Skip to main content
PLOS One logoLink to PLOS One
. 2024 May 16;19(5):e0303528. doi: 10.1371/journal.pone.0303528

Analysis of arsenic-modulated expression of hypothalamic estrogen receptor, thyroid receptor, and peroxisome proliferator-activated receptor gamma mRNA and simultaneous mitochondrial morphology and respiration rates in the mouse

Daiana Alymbaeva 1, Csaba Szabo 2, Gergely Jocsak 1,*, Tibor Bartha 1, Attila Zsarnovszky 1,2,3, Csaba Kovago 4, Silvia Ondrasovicova 5, David Sandor Kiss 1
Editor: Abeer El Wakil6
PMCID: PMC11098319  PMID: 38753618

Abstract

Arsenic has been identified as an environmental toxicant acting through various mechanisms, including the disruption of endocrine pathways. The present study assessed the ability of a single intraperitoneal injection of arsenic, to modify the mRNA expression levels of estrogen- and thyroid hormone receptors (ERα,β; TRα,β) and peroxisome proliferator-activated receptor gamma (PPARγ) in hypothalamic tissue homogenates of prepubertal mice in vivo. Mitochondrial respiration (MRR) was also measured, and the corresponding mitochondrial ultrastructure was analyzed. Results show that ERα,β, and TRα expression was significantly increased by arsenic, in all concentrations examined. In contrast, TRβ and PPARγ remained unaffected after arsenic injection. Arsenic-induced dose-dependent changes in state 4 mitochondrial respiration (St4). Mitochondrial morphology was affected by arsenic in that the 5 mg dose increased the size but decreased the number of mitochondria in agouti-related protein- (AgRP), while increasing the size without affecting the number of mitochondria in pro-opiomelanocortin (POMC) neurons. Arsenic also increased the size of the mitochondrial matrix per host mitochondrion. Complex analysis of dose-dependent response patterns between receptor mRNA, mitochondrial morphology, and mitochondrial respiration in the neuroendocrine hypothalamus suggests that instant arsenic effects on receptor mRNAs may not be directly reflected in St3-4 values, however, mitochondrial dynamics is affected, which predicts more pronounced effects in hypothalamus-regulated homeostatic processes after long-term arsenic exposure.

1. Introduction

Based on the latest analysis conducted by the Agency for Toxic Substances and Disease Registry (ATSDR), and the Environmental Protection Agency, which accounts for factors such as substance toxicity, potential human exposure, and frequency of occurrence, arsenic (As) has been identified as the most hazardous substance. This recent (2022) ranking is documented in their priority list, accessible at https://www.atsdr.cdc.gov/spl/index.html#2019spl. The issue of arsenic poisoning has become increasingly prevalent in many developing nations, often stemming from contamination of drinking water sources due to natural environmental factors, such as seepage into aquifers, mining activities, and industrial operations [1]. Globally, more than 150 million people are impacted by arsenic-contaminated drinking water, posing a substantial public health concern due to the chronic nature of exposure [2]. Both human population studies and animal research have consistently demonstrated that chronic exposure to As leads to cognitive impairment [3, 4].

Many of the environmental toxicants act as agonists or antagonists of certain hormones (mainly estrogens and/or thyroid hormones [511]); interfering with the physiological regulation of the homeostatic system. These chemicals have been termed endocrine disruptors (EDs) [12].

In general, within the frame of ED-modulated intracellular hormone actions, there are two major and fundamentally interrelated types of intracellular mechanisms. №.1.: regulatory pathways that regulate physiological balance, optimizing responses to the environment [1316]; №.2.: adjustment of cellular energy expenditure to homeostatic processes [1719].

Emerging evidence suggests that As exposure disrupts the normal functioning of the endocrine system, influencing some intricate regulatory processes related to hormone synthesis and secretion [20]. Given their integral roles in orchestrating diverse physiological processes, the thorough investigation of the thyroid and estrogen systems is of fundamental importance in the assessment of a substance demonstrating potential endocrine-disrupting properties [2123]. Apart from these “traditional” hormone receptors, some studies highlight that the adipogenesis activators like peroxisome proliferator‐activated receptor gamma (PPARγ), a crucial regulator of energy metabolism [24, 25], is also sensitive to As exposure [26] that raises the question of whether PPARγ is also a potential target for endocrine disruptors [27].

While the toxicological impact of As is widely acknowledged in various aspects, the mechanisms through which As elicits its endocrine disrupting effects on the above hormonal systems, remain largely unknown. Nevertheless, our earlier studies have already demonstrated some aspects of its disrupting potential in in vitro conditions [2831] however, until now it remained questionable whether these phenomena may also manifest in vivo.

Located in the basal region of the brain, the hypothalamus serves as a key regulator of the above-mentioned hormonal systems, functioning as an intricate network comprising specialized neurosecretory cells [32]. Receiving a myriad of external and internal signals through hypothalamus-pituitary-end-organ axes [33, 34], these cells compose the major regulatory center for homeostatic processes [35]. Among the numerous hypothalamic nuclei that integrate the processes related to energy balance, nutrient intake, and feeding behavior, the arcuate nucleus (ARC) receives special attention in our present research since it is positioned at a region with higher local permeability in the blood-brain barrier, making it readily exposed to circulating factors, such as chemicals expressing endocrine disrupting capability [3639]. ARC neurons, particularly those expressing proopiomelanocortin (POMC; termed as “satiety neurons”), and agouti-related peptide (AgRP; so-called “hunger cells”) not only play a vital role in sensing the overall energy status of the organism as key members of the melanocortin system but also integrate signals from both central and peripheral pathways [4042].

Mitochondria in somatic cells typically serve general cellular metabolic functions, producing ATP, while–quite interestingly–mitochondria located in AgRP and POMC neurons are suggested to play a specific role in regulating the energy balance of the whole organism through dynamic changes in their activity that are believed to directly mimic and modulate their hosting cells’ function; i.e. they act as energy sensors of the hosting neurons [4345]. Regarding the context between hormone receptors and cellular energy management, the influence of ER, TR, and PPAR on mitochondria is unequivocal, manifesting through both direct and indirect pathways across diverse cellular contexts [4649].

These effects encompass interactions with nuclear and extranuclear receptors, as well as non-genomic interactions with other organelles and intracellular events. Given the heightened presence of ER and TR within the hypothalamus, coupled with their capacity to traverse the blood-brain barrier and undergo local synthesis within the brain, it is crucial to comprehend the individual and collective impact of ER and TR on mitochondrial function [50, 51]. Considering the distinctive roles played by AgRP- and POMC-associated mitochondria and their profound impact on the overall organism, it becomes apparent that EDs acting upon these hormone receptor pathways possess the capacity to modulate an extensive array of systemic mechanisms. This modulation may result in altered physiological functions or even contribute to the onset of different diseases.

We hypothesize that As can act as an endocrine disruptor in low doses and can also in vivo modulate the expression of different hormone receptors (ERα,β, TRα,β and PPARγ) within the hypothalamus. Coupled with that, different intracellular pathways are suggested to be affected leading to altered function of organelles, most prominently those including mitochondria, which–in the case of AgRP and POMC cells–are remarkably sensitive to circulating factors, like hormone-mimicking chemicals. Therefore, beyond the above we investigated the mitochondrial respiration rates (MRR), a generally accepted parameter for the assessment of the intensity of mitochondrial metabolism, moreover, we examined the mitochondrial morphology in AgRP and POMC cells to offer insights into the broader mechanisms governing metabolic regulation.

In determining our arsenic dosage, we referenced several key studies. The selection of the quantities of hormones and EDs, specifically 40 μg, 5 mg, and 10 mg, was based on several factors. Firstly, these dosages were chosen according to the sensitivity of the applied neuronal cell culture system and our previous experiments where the most effective dosage in proving ED effects was determined [52, 53]. These quantities represent varying levels of arsenic exposure, reflecting different degrees of environmentally relevant concentrations of 40 μg, 5 mg, and 10 mg were chosen to represent varying exposure levels, with 40 μg falling within WHO limits (permissible limit of 10–50 μg) [54]. Chang et al. (2007) [55] noted reproductive effects in mice at 20–40 mg/l sodium arsenite exposure. Jana et al. (2006) [56] found neuroendocrine effects in rats at 5 mg/kg/day sodium arsenite. Moreover, Stump et al. (1999) [57] administered intraperitoneal doses (0, 5, 10, 20, 35 mg/kg) during in-utero development, observing the effects of As exposure during critical developmental periods. On the other hand, the concentrations of 5 mg and 10 mg, as cited in studies by Smedley and Kinniburgh, (2012) [58] and Shankar, (2014) [59], respectively, demonstrate higher exposure levels that exceed permissible limits. Our approach to determining intervention time in our animal model was based on insights from our previous comprehensive pilot study [28]. Initially, we conducted in vitro experiments assessing TR and ER receptor expressions with their ligands using cerebellar granule cells. We maintained a consistent intervention time of 6 hours for PCR analysis to ensure reliable results, aligning with the peak expression period observed, we also noted a secondary peak for Western blot analysis around 18 hours. Subsequently, we refined these time windows through further in vitro experiments examining hormone receptor expressions post-exposure to endocrine disruptors [30, 31, 60]. By systematically progressing through these stages and aligning intervention times across in vitro and in vivo experiments, we aimed to ensure consistency and reliability in our findings while effectively investigating the effects of arsenic exposure on hormone receptor expression in the hypothalamus.

We believe that exposure to elevated levels of As may exert modulatory effects on homeostatic functions by disrupting the integrity of the hypothalamic melanocortin system. This perturbation has the potential to induce enduring physiological consequences, including but not limited to conditions such as diabetes, accelerated aging, and cognitive impairments (Fig 1).

Fig 1. The hypothesized mechanisms underlying arsenic-induced endocrine disruption, emphasizing its potential impact on the primary endocrine center.

Fig 1

The melanocortin system within the hypothalamus emerges as a key target of arsenic, with adverse effects anticipated across diverse cellular processes, including mitochondrial respiration and morphology, as well as hormonal signaling pathways. Persistent dysfunction of the melanocortin system is implicated in the etiology of various systemic disorders, such as metabolic syndromes, reproductive dysfunction, mood disorders, and neurodegenerative diseases.

2. Materials and methods

2.1. Animals and treatments

18-day-old C57BL/6 mice were used for this study, (purchased from HAS Biological Research Centre, Szeged, Hungary). Since pilot studies showed no differences between the examined parameters in the two sexes, and we found no indication in the relevant literature for hypothalamic gender differences in this respect, pups of both sexes were used (weighing 9–10 g, n = 6 per treatment group, separately for PCR, MRR and electron microscopic stereological measurements). The age chosen represents a pre-pubertal state when the hypothalamus is not yet sexually active, however, being just before the onset of reproductive life, it is responsive to sexual steroids. This period marks a crucial transitional phase, highlighted by Sengupta P. et al., (2017 a,b) [61, 62] and Sengupta T. et al., (2021) [63], and Brydges N. (2016) [64]. Rebuli and Patisaul, (2016) [65] also emphasized the significant impacts of EDs on the hypothalamus during this developmental stage. While behavioral effects of EDs have been extensively studied, neural changes in pre-pubertal animals have received comparatively less attention.

Animals were kept under a 12/12-h light/dark cycle illumination program. Animals were fed with regular chow (vendor: FarmerMix Kft., Zsambek, Hungary) and ad libitum tap water. All experimental procedures were conducted at the University of Veterinary Medicine (Budapest, Hungary) in accordance to ARRIVE guidelines and EU Directive 2010/63/EU, and was reviewed and approved by the Animal Health and Animal Welfare Directorate of the National Food Chain Safety Office (permit no.: XIV-I-001/2202-4/2012, PEI/001/665-8/2015 and PE/EA/1252-6/2016), as well conformed to ARRIVE guidelines and EU Directive 2010/63/EU.

Animals were sorted into experimental groups according to doses administered as mentioned below, plus a control group injected with the vehicle only (non-treated control, ntC). Hypothalamic samples were measured for the following parameters: mRNA expression level of ERα, ERβ, TRα TRβ, PPARγ.

Six hours before sampling, animals received a single intraperitoneal injection of sodium (meta)arsenite (arsenic, As; purity: ≥90%; CAS: 7784-46-5), purchased from Sigma-Aldrich (St. Louis, MO, USA). Injections were given in three different doses: 40 μg, 5 mg and 10 mg As per 1000 g body weight, dissolved in 0.9% NaCl + dimethyl sulfoxide (0.1%) and DMSO solution. Non-treated controls (ntC) were injected with the solvent as vehicle only.

2.2. Sampling

2.2.1. Sampling for tissue homogenates

Sampling was performed through quick guillotine decapitation under deep isoflurane narcosis 6 hours after the treatments, as described earlier [28, 29, 53]. Removal of the hypothalamic tissue followed the anatomical protocol previously described [66]. Briefly, hypothalami were dissected from the removed brains as follows: in anterio-posterior direction: between the caudal margin of the optic chiasm and the rostral margin of the mamillary body and in dorsoventral direction: below the upper margin of the fornix. Both sides of the hypothalami were used for preparing the tissue homogenates (although we have taken measures to develop a suitable method for the measurement of mitochondrial respiration rates, in relatively small samples [67], still, using only half of the mouse hypothalamus provided a sample size too small to reliably obtain MRR data). Samples were then used for the determination of the level of receptor mRNAs and mitochondrial respiration rates, as detailed below.

2.2.2. Sampling for electron microscopy

Anesthetized animals (details above) were perfused through the left cardiac ventricle, first with 50 ml of 0.9% NaCl, followed by 250 ml of 4% paraformaldehyde and 0.5% glutaraldehyde in 0.1 M phosphate buffer (PB), pH 7.4. After perfusion, the isolated brains were placed in 4°C glutaraldehyde-free fixative (4% paraformaldehyde) for an additional 3 h. Samples were prepared for immunohistochemistry (IHC) by cutting the isolated brain blocks into 50-μm-thick sections (this thickness ensures optimal tissue morphology preservation and staining efficiency. Specifically, when employing 4% paraformaldehyde fixation, sections within the 20–50 μm range facilitate efficient antibody penetration and antigen retrieval).

2.3. Quantitative-RT-PCR

Total RNAs were isolated from mouse hypothalami using TRI reagent following the manufacturer’s protocol (Invitrogen, Carlsbad, CA, USA, #AM9738) and purified from samples with the Direct-zol RNA miniprep kit (Zymo Research, Irvine, CA, USA, #R2051). RNA levels were determined spectrophotometrically (NanoDrop™ ND-1000, Wilmington, NC, USA) at 260–280 nm absorption. 3 μg/μL of total RNA was reverse transcribed by RT-PCR (Amplitron II., Barnstead/Thermolyne, Dubuque, IA, USA) using M-MLV reverse transcriptase (Promega Corporation, Wisconsin, USA, #M1701), oligo (dt) primers and dNTPmix.

Subsequently, 2 μL of the resulting cDNA samples were analyzed by qRT-PCR (Master SYBRGreen, F. Hoffmann-La Roche, Basel, Switzerland) in triplicates, in a LightCycler 2.0 device (F. Hoffmann-La Roche, Basel, Switzerland). To evaluate qPCR reactions, constitutively expressed genes β-actin (ACTB), beta-glucuronidase (GUSB), glyceraldehyde 3-phosphate dehydrogenase (GAPDH), β-2-microglobulin (B2M) and transferrin receptor protein 1 (TFRC) were evaluated for stability. Transcription levels of the genes under investigation were normalized using the expression values of GAPDH, which was identified as the most consistently expressed gene across the experimental conditions, as determined by the GeNorm and NormFinder algorithms. These algorithms were accessed through the online platform RefFinder [68]. Primer pairs were designed using NCBI’s primer designer Primer-BLAST, or were taken from literature, and used at 2 μM concentration. The primer sequences used for GAPDH, ERα, ERβ, TRα, TRβ and PPARγ are given in Jocsak et al., 2019 [29], (please see Table 1). qPCR cycles and controls were planned according to the manufacturer’s instructions and were optimized for the primer pair as described by Jocsak et al., 2016 [31]. Real-time PCR threshold cycle (Ct) data were analyzed using the REST-XL software version 2.0 (GenEx-BioMcc, TUM, München, Germany) [69]. Cycle threshold values were normalized to those of GAPDH. The relative expression ratios of mRNA (fold changes) were calculated using the 2-ΔΔCt method.

Table 1. Primer sequences used for qRT-PCR analysis.

Target gene (mouse) Primer Sequence 5’–3’
ERα Forw. GGA ACT GTC TGC CCA TCG TT
Rev. GAA CCC AGG GCT GCC TTA C
ERβ Forw. AAC CTT CCT CTT GGG CAT CG
Rev. TTT CAT CCG GTT CTC CCA CC
TRα Forw. ACC GCA AAC ACA ACAT TCC G
Rev. GGG CCA GCC TCA GCT AAT AA
TRβ Forw. CGA GGC CAG CTG AAA AAT GG
Rev. CTC AGC ACA CTC ACC TGA AGA
PPARγ Forw. TTGGTGGGATTGTGTCTCGG
Rev. GGCCAAGATCTCACAGTGCT
GAPDH Forw. TGA AAT GTG CAC GCA CCA AG
Rev. GGG AAG CAG CAT TCA GGT CT

2.4. Mitochondria metabolism

2.4.1. Isolation of mitochondria

Hypothalamic samples were prepared for measurement of oxygen consumption as described by Kiss et al., 2016 [69]. Briefly, unit amounts of hypothalamic tissue homogenates (in isolation buffer) were used for mitochondrial purification by a Percoll gradient fractionation. Mitochondrial oxygen-consumption was measured by a Clark-type oxygen electrode (Oxytherm, Hansatech Instruments, Norfolk, UK) at 37°C. Measured values represent the mitochondrial respiration rate (MRR, given in consumed nmol O2 per ml of final volume in one minute). Mitochondrial sample homogenates included both the left and right sides of the hypothalami (i.e., the left and right sides were not separately processed and measured).

2.4.2. Definition of mitochondrial respiration states

As the name and numeral marking of different mitochondrial respiration states varies in the relevant literature, subsequently we explain our nomenclature as used in the present study. Explanation of mitochondrial respiration states as sequentially measured (60 seconds for each respiration state (please also see: [66]).

First step: the mitochondrial oxygen consumption was measured in respiration buffer only, without the addition of any substrates that may affect mitochondrial respiration. Under such conditions, oxygen consumption per unit time depends on the actual metabolic state of the hypothalamic sample and the sample’s original oxygen supply. We termed this experimental setup as state 1 mitochondrial respiration (St1).

Second step: to fuel the Krebs cycle, 5 μL pyruvate (P; comprising the following mixture: 275 mg pyruvate/5 mL distilled water + 100 μL 1 mol/L HEPES) and 2.5 μL malate (M; comprised the following mixture: 335.25 mg malate/5 mL distilled water +100 μL 1 mol/L HEPES) were added to the sample. Under such conditions, the Krebs cycle intensifies, and oxygen consumption increases due to consequential facilitation of the terminal oxidation and oxidative phosphorylation, if the prior (in vivo) blood/oxygen supply of the hypothalamic tissue was sufficient and downregulating mechanisms were not active. We termed this experimental setup as state 2 mitochondrial respiration (St2).

Third step: adenosine diphosphate (ADP; comprising of the following mixture: 64.1 mg ADP/5 mL distilled water + 100 μL 1 mol/L HEPES) of 2.5 μL was added to the sample. Since ADP is a major upregulator of mitochondrial respiration, under such conditions MRR increases if the prior (in vivo) blood/fuel supply of the hypothalamic tissue was sufficient. We termed this experimental setup as (ADP-dependent) state 3 mitochondrial respiration (St3).

Fourth step: oligomycin (comprised of the following mixture: 1 mg oligomycin/1 mL ethanol) of 1 μL was added to the sample. Oligomycin is an ATP-synthase blocker, therefore, it inhibits the oxidative phosphorylation (ATP synthesis), while terminal oxidation continues. Under such conditions, oxygen consumption depends on the actual uncoupled stage and alternative oxidation in mitochondria. Uncoupling and alternative oxidation play important roles in transient downregulation of ATP biosynthesis when cellular energy needs drop. Therefore, increased oxygen consumption in this case refers to the decline of a process (that was previously upregulated) or the attempt by the mitochondrion to downregulate ATP synthesis. We termed this experimental setup as state 4 mitochondrial respiration (St4).

Fifth step: carbonylcyanide-4-(trifluoromethoxy)-phenyl-hydrazone (FCCP; comprised the following mixture: 1.271 mg FCCP/5 mL dimethyl sulfoxide) of 2.5 μL was added to the sample. The FCCP is a cyanide derivative; therefore, by binding to, and blocking cytochrome C oxidase, it depletes all remaining oxygen from the sample. A decrease in oxygen level under such conditions depends on the actual/initial (in vivo) metabolic state of the sampled tissue and the amount of oxygen consumed during St1-4. Thus, the total amount of oxygen consumed in St1-4 plus the amount of remaining oxygen depleted by FCCP gives good reference to the blood/oxygen supply of the tissue at the time of the animal’s sacrifice. Therefore, this experimental setup is also known as total mitochondrial respiratory capacity, hereby referred to as state 5 mitochondrial respiration (St5).

Although all 5 mitochondrial respiration states (as explained above) were sequentially measured and evaluated, only MRR from state 3 (St3) and state 4 (St4) mitochondrial respiration values are presented in this study, since these MRR parameters are most appropriate to characterize the intensity of mitochondrial/tissue metabolism. St3 gives a plausible insight into mitochondrial metabolism, since the ADP/ATP ratio potently regulates mitochondrial activity, while St4 indicates the degree of uncoupling and the activity of alternative oxidases, two factors that play an important role in transient down-regulation of ATP biosynthesis when cellular energy needs drop.

2.5. Immunocytochemistry & electron microscopy

50 μm thick tissue slices [70] were rinsed in 1% sodium borohydride in PB for 15 min to eliminate unbound aldehydes. The primary antibodies used for labelling included a polyclonal primary POMC (Invitrogen PA5-18368, 1:1500, incubated overnight) and AgRP antibodies (Invitrogen PA5-78739, 1:200, incubated overnight). As a secondary antibody a biotinylated polyclonal goat anti-rabbit antibody was applied (Invitrogen 65–6140, 1:200, incubated for 1 hr.). Finally, ABC kits (avidin-biotin complex kits, # Pk.4001) and DAB (diaminobenzidine, CAS: 91-95-2) were used for development.

For electron microscopic analysis, sections were osmicated (1% OsO4 in PB) for 30 min, dehydrated through increasing ethanol concentrations, and embedded into Durcupan ACM resin (Sigma-Aldrich, 44610). Following embedding, ultrathin sections with a thickness of 50 nm were cut for precise examination, (see above the localization of the area in question).

The prepared sections were examined with a JEOL-1011 TEM (JEOL USA, Peabody, MA, USA). The calibrated electron micrographs were analyzed using the NIH Image J (ver. 1.52n) software. The measured parameters included the occurrence (number per area) and size of mitochondria, matrix/entire mitochondrion ratio [71].

2.6. Data analysis

In this study, statistical analysis was conducted using GraphPad Prism version 9 (GraphPad Software, San Diego, CA, USA) and carried out according to the guidance of the Department of Biomathematics, University of Veterinary Medicine, Budapest, Hungary.

Prior to analysis, the normality of the data distribution was assessed using the Shapiro-Wilk test. As data followed a normal distribution in the data sets, one-way ANOVA was performed followed by Bonferroni or Dunnett’s test for post hoc comparisons, for PCR, and MRR and for structure measurements, respectively. Additionally, the homogeneity of variances was examined using Levene’s test.

Results are presented as mean ± standard deviation (SD). All statistical tests were two-tailed, and p-values less than 0.05 were considered statistically significant.

3. Results

In the present study, we examined the modulatory effects of three environmentally relevant doses of As on mRNA expression levels of ERα,β, TRα,β, and PPARγ, respectively; simultaneous MRR in mouse hypothalamic tissue homogenates and mitochondrial morphology (number and size of mitochondria, and the proportional mitochondrial matrix area to the area of the entire mitochondrial crossection) were also determined. Concerning the MRR, although measurements have been carried out according to our description in the Materials and Methods section (i.e., all St1-5 were recorded), here we only present the results of St3 and St4 measurements, since these two parameters are directly relevant to demonstrate the intensity of mitochondrial metabolism (and №.2 reactions).

3.1. Receptor mRNA levels

In general, As treatment evoked elevated receptor expression in ERs and TRs, and even reached significance depending on the receptor subtypes. In the case of both ERα and TRα (Fig 2A and 2C.), As caused a remarkable dose-dependent increase in the receptor expressions (i.e., higher dose led to higher expression), where the treatment groups showed significant difference compared to the non-treated control and to each other, too. In contrast to TRα (Fig 2A), expression of TRβ (Fig 2B) was not affected by the As injection, since treated groups remained in the expression range observed in the case of the control. Also, the ERβ expression levels (Fig 2C) were drastically changed in a dose-dependent manner as a result of As-treatment, however, the pattern of the different doses was also different from that of ERα (Fig 2D): the expression in all treatment groups increased significantly compared to the control and each other. However, the highest peak was observed in the intermediate dose. Similarly to the TRβ groups, As only negligibly altered the PPARγ expression, without regard to the treatment doses (Fig 2E).

Fig 2. Effects of 3 different doses of a single intraperitoneal injection of sodium (meta)arsenite (arsenic, As) on the expression of hypothalamic nuclear hormone receptors (ERα,β; TRα,β, PPARγ), and hypothalamic mitochondrial state 3–4 respiration.

Fig 2

(A-E): Relative expression level of receptor genes was analyzed by qRT-PCR and normalized to the average of the control gene GAPDH. (F): ADP-dependent (St3) and oligomycin-induced (St4) mitochondrial respiration rates were given in consumed nmol O2 per ml of final volume in one minute. The data shown here are the mean ± standard deviation (SD) and compared to the non-treated control (ntC). P-values are represented by asterisk (*) where p < 0.05.

3.2. Mitochondrial respiration St3-4

Arsenic induced dose-dependent changes in MRR in both St3 and St4 values, their pattern showing a broad similarity to each other; however, that of the St4 was stronger (Fig 2F). Administration of the highest As dose (10 mg) decreased the mitochondrial activity (significantly in the case of St4), while the further two doses showed roughly the same activity as observed in the control. It is also important to note that St4 MRR values demonstrate a clear (and significant) inverse concentration dependence, i.e. as the As dose was reduced, the MRR increased to near the control mean (please see 40 μg group).

3.3. Mitochondrial ultrastructure

In both the AgRP (Fig 3A, 3C, 3E, 3G) and the POMC (Fig 3B, 3D, 3F, 3H) neurons, the size of mitochondria increased exclusively in animals treated with 5 mg of As (p < 0.001 and p < 0.05, respectively), while in the 40 μg and 10 mg group no significant change could been observed compared to the control (Fig 3A and 3B.).

Fig 3. The effects of 3 different doses of a single intraperitoneal injection of sodium (meta)arsenite (As) on the mitochondrial microstructure in AgRP and POMC neurons.

Fig 3

(A-B) the size of mitochondria, (C-D) the number of mitochondria is measured per unit section area. (E-F) the proportional mitochondrial matrix (matrix-to-total mitochondrial ratio) area is expressed as a percentage. (G-H) the images of electron micrographs of AgRP and POMC neurons, where arrows indicate examples of POMC-IR and AgRP-IR neurons. The micrographs showcase the effectiveness of the staining technique employed in this study, providing high-quality visualization of the cellular structures of interest. The data presented above (A-F) are expressed as the mean ± standard deviation (SD) and compared to the non-treated control (ntC) group. P-values are represented by asterisks (*,**) where p < 0.05 and p < 0.001, respectively.

The number (per 10 μm2 section area) of mitochondria has changed only in the 5 mg group, demonstrating a slight decrease (p < 0.1) in AgRP neurons (Fig 3C).

Proportional mitochondrial matrix area (matrix area per entire mitochondrion area in electron microscopic section) showed a drastic increase (p < 0.001) in a dose-dependent manner (higher As concentration caused higher density) (Fig 3E and 3F).

We also examined the juxtaposition of mitochondria and the endoplasmic reticulum in both the AgRP-IR (Fig 3G) and POMC-IR neurons (Fig 3H), however, neither the average distance between the two organelles, nor the number of mitochondrion-endoplasmic reticulum contacts were changed in any of the experimental groups.

3.4. Behavior of experimental animals

Mice that received the highest dose (10 mg) showed loss of physical activity, depression-like behavior, ataxia, loss of interest for feed and mild trembling 1.5–3 hours after receiving the intraperitoneal injection of As (similar administration of other EDs did not evoke such behavioral responses [unpublished observation]). Autopsy of these animals revealed no macroscopic pathological symptoms.

4. Discussion

4.1. Receptor mRNA levels

The literature abundantly reports how As exposure is associated with different endocrine disorders; however, a mechanistic investigation is still lacking [72, 73]. Arsenics’s impact on the hypothalamic-pituitary-ovarian axis is already unquestionable. Nevertheless, the impact of As on the estrogen signalization remains, at least in some senses, inconsistent, since As can either trigger or inhibit ER expression and ER-related pathways, with or without regard to its doses, tissues and species drawn into investigation [7479]. In our case, As treatment generally resulted in an elevated expression in both ER receptors. For example, ERβ expression levels (Fig 2D) were drastically changed in a dose-dependent manner, however, this change was different from that observed regarding ERα expression (Fig 2C). It is to note here, that ERα is recognized as the primary mediator of estrogenic effects on energy balance and an essential factor in the regulation of energy homeostasis, is mainly found in brain regions associated with the control of energy balance more precisely in the melanocortin system [80]. Our present findings are in accord with some previous literature data, however, our assessment of acute As exposure adds new findings to the existing knowledge from long-term exposure studies. Non-cytotoxic doses of As have already proved to strongly suppress ER-dependent gene transcription in in vivo poultry and in vitro human models [81]. This suggests that As may bind to ERs rendering them inaccessible for their natural ligands, a mechanism that can lead to an upregulation of ER expression in trying to regain the native functions. Regardless of whether As increases or decreases receptor expression levels, it is evident that ER-mediated №.1 intracellular processes are affected. Since ERs are not only present in the host cell but also in the mitochondria, it is likely that №.2 reactions are also promptly affected by AS exposure. This idea could be paralleled with our finding that As likewise seemed to depress dose-dependently St4: considering the interpretation of St values described in the Materials and Methods section, a decrease in St4 is a sign of an attempt of the mitochondria to produce more ATP. In contrast to this, Chen et al. [82] detected significant inhibition of ERα expression after long-term exposure to low doses of As2O3, but no effects have been detected with regard to ERβ. Another long-term study also reported the downregulation of ERα along with the decrease of the estrogen-responsive gene activities after a 4 μg/ml exposure to As [79]. Thus, our present results suggest that the immediate effects of As on ERs expression might differ from the long-term effects.

Studies on the relation of thyroid receptors and exposure to As are scarcer than those on estrogen-driven mechanisms. The majority of the reported studies examined thyroid markers other than the relevant receptors (in human patients), however, only a single article discussed the receptor expression (in amphibians). They reported that even very low As level can profoundly affect TR-dependent developmental processes in vertebrates [83]. According to some other studies, sub-chronic As exposure can significantly reduce TRβ expression in the rodent cerebellum both on mRNA and protein levels [84]. In humans, As treatment resulted in an elevated expression of TR mRNA, and also TSH was positively associated with the plasma As [85], however, the data on thyroid hormones (T3, T4) are again controversial [86, 87]. Compared to these, in our experiments, TRα mRNA was, significantly increased, regardless of the doses that we applied (Fig 2A); meanwhile, however, TRβ (Fig 2B) was not affected by the As-injection. The observed differences between As effects on TRs may lie on the grounds of several reasons: besides interspecies differences, direct hormonal and As environment of cells in question may significantly differ; and it is highly likely that significant differences between immediate and chronic effects of exposure also exist.

In this study, As altered only negligibly the PPARγ expression, with no regard to the injected doses (Fig 2E). In contrast, the literature reports variegated outcomes. Arsenic (3.80 ppm) increased PPARγ expression levels, followed by apoptosis, in in vitro cultured rat astrocytes [88]. PPARγ has also been reported to mediate the impact of As on increasing the incidence of type 2 diabetes through the PPARγ-mTORC2 signaling pathway [89, 90]. In preadipocytes, arsenic trioxide inhibited cell differentiation into fat cells by suppressing the PPARγ expression [9193]. In our present study, we detected no immediate effect of As exposure on PPARγ expression. However, this finding does not mean that PPARγ is insensitive to all EDs: our simultaneous experiments show that some other EDs like bisphenol A and zearalenone, in low concentrations, are able to increase PPARγ expression under similar experimental conditions that we applied in the present study (unpublished observations).

4.2. Mitochondrial respiration St3-4

Arsenic neurotoxicity and the adverse (usually drastic inhibitory) effects of As on brain mitochondria are already well-established. Toxic As doses, as a result of chronic exposure, induce super-oxide anion and ROS formation [83, 94, 95], decrease the activity of mitochondrial complexes I, II and IV [96], inhibition of ATPase activity [9698], dissipated mitochondrial transmembrane potentials [99102], as well as affect the mitochondrial proton motive force [95, 97, 103], moreover, leads to malformation of mitochondria [83, 104]. However, it is important to note that our current understanding does not include an examination of low (or non-toxic) doses of As in relation to mitochondrial function. Despite this, our results align with existing literature as we observed a significant inhibition of mitochondrial uncoupling activity at the highest dose of As used. In contrast, the lower two doses showed minimal deviation from the control group, demonstrating an inverse concentration-dependence. This pattern resembles the effects of certain E2 agonists [105107] (see Fig 2C and 2D). Interestingly, the effects of long-term exposure (days to weeks [102, 104]) to As, i.e., inhibition of ATP synthesis by mitochondria is preceded by an increase in ATP production as indicated by the observed low levels of St4 in the 10 mg group. With this regard, an additional important finding demonstrates that alterations in the uncoupling potency of the mitochondria in the melanocortin system are implicated in the intracellular processes that determine the output signals of the AgRP and POMC cells [108].

If this is the case, it suggests that 10 mg of As instantly propels mitochondrial ATP production, followed by the exhaustion of the mitochondria. This observation adds important information to our knowledge of As effects when discussing these effects in the context of №.1 and №.2 reactions. As mentioned in the case of the nuclear receptor expression, the finding that As only affected MRR in its St4 and only in 10 mg dose does not mean that mitochondrial metabolism is not sensitive to other EDs: various doses of bisphenol A and zearalenone had profound effects on St3-4 under experimental conditions that we applied for As exposure (unpublished observations). Those studies will further broaden our view of the temporal relationship of №.1 and №.2 reactions in response to various ED toxicants.

4.3. Mitochondrial ultrastructure

When discussing the ultrastructural features of mitochondria, we took into consideration the generally accepted interpretation of their morphological parameters. Accordingly, increased size, as well as decreased overall numbers of mitochondria (in AgRP and POMC cells) suggest organelle fusion. Increased fusion and/or decreased fission helps overcome low levels of stress (e.g., starvation [109, 110] or toxic effects [111]). Decreased fusion and/or increased fission occurs with high levels of stress (e.g., during apoptosis [112, 113] Increase in the size of the mitochondrial matrix per the entire mitochondrial crossection area is commonly associated with mitochondrial swelling, specifically in the context of oncotic or apoptotic mitochondria. This event is accompanied by a concomitant suppression of oxidative phosphorylation and a consequent decrease in ATP production [114]. More precisely, dynamic changes of mitochondria located in the melanocortin system are suggested to mimic the energy status of the organism (i.e. acting as energy sensors) and are actively participating in the modulation of the cellular activity (neurotransmission and release of cell-specific peptides) of their hosts, the AgRP and POMC neurons. Originating from their mitochondrial responses, these neurons are subsequently engaged in orchestrating distinctive processes governing energy expenditure across the organism and conveying pertinent information to higher brain centers [108].

In our present study As caused the increase of mitochondrial size at 5 mg dose only. With the interpretation described above in mind, this finding suggests that the aforementioned dose of As imposed a mild stress on mitochondria; however, definitions like those above, as taken from the literature, do not count with the versatility in the multitude of parameters of the cellular environment and the length of exposure. Thus, if we want to hold to generally accepted ways of interpretation, we have to emphasize the likely temporal changes in ED effects as cells try to acclimate to the various concentrations of EDs in the short- and long run. This means that, in our case, 5 mg of As may instantly cause a compensating adaptive response in mitochondria, but we cannot rule out that later the same dose could result in the exhaustion of mitochondria with inversed intensity in its metabolic activity [26, 115, 116]. At the same time, doses other than 5 mg may also be considered effective when examined after long-term exposure, although the present study does not answer this question.

The number (per section area) of mitochondria has changed only in the 5 mg group, demonstrating a slight decrease (p < 0.1) in AgRP neurons. It is not easy to interpret the meaning of this observation, considering that the number of mitochondria may increase as a physiological need for more ATP production (e.g., mitochondrial division in hypothalamic neurons after the mid-cycle estrogen surge [43, 108]) but it may also increase as a result of fission, latter which is the result of various stress effects and carries morphological signs such as mitochondrial swelling. With this in mind, our finding that the proportional size of the mitochondrial matrix (indicative of the level of mitochondrial swelling) increased linearly with the injected doses of As, suggests that the increase in the size of mitochondria with a simultaneous decrease in their numbers clearly shows immediate stressful and deleterious effect of As on AgRP neurons. The latter phenomenon may be an adaptive process aimed at mitigating the deleterious effects that pose a threat to the survival of the cell by compromising its metabolic and structural integrity. Beyond this, a decreased number of mitochondria paralleled with higher size of them, is characteristic of AgRP cells in the case of high-fat diet conditions [108]. This further supports the idea, that As may modulate the regulation of the energy homeostasis through affecting melanocortin neurons.

Arsenic’s immediate effect on POMC cells, at the same time, did not affect the number of mitochondria; this, however, does not exclude the possibility of effects on morphology after longer As exposure. The latter, seemingly speculative statement is supported by our findings that mitochondrial swelling occurred in a dose-linked manner (Fig 3A). It seems to be evident that As can alter the ultrastructure of mitochondria in hypothalamic neurons; however, here we only examined two distinct types of versatile neuron populations of the hypothalamus. Therefore, results from our studies do not mean that similar results occur in hypothalamic neurons other than AgRP and POMC cells; on the other hand, considering the rich neuronal interconnectivity between the hypothalamic neurons, it is highly likely that functional alterations in neuronal functions in response to As exposure may also play a role in the appearance of mitochondrial ultrastructure be it in the cell types examined in the present study or other cells of the hypothalamus.

Finally, the effects of arsenic, as observed and described so far, raise the question of how mitochondrial functions and related cellular metabolism in the hypothalamus could be affected after such changes. Firstly, animals showed signs of anxiety -/depression-like behavior. Numerous reports describe such effects of arsenic [117119], although those effects were observed after a longer time of exposure. These findings indicate the complexity of As’s effects, in that the aforementioned behavioral symptoms are regulated by a wide variety of brain regions, among them the loss of interest in feed-uptake involves the hypothalamus and its “hunger cells” (AgRP neurons) and “satiety cells” (POMC neurons). At the same time, however, among the ultrastructural results, the changes in size and number of mitochondria were observed, nevertheless, these morphological alterations were found in the 5 mg group instead of the 10 mg group. In the 10 mg group, as shown by the dose-dependent values of matrix-entire mitochondrion ratio (Fig 3E and 3F), the most severe changes appeared in the form of alterations of the behavior, as described above (3.4. of the Results section). Thus, it seems that initial changes in the number and size of mitochondria can be interpreted as compensational reactions to less toxic doses of As, whereas mitochondrial swelling (also known as the initial sign towards mitochondrial apoptosis) occurs at higher doses of As and such a toxic dose also manifests in the form of altered behavior. The finding that no changes occurred in the juxtaposition of mitochondria and endoplasmic reticulum suggests that immediate effects of As do not include mass growth in the volume of interaction between these organelles, namely, toxically affected mitochondria are likely to enter apoptosis [120123] instead of a generalized mitochondrial compensatory action in cellular metabolism.

4. Conclusion

The study confirms our hypothesis that even low doses of arsenic prompt immediate biological effects within hours of exposure. These effects involve rapid alterations in major hormone systems, particularly estrogen receptor (ER) and thyroid receptor (TR) expression, suggesting arsenic’s role as an endocrine disruptor targeting the neuroendocrine center. Considering these, arsenic influences energy expenditure by modulating cellular metabolism, appetite, and thermogenesis through hypothalamic estrogen and thyroid signaling pathways. Arsenic also affects mitochondrial dynamics in the hypothalamic melanocortin system, resembling the effects of a chronic high-fat diet.

Altogether, arsenic’s significant influence extends to the delicate and bidirectional interaction between the periphery and the central nervous system governed by the hypothalamus, potentially leading to endocrine dysfunction and the onset of metabolic diseases over time. These effects involve №.1 (intracellular regulatory pathways) and №.2 (adjustments of cellular energy expenditure) mechanisms, highlighting the complexity of arsenic’s impact on cellular and systemic processes.

The findings underscore the importance of further exploring the dose- and time-dependent effects of arsenic alone and in combination with other endocrine disruptors in the environment, to better understand their implications for human health and well-being.

Supporting information

S1 Raw data. It was obtained from electron microscopy (JEOL USA, Peabody, MA, USA).

(XLSX)

pone.0303528.s001.xlsx (161.6KB, xlsx)
S2 Raw data. It was measured by a Clark-type oxygen electrode (Oxytherm, Hansatech Instruments, Norfolk, UK).

(XLSX)

pone.0303528.s002.xlsx (20.1KB, xlsx)
S3 Raw data. It obtained from PCR.

(XLSX)

pone.0303528.s003.xlsx (48.6KB, xlsx)

Data Availability

All relevant data are within the paper and its Supporting Information files.

Funding Statement

This work was funded by the Hungarian Scientific Research Fund, OTKA K-115613 (to Attila Zsarnovszky; https://www.otka-palyazat.hu/) and was partly funded by the Ministry of Human Resources, EFOP-3.6.3-VEKOP-16-2017-00005 (to Attila Zsarnovszky; https://univet.hu/hu/egyetem/palyazati-projektek/eu-projektek/tudomanyos-utanpotlas-erositese/efop-3-6-3-vekop-16-2017-00005-mentorprogram/). This work was also supported by the Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, 7400 Kaposvár, Hungary) no. RRF-2.3.1-21-2022-00007 (to Attila Zsarnovszky; https://nkfih.gov.hu/national-laboratories). The sponsors or funders have not been involved in the study design, data collection and analysis, decision to publish, or preparation of the manuscript

References

  • 1.Ali W.; Rasool A.; Junaid M.; Zhang H. A Comprehensive Review on Current Status, Mechanism, and Possible Sources of Arsenic Contamination in Groundwater: A Global Perspective with Prominence of Pakistan Scenario. Environ. Geochem. Health 2019, 41, 737–760. doi: 10.1007/s10653-018-0169-x [DOI] [PubMed] [Google Scholar]
  • 2.Mukherjee A.; Sarkar S.; Coomar P.; Bhattacharya P. Towards Clean Water: Managing Risk of Arsenic-Contaminated Groundwater for Human Consumption. Curr. Opin. Environ. Sci. Heal. 2023, 36, 100509, doi: 10.1016/j.coesh.2023.100509 [DOI] [Google Scholar]
  • 3.Wasserman G.A.; Liu X.; Parvez F.; Chen Y.; Factor-Litvak P.; LoIacono N.J.; et al. A Cross-Sectional Study of Water Arsenic Exposure and Intellectual Function in Adolescence in Araihazar, Bangladesh. Environ. Int. 2018, 118, 304–313. doi: 10.1016/j.envint.2018.05.037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wang X.; Huang X.; Zhou L.; Chen J.; Zhang X.; Xu K.; et al. Association of Arsenic Exposure and Cognitive Impairment: A Population-Based Cross-Sectional Study in China. Neurotoxicology 2021, 82, 100–107. doi: 10.1016/j.neuro.2020.11.009 [DOI] [PubMed] [Google Scholar]
  • 5.Benvenga S.; Elia G.; Ragusa F.; Paparo S.R.; Sturniolo M.M.; Ferrari S.M.; et al. Endocrine Disruptors and Thyroid Autoimmunity. Best Pract. Res. Clin. Endocrinol. Metab. 2020, 34, 101377, doi: 10.1016/j.beem.2020.101377 [DOI] [PubMed] [Google Scholar]
  • 6.Alsen M.; Sinclair C.; Cooke P.; Ziadkhanpour K.; Genden E.; van Gerwen M. Endocrine Disrupting Chemicals and Thyroid Cancer: An Overview. Toxics 2021, 9, 1–26, doi: 10.3390/toxics9010014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Jugan M.L.; Levi Y.; Blondeau J.P. Endocrine Disruptors and Thyroid Hormone Physiology. Biochem. Pharmacol. 2010, 79, 939–947, doi: 10.1016/j.bcp.2009.11.006 [DOI] [PubMed] [Google Scholar]
  • 8.Boas M.; Feldt-Rasmussen U.; Main K.M. Thyroid Effects of Endocrine Disrupting Chemicals. Mol. Cell. Endocrinol. 2012, 355, 240–248, doi: 10.1016/j.mce.2011.09.005 [DOI] [PubMed] [Google Scholar]
  • 9.Lee H.R.; Jeung E.B.; Cho M.H.; Kim T.H.; Leung P.C.K.; Choi K.C. Molecular Mechanism(s) of Endocrine-Disrupting Chemicals and Their Potent Oestrogenicity in Diverse Cells and Tissues That Express Oestrogen Receptors. J. Cell. Mol. Med. 2013, 17, 1–11, doi: 10.1111/j.1582-4934.2012.01649.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Watson C.S.; Jeng Y.J.; Guptarak J. Endocrine Disruption via Estrogen Receptors That Participate in Nongenomic Signaling Pathways. J. Steroid Biochem. Mol. Biol. 2011, 127, 44–50, doi: 10.1016/j.jsbmb.2011.01.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Shanle E.K.; Xu W. Endocrine Disrupting Chemicals Targeting Estrogen Receptor Signaling: Identification and Mechanisms of Action. Chem. Res. Toxicol. 2011, 24, 6–19, doi: 10.1021/tx100231n [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kabir E.R.; Rahman M.S.; Rahman I. A Review on Endocrine Disruptors and Their Possible Impacts on Human Health. Environ. Toxicol. Pharmacol. 2015, 40, 241–258, doi: 10.1016/j.etap.2015.06.009 [DOI] [PubMed] [Google Scholar]
  • 13.Brix K. V; Baken S.; Poland C.A.; Blust R.; Pope L.J.; Tyler C.R. Challenges and Recommendations in Assessing Potential Endocrine‐Disrupting Properties of Metals in Aquatic Organisms. Environ. Toxicol. Chem. 2023, 42, 2564–2579. doi: 10.1002/etc.5741 [DOI] [PubMed] [Google Scholar]
  • 14.Britto T.I. Endocrine Equilibrium: Navigating the Hormonal Seas: Navigating the Whispers of Hormones: A Delicate Dance of Balance; BookRix, 2023; ISBN 3755446073. [Google Scholar]
  • 15.Pisapia L.; Del Pozzo G.; Barba P.; Caputo L.; Mita L.; Viggiano E.; et al U. Effects of Some Endocrine Disruptors on Cell Cycle Progression and Murine Dendritic Cell Differentiation. Gen. Comp. Endocrinol. 2012, 178, 54–63. doi: 10.1016/j.ygcen.2012.04.005 [DOI] [PubMed] [Google Scholar]
  • 16.Rutkowska A.Z.; Olsson A.; Rutkowski J.; Milewicz A. The Role of the Environment in Neuroendocrine Tumors. In Environmental Endocrinology and Endocrine Disruptors: Endocrine and Endocrine-targeted Actions and Related Human Diseases; Springer, 2023; pp. 1–30. [Google Scholar]
  • 17.Chen J.-Q.; Brown T.R.; Russo J. Regulation of Energy Metabolism Pathways by Estrogens and Estrogenic Chemicals and Potential Implications in Obesity Associated with Increased Exposure to Endocrine Disruptors. Biochim. Biophys. Acta (BBA)-Molecular Cell Res. 2009, 1793, 1128–1143. doi: 10.1016/j.bbamcr.2009.03.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Marroqui L.; Tudurí E.; Alonso-Magdalena P.; Quesada I.; Nadal Á.; Dos Santos R.S. Mitochondria as Target of Endocrine-Disrupting Chemicals: Implications for Type 2 Diabetes. J. Endocrinol. 2018, 239, R27–R45. doi: 10.1530/JOE-18-0362 [DOI] [PubMed] [Google Scholar]
  • 19.Mukherjee U.; Maitra S. Impact of Metabolic Disrupting Chemicals on Redox Homeostasis, Energy Sensors, Receptor Modulation, and Hormone Metabolism: A Comparative Account in Teleost and Mammalian Model Organisms. Aquac. Fish. 2023. [Google Scholar]
  • 20.Mohammed Abdul K.S.; Jayasinghe S.S.; Chandana E.P.S.; Jayasumana C.; De Silva P.M.C.S. Arsenic and Human Health Effects: A Review. Environ. Toxicol. Pharmacol. 2015, 40, 828–846, doi: 10.1016/j.etap.2015.09.016 [DOI] [PubMed] [Google Scholar]
  • 21.Kucheryavenko O.; Vogl S.; Marx-Stoelting P. Endocrine Disruptor Effects on Estrogen, Androgen and Thyroid Pathways: Recent Advances on Screening and Assessment. 2020. [Google Scholar]
  • 22.Köhrle J.; Frädrich C. Thyroid Hormone System Disrupting Chemicals. Best Pract. Res. Clin. Endocrinol. Metab. 2021, 35, 101562. doi: 10.1016/j.beem.2021.101562 [DOI] [PubMed] [Google Scholar]
  • 23.Oliveira K.J.; Chiamolera M.I.; Giannocco G.; Pazos-Moura C.C.; Ortiga-Carvalho T.M. Thyroid Function Disruptors: From Nature to Chemicals. J. Mol. Endocrinol. 2019, 62, R1–R19. [DOI] [PubMed] [Google Scholar]
  • 24.Tontonoz P.; Hu E.; Spiegelman B.M. Stimulation of Adipogenesis in Fibroblasts by PPARγ2, a Lipid-Activated Transcription Factor. Cell 1994, 79, 1147–1156, doi: 10.1016/0092-8674(94)90006-X [DOI] [PubMed] [Google Scholar]
  • 25.Yousefnia S.; Momenzadeh S.; Seyed Forootan F.; Ghaedi K.; Nasr Esfahani M.H. The Influence of Peroxisome Proliferator-Activated Receptor γ (PPARγ) Ligands on Cancer Cell Tumorigenicity. Gene 2018, 649, 14–22, doi: 10.1016/j.gene.2018.01.018 [DOI] [PubMed] [Google Scholar]
  • 26.Ambrosio F.; Brown E.; Stolz D.; Ferrari R.; Goodpaster B.; Deasy B.; et al. Arsenic Induces Sustained Impairment of Skeletal Muscle and Muscle Progenitor Cell Ultrastructure and Bioenergetics. Free Radic. Biol. Med. 2014, 74, 64–73. doi: 10.1016/j.freeradbiomed.2014.06.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Janesick A.; Blumberg B. Minireview: PPARγ as the Target of Obesogens. J. Steroid Biochem. Mol. Biol. 2011, 127, 4–8, doi: 10.1016/j.jsbmb.2011.01.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Scalise T.; Györffy A.; Tóth I.; Kiss D.; Somogyi V.; Goszleth G.; et al. Ligand-Induced Changes in Oestrogen and Thyroid Hormone Receptor Expression in the Developing Rat Cerebellum: A Comparative Quantitative PCR and Western Blot Study. Acta Vet. Hung. 2012, 60, 263–284, doi: 10.1556/AVet.2012.023 [DOI] [PubMed] [Google Scholar]
  • 29.Jocsak G.; Ioja E.; Kiss D.S.; Toth I.; Barany Z.; Bartha T.; et al. Endocrine Disruptors Induced Distinct Expression of Thyroid and Estrogen Receptors in Rat versus Mouse Primary Cerebellar Cell Cultures. Brain Sci. 2019, 9, doi: 10.3390/brainsci9120359 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Somogyi V.; Horváth T.L.; Tóth I.; Bartha T.; Frenyó L.V.; Kiss D.S.; et al. Bisphenol a Influences Oestrogen- and Thyroid Hormone-Regulated Thyroid Hormone Receptor Expression in Rat Cerebellar Cell Culture. Acta Vet. Hung. 2016, 64, 497–513, doi: 10.1556/004.2016.046 [DOI] [PubMed] [Google Scholar]
  • 31.Jocsak G.; Kiss D.S.; Toth I.; Goszleth G.; Bartha T.; Frenyo L. V.; et al. Comparison of Individual and Combined Effects of Four Endocrine Disruptors on Estrogen Receptor Beta Transcription in Cerebellar Cell Culture: The Modulatory Role of Estradiol and Triiodo-Thyronine. Int. J. Environ. Res. Public Health 2016, 13, 1–14, doi: 10.3390/ijerph13060619 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Palkovits M. Hypothalamic Regulation of Food Intake. Ideggyogy. Sz. 2003, 56, 288–302. [PubMed] [Google Scholar]
  • 33.Toni R. The Neuroendocrine System: Organization and Homeostatic Role. J. Endocrinol. Invest. 2004, 27, 35–47. [PubMed] [Google Scholar]
  • 34.Patisaul H.B.; Belcher S.M. 73 The Neuroendocrine System and General Mechanisms of Endocrine Disruption. In Endocrine Disruptors, Brain, and Behavior; Oxford University Press, 2017. ISBN 9780199935734. [Google Scholar]
  • 35.Chen T.T.; Maevsky E.I.; Uchitel M.L. Maintenance of Homeostasis in the Aging Hypothalamus: The Central and Peripheral Roles of Succinate. Front. Endocrinol. (Lausanne). 2015, 6, 7, doi: 10.3389/fendo.2015.00007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Hall K.D.; Heymsfield S.B.; Kemnitz J.W.; Klein S.; Schoeller D.A.; Speakman J.R. Energy Balance and Its Components: Implications for Body Weight Regulation. Am. J. Clin. Nutr. 2012, 95, 989–994, doi: 10.3945/ajcn.112.036350 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Timper K.; Brüning J.C. Hypothalamic Circuits Regulating Appetite and Energy Homeostasis: Pathways to Obesity. Dis. Model. Mech. 2017, 10, 679–689, doi: 10.1242/dmm.026609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Haddad-Tóvolli R.; Dragano N.R. V; Ramalho A.F.S.; Velloso L.A. Development and Function of the Blood-Brain Barrier in the Context of Metabolic Control. Front. Neurosci. 2017, 11, 224, doi: 10.3389/fnins.2017.00224 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Langlet F. Tanycytes: A Gateway to the Metabolic Hypothalamus. J. Neuroendocrinol. 2014, 26, doi: 10.1111/jne.12191 [DOI] [PubMed] [Google Scholar]
  • 40.Jais A.; Brüning J.C. Arcuate Nucleus-Dependent Regulation of Metabolism-Pathways to Obesity and Diabetes Mellitus. Endocr. Rev. 2022, 43, 314–328, doi: 10.1210/endrev/bnab025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Coll A.P.; Yeo G.S.H. The Hypothalamus and Metabolism: Integrating Signals to Control Energy and Glucose Homeostasis. Curr. Opin. Pharmacol. 2013, 13, 970–976, doi: 10.1016/j.coph.2013.09.010 [DOI] [PubMed] [Google Scholar]
  • 42.Vohra M.S.; Benchoula K.; Serpell C.J.; Hwa W.E. AgRP/NPY and POMC Neurons in the Arcuate Nucleus and Their Potential Role in Treatment of Obesity. Eur. J. Pharmacol. 2022, 915, 174611, doi: 10.1016/j.ejphar.2021.174611 [DOI] [PubMed] [Google Scholar]
  • 43.Dietrich M.O.; Liu Z.-W.; Horvath T.L. Mitochondrial Dynamics Controlled by Mitofusins Regulate Agrp Neuronal Activity and Diet-Induced Obesity. Cell 2013, 155, 188–199. doi: 10.1016/j.cell.2013.09.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Drougard A.; Fournel A.; Valet P.; Knauf C. Impact of Hypothalamic Reactive Oxygen Species in the Regulation of Energy Metabolism and Food Intake. Front. Neurosci. 2015, 9, 56, doi: 10.3389/fnins.2015.00056 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Rochford J.J.; Myers M.G.; Heisler L.K. Setting the Tone: Reactive Oxygen Species and the Control of Appetitive Melanocortin Neurons. Cell Metab. 2011, 14, 573–574, doi: 10.1016/j.cmet.2011.10.004 [DOI] [PubMed] [Google Scholar]
  • 46.Chen J.Q.; Cammarata P.R.; Baines C.P.; Yager J.D. Regulation of Mitochondrial Respiratory Chain Biogenesis by Estrogens/Estrogen Receptors and Physiological, Pathological and Pharmacological Implications. Biochim. Biophys. Acta—Mol. Cell Res. 2009, 1793, 1540–1570, doi: 10.1016/j.bbamcr.2009.06.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Irwin R.W.; Yao J.; Hamilton R.T.; Cadenas E.; Brinton R.D.; Nilsen J. Progesterone and Estrogen Regulate Oxidative Metabolism in Brain Mitochondria. Endocrinology 2008, 149, 3167–3175, doi: 10.1210/en.2007-1227 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Goglia F.; Silvestri E.; Lanni A. Thyroid Hormones and Mitochondria. Biosci. Rep. 2002, 22, 17–32, doi: 10.1023/a:1016056905347 [DOI] [PubMed] [Google Scholar]
  • 49.Wrutniak-Cabello C.; Casas F.; Cabello G. Thyroid Hormone Action in Mitochondria. J. Mol. Endocrinol. 2001, 26, 67–77, doi: 10.1677/jme.0.0260067 [DOI] [PubMed] [Google Scholar]
  • 50.Kowalska K.; Habrowska-Górczyńska D.E.; Piastowska-Ciesielska A.W. Zearalenone as an Endocrine Disruptor in Humans. Environ. Toxicol. Pharmacol. 2016, 48, 141–149, doi: 10.1016/j.etap.2016.10.015 [DOI] [PubMed] [Google Scholar]
  • 51.Chatterjee A.; Jamal Z.; Das J.; Banerjee A.; Chatterji U. The Imperceptible Contagion: Arsenic as a Neuro-Immune-Endocrine Disruptor. Proc. Zool. Soc. 2021 744 2021, 74, 387–407, doi: 10.1007/S12595-021-00393-3 [DOI] [Google Scholar]
  • 52.Wong J.K.; Le H.H.; Zsarnovszky A.; Belcher S.M. Estrogens and ICI182,780 (Faslodex) Modulate Mitosis and Cell Death in Immature Cerebellar Neurons via Rapid Activation of P44/P42 Mitogen-Activated Protein Kinase. J. Neurosci. 2003, 23, 4984–4995, doi: 10.1523/JNEUROSCI.23-12-04984.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kiss D.S.; Ioja E.; Toth I.; Barany Z.; Jocsak G.; Bartha T.; et al. Comparative Analysis of Zearalenone Effects on Thyroid Receptor Alpha (TRα) and Beta (TRβ) Expression in Rat Primary Cerebellar Cell Cultures. Int. J. Mol. Sci. 2018, 19, doi: 10.3390/ijms19051440 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mandal B.K.; Suzuki K.T. Arsenic Round the World: A Review. Talanta 2002, 58, 201–235, doi: 10.1016/S0039-9140(02)00268-0 [DOI] [PubMed] [Google Scholar]
  • 55.Chang S.I.; Jin B.; Youn P.; Park C.; Park J.-D.; Ryu D.-Y. Arsenic-Induced Toxicity and the Protective Role of Ascorbic Acid in Mouse Testis. Toxicol. Appl. Pharmacol. 2007, 218, 196–203, doi: 10.1016/j.taap.2006.11.009 [DOI] [PubMed] [Google Scholar]
  • 56.Jana K.; Jana S.; Samanta P.K. Effects of Chronic Exposure to Sodium Arsenite on Hypothalamo-Pituitary-Testicular Activities in Adult Rats: Possible an Estrogenic Mode of Action. Reprod. Biol. Endocrinol. 2006, 4, 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Stump D.G.; Holson J.F.; Fleeman T.L.; Nemec M.D.; Farr C.H. Comparative Effects of Single Intraperitoneal or Oral Doses of Sodium Arsenate or Arsenic Trioxide during in Utero Development. Teratology 1999, 60, 283–291. doi: [DOI] [PubMed] [Google Scholar]
  • 58.Smedley P.L.; Kinniburgh D.G. Arsenic in Groundwater and the Environment. In Essentials of Medical Geology: Revised Edition; Springer, 2012; pp. 279–310. [Google Scholar]
  • 59.Shankar S.; Shanker U. Arsenic Contamination of Groundwater: A Review of Sources, Prevalence, Health Risks, and Strategies for Mitigation. Sci. world J. 2014, 2014. doi: 10.1155/2014/304524 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Zsarnovszky A.; Kiss D.; Jocsak G.; Nemeth G.; Toth I.; Horvath T.L. Thyroid Hormone- and Estrogen Receptor Interactions with Natural Ligands and Endocrine Disruptors in the Cerebellum. Front. Neuroendocrinol. 2018, 48, 23–36, doi: 10.1016/j.yfrne.2017.10.001 [DOI] [PubMed] [Google Scholar]
  • 61.Sengupta P. The Laboratory Rat: Relating Its Age with Human’s. Int. J. Prev. Med. 2013, 4, 624. [PMC free article] [PubMed] [Google Scholar]
  • 62.Sengupta P. Environmental and Occupational Exposure of Metals and Their Role in Male Reproductive Functions. Drug Chem. Toxicol. 2013, 36, 353–368. doi: 10.3109/01480545.2012.710631 [DOI] [PubMed] [Google Scholar]
  • 63.Sengupta T.; Ghosh S.; Gaur T A.; Nayak P. Dissimilar Anxiety-like Behavior in Prepubertal and Young Adult Female Rats on Acute Exposure to Aluminium. Cent. Nerv. Syst. Agents Med. Chem. (Formerly Curr. Med. Chem. Nerv. Syst. Agents) 2021, 21, 187–194. doi: 10.2174/1871524922666211231095507 [DOI] [PubMed] [Google Scholar]
  • 64.Brydges N.M. Pre-Pubertal Stress and Brain Development in Rodents. Curr. Opin. Behav. Sci. 2016, 7, 8–14. [Google Scholar]
  • 65.Rebuli M.E.; Patisaul H.B. Assessment of Sex Specific Endocrine Disrupting Effects in the Prenatal and Pre-Pubertal Rodent Brain. J. Steroid Biochem. Mol. Biol. 2016, 160, 148–159. doi: 10.1016/j.jsbmb.2015.08.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Toth I.; Kiss D.S.; Goszleth G.; Bartha T.; Frenyo L. V.; Naftolin F.; et al. Hypothalamic Sidedness in Mitochondrial Metabolism: New Perspectives. Reprod. Sci. 2014, 21, 1492–1498, doi: 10.1177/1933719114530188 [DOI] [PubMed] [Google Scholar]
  • 67.Kiss D.S.; Toth I.; Jocsak G.; Sterczer A.; Bartha T.; Frenyo L. V.; Zsarnovszky A. Preparation of Purified Perikaryal and Synaptosomal Mitochondrial Fractions from Relatively Small Hypothalamic Brain Samples. MethodsX 2016, 3, 417–429, doi: 10.1016/j.mex.2016.05.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Berruien N.N.A.; Murray J.F.; Smith C.L. Pregnancy Influences the Selection of Appropriate Reference Genes in Mouse Tissue: Determination of Appropriate Reference Genes for Quantitative Reverse Transcription PCR Studies in Tissues from the Female Mouse Reproductive Axis. Gene 2021, 801, 145855, doi: 10.1016/j.gene.2021.145855 [DOI] [PubMed] [Google Scholar]
  • 69.Pfaffl M.W.; Horgan G.W.; Dempfle L. Relative Expression Software Tool (REST) for Group-Wise Comparison and Statistical Analysis of Relative Expression Results in Real-Time PCR. Nucleic Acids Res. 2002, 30, doi: 10.1093/nar/30.9.e36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Kiss D.S.; Zsarnovszky A.; Horvath K.; Gyorffy A.; Bartha T.; Hazai D.; et al. Ecto-Nucleoside Triphosphate Diphosphohydrolase 3 in the Ventral and Lateral Hypothalamic Area of Female Rats: Morphological Characterization and Functional Implications. Reprod. Biol. Endocrinol. 2009, 7, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Patra M.; Mahata S.K.; Padhan D.K.; Sen M. CCN6 Regulates Mitochondrial Function. J. Cell Sci. 2016, 129, 2841–2851. doi: 10.1242/jcs.186247 [DOI] [PubMed] [Google Scholar]
  • 72.Naghizadeh A.; Esform A.; Rezaei M.; Farkhondeh T.; Samarghandian S. Environmental Arsenic Exposure and Its Toxicological Effect on Thyroid Function: A Systematic Review. Rev. Environ. Health 2022, 37, 281–289, doi: 10.1515/reveh-2021-0025 [DOI] [PubMed] [Google Scholar]
  • 73.Sun H.J.; Xiang P.; Luo J.; Hong H.; Lin H.; Li H.B.; et al. Mechanisms of Arsenic Disruption on Gonadal, Adrenal and Thyroid Endocrine Systems in Humans: A Review. Environ. Int. 2016, 95, 61–68, doi: 10.1016/j.envint.2016.07.020 [DOI] [PubMed] [Google Scholar]
  • 74.Miller W.H.; Schipper H.M.; Lee J.S.; Singer J.; Waxman S. Mechanisms of Action of Arsenic Trioxide. Cancer Res. 2002, 62, 3893–3903. [PubMed] [Google Scholar]
  • 75.Stoica A.; Pentecost E.; Martin M.B. Effects of Arsenite on Estrogen Receptor-α Expression Activity in MCF-7 Breast Cancer Cells. Endocrinology 2000, 141, 3595–3602, doi: 10.1210/endo.141.10.7704 [DOI] [PubMed] [Google Scholar]
  • 76.Davey J.C.; Nomikos A.P.; Wungjiranirun M.; Sherman J.R.; Ingram L.; Batki C.; et al. Arsenic as an Endocrine Disruptor: Arsenic Disrupts Retinoic Acid Receptor- and Thyroid Hormone Receptor-Mediated Gene Regulation and Thyroid Hormone-Mediated Amphibian Tail Metamorphosis. Environ. Health Perspect. 2008, 116, 165–172, doi: 10.1289/ehp.10131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Chow S.K.Y.; Chan J.Y.W.; Fung K.P. Suppression of Cell Proliferation and Regulation of Estrogen Receptor α Signaling Pathway by Arsenic Trioxide on Human Breast Cancer MCF-7 Cells. J. Endocrinol. 2004, 182, 325–337, doi: 10.1677/joe.0.1820325 [DOI] [PubMed] [Google Scholar]
  • 78.Waalkes M.P.; Liu J.; Chen H.; Xie Y.; Achanzar W.E.; Zhou Y.S.; et al. Estrogen Signaling in Livers of Male Mice with Hepatocellular Carcinoma Induced by Exposure to Arsenic in Utero. J. Natl. Cancer Inst. 2004, 96, 466–474, doi: 10.1093/jnci/djh070 [DOI] [PubMed] [Google Scholar]
  • 79.Chatterjee A.; Chatterji U. Arsenic Abrogates the Estrogen-Signaling Pathway in the Rat Uterus. Reprod. Biol. Endocrinol. 2010, 8, 1–11, doi: 10.1186/1477-7827-8-80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Xu Y.; Elmquist J.K.; Fukuda M. Central Nervous Control of Energy and Glucose Balance: Focus on the Central Melanocortin System. Ann. N. Y. Acad. Sci. 2011, 1243, 1–14, doi: 10.1111/j.1749-6632.2011.06248.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Rzemieniec J.; Litwa E.; Wnuk A.; Lason W.; Kajta M. Bazedoxifene and Raloxifene Protect Neocortical Neurons Undergoing Hypoxia via Targeting ERα and PPAR-γ. Mol. Cell. Endocrinol. 2018, 461, 64–78, doi: 10.1016/j.mce.2017.08.014 [DOI] [PubMed] [Google Scholar]
  • 82.Chen G.C.; Guan L.S.; Hu W.L.; Wang Z.Y. Functional Repression of Estrogen Receptor α by Arsenic Trioxide in Human Breast Cancer Cells. Anticancer Res. 2002, 22, 633–638. [PubMed] [Google Scholar]
  • 83.Prakash C.; Soni M.; Kumar V. Biochemical and Molecular Alterations Following Arsenic-Induced Oxidative Stress and Mitochondrial Dysfunction in Rat Brain. Biol. Trace Elem. Res. 2015, 167, 121–129, doi: 10.1007/s12011-015-0284-9 [DOI] [PubMed] [Google Scholar]
  • 84.Guan H.; Li S.; Guo Y.; Liu X.; Yang Y.; Guo J.; et al. Subchronic Exposure to Arsenic Represses the TH/TRβ1-CaMK IV Signaling Pathway in Mouse Cerebellum. Int. J. Mol. Sci. 2016, 17, 157, doi: 10.3390/ijms17020157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Molin M.; Ulven S.M.; Dahl L.; Lundebye A.K.; Holck M.; Alexander J.; et al. Arsenic in Seafood Is Associated with Increased Thyroid-Stimulating Hormone (TSH) in Healthy Volunteers–A Randomized Controlled Trial. J. Trace Elem. Med. Biol. 2017, 44, 1–7, doi: 10.1016/j.jtemb.2017.05.004 [DOI] [PubMed] [Google Scholar]
  • 86.Meltzer H.M.; Maage A.; Ydersbond T.A.; Haug E.; Glattre E.; Holm H. Fish Arsenic May Influence Human Blood Arsenic, Selenium, and T4:T3 Ratio. Biol. Trace Elem. Res. 2002, 90, 83–98, doi: 10.1385/BTER:90:1-3:83 [DOI] [PubMed] [Google Scholar]
  • 87.Glattre E.; Mravcova A.; Lener J.; Vobecky M.; Egertova E.; Mysliveckova M. Study of Distribution and Interaction of Arsenic and Selenium in Rat Thyroid. Biol. Trace Elem. Res. 1995, 49, 177–186, doi: 10.1007/BF02788967 [DOI] [PubMed] [Google Scholar]
  • 88.Kushwaha R.; Mishra J.; Tripathi S.; Khare P.; Bandyopadhyay S. Arsenic, Cadmium, and Lead Like Troglitazone Trigger PPARγ-Dependent Poly (ADP-Ribose) Polymerase Expression and Subsequent Apoptosis in Rat Brain Astrocytes. Mol. Neurobiol. 2018, 55, 2125–2149, doi: 10.1007/s12035-017-0469-7 [DOI] [PubMed] [Google Scholar]
  • 89.Kirkley A.G.; Carmean C.M.; Ruiz D.; Ye H.; Regnier S.M.; Poudel A.; et al. Arsenic Exposure Induces Glucose Intolerance and Alters Global Energy Metabolism. Am. J. Physiol.—Regul. Integr. Comp. Physiol. 2018, 314, R294–R303, doi: 10.1152/ajpregu.00522.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Gao N.; Yao X.; Jiang L.; Yang L.; Qiu T.; Wang Z.; et al. Taurine Improves Low-Level Inorganic Arsenic-Induced Insulin Resistance by Activating PPARγ-MTORC2 Signalling and Inhibiting Hepatic Autophagy. J. Cell. Physiol. 2019, 234, 5143–5152, doi: 10.1002/jcp.27318 [DOI] [PubMed] [Google Scholar]
  • 91.Ceja-Galicia Z.A.; Daniel A.; Salazar A.M.; Pánico P.; Ostrosky-Wegman P.; Díaz-Villaseñor A. Effects of Arsenic on Adipocyte Metabolism: Is Arsenic an Obesogen? Mol. Cell. Endocrinol. 2017, 452, 25–32, doi: 10.1016/j.mce.2017.05.008 [DOI] [PubMed] [Google Scholar]
  • 92.Wauson E.M.; Langan A.S.; Vorce R.L. Sodium Arsenite Inhibits and Reverses Expression of Adipogenic and Fat Cell-Specific Genes during in Vitro Adipogenesis. Toxicol. Sci. 2002, 65, 211–219, doi: 10.1093/toxsci/65.2.211 [DOI] [PubMed] [Google Scholar]
  • 93.Wang Z.X.; Jiang C.S.; Liu L.; Wang X.H.; Jin H.J.; Wu Q.; et al. The Role of Akt on Arsenic Trioxide Suppression of 3T3-L1 Preadipocyte Differentiation. Cell Res. 2005, 15, 379–386, doi: 10.1038/sj.cr.7290305 [DOI] [PubMed] [Google Scholar]
  • 94.Srivastava P.; Yadav R.S.; Chandravanshi L.P.; Shukla R.K.; Dhuriya Y.K.; Chauhan L.K.S.; et al. Unraveling the Mechanism of Neuroprotection of Curcumin in Arsenic Induced Cholinergic Dysfunctions in Rats. Toxicol. Appl. Pharmacol. 2014, 279, 428–440, doi: 10.1016/j.taap.2014.06.006 [DOI] [PubMed] [Google Scholar]
  • 95.Ghosh S.; Dungdung S.R.; Chowdhury S.T.; Mandal A.K.; Sarkar S.; Ghosh D.; et al. Encapsulation of the Flavonoid Quercetin with an Arsenic Chelator into Nanocapsules Enables the Simultaneous Delivery of Hydrophobic and Hydrophilic Drugs with a Synergistic Effect against Chronic Arsenic Accumulation and Oxidative Stress. Free Radic. Biol. Med. 2011, 51, 1893–1902, doi: 10.1016/j.freeradbiomed.2011.08.019 [DOI] [PubMed] [Google Scholar]
  • 96.Dwivedi N.; Mehta A.; Yadav A.; Binukumar B.K.; Gill K.D.; Flora S.J.S. MiADMSA Reverses Impaired Mitochondrial Energy Metabolism and Neuronal Apoptotic Cell Death after Arsenic Exposure in Rats. Toxicol. Appl. Pharmacol. 2011, 256, 241–248, doi: 10.1016/j.taap.2011.04.004 [DOI] [PubMed] [Google Scholar]
  • 97.Hughes M.F. Arsenic Toxicity and Potential Mechanisms of Action. Toxicol. Lett. 2002, 133, 1–16, doi: 10.1016/s0378-4274(02)00084-x [DOI] [PubMed] [Google Scholar]
  • 98.Gopalkrishnan A.; Rao M.V. Amelioration by Vitamin A upon Arsenic Induced Metabolic and Neurotoxic Effects. J. Heal. Sci. 2006, 52, 568–577, doi: 10.1248/jhs.52.568 [DOI] [Google Scholar]
  • 99.Rocha R.A.; Gimeno-Alcañiz J. V; Martín-Ibañez R.; Canals J.M.; Vélez D.; Devesa V. Arsenic and Fluoride Induce Neural Progenitor Cell Apoptosis. Toxicol. Lett. 2011, 203, 237–244, doi: 10.1016/j.toxlet.2011.03.023 [DOI] [PubMed] [Google Scholar]
  • 100.Pachauri V.; Mehta A.; Mishra D.; Flora S.J.S. Arsenic Induced Neuronal Apoptosis in Guinea Pigs Is Ca2+ Dependent and Abrogated by Chelation Therapy: Role of Voltage Gated Calcium Channels. Neurotoxicology 2013, 35, 137–145, doi: 10.1016/j.neuro.2013.01.006 [DOI] [PubMed] [Google Scholar]
  • 101.Lu T.H.; Tseng T.J.; Su C.C.; Tang F.C.; Yen C.C.; Liu Y.Y.; et al. Arsenic Induces Reactive Oxygen Species-Caused Neuronal Cell Apoptosis through JNK/ERK-Mediated Mitochondria-Dependent and GRP 78/CHOP-Regulated Pathways. Toxicol. Lett. 2014, 224, 130–140, doi: 10.1016/j.toxlet.2013.10.013 [DOI] [PubMed] [Google Scholar]
  • 102.Rojewski M.T.; Korper S.; Thiel E.; Schrezenmeier H. Arsenic Trioxide-Induced Apoptosis Is Independent of CD95 in Lymphatic Cell Lines. Oncol. Rep. 2004, 11, 509–513, doi: 10.3892/or.11.2.509 [DOI] [PubMed] [Google Scholar]
  • 103.Hong Y.; Piao F.; Zhao Y.; Li S.; Wang Y.; Liu P. Subchronic Exposure to Arsenic Decreased Sdha Expression in the Brain of Mice. Neurotoxicology 2009, 30, 538–543, doi: 10.1016/j.neuro.2009.04.011 [DOI] [PubMed] [Google Scholar]
  • 104.Hassani S.; Yaghoubi H.; Khosrokhavar R.; Jafarian I.; Mashayekhi V.; Hosseini M.J.; et al. Mechanistic View for Toxic Effects of Arsenic on Isolated Rat Kidney and Brain Mitochondria. Biol. 2015, 70, 683–689, doi: 10.1515/biolog-2015-0081 [DOI] [Google Scholar]
  • 105.Haczynski J.; Tarkowski R.; Jarzabek K.; Wolczynski S.; Magoffin D.A.; Czarnocki K.J.; et al. Differential Effects of Estradiol, Raloxifene and Tamoxifen on Estrogen Receptor Expression in Cultured Human Skin Fibroblasts. Int. J. Mol. Med. 2004, 13, 903–908, doi: 10.3892/ijmm.13.6.903 [DOI] [PubMed] [Google Scholar]
  • 106.Razmara A.; Sunday L.; Stirone C.; Xiao B.W.; Krause D.N.; Duckles S.P.; et al. Mitochondrial Effects of Estrogen Are Mediated by Estrogen Receptor α in Brain Endothelial Cells. J. Pharmacol. Exp. Ther. 2008, 325, 782–790, doi: 10.1124/JPET.107.134072 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Gorgogietas V.A.; Tsialtas I.; Sotiriou N.; Laschou V.C.; Karra A.G.; Leonidas D.D.; et al. Potential Interference of Aluminum Chlorohydrate with Estrogen Receptor Signaling in Breast Cancer Cells. J. Mol. Biochem. 2018, 7, 1–13. [PMC free article] [PubMed] [Google Scholar]
  • 108.Nasrallah C.M.; Horvath T.L. Mitochondrial Dynamics in the Central Regulation of Metabolism. Nat. Rev. Endocrinol. 2014, 10, 650–658. doi: 10.1038/nrendo.2014.160 [DOI] [PubMed] [Google Scholar]
  • 109.Gomes L.C.; Benedetto G. Di; Scorrano L. During Autophagy Mitochondria Elongate, Are Spared from Degradation and Sustain Cell Viability. Nat. Cell Biol. 2011, 13, 589–598, doi: 10.1038/ncb2220 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Rambold A.S.; Kostelecky B.; Elia N.; Lippincott-Schwartz J. Tubular Network Formation Protects Mitochondria from Autophagosomal Degradation during Nutrient Starvation. Proc. Natl. Acad. Sci. 2011, 108, 10190–10195, doi: 10.1073/pnas.1107402108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Tondera D.; Grandemange S.; Jourdain A.; Karbowski M.; Mattenberger Y.; Herzig S.; et al. SLP‐2 Is Required for Stress‐induced Mitochondrial Hyperfusion. EMBO J. 2009, 28, 1589–1600. doi: 10.1038/emboj.2009.89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Ehses S.; Raschke I.; Mancuso G.; Bernacchia A.; Geimer S.; Tondera D.; et al. Regulation of OPA1 Processing and Mitochondrial Fusion by M-AAA Protease Isoenzymes and OMA1. J. Cell Biol. 2009, 187, 1023–1036, doi: 10.1083/jcb.200906084 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Head B.; Griparic L.; Amiri M.; Gandre-Babbe S.; van der Bliek A.M. Inducible Proteolytic Inactivation of OPA1 Mediated by the OMA1 Protease in Mammalian Cells. J. Cell Biol. 2009, 187, 959–966, doi: 10.1083/jcb.200906083 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Wojtczak L.; Lehninger A.L. Formation and Disappearance of an Endogenous Uncoupling Factor during Swelling and Contraction of Mitochondria. Biochim. Biophys. Acta 1961, 51, 442–456, doi: 10.1016/0006-3002(61)90600-x [DOI] [PubMed] [Google Scholar]
  • 115.Lin C.; Chang L.W.; Wei Y.; Wu S.; Yang C.; Chang W.; et al. Electronic Microscopy Evidence for Mitochondria as Targets for Cd/Se/Te‐based Quantum Dot 705 Toxicity in Vivo. Kaohsiung J. Med. Sci. 2012, 28, S53–S62. doi: 10.1016/j.kjms.2012.05.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Prakash C.; Kumar V. Arsenic-Induced Mitochondrial Oxidative Damage Is Mediated by Decreased PGC-1α Expression and Its Downstream Targets in Rat Brain. Chem. Biol. Interact. 2016, 256, 228–235. [DOI] [PubMed] [Google Scholar]
  • 117.Samad N.; Rao T.; Rehman M.H. ur; Bhatti S.A.; Imran I. Inhibitory Effects of Selenium on Arsenic-Induced Anxiety-/Depression-Like Behavior and Memory Impairment. Biol. Trace Elem. Res. 2022, 200, 689–698, doi: 10.1007/s12011-021-02679-1 [DOI] [PubMed] [Google Scholar]
  • 118.Chang C.-Y.; Guo H.-R.; Tsai W.-C.; Yang K.-L.; Lin L.-C.; Cheng T.-J.; et al. Subchronic Arsenic Exposure Induces Anxiety-Like Behaviors in Normal Mice and Enhances Depression-Like Behaviors in the Chemically Induced Mouse Model of Depression. Biomed Res. Int. 2015, 2015, 159015, doi: 10.1155/2015/159015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Samad N.; Jabeen S.; Imran I.; Zulfiqar I.; Bilal K. Protective Effect of Gallic Acid against Arsenic-Induced Anxiety−/Depression- like Behaviors and Memory Impairment in Male Rats. Metab. Brain Dis. 2019, 34, 1091–1102, doi: 10.1007/s11011-019-00432-1 [DOI] [PubMed] [Google Scholar]
  • 120.Chattopadhyay S.; Bhaumik S.; Chaudhury A.N.; Gupta S. Das Arsenic Induced Changes in Growth Development and Apoptosis in Neonatal and Adult Brain Cells in Vivo and in Tissue Culture. Toxicol. Lett. 2002, 128, 73–84. [DOI] [PubMed] [Google Scholar]
  • 121.Chattopadhyay S.; Bhaumik S.; Purkayastha M.; Basu S.; Chaudhuri A.N.; Gupta S. Das Apoptosis and Necrosis in Developing Brain Cells Due to Arsenic Toxicity and Protection with Antioxidants. Toxicol. Lett. 2002, 136, 65–76. [DOI] [PubMed] [Google Scholar]
  • 122.Santra A.; Chowdhury A.; Ghatak S.; Biswas A.; Dhali G.K. Arsenic Induces Apoptosis in Mouse Liver Is Mitochondria Dependent and Is Abrogated by N-Acetylcysteine. Toxicol. Appl. Pharmacol. 2007, 220, 146–155. doi: 10.1016/j.taap.2006.12.029 [DOI] [PubMed] [Google Scholar]
  • 123.Shen Z.Y.; Shen J.; Cai W.J.; Hong C.; Zheng M.H. The Alteration of Mitochondria Is an Early Event of Arsenic Trioxide Induced Apoptosis in Esophageal Carcinoma Cells. Int. J. Mol. Med. 2000, 5, 155–163. doi: 10.3892/ijmm.5.2.155 [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Abeer El Wakil

12 Feb 2024

PONE-D-23-41249Analysis of arsenic-modulated expression of hypothalamic estrogen receptor, thyroid receptor, and peroxisome proliferator-activated receptor gamma mRNA and simultaneous mitochondrial morphology and respiration rates in the mouse.PLOS ONE

Dear Dr. Jocsak,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

Please submit your revised manuscript by Mar 28 2024 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols.

We look forward to receiving your revised manuscript.

Kind regards,

Abeer El Wakil, PhD

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at 

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and 

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. Please include captions for your Supporting Information files at the end of your manuscript, and update any in-text citations to match accordingly. Please see our Supporting Information guidelines for more information: http://journals.plos.org/plosone/s/supporting-information.

Additional Editor Comments:

The concept of the present study seems interesting. The aim of the study is to investigate the subtoxic effects of the environmental toxicant arsenic exerted on the main neuroendocrine centre in mice.

The structure of the article fulfills the structure of a research article. The paper's title reflects the main theme of the paper. Generally speaking, the manuscript is well written and well presented. However, it does not meet the standards set forth by Plos One in its actual form. I highly recommend the authors to precisely address the feedback I provided on how to improve the manuscript as well as the reviewers’ concerns.

My suggestions include:

- The introductory part must be reduced.

- The abbreviations must be revised.

- The 50�m thickness of tissue sections needs to be justified.

- The conclusion must be concise.

- The references must be updated and reduced.

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Partly

Reviewer #2: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: No

Reviewer #2: I Don't Know

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: No

Reviewer #2: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: No

Reviewer #2: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: Reviewers' comments to editor:

This manuscript describes "Analysis of arsenic-modulated expression of hypothalamic estrogen receptor, thyroid receptor, and peroxisome proliferator-activated receptor gamma mRNA and simultaneous mitochondrial morphology and respiration rates in the mouse.".

The topic is of interest. However, there are several concerns about the study, and needs to be largely improved.

1- The part of the review lacks epidemiological research evidence and systematic review of this research field, especially concerning the mechanism. It is suggested to supplement relevant studies to better propose research hypotheses. In addition, the length of the introduction should be reduced and logical.

2- Some words and paragraph are not accurate and normal, please check and modify. Such as Fig 1. at line165 should be revised. There is no space between segments. In addition, the first time the abbreviation appears, the full name should be given. Please check and modify.

3- Why 18-day-old C57BL/6 mice were designed in animal experiments? Representativeness or not?

4- In the part of animal model, please state the basis for the design of As dosage and intervention time or supplement references. In addition, how are experiments grouped in this study? And the basis for grouping is not clearly stated.

5-In the part of materials and methods, the description of main reagents and determination indicators is missing, such as reagent model, manufacturer and other information. In addition, please added the IHC and PCR indexs, etc.

6- In the materials and methods section 2.9., there is a lack of detailed description of statistical analysis, Whether the statistical analysis of normal and non-normal needs to distinguish the description, please supplement the description of the homogeneity of variance and non-homogeneity.

7- The writing of relevant statistical indicators should be standardized, with Spaces between symbols P<0.05 and P<0.01 in this text should be P < 0.05 and P < 0.01, respectively.

8- From the perspective of in vivo experiment, the basic exposure time description and design are lacking. In addition, it lacks the experimental support of basic pathophysiological indicators, biochemical and WB detection of key indicators in animal experiments.

9- Unit animal ethics certificate approval number needs to be added.

10- It is suggested that partial consolidation be discussed. Discussion according to experimental categories in international journals is usually unreasonable and cannot reflect the integrity and logic of scientific research

Other comments:

1. The formatting of the article is irregular and needs to be adjusted, such as paragraph alignment, abbreviations, etc. The first occurrence of the abbreviation should be the full name outside the parentheses and the abbreviation inside the parentheses.

2. The case format of p-values should be uniform, p-values should be italics in all Figures.

3. It is recommended to verify the appropriate selection and use of statistical methods.

4. All PCR result should be supplemented with gene primers information provided in the method section. The sequence of gene primers should be clearly labeled.

5. This paper lacks the latest research literature, which is almost absent in the past three years, especially the preface. The latest related research progress and description should be added in the frontier and discussion section. Without the latest research progress in this field, the research value of this topic cannot be explained.

6.It is suggested that this article should be edited and polished by native English speaking experts, or by a professional company, so as to meet the requirements of magazine publication.

Reviewer #2: The idea of ms is interesting and urgently needed to check, still not much is known on chemicals that can act on hormonal signaling. Here not only the route sex steroids mechanism of action but also thyroid signaling one is studied on hypothalamus levels. With special attention on mitochondria All the above are strong sides of the report.

40 µg, 5 mg and 10 mg- please explain the choose and calculation.

Provide histological microscopic documentation

50 µm thick tissue slices for ihc ?give robust explanation

“The age chosen represents a pre-pubertal state when the hypothalamus …” add citation.

Add study limitations.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2024 May 16;19(5):e0303528. doi: 10.1371/journal.pone.0303528.r002

Author response to Decision Letter 0


2 Apr 2024

Dear Editor,

dear Prof. Abeer El Wakil,

Thank you for your thoughtful evaluation of our manuscript. We appreciate your recognition of the interesting concept of our study. Your feedback regarding the article's structure was valuable to us. We acknowledge your suggestions for improvement, we diligently addressed each of these points to enhance the quality and alignment of our manuscript with the standards set forth by Plos One.

The abbreviations in the text have been carefully revised based on your recommendation.

In response to the suggestion to reduce the introductory part, we have made appropriate revisions to streamline the content while retaining essential information. Our article focuses on several critical aspects, and while the introduction has been condensed, we have ensured that the most important parts are retained. Nevertheless, efforts were made to trim it down as much as possible without compromising clarity or completeness. Thank you for highlighting this area for improvement.

We have addressed the need for justification regarding the 50 µm thickness of tissue sections by incorporating the necessary information (please see lines 208-211; 309-312).

Recognizing the importance of conciseness in the conclusion, we have revised it accordingly to ensure alignment with your expectations (lines 565-582).

The reviewers kindly suggested updating the reference list and including research conducted no later than three years ago. As a result, while our reference list has grown slightly, we have ensured the inclusion of recent studies and removed outdated references to maintain the relevance and currency of the literature cited.

---------------------------------------

Response to Reviewer 1 Comments

We appreciate the time and efforts by the editor and referee in reviewing this manuscript. We have addressed all issues indicated in the review report, and hope that the revised version meets the journal publication requirements.

Question 1: “The part of the review lacks epidemiological research evidence and a systematic review of this research field, especially concerning the mechanism. It is suggested to supplement relevant studies to better propose research hypotheses. In addition, the length of the introduction should be reduced and logical.”

Answer: Thank you for your valuable feedback. We appreciate your suggestion regarding the need for additional epidemiological research evidence and a systematic review of our manuscript. We have carefully considered your recommendation and have made the necessary revisions by incorporating recent epidemiological data to support our proposed research hypotheses. Additionally, we have taken steps to streamline and reduce the length of the introduction section to ensure clarity and logical flow. (please, see “Introduction”, lines 44-147)

Question 2: “Some words and paragraphs are not accurate and normal, please check and modify them. Such as Fig 1. at line 165 should be revised. There is no space between segments. In addition, the first time the abbreviation appears, the full name should be given. Please check and modify.”

Answer: We have made the necessary modifications as per your suggestions.

Question 3: “Why 18-day-old C57BL/6 mice were designed in animal experiments? Representativeness or not?”

Answer: The selection of 18-day-old mice for our study aligns with the findings of our previous investigation (please see: https://doi.org/10.1016/S0165-3806(01)00180-8) into ER expression dynamics in the developing rat primary somatosensory cortex. In both studies, we aim to elucidate the role of ERs (and TR in later studies) during critical developmental periods, albeit in different brain regions and under distinct experimental conditions.

In our previous study, we observed widespread ERα expression in the developing rat cortex at PN3, with a transition to predominantly nuclear localization observed by PN18. This developmental switch in ERα expression coincided with key neurodevelopmental events, suggesting a role for ERα in cortical maturation and organization.

Similarly, in the present study assessing the effects of arsenic exposure on hypothalamic tissue homogenates of prepubertal mice, we aim to understand the impact of environmental insults on neuroendocrine regulation during a critical developmental window. By choosing 18-day-old mice, we target a developmental stage characterized by heightened sensitivity to environmental toxins and significant maturation of hypothalamic functions.

Furthermore, both studies share a common focus on elucidating the molecular mechanisms underlying neuroendocrine regulation and homeostasis. While our previous study focused on ER expression dynamics in the cerebral cortex, the current study investigates the modulation of ER and TR mRNA expression levels, as well as PPARγ, in response to arsenic exposure in the hypothalamus.

Overall, the selection of 18-day-old mice for our present study is justified by the critical developmental stage of the hypothalamus and its susceptibility to environmental insults, similar to the rationale employed in our previous investigation of ER expression dynamics in the developing cortex. This alignment ensures consistency in experimental design and enhances the comparability of results across studies, ultimately advancing our understanding of the neurodevelopmental effects of environmental toxins such as arsenic.

Question 4: “In the part of the animal model, please state the basis for the design of As dosage and intervention time or supplement references. In addition, how are experiments grouped in this study? And the basis for grouping is not clearly stated.”

Answer: Thank you for your question, we will fully cover and address the concerns.

• As dosage: In addressing the question regarding the basis for the design of As dosage and intervention time, we provided and expanded modifications within the main text (please, see lines 118-142).

• Intervention time: To address the question concerning intervention time, it's imperative to delve into the comprehensive process of our study, which commenced with a pilot study in 2012. We`ve incorporated the necessary links of the studies used, please see below.

Stages of the Study:

• 1st Stage:

In the initial phase, a pilot study (Scalise et al., 2012; https://doi.org/10.1556/avet.2012.023) was conducted under in vitro conditions to examine the expressions of receptors TR and ER with their respective ligands. The primary objective of this stage was to establish the relevant intervention time window, which ranged from 6 to 16 hours for PCR; 18 hours for Western blot analyses. This stage provided crucial insights into the experimental design.

• 2nd Stage:

Building upon the insights gained from the pilot study, the research progressed to the next phase. Utilizing the same experimental conditions and setup, cerebellar granule cells were employed to further investigate the expression of hormone receptors resulting from exposure to various endocrine disruptors (Jocsak et al., 2016; https://doi.org/10.3390/ijerph13060619; Kiss et al., 2018; https://doi.org/10.3390/ijms19051440; Somogy V., 2016; http://hdl.handle.net/10832/1504; Jocsak et al., 2019; https://doi.org/10.3390/brainsci9120359). This phase aimed to refine the expression time window established in the pilot study, incorporating additional data from studies on endocrine disruptors.

• 3rd Stage:

Following the investigations with cerebellar granule cells, the study transitioned to in vitro experiments involving hypothalamic cells. This marked the third stage of the research, wherein the focus shifted to assessing hormone receptor expression in a relevant cellular context.

• 4th Stage:

In the final stage, the research moved to an in vivo setting, specifically focusing on hypothalamic cells. By administering As in vivo, we aimed to replicate the conditions observed in the earlier stages while investigating the effects on hormone receptor expression. The intervention time of 6 hours was deemed appropriate for obtaining reliable PCR results, ensuring consistency across in vitro and in vivo experiments.

• Grouping: In this study, a total of 72 animals were employed to investigate the effects of varying doses on multiple biological endpoints. Each treatment group consisted of 6 animals, with three distinct doses administered: 10 µm, 5 mg, and 10 mg.

Therefore, the total number of animals utilized can be calculated as follows:

Total number of animals: n=6 animals/ group × 4 treatments (3 doses+ctrl) × 3 methods (PCR, MRR, EM) making = 6 × 4 × 3 = 72 animals.

Question 5: “In the part of materials and methods, the description of main reagents and determination indicators is missing, such as reagent model, manufacturer and other information. In addition, please added the IHC and PCR indexs, etc.”

Answer: Thank you for your feedback. We have carefully revised the materials and methods section of the manuscript, ensuring that all necessary information regarding main reagents, including reagent models, manufacturers, and catalogue numbers, has been added. Additionally, we have included details such as PCR primer sequences (please see Table 1, lines 239-240) to enhance the clarity and completeness of the experimental procedures.

Regarding your mention of IHC and PCR indexes, we would appreciate further clarification on what specific information you are referring to.

Questions 6: “In the materials and methods section 2.9., there is a lack of a detailed description of statistical analysis, Whether the statistical analysis of normal and non-normal needs to distinguish the description, please supplement the description of the homogeneity of variance and non-homogeneity.”

Answer: Thank you for bringing this to our attention. We have carefully reviewed the materials and methods section, specifically the part regarding statistical analysis. Following your suggestion, we have provided a more detailed description of the statistical analysis (please see lines 318-327).

Question 7: “The writing of relevant statistical indicators should be standardized, with Spaces between symbols P<0.05 and P<0.01 in this text should be P < 0.05 and P < 0.01, respectively”.

Answer: Thank you for your feedback. We have taken your suggestion into consideration and made the necessary adjustments to the writing of relevant statistical indicators in the text.

Question 8: “From the perspective of in vivo experiments, the basic exposure time description and design are lacking. In addition, it lacks the experimental support of basic pathophysiological indicators, and biochemical and WB detection of key indicators in animal experiments.”

Answer: Thank you for the question, we have duly addressed the concern by providing a comprehensive explanation. We acknowledge that our study was primarily focused on basic research, which may explain the lack of inclusion of pathophysiological indicators. In our previous research (please, see above), we utilized both Western blot and PCR analyses, revealing occasional discrepancies between the two methods, a phenomenon observed in other studies as well. These disparities likely stem from the intricate cellular responses to external stimuli, which prompt diverse mechanisms within cells. One such initial response is the modulation of mRNA expression levels, a parameter assessable through PCR. Obviously, changes in mRNA levels may not consistently correlate with alterations in protein abundance, illustrating the complexity of cellular regulation. This discrepancy may arise due to various automatic mechanisms governing mRNA dynamics, independent of concurrent protein changes. For instance, cellular responses to stimuli may entail rapid adjustments in mRNA levels, followed by delayed or no changes in protein modifications (Zeisel et al., 2011 [doi.org/10.1038/msb.2011.62]). Notably, PCR often offers a more immediate and determined reflection of these responses compared to WB, particularly evident in the context of certain receptors such as estrogen receptors and thyroid hormone receptors. These receptors, partially housed within vesicles in an active state, exhibit swift mRNA expression changes in response to stimuli, potentially preceding discernible alterations in protein levels (Dominguez & Micevych, 2010 [doi.org/10.1523/JNEUROSCI.1038-10.2010]). Consequently, while external stimuli induce mRNA expression changes, protein levels may remain unaltered, highlighting the distinct temporal dynamics between mRNA and protein responses. In other words, the biological target of a chemical stimulus can be better determined by the activation of transcription (than by the end product protein, the latter which is produced in actually relevant amounts), even more so if we examine transcription factors, like ERs and TRs. Thus, PCR emerges as a valuable tool for capturing immediate cellular responses to stimuli, complementing WB analyses which may not always align with PCR findings. This preference for PCR underscores its capacity to offer prompt and informative insights into cellular dynamics in response to external factors, including endocrine disruptors.

Question 9: “Unit animal ethics certificate approval number needs to be added.”

Answer: Thank you for your attention to detail. The requested approval numbers can be found in the text at lines 177-178. We understand the importance of ensuring clarity and transparency in our reporting. To facilitate your review, we will ensure that the approval number is highlighted for your convenience in the revised manuscript.

Question 10: “It is suggested that partial consolidation be discussed. Discussion according to experimental categories in international journals is usually unreasonable and cannot reflect the integrity and logic of scientific research.”

Answer: Thank you for providing feedback on the discussion section. We would like to inform you that our paper faced rejection from another journal, where we received advice to divide the discussion into segments. Considering the previous feedback, we decided to adopt a segmented format in our current submission to ensure alignment with the preferences of the journal and its reviewers. We recognize that different journals may have varying preferences regarding the organization of discussion sections. If the Editor and the Reviewers agree, we are certainly open to considering a merged discussion format again. Our priority is to ensure that the manuscript meets the standards and preferences of the journal while effectively communicating the scientific findings.

Other comments:

1. “The formatting of the article is irregular and needs to be adjusted, such as paragraph alignment, abbreviations, etc. The first occurrence of the abbreviation should be the full name outside the parentheses and the abbreviation inside the parentheses.”

We have addressed the irregularities in the article's formatting.

2. “The case format of p-values should be uniform, p-values should be italics in all Figures.”

Your attention to detail is greatly appreciated, we have addressed the concerns.

3. “It is recommended to verify the appropriate selection and use of statistical methods.”

We have carefully reviewed and verified the selection and application of statistical methods in the text body to ensure accuracy and reliability.

4. “All PCR result should be supplemented with gene primers information provided in the method section. The sequence of gene primers should be clearly labeled.”

We have addressed your concern by supplementing all PCR results with gene primer information provided in the method section (please see Table 1., lines 239-240).

5. “This paper lacks the latest research literature, which is almost absent in the past three years, especially the preface. The latest related research progress and description should be added in the frontier and discussion section. Without the latest research progress in this field, the research value of this topic cannot be explained.”

We have taken your comments into account and updated the literature review to incorporate the latest research findings from the past three years.

6. “It is suggested that this article should be edited and polished by

Attachment

Submitted filename: PLOS_RESPONSE_RVW2_FINAL.docx

pone.0303528.s004.docx (18.8KB, docx)

Decision Letter 1

Abeer El Wakil

26 Apr 2024

Analysis of arsenic-modulated expression of hypothalamic estrogen receptor, thyroid receptor, and peroxisome proliferator-activated receptor gamma mRNA and simultaneous mitochondrial morphology and respiration rates in the mouse.

PONE-D-23-41249R1

Dear Dr. Jocsak,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice will be generated when your article is formally accepted. Please note, if your institution has a publishing partnership with PLOS and your article meets the relevant criteria, all or part of your publication costs will be covered. Please make sure your user information is up-to-date by logging into Editorial Manager at Editorial Manager® and clicking the ‘Update My Information' link at the top of the page. If you have any questions relating to publication charges, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Abeer El Wakil, PhD

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #2: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #2: My comments were included in the revision. Based on other opinions the decision will be made. ...................................................................................................................................................................................................................................................

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #2: No

**********

Acceptance letter

Abeer El Wakil

3 May 2024

PONE-D-23-41249R1

PLOS ONE

Dear Dr. Jocsak,

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now being handed over to our production team.

At this stage, our production department will prepare your paper for publication. This includes ensuring the following:

* All references, tables, and figures are properly cited

* All relevant supporting information is included in the manuscript submission,

* There are no issues that prevent the paper from being properly typeset

If revisions are needed, the production department will contact you directly to resolve them. If no revisions are needed, you will receive an email when the publication date has been set. At this time, we do not offer pre-publication proofs to authors during production of the accepted work. Please keep in mind that we are working through a large volume of accepted articles, so please give us a few weeks to review your paper and let you know the next and final steps.

Lastly, if your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

If we can help with anything else, please email us at customercare@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Professor Abeer El Wakil

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Raw data. It was obtained from electron microscopy (JEOL USA, Peabody, MA, USA).

    (XLSX)

    pone.0303528.s001.xlsx (161.6KB, xlsx)
    S2 Raw data. It was measured by a Clark-type oxygen electrode (Oxytherm, Hansatech Instruments, Norfolk, UK).

    (XLSX)

    pone.0303528.s002.xlsx (20.1KB, xlsx)
    S3 Raw data. It obtained from PCR.

    (XLSX)

    pone.0303528.s003.xlsx (48.6KB, xlsx)
    Attachment

    Submitted filename: PLOS_RESPONSE_RVW2_FINAL.docx

    pone.0303528.s004.docx (18.8KB, docx)

    Data Availability Statement

    All relevant data are within the paper and its Supporting Information files.


    Articles from PLOS ONE are provided here courtesy of PLOS

    RESOURCES