]. We show here that F1 generation male offspring developed metabolism, and cysteine and methionine metabolism, are
higher adiposity and had lower HDL cholesterol and higher TG enriched in VR males. VR females show alterations in metabolic
levels under maternal vitamin B12 deficiency at 3 mo, whereas pathways related to carcinogenesis, oxidative phosphorylation,
females did not show these abnormalities. These differences in and cytochrome P450-related processes. Metabolomics study
biochemical parameters and body composition indicate an targeting energy metabolism (glycolysis and TCA cycle), amino
increased cardiometabolic disease risk in adulthood in vitamin acids, arginine metabolism, and steroid hormone biosynthesis
B12-restricted male offspring but not in female offspring, similar shows some sex-specific alterations. Dysregulated glucose
to the observations in human longitudinal studies. A recent study metabolism is suggestive of lower glycolytic flux in liver of male
in the Pune Maternal Nutrition Study cohort from India shows offspring born to vitamin B12-restricted females than controls.
similar findings; in children born to mothers with nutritional Alteration in amino acids levels in liver is more prominent in VR
deficiency (vitamin B12 deficiency among ~70% of mothers), male compared with VR female liver. OCM and arginine
28% of children were glucose intolerant at the age of 18 y. The biosynthesis intermediates also show alteration only in VR male
glucose intolerance was more common in men (37%) compared liver. Elevated BHMT enzyme expression in the VR male

3392
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

offspring from proteomics data and lower levels of betaine in with elevated hepatic TGs only in males, which is reversed by
metabolomics experiments suggest possible utilization of betaine estrogen replacement [78]. Etomoxir treatment in PPARα null
through BHMT pathway to maintain the flux of SAM/SAH under mice with steatosis phenotype causes 100% mortality in males but
vitamin B12 deficient conditions in the male offspring. Inter- only 25% in females. However, 17β-estradiol treatment reduces
estingly, metabolic profiling also shows increased SAM levels in the mortality in males to 20% [79]. We speculate that steroid
both male and female offspring. Increased homocysteine levels hormone biosynthesis alteration in VR offspring liver could
in male offspring can activate the transsulfuration pathway, change the concentration of estrogen, which may lead to
resulting in increased cysteine levels. Earlier reports have linked sex-specific outcomes.
higher homocysteine levels with DNA hypermethylation, which In summary, our study shows that vitamin B12 deficiency
supports our present findings of increased hypermethylation causes a sexually dimorphic dysregulation of the OCM pathway,
signatures in the liver of VR male offspring [68]. Steroid hor- altering DNA methylation pattern, which leads to differential
mones show elevated levels in VR male whereas they are unal- proteomic and metabolic alterations in key metabolic pathways.
tered in VR female. Cortisol, a glucocorticoid associated with These signatures correct to near-normal on vitamin B12 RC, and
stress is also increased in VR male liver. Cortisol regulates blood thus are likely to be causally associated.
glucose level by regulating the release of insulin and glucagon This study has several strengths and a few limitations. The
[69,70]. In the liver, increased cortisol levels can increase major strength is its comprehensive nature, both in terms of in-
gluconeogenesis from nonhexose precursors like pyruvate and clusion of a group of pregnant animals, repleted with vitamin
glycogenic amino acids like α-ketoglutarate, decrease glycogen B12 at conception and a multiomic data analysis approach from
synthesis, and induce insulin resistance and hepatic steatosis liver, the single most important tissue associated with metabolic
[71,72]. Evidence from the literature indicates an association of abnormalities. Overall, these approaches allow identification of
vitamin B12 deficiency with increased stress levels and secretion correlated differential signatures highlighting strong corrobora-
of corticosteroids [62]. In the present study, we show that tion of findings from one to another and propose a causal
upregulation of intermediates of the corticosteroid biosynthesis inference based on reversion of changes upon vitamin B12
pathway and cortisol levels in the liver of male offspring, which repletion. Due to limited availability of blood samples, some
could also be due to impaired glucose metabolism in the males, limitations such as unavailability of data on plasma glucose,
whereas, in females cortisol levels are unchanged. All these ob- glucose tolerance tests, and estrogen levels have crept in, which
servations from body composition, plasma biochemical param- could help emphasize the hormonal regulation of sexually
eter analysis, and omics analysis (DNA methylomics, proteomics, dimorphic phenotypes with MetS-like outcomes only in male
and metabolomics) demonstrate a sexually dimorphic outcome offspring born to vitamin B12-restricted mothers.
in which VR male offspring in the F1 generation show an obese Further, investigations utilizing metabolic tracer techniques
phenotype with metabolic dysregulation, indicative of insulin would be required to understand the detailed mechanisms of
resistance and MetS, whereas VR females present with relatively metabolic dysregulation in offspring born to vitamin B12-
less prominent outcomes. restricted mothers. Studies with vitamin B12 restriction in
Energy homeostasis in liver is tightly controlled by the family PPARα null rodents and application of steroid hormones like
of transcription factors that perform the task of nutrient sensing. estrogen could specifically ascertain the role of sex-ster-
PPARs are ligand-activated nuclear transcription factors that oid–mediated PPAR regulations and consequent sexually
play a key role in nutrient homeostasis [73]. In the current study, dimorphic outcomes as observed in this study. Identifying
we find gene promoter hypermethylation in all the 3 PPAR iso- detailed signaling mechanisms playing a causal role in the
types [PPARα, PPARδ (also known as PPARβ), and PPARγ] in the metabolic abnormalities due to in utero vitamin B12 deficiency
VR male liver, which indicates their lower protein expression. is relevant to future studies.
PPARs control expression of genes involved in carbohydrate,
lipid, lipoprotein, and energy metabolism [74]. PPARα is the Author contributions
predominant PPAR isotype in liver that controls mitochondrial
and peroxisomal β-oxidation pathways, TG metabolism, and The authors’ responsibilities were as follows – PS: per-
fatty acid uptake, especially during fasting [75]. This observa- formed the metabolomics experiment; SV: performed the
tion is in line with our previous study showing PPARα hyper- biochemical analysis and proteomics work; SG (Subhoshree):
methylation in F1 generation 12-mo male offspring under performed the MeDIP experiments and was involved in data
maternal vitamin B12 deficiency and PPAR signaling pathway to analysis; SG (Sourav): was involved in establishing the
be a key modulator of liver proteome in pups born to vitamin MeDIP-Seq analysis pipeline on Wistar rat tissues; VS: pro-
B12 deficient rats [14,15]. vided valuable inputs in MeDIP-Seq data analysis; SKV: hel-
The sexually dimorphic perturbation in metabolic processes ped in troubleshooting and running the sequencing workflow;
and phenotypic outcomes as observed in this study can result from SSB: supervised design of the sequencing experiments and
sex hormones and/or sex-specific developmental programming. provided critical inputs to the study; LK, VJ, PK: were
Genes involved in steroid hormone metabolism like androgen and involved in all the animal experiments; PS, SG, SV, LK: per-
estrogen metabolism have been shown to exhibit PPARα- depen- formed the integrated multiomics data analysis; PS: wrote the
dent sexual dimorphism [76]. Estrogen, a sex-steroid hormone, initial draft of the manuscript; SG, LK: provided critical input
has been shown to inhibit the action of PPARα on obesity and lipid in improving the manuscript; SS, GRC: conceptualized the
metabolism by regulating PPARα-dependent target genes [77]. study and designed the experiments and were also involved
Impaired estrogen synthesis in aromatase-null mice causes inac- in data analysis and writing the manuscript; and all authors:
tivation of estrogen receptors, which induces hepatic steatosis, read and approved the final manuscript.

3393
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

Conflicts of interest [12] S. Ghosh, J.K. Sinha, U.K. Putcha, M. Raghunath, Severe but not
moderate vitamin B12 deficiency impairs lipid profile, induces
adiposity, and leads to adverse gestational outcome in female C57BL/6
The authors report no conflicts of interest. mice, Front. Nutr. 3 (2016) 1, https://doi.org/10.3389/
fnut.2016.00001.
Funding [13] K.A. Kumar, A. Lalitha, U. Reddy, G.R. Chandak, S. SenGupta,
M. Raghunath, Chronic maternal vitamin B12 restriction induced
changes in body composition and glucose metabolism in the Wistar rat
The study was supported by grant no. MLP1804 and offspring are partly correctable by rehabilitation, PLOS ONE 9 (11)
MLP2014 from Council of Scientific and Industrial Research (2014) e112991, https://doi.org/10.1371/journal.pone.0112991.
[14] S. Ahmad, K.A. Kumar, T. Basak, G. Bhardwaj, D.K. Yadav, A. Lalitha, et
(CSIR), India, to SS and GRC. PS and SG (Subhoshree) were al., PPAR signaling pathway is a key modulator of liver proteome in
supported by research fellowships from Council of Scientific and pups born to vitamin B(12) deficient rats, J. Proteomics 91 (2013)
Industrial Research (CSIR), India, and SV was supported by 297–308, https://doi.org/10.1016/j.jprot.2013.07.027.
research fellowships from University Grants Commission, India. [15] V.S. Tanwar, S. Ghosh, S. Sati, S. Ghose, L. Kaur, K.A. Kumar, et al.,
Maternal vitamin B12 deficiency in rats alters DNA methylation in
metabolically important genes in their offspring, Mol. Cell. Biochem.
Data availability 468 (1–2) (2020) 83–96, https://doi.org/10.1007/s11010-020-
03713-x.
[16] R. Carmel, R. Green, D.W. Jacobsen, K. Rasmussen, M. Florea, C. Azen,
All the data supporting the finding is available within the Serum cobalamin, homocysteine, and methylmalonic acid
article and its supplementary materials. Raw data that supports concentrations in a multiethnic elderly population: ethnic and sex
differences in cobalamin and metabolite abnormalities, Am. J. Clin.
the findings of this study are available from the corresponding Nutr. 70 (5) (1999) 904–910, https://doi.org/10.1093/ajcn/70.5.904.
author, upon reasonable request. [17] H.S. Lim, Y.R. Heo, Plasma total homocysteine, folate, and vitamin B12
status in Korean adults, J. Nutr. Sci. Vitaminol. (Tokyo) 48 (4) (2002)
290–297, https://doi.org/10.3177/jnsv.48.290.
Appendix A. Supplementary data [18] S. Azimi, E. Faramarzi, P. Sarbakhsh, A. Ostadrahimi, M.H. Somi,
Supplementary data to this article can be found online at M. Ghayour, Folate and vitamin B12 status and their relation to
https://doi.org/10.1016/j.tjnut.2023.08.032. hematological indices in healthy adults of Iranians: Azar cohort study,
Nutr. Health 25 (1) (2019) 29–36, https://doi.org/10.1177/
0260106018815392.
References [19] R. Xu, F. Huang, Y. Wang, Q. Liu, Y. Lv, Q. Zhang, Gender- and age-
related differences in homocysteine concentration: a cross-sectional
[1] G. Wu, F.W. Bazer, T.A. Cudd, C.J. Meininger, T.E. Spencer, Maternal study of the general population of China, Sci. Rep. 10 (1) (2020) 17401,
nutrition and fetal development, J. Nutr. 134 (9) (2004) 2169–2172, https://doi.org/10.1038/s41598-020-74596-7.
https://doi.org/10.1093/jn/134.9.2169. [20] F. Sadre-Marandi, T. Dahdoul, M.C. Reed, H.F. Nijhout, Sex differences
[2] D.J. Barker, Fetal origins of coronary heart disease, BMJ 311 (6998) in hepatic one-carbon metabolism, BMC Syst. Biol. 12 (1) (2018) 89,
(1995) 171–174, https://doi.org/10.1136/bmj.311.6998.171. https://doi.org/10.1186/s12918-018-0621-7.
[3] D.J. Barker, The origins of the developmental origins theory, J. Intern. [21] R. Kim, H.F. Nijhout, M.C. Reed, One-carbon metabolism during the
Med. 261 (5) (2007) 412–417, https://doi.org/10.1111/j.1365- menstrual cycle and pregnancy, PLOS Comput. Biol. 17 (12) (2021)
2796.2007.01809.x. e1009708, https://doi.org/10.1371/journal.pcbi.1009708.
[4] D.J. Hoffman, R.M. Reynolds, D.B. Hardy, Developmental origins of [22] K.A. Kumar, A. Lalitha, D. Pavithra, I.J. Padmavathi, M. Ganeshan,
health and disease: current knowledge and potential mechanisms, Nutr. K.R. Rao, et al., Maternal dietary folate and/or vitamin B12 restrictions
Rev. 75 (12) (2017) 951–970, https://doi.org/10.1093/nutrit/nux053. alter body composition (adiposity) and lipid metabolism in Wistar rat
[5] M.S. Kramer, R. Kakuma, Energy and protein intake in pregnancy, offspring, J. Nutr. Biochem. 24 (1) (2013) 25–31, https://doi.org/
Cochrane Database Syst. Rev. (4) (2003) CD000032, https://doi.org/ 10.1016/j.jnutbio.2012.01.004.
10.1002/14651858.CD000032, 2003. [23] D.W. Jacobsen, V.J. Gatautis, R. Green, K. Robinson, S.R. Savon,
[6] B. Koletzko, I. Cetin, J.T. Brenna, Perinatal Lipid Intake Working Group, M. Secic, et al., Rapid HPLC determination of total homocysteine and
Child Health Foundation, Diabetic Pregnancy Study Group, et al., other thiols in serum and plasma: sex differences and correlation with
Dietary fat intakes for pregnant and lactating women, Br. J. Nutr. 98 (5) cobalamin and folate concentrations in healthy subjects, Clin. Chem. 40
(2007) 873–877, https://doi.org/10.1017/S0007114507764747. (6) (1994) 873–881, https://doi.org/10.1093/clinchem/40.6.873.
[7] P. Christian, C.P. Stewart, Maternal micronutrient deficiency, fetal [24] S. Sati, V.S. Tanwar, K.A. Kumar, A. Patowary, V. Jain, S. Ghosh, et al.,
development, and the risk of chronic disease, J. Nutr. 140 (3) (2010) High resolution methylome map of rat indicates role of intragenic DNA
437–445, https://doi.org/10.3945/jn.109.116327. methylation in identification of coding region, PLOS ONE 7 (2) (2012)
[8] A. Mahajan, D. Sapehia, S. Thakur, P.S. Mohanraj, R. Bagga, J. Kaur, e31621, https://doi.org/10.1371/journal.pone.0031621.
Effect of imbalance in folate and vitamin B12 in maternal/parental diet [25] T. Basak, A. Bhat, D. Malakar, M. Pillai, S. SenGupta, In-depth
on global methylation and regulatory miRNAs, Sci. Rep. 9 (1) (2019) comparative proteomic analysis of yeast proteome using iTRAQ and
17602, https://doi.org/10.1038/s41598-019-54070-9. SWATH based MS, Mol. Biosyst. 11 (8) (2015) 2135–2143, https://
[9] C.E. Visentin, S.P. Masih, L. Plumptre, T.H. Schroder, K.J. Sohn, A. Ly, et doi.org/10.1039/c5mb00234f.
al., Low serum vitamin B-12 concentrations are prevalent in a cohort of [26] A. Ferrarini, C. Di Poto, S. He, C. Tu, R.S. Varghese, A. Kara Balla, et al.,
pregnant Canadian women, J. Nutr. 146 (5) (2016) 1035–1042, https:// Metabolomic analysis of liver tissues for characterization of
doi.org/10.3945/jn.115.226845. hepatocellular carcinoma, J. Proteome Res. 18 (8) (2019) 3067–3076,
[10] E. Peker, N. Demir, O. Tuncer, L. Üstyol, R. Balahoro glu, S. Kaba, et al., https://doi.org/10.1021/acs.jproteome.9b00185.
The levels of vitamin B12, folate and homocysteine in mothers and their [27] A.M. Bolger, M. Lohse, B. Usadel, Trimmomatic: a flexible trimmer for
babies with neural tube defects, J. Matern. Fetal Neonatal Med. 29 (18) Illumina sequence data, Bioinformatics 30 (15) (2014) 2114–2120,
(2016) 2944–2948, https://doi.org/10.3109/ https://doi.org/10.1093/bioinformatics/btu170.
14767058.2015.1109620. [28] R.M. Kuhn, D. Karolchik, A.S. Zweig, T. Wang, K.E. Smith,
[11] T. Rogne, M.J. Tielemans, M.F. Chong, C.S. Yajnik, G.V. Krishnaveni, K.R. Rosenbloom, et al., The UCSC Genome Browser Database: update
L. Poston, et al., Associations of maternal vitamin B12 concentration in 2009, Nucleic Acids Res 37 (database issue) (2009) D755–D761,
pregnancy with the risks of preterm birth and low birth weight: a https://doi.org/10.1093/nar/gkn875.
systematic review and meta-analysis of individual participant data, Am. [29] B. Langmead, S.L. Salzberg, Fast gapped-read alignment with Bowtie 2,
J. Epidemiol. 185 (3) (2017) 212–223, https://doi.org/10.1093/aje/ Nat. Methods 9 (4) (2012) 357–359, https://doi.org/10.1038/
kww212. nmeth.1923.

3394
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

[30] W.J. Kent, C.W. Sugnet, T.S. Furey, K.M. Roskin, T.H. Pringle, [51] P. Martínez, G. G omez-L
opez, F. García, E. Mercken, S. Mitchell,
A.M. Zahler, et al., The human genome browser at UCSC, Genome Res J.M. Flores, et al., RAP1 protects from obesity through its
12 (6) (2002) 996–1006, https://doi.org/10.1101/gr.229102. extratelomeric role regulating gene expression, Cell Rep 3 (6) (2013)
[31] H. Li, B. Handsaker, A. Wysoker, T. Fennell, J. Ruan, N. Homer, et al., 2059–2074, https://doi.org/10.1016/j.celrep.2013.05.030.
The Sequence Alignment/Map format and SAMtools, Bioinformatics 25 [52] S. Spydevold, E.J. Davis, J. Bremer, Replenishment and depletion of
(16) (2009) 2078–2079, https://doi.org/10.1093/bioinformatics/ citric acid cycle intermediates in skeletal muscle. Indication of pyruvate
btp352. carboxylation, Eur. J. Biochem. 71 (1) (1976) 155–165, https://
[32] J. Feng, T. Liu, Y. Zhang, Using MACS to identify peaks from ChIP-Seq doi.org/10.1111/j.1432-1033.1976.tb11101.x.
data, Curr. Protoc. Bioinformatics 34 (1) (2011) 2, https://doi.org/ [53] I. Martinez-Reyes, N.S. Chandel, Mitochondrial TCA cycle metabolites
10.1002/0471250953.bi0214s34, 14.1–2.14.14. control physiology and disease, Nat. Commun. 11 (1) (2020) 102,
[33] J.P. Thomson, R. Ottaviano, R. Buesen, J.G. Moggs, M. Schwarz, https://doi.org/10.1038/s41467-019-13668-3.
R.R. Meehan, Defining baseline epigenetic landscapes in the rat liver, [54] N. Frery, G. Huel, M. Leroy, T. Moreau, R. Savard, P. Blot, et al., Vitamin
Epigenomics 9 (12) (2017) 1503–1527, https://doi.org/10.2217/epi- B12 among parturients and their newborns and its relationship with
2017-0029. birthweight, Eur. J. Obstet. Gynecol. Reprod. Biol. 45 (3) (1992)
[34] W. da Huang, B.T. Sherman, R.A. Lempicki, Systematic and integrative 155–163, https://doi.org/10.1016/0028-2243(92)90076-b.
analysis of large gene lists using DAVID bioinformatics resources, Nat. [55] B. Lindblad, S. Zaman, A. Malik, H. Martin, A.M. Ekstr€ om, S. Amu, et al.,
Protoc. 4 (1) (2009) 44–57, https://doi.org/10.1038/nprot.2008.211. Folate, vitamin B12, and homocysteine levels in South Asian women
[35] B.T. Sherman, M. Hao, J. Qiu, X. Jiao, M.W. Baseler, H.C. Lane, et al., with growth-retarded fetuses, Acta Obstet. Gynecol. Scand. 84 (11)
DAVID: a web server for functional enrichment analysis and functional (2005) 1055–1061, https://doi.org/10.1111/j.0001-
annotation of gene lists (2021 update), Nucleic Acids Res 50 (W1) 6349.2005.00876.x.
(2022), https://doi.org/10.1093/nar/gkac194. W216–W221. [56] S. Muthayya, A.V. Kurpad, C.P. Duggan, R.J. Bosch, P. Dwarkanath,
[36] The UniProt Consortium, UniProt: the universal protein knowledgebase, A. Mhaskar, et al., Low maternal vitamin B12 status is associated with
Nucleic Acids Res 46 (5) (2018) 2699, https://doi.org/10.1093/nar/ intrauterine growth retardation in urban South Indians, Eur. J. Clin.
gky092. Nutr. 60 (6) (2006) 791–801, https://doi.org/10.1038/
[37] Y. Zhang, M. Ou, H. Lin, L. Lai, H. Chen, J. Chen, et al., Proteomic sj.ejcn.1602383.
analysis of differentially expressed proteins in the serum of patients [57] C.S. Yajnik, S.S. Deshpande, A.V. Panchanadikar, S.S. Naik,
with acute renal allograft rejection using iTRAQ labelling technology, J.A. Deshpande, K.J. Coyaji, et al., Maternal total homocysteine
Mol. Med. Rep. 22 (3) (2020) 2329–2341, https://doi.org/10.3892/ concentration and neonatal size in India, Asia Pac, J. Clin. Nutr. 14 (2)
mmr.2020.11299. (2005) 179–181.
[38] Y. Cheng, Q. Meng, L. Huang, X. Shi, J. Hou, X. Li, et al., iTRAQ-based [58] R.V. Behere, A.S. Deshmukh, S. Otiv, M.D. Gupte, C.S. Yajnik, Maternal
quantitative proteomic analysis and bioinformatics study of proteins in vitamin B12 status during pregnancy and its association with outcomes
retinoblastoma, Oncol. Lett. 14 (6) (2017) 8084–8091, https://doi.org/ of pregnancy and health of the offspring: a systematic review and
10.3892/ol.2017.7221. implications for policy in India, Front. Endocrinol. (Lausanne) 12
[39] P. SenGupta, The laboratory rat: relating its age with human’s, Int. J. (2021) 619176, https://doi.org/10.3389/fendo.2021.619176.
Prev. Med. 4 (6) (2013) 624–630. [59] U. Hübner, A. Alwan, M. Jouma, M. Tabbaa, H. Schorr, W. Herrmann,
[40] P. Sharma, J. Kumar, G. Garg, A. Kumar, A. Patowary, G. Karthikeyan, Low serum vitamin B12 is associated with recurrent pregnancy loss in
et al., Detection of altered global DNA methylation in coronary artery Syrian women, Clin. Chem. Lab. Med. 46 (9) (2008) 1265–1269,
disease patients, DNA Cell Biol 27 (7) (2008) 357–365, https://doi.org/ https://doi.org/10.1515/CCLM.2008.247.
10.1089/dna.2007.0694. [60] J.G. Ray, P.R. Wyatt, M.D. Thompson, M.J. Vermeulen, C. Meier,
[41] A.T. Nguyen-Lefebvre, N. Selzner, J.L. Wrana, M. Bhat, The hippo P.Y. Wong, et al., Vitamin B12 and the risk of neural tube defects in a
pathway: a master regulator of liver metabolism, regeneration, and folic-acid-fortified population, Epidemiology 18 (3) (2007) 362–366,
disease, FASEB J 35 (5) (2021) e21570, https://doi.org/10.1096/ https://doi.org/10.1097/01.ede.0000257063.77411.e9.
fj.202002284RR. [61] C.S. Yajnik, S. Bandopadhyay, A. Bhalerao, D.S. Bhat, S.B. Phatak,
[42] Y. Liu, X. Wang, Y. Yang, Hepatic Hippo signaling inhibits development R.H. Wagh, et al., Poor in utero growth, and reduced beta-cell
of hepatocellular carcinoma, Clin. Mol. Hepatol. 26 (4) (2020) compensation and high fasting glucose from childhood, are harbingers
742–750, https://doi.org/10.3350/cmh.2020.0178. of glucose intolerance in young Indians, Diabetes Care 44 (12) (2021)
[43] W. Link, P.J. Fernandez-Marcos, FOXO transcription factors at the 2747–2757, https://doi.org/10.2337/dc20-3026.
interface of metabolism and cancer, Int. J. Cancer 141 (12) (2017) [62] G.V. Krishnaveni, S.R. Veena, M. Johnson, K. Kumaran, A. Jones,
2379–2391, https://doi.org/10.1002/ijc.30840. D.S. Bhat, et al., Maternal B12, folate and homocysteine concentrations
[44] A. Lawan, A.M. Bennett, Mitogen-activated protein kinase regulation in and offspring cortisol and cardiovascular responses to stress, J. Clin.
hepatic metabolism, Trends Endocrinol. Metab. 28 (12) (2017) Endocrinol. Metab. 105 (7) (2020) e2591–e2599, https://doi.org/
868–878, https://doi.org/10.1016/j.tem.2017.10.007. 10.1210/clinem/dgz114.
[45] W. Wang, Z.D. Xiao, X. Li, K.E. Aziz, B. Gan, R.L. Johnson, et al., AMPK [63] D.S. Goodsell, The molecular perspective: the ras oncogene, Oncologist
modulates Hippo pathway activity to regulate energy homeostasis, 4 (3) (1999) 263–264, https://doi.org/10.1634/theoncologist.4-3-263.
Nat. Cell Biol. 17 (4) (2015) 490–499, https://doi.org/10.1038/ [64] I. Ferrara-Romeo, P. Martínez, M.A. Blasco, Mice lacking RAP1 show
ncb3113. early onset and higher rates of DEN-induced hepatocellular carcinomas
[46] J.S. Mo, Z. Meng, Y.C. Kim, H.W. Park, C.G. Hansen, S. Kim, et al., in female mice, PLOS ONE 13 (10) (2018) e0204909, https://doi.org/
Cellular energy stress induces AMPK-mediated regulation of YAP and 10.1371/journal.pone.0204909.
the Hippo pathway, Nat. Cell Biol. 17 (4) (2015) 500–510, https:// [65] C.K. Looi, L.W. Hii, S.C. Ngai, C.O. Leong, C.W. Mai, The role of Ras-
doi.org/10.1038/ncb3111. associated protein 1 (Rap1) in cancer: bad actor or good player?
[47] P. Lefebvre, G. Chinetti, J.C. Fruchart, B. Staels, Sorting out the roles of Biomedicines 8 (9) (2020) 334, https://doi.org/10.3390/
PPAR alpha in energy metabolism and vascular homeostasis, J. Clin. biomedicines8090334.
Invest. 116 (3) (2006) 571–580, https://doi.org/10.1172/JCI27989. [66] N.A. Babenko, V.S. Kharchenko, Modulation of insulin sensitivity of
[48] A. Peeters, M. Baes, Role of PPARalpha in hepatic carbohydrate hepatocytes by the pharmacological downregulation of phospholipase
metabolism, PPAR Res (2010) 572405, https://doi.org/10.1155/2010/ D, Int. J. Endocrinol. (2015) 794838, https://doi.org/10.1155/2015/
572405, 2010. 794838, 2015.
[49] M. Pawlak, P. Lefebvre, B. Staels, Molecular mechanism of PPARalpha [67] Z. Yang, K. Roth, M. Agarwal, W. Liu, M.C. Petriello, The transcription
action and its impact on lipid metabolism, inflammation and fibrosis in factors CREBH, PPARa, and FOXO1 as critical hepatic mediators of diet-
non-alcoholic fatty liver disease, J. Hepatol. 62 (3) (2015) 720–733, induced metabolic dysregulation, J. Nutr. Biochem. 95 (2021) 108633,
https://doi.org/10.1016/j.jhep.2014.10.039. https://doi.org/10.1016/j.jnutbio.2021.108633.
[50] D. Eberle, B. Hegarty, P. Bossard, P. Ferre, F. Foufelle, SREBP [68] D. B€onsch, B. Lenz, U. Reulbach, J. Kornhuber, S. Bleich, Homocysteine
transcription factors: master regulators of lipid homeostasis, Biochimie associated genomic DNA hypermethylation in patients with chronic
86 (11) (2004) 839–848, https://doi.org/10.1016/ alcoholism, J. Neural Transm. (Vienna) 111 (12) (2004) 1611–1616,
j.biochi.2004.09.018. https://doi.org/10.1007/s00702-004-0232-x.

3395
P. Singh et al. The Journal of Nutrition 153 (2023) 3382–3396

[69] J.K. Wise, R. Hendler, P. Felig, Influence of glucocorticoids on glucagon [75] L.M. Sanderson, M.V. Boekschoten, B. Desvergne, M. Müller, S. Kersten,
secretion and plasma amino acid concentrations in man, J. Clin. Invest. Transcriptional profiling reveals divergent roles of PPARalpha and
52 (11) (1973) 2774–2782, https://doi.org/10.1172/JCI107473. PPARbeta/delta in regulation of gene expression in mouse liver,
[70] A. Rafacho, L.M. Gonçalves-Neto, J.C. Santos-Silva, P. Alonso-Magdalena, Physiol. Genomics 41 (1) (2010) 42–52, https://doi.org/10.1152/
B. Merino, S.R. Taboga, et al., Pancreatic alpha-cell dysfunction physiolgenomics.00127.2009.
contributes to the disruption of glucose homeostasis and compensatory [76] N. Leuenberger, S. Pradervand, W. Wahli, SUMOylated PPARalpha
insulin hypersecretion in glucocorticoid-treated rats, PLOS ONE 9 (4) mediates sex-specific gene repression and protects the liver from
(2014) e93531, https://doi.org/10.1371/journal.pone.0093531. estrogen-induced toxicity in mice, J. Clin. Invest. 119 (10) (2009)
[71] T. Kuo, A. McQueen, T.C. Chen, J.C. Wang, Regulation of glucose 3138–3148, https://doi.org/10.1172/JCI39019.
homeostasis by glucocorticoids, Adv. Exp. Med. Biol. 872 (2015) [77] M. Yoon, The role of PPARalpha in lipid metabolism and obesity: focusing
99–126, https://doi.org/10.1007/978-1-4939-2895-8_5. on the effects of estrogen on PPARalpha actions, Pharmacol. Res. 60 (3)
[72] E.B. Geer, J. Islam, C. Buettner, Mechanisms of glucocorticoid-induced (2009) 151–159, https://doi.org/10.1016/j.phrs.2009.02.004.
insulin resistance: focus on adipose tissue function and lipid [78] K.N. Hewitt, K. Pratis, M.E. Jones, E.R. Simpson, Estrogen replacement
metabolism, Endocrinol. Metab. Clin. North Am. 43 (1) (2014) 75–102, reverses the hepatic steatosis phenotype in the male aromatase
https://doi.org/10.1016/j.ecl.2013.10.005. knockout mouse, Endocrinology 145 (4) (2004) 1842–1848, https://
[73] S. Kersten, B. Desvergne, W. Wahli, Roles of PPARs in health and doi.org/10.1210/en.2003-1369.
disease, Nature 405 (6785) (2000) 421–424, https://doi.org/10.1038/ [79] F. Djouadi, C.J. Weinheimer, J.E. Saffitz, C. Pitchford, J. Bastin,
35013000. F.J. Gonzalez, et al., A gender-related defect in lipid metabolism and
[74] Y. Kidani, S.J. Bensinger, Liver X receptor and peroxisome proliferator- glucose homeostasis in peroxisome proliferator- activated receptor
activated receptor as integrators of lipid homeostasis and immunity, alpha- deficient mice, J. Clin. Invest. 102 (6) (1998) 1083–1091,
Immunol. Rev. 249 (1) (2012) 72–83, https://doi.org/10.1111/j.1600- https://doi.org/10.1172/JCI3949.
065X.2012.01153.x.

3396

You might also like