The A140D mutant exhibits distinct biochemical properties compared to the wild-type enzyme:
Activity | Wild-Type GSTO1 | A140D Mutant | Source |
---|---|---|---|
Arsenic reduction | High | Reduced | |
Dehydroascorbate reductase | Active | Partially active | |
Deglutathionylation | Robust | Diminished |
This polymorphism also affects interactions with signaling proteins like β-actin, impairing redox homeostasis in cellular models .
Cancer: GSTO1 overexpression in tumors (e.g., colorectal, breast) correlates with chemoresistance. The A140D variant may alter drug metabolism, though its direct role remains under investigation .
Neurodegeneration: Reduced GSTO1 activity is linked to Alzheimer’s and Parkinson’s diseases due to impaired oxidative stress management .
Inflammation: GSTO1 regulates TLR4-mediated inflammatory responses. Mutants show attenuated ROS production in macrophages, impacting innate immunity .
Inhibitor Development: Small-molecule inhibitors (e.g., C1-27 and 25) covalently bind GSTO1’s active site, suppressing cancer cell growth and enhancing cisplatin efficacy .
Cellular Models: siRNA knockdown of GSTO1 reduces viability in HCT116 colon cancer cells, validating it as a therapeutic target .
Biomarker Potential: GSTO1 expression levels are reduced in chronic obstructive pulmonary disease (COPD) and linked to arsenic toxicity susceptibility .
Polymorphism Studies: The A140D variant is associated with altered drug metabolism and increased risk of renal cell carcinoma .
GSTO1 (Glutathione S-transferase Omega 1) is an atypical member of the GST superfamily with distinct structural and functional attributes. Unlike other GSTs, GSTO1 contains an active site cysteine residue that confers unique catalytic capabilities, particularly for redox reactions . This structural distinction enables GSTO1 to perform novel roles unrelated to the detoxification functions of typical GSTs, including deglutathionylation and thioltransferase activities that contribute to cellular redox regulation . The enzyme's structure incorporates an N-terminal thioredoxin domain and a C-terminal α-helical domain, forming a unique active site that explains its functional versatility in various biological contexts .
Several polymorphisms have been identified in the human GSTO1 gene, with the most extensively studied being rs4925 (A140D), which causes the substitution of alanine with aspartic acid at position 140. This polymorphism significantly affects the enzyme's deglutathionylation kinetics, as demonstrated in studies showing differential reaction rates among GSTO1 genetic variants . Other important polymorphisms include those affecting the gene's regulatory regions that may influence expression levels. These genetic variations have been associated with susceptibility to various conditions, most notably Alzheimer's disease (AD), where certain GSTO1 polymorphisms have been linked to age of disease onset . The rs4925 polymorphism has also been investigated in relation to cancer risk, with studies examining its association with childhood acute lymphoblastic leukemia and urothelial carcinoma .
GSTO1 expression is regulated through complex mechanisms involving both cis and trans-acting elements. Studies using BXD recombinant inbred mice have confirmed that variation in Gsto1 expression is primarily controlled by cis-expression quantitative trait loci (eQTLs) . The gene shows tissue-specific expression patterns, with particularly notable levels in the central nervous system, where its expression correlates with several neuroanatomical traits including cortical gray matter volume and hippocampus mossy fiber pathway volume . In disease states such as Alzheimer's disease, GSTO1 expression levels are significantly decreased compared to healthy controls . Regulatory analysis has identified over 2,000 genes whose expression correlates with GSTO1, suggesting complex transcriptional networks that respond to cellular stressors such as oxidative stress and inflammation . Experimental approaches to study this regulation include allele-specific expression assays such as SNaPshot, which have been used to quantify differential expression between Gsto1 alleles in hybrid offspring .
Mechanistically, GSTO1's role in modulating oxidative stress and inflammation—both critical processes in neurodegeneration—provides biological plausibility for its contribution to AD pathology. Transcriptomic analyses identified 2,168 genes correlated with GSTO1 expression, many enriched in pathways relevant to neurodegenerative diseases . Furthermore, GSTO1 expression levels correlated with several central nervous system traits in mouse models, including glial acidic fibrillary protein levels in the caudate putamen and hippocampal structural features, suggesting its involvement in maintaining neuronal integrity .
GSTO1 knockout models have provided crucial insights into neuroinflammatory processes, revealing its complex role in modulating inflammation across different contexts. In inflammation studies, GSTO1-deficient mice show altered responses to inflammatory stimuli, with the direction of effect depending on the specific disease model . For instance, GSTO1 deficiency ameliorates the inflammatory response stimulated by lipopolysaccharide (LPS) and attenuates inflammation-induced metabolic dysfunction in high-fat diet models . These findings align with GSTO1's identified pro-inflammatory role in Toll-like receptor 4 (TLR4) signaling.
Conversely, in a dextran sodium sulfate model of inflammatory bowel disease, GSTO1-deficient mice exhibited more severe inflammation than wild-type counterparts, with increased bacterial escape from the colon into the lymphatic system . This response pattern mirrors that observed in TLR4 and MyD88 deficient mice, confirming GSTO1's importance in TLR4-like pro-inflammatory responses in vivo . In neuroinflammation specifically, studies highlight GSTO1's role in astrocyte-mediated inflammatory processes, which are crucial in various neurological diseases including AD, Parkinson's disease, and amyotrophic lateral sclerosis . The knockout models thus demonstrate that GSTO1's influence on inflammation is context-dependent and tissue-specific, suggesting that therapeutic targeting must consider these nuanced effects.
For studying GSTO1 in primary astrocyte cultures, several experimental approaches have proven effective. The isolation protocol described in the literature involves obtaining cortical astrocytes from newborn mice, with careful removal of meninges followed by mechanical and enzymatic digestion of cortical tissue . Cells are then cultured in poly-L-lysine-coated flasks using Dulbecco's modified Eagle's medium supplemented with antibiotics and fetal bovine serum. This culture system maintains mixed glial cultures for 2-3 weeks at 37°C in a humidified atmosphere (5% CO2, 95% air) .
For manipulating GSTO1 expression in these cultures, RNA interference techniques have been successfully employed. Specifically, Gsto1 knockdown in primary astrocytes has been achieved and validated using quantitative PCR (qPCR) . This approach allows researchers to examine the consequences of reduced GSTO1 expression on astrocyte function, inflammatory responses, and potential downstream genes.
To comprehensively assess GSTO1's role, researchers should combine these approaches with:
Transcriptomic analysis (RNA-seq) to identify genes affected by GSTO1 modulation
Immunocytochemical characterization to confirm astrocyte purity and GSTO1 expression patterns
Functional assays measuring inflammatory mediator production (e.g., cytokine ELISAs)
Oxidative stress assessments (e.g., ROS measurements, glutathione ratio determination)
Co-culture systems with neurons to evaluate non-cell autonomous effects
These methodologies collectively enable detailed investigation of GSTO1's functions in astrocytes and its potential contributions to neuroinflammatory processes relevant to neurodegenerative diseases .
GSTO1 deletion profoundly impacts tumor growth in xenograft models, demonstrating its significant role in cancer progression. In studies using CRISPR/Cas9 to knockout GSTO1 in cancer cell lines (HCT116 and U-87 MG), researchers observed remarkable inhibition of tumor growth following implantation in mouse models . HCT116 GSTO1 knockout cells showed significantly reduced tumor burden compared to non-targeted wild-type cells, with survival significantly prolonged (log-rank p=0.032) by up to four weeks in the knockout group . Even more strikingly, U-87 MG cells lacking GSTO1 completely failed to form tumors, with no detectable growth even after reinjection and extended monitoring for an additional four weeks .
These dramatic in vivo effects contrast with more modest impacts observed in standard 2D cell culture. While U-87 MG GSTO1 knockout cells showed reduced proliferation and colony formation in vitro, HCT116 cells lacking GSTO1 exhibited minimal growth differences in 2D culture despite their substantial tumor suppression in vivo . This discrepancy highlights the importance of the tumor microenvironment in mediating GSTO1's effects on cancer progression and underscores the limitations of conventional in vitro systems for fully capturing GSTO1's role in tumor biology .
GSTO1 influences cancer progression through multiple molecular pathways revealed by comprehensive multi-omics analyses. Transcriptional profiling of GSTO1 knockout cells identified significant suppression of genes involved in cell cycle checkpoints and target genes for Myc and E2F transcription factors, both critical regulators of cell proliferation . A strong negative correlation was observed between GSTO1 deletion and Myc transcriptional signatures, suggesting that GSTO1 may support cancer progression partly through sustaining Myc-dependent gene expression programs .
Simultaneously, GSTO1 knockout led to enrichment of genes and proteins involved in cell adhesion and interferon response pathways (both IFNα and IFNγ), indicating GSTO1 may normally suppress anti-tumor immune responses . One particularly notable downstream effect is GSTO1's regulation of tissue factor (F3) expression. F3 is associated with poor patient survival and tumor progression in multiple cancers and represents a known risk factor for metastasis . Mechanistically, F3 transcription appears regulated by IL-1β, whose secretion decreases upon GSTO1 inhibition, establishing a GSTO1-IL-1β-F3 signaling axis that promotes cancer progression .
Regarding drug resistance, GSTO1 has been implicated in detoxification processes and protection against oxidative stress induced by treatments. Notably, synthetic lethality screens identified several FDA-approved drugs showing enhanced efficacy when combined with GSTO1 inhibition or deletion, suggesting GSTO1 targeting could overcome resistance mechanisms and enhance therapeutic efficacy in combination treatments .
Effective measurement of GSTO1 expression for patient stratification in oncology requires a multi-layered approach incorporating genomic, transcriptomic, and proteomic analyses. Based on the research findings, the following methodological framework is recommended:
Genotyping for key GSTO1 polymorphisms (e.g., rs4925) that may affect enzyme activity and cancer susceptibility
Analysis of copy number variations that might contribute to GSTO1 overexpression in tumors
Quantitative RT-PCR for precise measurement of GSTO1 mRNA levels in tumor samples
RNA-seq to assess not only GSTO1 expression but also its correlation with the expression of the 2,000+ genes identified as part of its regulatory network
Analysis of GSTO1-related gene signatures that could serve as more robust biomarkers than GSTO1 expression alone
Immunohistochemistry to assess GSTO1 protein expression and localization within tumor tissues
Western blotting for quantitative determination of GSTO1 protein levels
Activity assays to measure functional GSTO1 enzyme activity, which may more accurately reflect its biological impact than expression alone
Measurement of downstream effectors like tissue factor (F3) that have been directly linked to GSTO1's effects on tumor progression
Assessment of interferon response gene expression, which shows strong positive correlation with GSTO1 knockout and may serve as an inverse marker of GSTO1 activity
This comprehensive profiling would enable effective patient stratification for clinical trials of GSTO1 inhibitors and potentially identify those most likely to benefit from GSTO1-targeted therapies or combination treatments exploiting synthetic lethality relationships .
Generating and validating GSTO1 knockout models requires rigorous methodology to ensure complete elimination of functional protein and appropriate controls. Based on published research, the following approach is recommended:
Generation of GSTO1 Knockout Models:
CRISPR/Cas9 gene editing has proven effective for creating GSTO1 knockouts in multiple cell lines (A172, HCT116, U87-MG) . Design guide RNAs targeting early exons (e.g., exon 2) of the GSTO1 gene to ensure functional disruption.
For animal models, either conventional gene targeting or CRISPR/Cas9-mediated germline editing can be employed, with careful consideration of genetic background effects on phenotypic manifestation .
Validation Strategy:
Genomic verification: Confirm modifications through Sanger sequencing to characterize the exact nature of genomic edits (deletions, frameshifts). Studies have validated that genome editing caused deletions and frameshifts in exon 2 of the GSTO1 gene .
Transcriptional verification: Quantitative RT-PCR to confirm absence or significant reduction of GSTO1 mRNA .
Protein verification: Western blotting using specific antibodies to demonstrate complete loss of GSTO1 protein .
Functional validation: Assess enzymatic activity using GSTO1-specific substrates to confirm loss of function.
Clone selection: Generate and characterize multiple independent knockout clones to avoid clone-specific artifacts. The published research examined three single clones for each cell line to avoid characterization of single clone behaviors .
Important Considerations:
Include appropriate non-targeted wild-type controls processed through the same procedures to account for potential effects of the gene editing process itself .
For in vivo studies, carefully select the appropriate animal model and housing conditions, as demonstrated in studies using ICRSC or NOD/SCID mice for xenograft experiments .
Phenotypic comparison across multiple knockout clones is essential, as the research noted variation in growth rates among HCT116 GSTO1 KO clones, highlighting potential clone-specific behaviors .
This comprehensive approach ensures the generation of reliable knockout models that can accurately reveal GSTO1's biological functions in different experimental contexts.
Studying GSTO1 mutant enzyme kinetics requires specialized methodological approaches that account for its unique catalytic properties. Based on research findings, the following methodological framework is recommended:
Enzyme Preparation:
Recombinant expression systems (E. coli, insect cells, or mammalian cells) for producing wild-type and mutant GSTO1 proteins
Affinity purification (e.g., GST-tag or His-tag) followed by size exclusion chromatography to ensure high purity
Verification of protein integrity through circular dichroism to assess proper folding
Kinetic Assays:
Deglutathionylation assays: The genetic variants of GSTO1-1 exhibit significantly different deglutathionylation reaction kinetics, as noted in Table 1 of the referenced research . Substrates like S-(4-nitrophenacyl)glutathione can be used with spectrophotometric detection.
Thiol transferase activity: Monitor the reaction rate using NADPH consumption as an indirect measure of activity
Monomethylarsonate reductase activity: Important for studying GSTO1's role in arsenic biotransformation, particularly relevant for cancer studies
Experimental Parameters to Determine:
Michaelis-Menten kinetics (Km, Vmax, kcat)
Substrate specificity profiles comparing different physiological and xenobiotic substrates
pH and temperature optima and stability
Effects of potential inhibitors on enzyme kinetics
Advanced Analytical Techniques:
Stopped-flow spectroscopy for rapid kinetics
Isothermal titration calorimetry (ITC) for binding thermodynamics
Hydrogen-deuterium exchange mass spectrometry to examine structural dynamics affecting catalysis
X-ray crystallography or cryo-EM to visualize structural changes in mutant enzymes
Comparative Analysis:
Create comprehensive tables comparing kinetic parameters between wild-type and various GSTO1 mutants, similar to the referenced Table 1 showing significantly different deglutathionylation reaction kinetics among genetic variants . This approach allows for systematic evaluation of how specific mutations affect the catalytic properties of GSTO1, providing insights into structure-function relationships and potential implications for disease susceptibility or therapeutic targeting.
Multi-omics approaches offer powerful means to comprehensively understand GSTO1 function across biological contexts. Based on published research, the following integrated framework is recommended:
Transcriptomic Analysis:
RNA sequencing (RNA-seq) to identify differentially expressed genes in GSTO1 knockout or overexpression models
Bru-seq (bromouridine sequencing) to measure nascent mRNA synthesis rates, providing insights into direct transcriptional effects of GSTO1 modulation
Correlation analysis to identify genes whose expression patterns mirror GSTO1, as demonstrated in studies that identified 2,168 genes highly correlated with Gsto1 expression
Proteomic Analysis:
Mass spectrometry-based proteomics to quantify protein-level changes in response to GSTO1 manipulation
Phosphoproteomics to identify signaling pathways affected by GSTO1
Interactome analysis using immunoprecipitation coupled with mass spectrometry to identify GSTO1-interacting proteins
Metabolomic/Redox Analysis:
Targeted metabolomics focusing on glutathione and related metabolites
Redox proteomics to identify proteins undergoing GSTO1-dependent modifications
Lipid peroxidation measurements to assess oxidative stress consequences
Integration Strategies:
Pathway Analysis: Integrate data across omics layers to identify consistently affected biological processes. Research has revealed enrichment in cell adhesion molecules and interferon response pathways in GSTO1 knockout cells .
Network Analysis: Construct multi-omics networks centered on GSTO1 to identify hub genes/proteins that may mediate its effects.
Causal Inference: Use statistical approaches like partial correlation analysis (as described in the research to exclude the effect of linkage disequilibrium) to distinguish direct from indirect effects .
Validation Approaches:
Functional assays targeting key pathways identified through multi-omics
CRISPR screens to identify synthetic lethal interactions with GSTO1, as demonstrated in research showing synthetic lethality between GSTO1 inhibition and several FDA-approved drugs
In vivo validation of key findings in appropriate disease models
This comprehensive multi-omics strategy provides a systems-level understanding of GSTO1 function, revealing its roles in diverse cellular processes including inflammation, oxidative stress response, cell cycle regulation, and immune modulation, while identifying potential therapeutic targets and biomarkers .
Developing selective GSTO1 inhibitors requires careful consideration of several critical factors based on the enzyme's unique structural and functional properties. The following framework outlines key considerations for successful inhibitor development:
Structural Considerations:
Active site targeting: GSTO1 contains a distinctive active site cysteine residue that differentiates it from other GSTs. Inhibitors that covalently bind to this site have demonstrated efficacy in modulating inflammatory responses, as shown in studies with small molecule inhibitors .
Selectivity: Design compounds that exploit structural differences between GSTO1 and other GST family members to achieve isoform selectivity, reducing off-target effects.
Binding mode: Consider both reversible and irreversible inhibitors, noting that covalent modifiers targeting the active site cysteine have shown promise in preclinical models .
Pharmacological Properties:
Potency: Optimize compounds for high binding affinity and functional inhibition of GSTO1 enzymatic activities.
Pharmacokinetics: Design inhibitors with appropriate bioavailability, tissue distribution (especially CNS penetration for neurodegenerative disease applications), and metabolic stability.
Safety profile: Consider potential toxicity related to off-target effects and consequences of long-term GSTO1 inhibition, particularly given its context-dependent roles in inflammation .
Disease Context Considerations:
Context-dependent roles: GSTO1 exhibits contrasting roles in different inflammatory conditions—ameliorating LPS-induced inflammation while exacerbating inflammatory bowel disease when deleted . Inhibitor development should account for these context-specific effects.
Tissue specificity: Consider developing inhibitors with tissue-targeted delivery to focus effects on relevant disease sites.
Combination potential: Explore synergistic combinations with existing therapies, as research has identified synthetic lethality between GSTO1 inhibition and several FDA-approved drugs in cancer models .
Validation Strategies:
Biomarker development: Establish pharmacodynamic markers to confirm target engagement in preclinical and clinical studies.
Phenotypic confirmation: Validate that GSTO1 inhibitors recapitulate key phenotypes observed in genetic knockout models, such as reduced tumor growth or modulated inflammatory responses .
These considerations provide a comprehensive framework for developing selective GSTO1 inhibitors as therapeutic agents for conditions including cancer and inflammatory disorders, where research has validated GSTO1 as a promising therapeutic target .
Designing clinical trials for GSTO1-targeted therapies in neurodegenerative diseases requires careful consideration of multiple factors informed by preclinical research findings. The following framework outlines key elements for effective trial design:
Patient Selection and Stratification:
Genetic stratification: Screen patients for GSTO1 polymorphisms associated with disease risk or age of onset, particularly those linked to Alzheimer's disease susceptibility .
Expression profiling: Consider measuring GSTO1 expression levels in accessible biospecimens as a potential enrichment strategy.
Disease stage: Initially focus on early-stage disease or even presymptomatic high-risk individuals (based on genetic and biomarker profiles) given GSTO1's potential role in disease onset rather than late-stage progression .
Biomarker Strategy:
Target engagement: Develop assays to confirm inhibition of GSTO1 activity in accessible tissues.
Pathway markers: Monitor inflammatory mediators and oxidative stress markers associated with GSTO1 function.
Disease-specific biomarkers: Include established markers of neurodegeneration (e.g., amyloid, tau, neurofilament light chain) to assess disease modification.
Neuroimaging: Incorporate structural and functional imaging given the correlation between GSTO1 expression and neuroanatomical traits like cortical gray matter volume and hippocampus mossy fiber pathway volume .
Trial Design Considerations:
Adaptive design: Implement interim analyses to identify responsive subgroups early.
Duration: Plan for extended observation periods given the slowly progressive nature of neurodegenerative diseases.
Endpoints: Include sensitive cognitive assessments and functional measures alongside biomarker outcomes.
Safety monitoring: Pay particular attention to inflammatory responses given GSTO1's context-dependent effects on inflammation .
Mechanistic Considerations:
Astrocyte-specific effects: Consider the role of GSTO1 in astrocyte-mediated inflammation and oxidative stress when designing outcome measures .
Pathway specificity: Assess effects on downstream genes identified in correlation studies (the 2,168 genes whose expression was highly correlated with GSTO1) .
Combination potential: Evaluate combinations with other disease-modifying approaches based on mechanistic rationale.
This framework provides a comprehensive approach to clinical trial design for GSTO1-targeted therapies in neurodegenerative diseases, leveraging insights from genetic, molecular, and preclinical studies to maximize the likelihood of detecting therapeutic benefit while ensuring patient safety.
Targeting GSTO1 therapeutically presents both opportunities and challenges that vary significantly across disease contexts. Understanding these context-specific considerations is essential for safe and effective therapeutic development:
Inflammatory Conditions:
Context-dependent effects: GSTO1 exhibits contrasting roles in different inflammatory models. While GSTO1 deficiency ameliorates LPS-induced inflammation and attenuates high-fat diet inflammatory impacts, it exacerbates inflammation in dextran sodium sulfate models of inflammatory bowel disease . These contrasting effects highlight the need for careful indication selection.
Bacterial defense considerations: GSTO1-deficient mice show increased escape of bacteria from the colon into the lymphatic system in inflammatory bowel disease models, suggesting potential infection risks with GSTO1 inhibition in certain contexts .
TLR4 pathway interactions: GSTO1's role in TLR4-like pro-inflammatory responses suggests that inhibition could impair certain immune defense mechanisms while benefiting inflammatory conditions where this pathway drives pathology .
Neurodegenerative Diseases:
Astrocyte-mediated effects: Given GSTO1's role in astrocyte-mediated inflammation and oxidative stress responses relevant to multiple neurodegenerative diseases, inhibition effects may vary across different neurological conditions .
Disease stage considerations: GSTO1 inhibition may have different effects at different disease stages, with potentially greater benefit in early disease before extensive neurodegeneration.
Genetic background influence: The impact of GSTO1 inhibition may vary based on patient genetic background, particularly regarding polymorphisms associated with disease risk or age of onset .
Cancer:
Tissue-specific effects: While GSTO1 knockout dramatically reduced tumor growth in multiple cancer models, the effect was more pronounced in U-87 MG (glioblastoma) cells than HCT116 (colorectal) cells in vitro, suggesting tissue-specific dependencies .
Combination therapy considerations: Synthetic lethality observed between GSTO1 inhibition and several FDA-approved drugs suggests promising combination therapy approaches, but also potential for enhanced toxicity .
Patient selection importance: GSTO1 expression varies across cancer types and individual tumors, necessitating biomarker-guided patient selection.
Universal Considerations:
Long-term inhibition risks: Given GSTO1's role in redox homeostasis and inflammation modulation, chronic inhibition could have unpredictable long-term consequences requiring careful monitoring.
Developmental implications: The roles of GSTO1 in normal development and tissue homeostasis should be considered, particularly for therapies intended for long-term administration.
These multifaceted considerations underscore the importance of disease-specific approaches to GSTO1 targeting, with careful attention to biomarker development, patient selection, and context-appropriate dosing strategies .
Glutathione S-Transferase Omega 1 (GSTO1) is an enzyme belonging to the omega class of the glutathione S-transferase (GST) family. These enzymes play a crucial role in the detoxification of endogenous and xenobiotic compounds by catalyzing the conjugation of glutathione to various substrates. GSTO1 is particularly notable for its involvement in the reduction of monomethylarsonate, an intermediate in the pathway of arsenic biotransformation .
GSTO1 is a homodimeric protein found in the cytoplasm and exhibits glutathione-dependent thiol transferase and dehydroascorbate reductase activities . The enzyme contains four conserved cysteine residues (C32, C90, C191, C236), which are essential for its catalytic activity . These residues are often mutated in patients with associated diseases, highlighting their importance in the enzyme’s function.
Several polymorphisms in the coding regions of the human GSTO1 gene have been identified. One notable variant is the A140D mutation, which results from an alanine-to-aspartate substitution at amino acid position 140 . This mutation produces a variant with altered enzymatic properties and is often studied in the context of its role in oxidative stress and its potential link to diseases such as Alzheimer’s, vascular dementia, and stroke .
Recombinant human GSTO1 is typically expressed in Escherichia coli (E. coli) systems. The recombinant protein is often fused with an amino-terminal hexahistidine tag to facilitate purification . The recombinant form retains the enzymatic activities of the native protein and is used extensively in research to study its biochemical properties and potential therapeutic applications .
GSTO1 is widely used in research due to its role in detoxification processes and its potential implications in various diseases. Studies have shown that GSTO1 protects cells from oxidative stress, a risk factor for several neurodegenerative diseases . Additionally, its role in arsenic biotransformation makes it a target for studies on arsenic toxicity and detoxification .