Next Article in Journal
Extraction Methods Shape the Phenolic Composition and Bioactivities of Defatted Moroccan Pistacia lentiscus L. Resin
Previous Article in Journal
Oxidative Stress in Diabetic Retinopathy: A Comprehensive Review of Mechanisms, Biomarkers, and Therapeutic Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Environmental Nephrotoxicity Across the Life Course: Oxidative Stress Mechanisms and Opportunities for Early Intervention

1
Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
2
Department of Pharmacy, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan
3
School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
4
Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan
5
Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
6
Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
7
Center for Environmental Toxin and Emerging-Contaminant Research, Cheng Shiu University, Kaohsiung 833, Taiwan
8
Super Micro Mass Research and Technology Center, Cheng Shiu University, Kaohsiung 833, Taiwan
9
Institute of Environmental Toxin and Emerging-Contaminant, Cheng Shiu University, Kaohsiung 833, Taiwan
10
Department of Pediatrics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan
11
College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
*
Author to whom correspondence should be addressed.
Antioxidants 2025, 14(10), 1205; https://doi.org/10.3390/antiox14101205
Submission received: 31 August 2025 / Revised: 24 September 2025 / Accepted: 29 September 2025 / Published: 4 October 2025
(This article belongs to the Special Issue The Role of Oxidative Stress in Environmental Toxicity—2nd Edition)

Abstract

Chronic kidney disease (CKD) affects nearly 10% of the global population, ranks among the top ten causes of death, and often progresses silently to end-stage disease without timely intervention. Increasing evidence indicates that many adult-onset cases originate in early life through adverse influences on kidney development, a process termed kidney programming within the Developmental Origins of Health and Disease (DOHaD) framework. Environmental pollutants are now recognized as key drivers of kidney injury across the life course. Heavy metals, air pollutants, plastic contaminants such as bisphenol A, phthalates, and micro/nanoplastics—as well as biocontaminants like mycotoxins and aristolochic acid—and chronic light pollution can accumulate in kidney tissue or act systemically to impair function. These exposures promote oxidative stress, inflammation, and endothelial and circadian disruption, culminating in tubular injury, glomerular damage, and fibrosis. Notably, early-life exposures can induce epigenetic modifications that program lifelong susceptibility to CKD and related complications. Oxidative stress is central to these effects, mediating DNA, lipid, and protein damage while influencing developmental reprogramming during gestation. Preclinical studies demonstrate that antioxidant-based interventions may mitigate these processes, providing both renoprotective and reprogramming benefits. This review explores the mechanistic links between environmental pollutants, oxidative stress, and kidney disease and highlights antioxidant strategies as promising avenues for prevention and intervention in vulnerable populations.

1. Introduction

Chronic kidney disease (CKD) poses a major global health burden, affecting approximately 10% of the world’s population and ranking among the top 10 leading causes of death worldwide [1,2]. Notably, CKD is largely preventable, and its progression to end-stage disease can be delayed through timely and effective interventions—particularly when initiated early [3]. In light of this, the global kidney health community has increasingly emphasized the importance of early identification of individuals at high risk—including children, pregnant women, and their fetuses—and the implementation of targeted prevention strategies across the life course [3,4]. Mounting evidence suggests that adult-onset CKD often originates in early life due to adverse influences on kidney development, a process known as kidney programming [5,6]. This concept is rooted in the broader framework of the Developmental Origins of Health and Disease (DOHaD) [7,8].
Common toxicants [9,10]—including heavy metals (e.g., lead, cadmium) [11], air pollutants (e.g., PM2.5) [12], emerging plastic contaminants such as bisphenol A (BPA), phthalates, and microplastics/nanoplastics (MPs/NPs) [13,14,15], as well as environmental stressors like chronic light pollution—can bioaccumulate in kidney tissues or exert systemic effects that impair kidney function. These exposures induce oxidative stress, trigger chronic inflammation, disrupt endothelial and circadian-regulated pathways, and contribute to tubular injury, glomerular damage, and interstitial fibrosis [9,10,11,12,13,14,15].
Early-life exposure to environmental pollutants is particularly concerning, as it can induce epigenetic alterations and developmental programming that elevate the long-term risk of CKD and related complications such as hypertension [10,16]. Moreover, pollutant exposure may exacerbate oxidative stress-mediated tissue injury. Epidemiological studies have consistently linked such exposures to reduced glomerular filtration rate (GFR), proteinuria, and accelerated CKD progression. Thus, environmental pollutants constitute a modifiable risk factor with important implications for both clinical management and public health policy.
Oxidative stress is a central pathogenic mechanism in pollutant-induced kidney disease [17,18,19]. Many environmental toxicants stimulate excessive production of reactive oxygen and nitrogen species (ROS/RNS), disrupting redox homeostasis and causing oxidative damage to DNA, proteins, and lipids in renal tissues. During pregnancy, elevated oxidative stress may also trigger epigenetic modifications that alter gene expression and contribute to the developmental programming of CKD [20]. Conversely, antioxidant therapies have demonstrated both renoprotective and reprogramming effects in preclinical models of kidney programming linked to early-life environmental exposures [21]. CKD is influenced not only by environmental toxicants but also by occupational exposures—metals, solvents, heat, and work-related stress—that cause renal injury through oxidative stress and mitochondrial dysfunction [22]. Targeting these oxidative stress pathways provides a promising strategy for early detection, prevention, and intervention, particularly in vulnerable populations such as children, pregnant women, and individuals with pre-existing kidney disease.
This narrative review summarizes current evidence on the role of environmental pollution in kidney disease arising from both well-known adult risk factors and early-life developmental exposures, with a particular focus on oxidative stress mechanisms. It also highlights the potential therapeutic benefits of antioxidant-based strategies for the treatment and prevention of pollution-related kidney injury.

2. Materials and Methods

Considering the breadth of disciplines and variability in the existing studies, a narrative review was selected over a systematic or scoping approach to allow for an integrative exploration of emerging concepts spanning nephrology, developmental biology, redox signaling, toxicology, and environmental health.
This review aims to collect current evidence on the role of oxidative stress in mediating the effects of environmental pollutants on kidney disease risk, with a particular focus on developmental programming and redox-targeted interventions. A comprehensive literature search was conducted using PubMed, Scopus, and Web of Science through June 2025. Search terms included combinations of: “oxidative stress,” “reactive oxygen species,” “reactive nitrogen species,” “nitric oxide,” “pollutants,” “endocrine-disrupting chemicals,” “organophosphate flame retardants,” “phthalates,” “microplastics,” “heavy metals,” “air pollution,” “PM2.5,” “light pollution,” “mycotoxins” “phytotoxins” “pregnancy,” “maternal,” “fetal programming,” “kidney development,” “nephrogenesis,” “kidney programming,” “DOHaD,” “reprogramming,” “antioxidants,” “melatonin,” “polyphenol,” “vitamin” “hypertension,” and “chronic kidney disease.”
Eligible studies included original research articles, reviews, and clinical studies involving both animal models and human populations that examined the impact of environmental pollutants on kidney disease through oxidative stress mechanisms. Special emphasis was placed on studies addressing redox imbalance, fetal and renal vulnerability, and the therapeutic potential of antioxidants. Reference lists of key articles were also screened to identify additional relevant studies. Figures were generated using Napkin AI software.

3. The Interplay Between Oxidative Stress and Kidney Disease

3.1. Mechanistic Basis of Oxidative Stress

Oxidative stress occurs when the generation of ROS and RNS surpasses the neutralizing capacity of antioxidant defenses, resulting in redox imbalance [23]. Under physiological conditions, ROS and RNS are generated in controlled amounts as part of normal cellular signaling and play important roles in regulating renal physiology and blood pressure (BP) [24,25]. Key free radicals include the superoxide anion (O2), produced mainly in mitochondria during oxidative phosphorylation, and nitric oxide (NO•), synthesized by nitric oxide synthases (NOS) [26]. In the kidney, major ROS sources include NADPH oxidases (notably NOX4 in tubules), which generate superoxide in renal and vascular cells, and mitochondrial electron transport chain complexes I and III, which leak superoxide during oxidative phosphorylation, especially with dysfunction or pollutant exposure. Xanthine oxidase produces superoxide and hydrogen peroxide during purine metabolism. Additionally, uncoupled NOS generates superoxide instead of NO when cofactors are deficient or when elevated levels of asymmetric dimethylarginine (ADMA), an endogenous NOS inhibitor, promote NOS uncoupling through substrate competition [27]. In inflammation, myeloperoxidase forms hypochlorous acid from hydrogen peroxide, and cytochrome P450 enzymes release ROS during xenobiotic metabolism. Dysregulation of these systems promotes oxidative injury in CKD. While NO• plays an essential role in vasodilation and the regulation of blood pressure and renal hemodynamics [28], its reaction with O2 forms peroxynitrite (ONOO)—a highly reactive RNS that can inflict widespread oxidative and nitrosative damage [29]. Antioxidant defense systems counteract these effects and include enzymatic components (e.g., SOD converting O2 to hydrogen peroxide) and non-enzymatic molecules (e.g., glutathione, vitamins C and E, uric acid) [30].
Excess free radicals cause oxidative modifications to biomolecules: lipid peroxidation disrupts membrane integrity; protein carbonylation alters enzyme activity and receptor function; and oxidative DNA lesions impair transcription and promote mutagenesis. In renal tissues, these molecular insults activate pro-inflammatory pathways (e.g., NF-κB) and pro-fibrotic cascades (e.g., transforming growth factor-β), driving tubular injury, glomerulosclerosis, and progressive nephron loss. Notably, persistent lipid peroxidation can trigger ferroptosis—an iron-dependent, non-apoptotic form of regulated cell death characterized by the accumulation of lethal lipid ROS [31]. Ferroptosis has been increasingly implicated in acute and chronic kidney injury, linking oxidative stress, disorganized iron metabolism, and membrane damage to irreversible nephron loss.

3.2. Role of Oxidative Stress in CKD

Oxidative stress plays a central role in the initiation and progression of CKD, functioning both as a pathogenic driver and a potential therapeutic target [32,33]. In CKD, excessive ROS generation arises from multiple sources, including uremic toxin accumulation, chronic inflammation, mitochondrial dysfunction, and activation of the renin–angiotensin system (RAS). This redox imbalance damages lipids, proteins, and nucleic acids, thereby promoting kidney fibrosis, endothelial dysfunction, and progressive nephron loss.
Commonly used oxidative stress markers in CKD encompass indicators of oxidative damage to lipids, proteins, and DNA, as well as measures of antioxidant defense [34,35]. Lipid peroxidation is frequently assessed by malondialdehyde (MDA) [36] or the more specific F2-isoprostanes [37]. Protein oxidation can be evaluated by protein carbonyl content or advanced oxidation protein products (AOPP) [38], while DNA oxidation is typically measured by urinary, serum, or tissue levels of 8-hydroxy-2′-deoxyguanosine (8-OHdG) [39]. These key biomarkers—MDA, F2-isoprostanes, 8-OHdG, and AOPP—are often elevated in CKD and correlate with disease severity and cardiovascular risk.
On the other side, antioxidant status is assessed through total antioxidant capacity (TAC), activities of enzymatic antioxidants such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) [40], and levels of non-enzymatic antioxidants, particularly the reduced-to-oxidized glutathione (GSH/GSSG) ratio [41]. Impairments in these antioxidant systems are common in CKD, further exacerbating oxidative injury and contributing to CKD-related morbidity and mortality.
Importantly, oxidative stress is not only a biomarker of kidney injury but also a modifiable therapeutic target. Antioxidant strategies—including dietary natural antioxidants, pharmacologic antioxidants, RAS blockade, and agents targeting mitochondrial ROS—have shown potential in experimental CKD models [42]. However, translating these findings into consistent clinical benefit remains challenging, highlighting the need for biomarker-guided, personalized antioxidant therapies [43]. Advances in non-invasive redox biomarkers and targeted delivery systems could pave the way for precision nephrology approaches aimed at restoring redox homeostasis in CKD.

3.3. Oxidative Stress as a Mediator of Kidney Programming

Oxidative stress plays a pivotal role in kidney programming [44,45], the process by which adverse conditions during fetal and early postnatal development shape long-term kidney structure and function, predisposing individuals to CKD later in life [5,6].
A controlled rise in ROS during gestation supports placental blood vessel formation, cellular differentiation, and the development of fetal organs. Conversely, excessive oxidative stress, as seen in complicated pregnancies, is linked to negative maternal and fetal outcomes [46,47]. Accordingly, excessive ROS and RNS can disrupt normal nephrogenesis, leading to low nephron endowment and a spectrum of defects namely congenital anomalies of the kidneys and urinary tracts (CAKUT) [48,49]. Oxidative stress can induce epigenetic modifications [50]—such as DNA methylation, histone modification, and microRNA expression changes—that program gene expression patterns governing nephron number, tubular function, and renal hemodynamics [51]. Transcriptome analysis of ADMA-exposed embryonic kidneys revealed 1221 differentially expressed genes related to kidney development and epigenetic regulation [51]. Likewise, maternal NO inhibition by L-NAME altered 2289 genes in neonatal kidneys [52]. These findings suggest oxidative stress and epigenetic gene regulation during gestation contribute significantly to kidney programming and future kidney disease risk.
Environmental insults such as maternal malnutrition [53], illness [54], inflammation [55], and exposure to pollutants [56] or medications such as glucocorticoids [57] commonly elevate oxidative stress in utero. This increased oxidative burden not only directly impairs nephrogenesis but also promotes hypertension, glomerular hypertrophy, glomerulosclerosis, tubulointerstitial injury, kidney dysfunction, and albuminuria—collectively setting the stage for accelerated CKD progression in adulthood [21].
Kidney programming involves multiple oxidative stress-related mechanisms, including increased ROS-producing enzymes [58], elevated ROS and peroxynitrite levels [58,59], decreased antioxidant capacity [60], higher ADMA concentrations [61], reduced NO bioavailability [61], and enhanced oxidative damage [52]. Biomarkers of lipid peroxidation—such as MDA and F2-isoprostanes—are elevated in offspring kidneys in various rodent models of kidney programming [20,21]. Additionally, 8-OHdG is highly expressed in these models and correlates with adverse renal outcomes. Numerous studies highlight the role of impaired ADMA/NO pathways in oxidative stress–induced kidney programming [62]. Overall, these findings support oxidative stress as a key contributor to kidney programming and CKD later in life, which can be assessed by related biomarkers.
Animal studies reviewed elsewhere [21] show that maternal supplementation with natural antioxidants—including vitamins C and E, amino acids such as arginine and taurine, melatonin, and polyphenols—during pregnancy and lactation can protect kidney development and mitigate the risk of renal programming. In addition to natural compounds, several synthetic antioxidants have also been tested in animal models [63,64,65,66], showing potential to mitigate oxidative stress and preserve kidney function in offspring. Therefore, oxidative stress is both a mediator and amplifier of developmental kidney injury, linking early-life environmental exposures to lifelong kidney health outcomes through the concept of DOHaD. Targeting oxidative stress during these sensitive periods may offer preventive strategies to mitigate programmed kidney disease risk.

4. Oxidative Stress Links Environmental Pollutants to Nephrotoxicity

Oxidative stress is a central mediator of kidney injury caused by environmental pollutants. Nephrotoxicity—characterized by increased urinary albumin excretion or reduced GFR—is increasingly linked to chronic exposure to toxicants such as MPs [67], phthalates [68], air pollution [69], and BPA [70]. These agents contribute to kidney dysfunction through oxidative and inflammatory mechanisms beyond traditional risk factors like diabetes and hypertension.
Pollutant exposure leads to cellular uptake via endocytosis, causing intracellular accumulation and excessive ROS production. This disrupts redox balance, triggering lipid peroxidation, mitochondrial dysfunction, DNA damage, and protein modifications, which collectively promote apoptosis and fibrosis [71]. Oxidative stress also weakens antioxidant defenses—including SOD and catalase—and disturbs key signaling pathways such as AMP-activated protein kinase (AMPK) and the phosphoinositide 3-kinase/protein kinase B/mechanistic target of rapamycin (PI3K/AKT/mTOR), aggravating renal tubular epithelial injury [72].
The mitochondrial apoptotic pathway, marked by cytochrome c release and activation of caspase-9 and caspase-3, plays a crucial role in pollutant-induced cell death [73]. As well, disturbances in ion homeostasis and organelle function exacerbate kidney cell damage. Emerging research highlights oxidative stress as a unifying driver of toxicant-associated nephropathy. Given the diverse toxicity profiles of environmental chemicals, each will be discussed in detail below.

4.1. Dioxins

Dioxin, most notably 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), is the most toxic and well-studied member of a group of structurally related compounds—including polychlorinated dibenzo-p-dioxins (PCDDs), dioxin-like polychlorinated biphenyls (PCBs), and polychlorinated dibenzofurans (PCDFs) [74]. These persistent organic pollutants are primarily released from anthropogenic sources such as pesticide production, waste incineration, and paper bleaching. Lipophilic in nature, dioxins bioaccumulate in the food chain and persist for years in adipose tissue, enabling chronic, low-level exposure [75]. Experimental and epidemiological evidence indicates that TCDD can induce kidney injury through multiple interrelated mechanisms: activation of the aryl hydrocarbon receptor (AhR) leading to altered gene expression [76], oxidative stress [77], mitochondrial dysfunction, and chronic inflammation; disruption of renal hemodynamics and glomerular filtration; and promotion of tubulointerstitial fibrosis [78].
Maternal exposure to these chemicals—through high-animal-fat diets or occupational contact—may induce epigenetic changes that predispose offspring to long-term CKD [79]. Elevated dioxin exposure has been associated with adverse offspring kidney outcomes in several animal models of kidney programming [80,81,82].

4.2. Plastic Chemical Pollutants

Plastics, composed of polymers with additives such as plasticisers, flame retardants, stabilizers, and colorants, are produced in massive volumes, exceeding 400 million metric tonnes annually and projected to triple by 2060 [83,84]. Most additives are not covalently bound and can leach into food, water, and air, entering the body via ingestion, inhalation, or skin contact [85,86]. Many are detected in human biosamples across all life stages [87], yet most of the >16,000 identified chemicals remain unregulated and poorly studied [88]. Major plastic chemical pollutants contribute to kidney disease through a range of toxicological pathways, including oxidative stress, chronic inflammation, endocrine disruption, and direct tubular injury [89,90,91].

4.2.1. Monomers

Major plastic monomers—including bisphenol A (BPA), vinyl chloride, and styrene—are integral to the manufacture of polycarbonate, PVC, and polystyrene, yet also function as endocrine-disrupting chemicals (EDCs) and metabolic toxins. In humans, BPA exposure has been associated with albuminuria and reduced estimated GFR (eGFR) in CKD patients [92], and cross-sectional studies across diverse populations suggest a possible link to hypertension [93]. These renal and vascular effects are thought to be mediated by oxidative stress, podocyte injury, and activation of profibrotic signaling pathways [70]. Mother–child cohort studies further indicate that maternal BPA exposure may be associated with elevated BP in offspring, although findings remain inconclusive [94,95,96]. Vinyl chloride and styrene, both recognized human carcinogens, have been shown in animal models to cause albuminuria, glomerulosclerosis, and tubular injury [97,98,99], yet their kidney-specific effects in humans remain largely uncharacterized.

4.2.2. Plasticizers

Plasticizers, particularly phthalates such as di(2-ethylhexyl) phthalate (DEHP), dibutyl phthalate (DBP), and butyl benzyl phthalate (BBP), are widely used in PVC products and can leach into food, water, and medical devices [100]. As reviewed elsewhere [101], accumulating evidence links phthalate metabolites to proteinuria, decreased kidney function, and higher CKD prevalence, mediated by oxidative stress, mitochondrial damage, altered renal hemodynamics, and activation of inflammatory pathways [102,103,104]. Phthalate metabolites readily cross the placenta, exposing the fetus [105]. Due to their estrogenic and antiandrogenic effects, prenatal phthalate exposure has been associated with adverse renal and cardiovascular outcomes in offspring; epidemiological studies report that elevated maternal urinary DEHP correlates with higher BP, lower estimated GFR, and albuminuria in children [95,106,107]. Regulatory thresholds, as summarized in the 15th Report on Carcinogens (NTP 2021) [108], include an EU tolerable daily intake of 0.05 mg/kg/day, occupational exposure limits of ~5 mg/m3, and medical exposures in infants that may reach several mg/kg/day, underscoring pregnancy and early life as the most vulnerable periods. These findings underscore the urgent need to investigate the impact of prenatal phthalate exposure on offspring kidney health.

4.2.3. Flame Retardants and Stabilizers

Flame retardants such as polybrominated diphenyl ethers (PBDEs) and hexabromocyclododecane (HBCD) persist in human tissues for years [109] and are widely used in vehicles, electronics, furnishings, plastics, building materials, polyurethane foams, and textiles [108]. These compounds have been linked to increased oxidative DNA damage in renal cells, disruption of the thyroid–kidney axis, and promotion of tubulointerstitial fibrosis [110,111,112,113]. Stabilizers, including organotins like tributyltin, exert nephrotoxic effects by inducing oxidative stress, apoptosis, and immune dysregulation, characterized by pro-inflammatory cytokine release and immune cell infiltration in the kidney [114]. Ultraviolet (UV) stabilizers such as benzophenones may contribute indirectly to kidney injury through endocrine and metabolic disturbances; in zebrafish models, benzophenones induce oxidative stress-associated renal damage [115,116]. Additionally, nonylphenol and related antioxidant additives display estrogenic activity and provoke oxidative stress in renal tissue [117]. Despite these findings, the impact of these chemicals on kidney developmental programming and the relationship between maternal exposure and offspring renal outcomes remains poorly understood.

4.2.4. Microplastics/Nanoplastics

Plastic degradation produces microplastics (MPs, <0.5 mm) and nanoplastics (NPs, 1–1000 nm) [118]. MPs/NPs can pollute drinking water and accumulate in food chain, which may cause detrimental effects to human body [119]. Collectively, considering both inhalation and ingestion, humans may be exposed to tens of thousands to millions of MPs daily, depending on individual behaviors and environmental factors [120]. Human and animal studies show MPs and NPs can cross the digestive epithelium, translocate to the kidney, and accumulate in kidney tissue [15,121]. Histological analyses confirm renal deposition of particles ranging from 50 nm to 20 μm, raising concerns about potential kidney toxicity.
Critically, MPs serve as carriers for various harmful chemicals and adsorbed environmental pollutants, such as persistent organic pollutants like dioxins and PCBs, facilitating their accumulation in the kidney. Once internalized, they can trigger oxidative stress injury, tubular inflammation, and fibrosis, ultimately contributing to CKD onset and progression [120]. In vitro and in vivo studies show that MPs enter renal cells, triggering mitochondrial ROS overproduction, ER stress, inflammation, apoptosis, and MAPK pathway activation [122]. MP exposure disrupts antioxidant defenses and elevates inflammatory mediators (TNF-α, IL-6, MCP-1), leading to structural damage, lipid accumulation, and fibrosis. Chronic MP exposure can induce ferroptosis—characterized by glutathione depletion, GPX4 suppression, lipid peroxidation, and Fe2+ overload—which promotes renal fibrosis [123]. Conversely, antioxidant interventions mitigate oxidative stress and inflammation, highlighting oxidative stress as a central driver of MP-related kidney injury [124,125].
The combined burden of these plastic-associated chemicals—including BPA, phthalates, flame retardants, plastic stabilizers, and MPs/NPs—represents an emerging yet underrecognized risk factor for CKD. The risk is particularly concerning for vulnerable populations such as children, pregnant women, and those with preexisting kidney disease, in whom early-life or cumulative exposures may accelerate CKD onset and progression.
Nanomaterials are not limited to plastics; they encompass metals, metal oxides, carbon-based structures, polymers, and hybrid systems. Nanomaterials are materials with at least one dimension in the nanometer range (1–100 nm) and unique physicochemical properties due to their size. While microplastics and nanoplastics are widely discussed, nanomaterials go far beyond plastics and include a broad variety of inorganic, organic, and hybrid materials. Nanomaterials beyond plastics can cause nephrotoxicity primarily through oxidative stress, inflammation, apoptosis, DNA damage, and structural disruption of renal tissue. The extent depends on physicochemical properties and exposure parameters. While some NPs (like lipid-based carriers) are relatively safe, inorganic and carbon-based NPs pose significant renal risks in preclinical studies.

4.3. Heavy Metals

Heavy metals represent a heterogeneous group of inorganic chemical hazards, with lead (Pb), cadmium (Cd), and mercury (Hg) being the most frequently implicated in nephrotoxicity [126,127]. In the general population, lead exposure primarily arises from contaminated air and food, historically linked to leaded petrol emissions and cooking utensils. Cadmium compounds, still used in certain stabilizers and rechargeable nickel–cadmium batteries, contribute to exposure mainly through contaminated household waste, food, and cigarette smoke. Mercury exposure is predominantly dietary, via consumption of contaminated fish, and from dental amalgam. A systematic review of 14 studies reported significant associations between proteinuria and exposure to arsenic, cadmium, lead, and chromium in drinking water [128].
These heavy metals induce oxidative stress through distinct but converging mechanisms that generate ROS/RNS, leading to DNA damage, lipid peroxidation, and protein oxidation [129]. Arsenic binds to critical thiols, produces hydrogen peroxide, and triggers NO–mediated genotoxicity [130]; cadmium depletes glutathione and binds sulfhydryl groups, impairing antioxidant defenses; lead inhibits antioxidant enzymes and disrupts calcium homeostasis, enhancing ROS formation; and chromium undergoes redox cycling, producing superoxide and hydroxyl radicals that form mutagenic lipid peroxidation products [131]. Beyond redox effects, toxic metals impair kidney mitochondria, as shown in 20 studies on arsenic, cadmium, and lead. Metal exposure disrupts mitochondrial membrane potential, electron transport, and bioenergetics, causing oxidative imbalance in a metal- and dose-dependent manner, with the proximal tubule being especially vulnerable [132]. These processes activate redox-sensitive signaling pathways (e.g., NF-κB, AP-1, p53), contributing to nephrotoxicity.
Human studies indicate that lead, cadmium, and mercury tend to accumulate more extensively in the fetal kidney than in the brain [133]. Long-term lead exposure has been associated with the onset of lead-induced nephropathy [134], while cadmium readily enters renal epithelial cells, triggering toxic injury [135]. Mercury, likewise, is capable of inducing acute kidney injury and damaging proximal tubules [136]. In pediatric populations, even sustained low-level exposure to multiple heavy metals has been linked to elevated risks of CKD and hypertension [137,138]. Given that heavy metals remain major environmental and occupational contaminants with well-documented nephrotoxic potential, it is increasingly important to determine whether maternal exposure during pregnancy influences kidney health trajectories into adulthood.

4.4. Polycyclic Aromatic Hydrocarbons

Polycyclic aromatic hydrocarbons (PAHs) are lipophilic organic contaminants consisting of two or more fused aromatic rings, generated from industrial processes, vehicular emissions, domestic combustion, and agricultural activities [139]. Their strong fat solubility promotes bioaccumulation in adipose tissue. Among them, benzo[a]pyrene (BaP) is one of the most extensively studied PAHs, well known for its mutagenic, carcinogenic, teratogenic, and immunotoxic effects in humans [140].
PAH-induced nephrotoxicity is driven by oxidative stress from CYP-mediated metabolism to ROS-generating intermediates, causing lipid peroxidation, DNA damage, and mitochondrial dysfunction in renal cells [141]. Activation of the AhR further amplifies pro-oxidant signaling, while depletion of glutathione and inhibition of key antioxidant enzymes weaken renal defense, leading to inflammation and kidney injury [142].
During pregnancy, maternal and cord blood show comparable PAH concentrations, with markedly lower levels in placental tissue, indicating transplacental transfer to the fetus [143]. Postnatally, breastfeeding has been reported to expose 30–95% of infants to PAHs [144]. Gestational PAH exposure has been linked to adverse outcomes including low birth weight, preterm delivery, stillbirth, and congenital malformations [145,146]. In adults, elevated urinary PAH metabolites correlate with reduced eGFR [147], increased BP [148], and albuminuria [149].

4.5. Per- and Polyfluoroalkyl Substances

Per- and polyfluoroalkyl substances (PFAS) comprise a broad class of synthetic chemicals widely used in consumer goods and industrial applications [150]. Exposure to PFAS is nearly universal, with compounds such as perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) detected in over 90% of the general population [151]. In adults, elevated serum concentrations of PFOA and PFOS have been correlated with chronic kidney disease (CKD) [152], and similarly, increased PFOA exposure has been linked to impaired renal function in pediatric populations [153]. For pregnant women, ingestion of contaminated food and water, along with airborne exposure, represents the primary routes of PFAS intake. These substances readily cross the placenta and are also transmitted to infants via breastfeeding [151].
Multiple biological pathways have been implicated in PFAS-related renal toxicity, including oxidative stress, disruption of endothelial barrier integrity, and induction of epithelial-to-mesenchymal transition in the kidneys [154]. PFAS exposure leads to increased ROS generation, which damages cellular components including lipids, proteins, and DNA. This oxidative imbalance activates signaling pathways such as peroxisome proliferator-activated receptors (PPARs) and NRF2, affecting cellular metabolism and inflammatory responses [155]. The resulting oxidative stress contributes to endothelial dysfunction, epithelial-to-mesenchymal transition, and tissue injury, especially in the kidneys, linking PFAS exposure to impaired renal function and CKD. Despite growing recognition of PFAS as environmental contributors to kidney dysfunction, the impact of maternal exposure on offspring renal health remains insufficiently understood.

4.6. Air Pollution

Air pollution encompasses a mixture of toxic gases and particulate matter in the atmosphere that threaten both human health and environmental quality. These pollutants include a mixture of gases—such as carbon monoxide, nitrogen oxides, sulfur dioxide, ozone—and particulate matter (PM), which consists of tiny solid or liquid particles suspended in the air. PM is categorized by size into coarse (PM10), fine (PM2.5), and ultrafine (PM0.1) particles. Air pollution originates from various sources, including industrial emissions, vehicle exhaust, combustion of fossil fuels, agricultural activities, and natural events like wildfires. Emerging studies have suggested that exposure to air pollution is closely relevant to increased risk of CKD, CKD progression, and ESKD [156,157,158,159]. The mechanism of PM2.5-induced kidney injury involves oxidative stress, inflammatory response, and cytotoxicity [160]. Air pollutant-induced oxidative stress activates redox-sensitive pathways (e.g., AP-1, Nrf2) that initially promote protective antioxidant responses but, with sustained exposure, lead to inflammation and cell death. Vulnerable populations, including the young, elderly, and those with pre-existing conditions, are especially susceptible due to impaired antioxidant capacity, making oxidative stress a central mediator linking air pollution to renal injury.
While the link between maternal exposure to air pollution and congenital anomalies has been documented [161], the impact of early-life exposure to PM on subsequent renal health in offspring remains inadequately understood.

4.7. Light Pollution

Unlike chemical or particulate exposures, light pollution primarily disrupts circadian rhythms [162]. Although artificial light is essential for visual performance and safety, growing concerns highlight its potential health and environmental consequences [163]. Chronic exposure to excessive light at night disturbs the sleep–wake cycle, hormonal rhythms (e.g., melatonin, cortisol), and metabolic regulation [162,163,164]. These systemic disturbances can indirectly impair kidney function, tubular reabsorption, and BP control. In addition, circadian disruption promotes oxidative stress and inflammation, directly damaging renal cells and accelerating dysfunction [165,166]. Light-induced circadian misalignment also worsens metabolic and cardiovascular risk factors—such as hypertension, obesity, and insulin resistance—that further exacerbate kidney injury [162,163]. Collectively, these mechanisms suggest that chronic light pollution contributes to kidney disease development and progression through circadian dysregulation and oxidative stress. Importantly, light also regulates circadian signaling pathways critical for pregnancy and fetal development [167,168]. Animal studies indicate that maternal chronodisruption has long-term adverse effects on offspring health, including increased risks of hypertension and kidney disease [169,170,171].

4.8. Natural Pollutants

In addition to anthropogenic pollutants, natural toxins such as mycotoxins and phytotoxins can adversely impact kidney health [172]. Mycotoxins, including ochratoxin A and citrinin from contaminated grains and nuts, primarily target the proximal tubules, leading to tubular injury, interstitial fibrosis, and progressive CKD through oxidative stress, mitochondrial dysfunction, and apoptotic pathways [173,174]. Phytotoxins, notably aristolochic acids from certain herbal remedies, induce aristolochic acid nephropathy (AAN), a well-defined nephropathy characterized by tubular atrophy and interstitial fibrosis via mutagenic DNA adduct formation, sustained oxidative stress, and epithelial apoptosis [175,176]. Collectively, these natural pollutants share mechanisms of oxidative stress, mitochondrial damage, and inflammatory activation, emphasizing their role in kidney injury. Although mycotoxin contamination of breast milk has been reported worldwide, data on the effects of lactational transfer on offspring health remain limited [177]. Maternal exposure to aristolochic acids also poses significant risks to fetal and maternal health [178,179], although the direct impact on offspring kidney function remains unclear. Importantly, both mycotoxins and phytotoxins converge on common nephrotoxic mechanisms, including reactive oxygen species generation, mitochondrial injury, impaired DNA repair, and activation of pro-inflammatory and profibrotic signaling cascades, which collectively drive CKD development.
AAN has emerged as a globally recognized form of environmentally induced CKD and urothelial malignancy. First described in the context of “Chinese herb nephropathy,” AAN results from exposure to herbal remedies containing Aristolochia species, and is now considered a distinct clinicopathologic entity [180]. Epidemiological studies have linked aristolochic acid exposure not only to cases in East Asia but also to Balkan endemic nephropathy, further highlighting its global importance [181]. The pathophysiology of AAN is driven by the formation of aristolochic acid-DNA adducts that cause characteristic A:T → T:A transversion mutations in tumor suppressor genes, together with persistent oxidative stress, tubular epithelial apoptosis, and inflammatory activation [182,183]. Clinically, AAN presents with an insidious decline in renal function, often progressing rapidly to ESKD. Of note, even brief or low-level exposure to aristolochic acids may cause irreversible nephropathy, emphasizing the urgency for stricter regulation of herbal supplements, food safety monitoring, and environmental surveillance. Recognition of AAN as a model of toxin-induced kidney fibrosis provides important mechanistic insight into how natural pollutants initiate CKD progression.

4.9. Other Contaminants

Emerging contaminants such as nanomaterials extend beyond plastics to include metals, metal oxides, carbon-based structures, polymers, and hybrid systems, each with at least one dimension in the nanometer range (1–100 nm) and unique physicochemical properties [184]. These materials can induce nephrotoxicity through oxidative stress, inflammation, apoptosis, DNA damage, and structural disruption of renal tissue, with the severity depending on their physicochemical characteristics and exposure conditions [185]. Figure 1 summarizes the impacts of environmental pollutants on kidney health.

5. Oxidative Stress in Kidney Programming Models of Early-Life Toxicant Exposure

Epidemiological evidence indicates an association between early-life exposure to environmental toxicants and the risk of kidney disease in offspring. However, detailed insights into how such exposures shape kidney programming and contribute to CKD in later life are still limited. Observational data alone cannot establish a direct causal link between prenatal toxicant exposure and adult kidney disease. In addition, these studies provide little understanding of the molecular pathways involved or guidance on potential interventions to counteract adverse programming.
Animal models provide essential platforms for dissecting how oxidative stress contributes to kidney programming following early-life toxicant exposure. These experimental systems not only clarify underlying biological mechanisms but also offer opportunities to explore preventive interventions. Table 1 summarizes representative studies linking toxicant exposure during critical developmental windows with an elevated risk of CKD in offspring [80,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186].
Table 1 shows that rodents are the primary species used in these studies, whereas larger animals have not yet been applied for comparable toxicant exposure experiments. Documented programming effects of environmental toxicants in rats span ages 4 to 21 weeks, roughly equivalent to human childhood through early adulthood [202].

5.1. Mechanisms Behind Early-Life Toxicant Exposure Induced Kidney Programming

Early-life exposure to environmental toxicants—including dioxins [80,186,187,188,189], plastic chemical pollutants [190,191,192,193,194], polystyrene nanoplastics [195], heavy metals [196,197], PAHs such as BaP [198], PFOS [199], and air pollution [200,201]—programs kidney development, leading to offspring outcomes characteristic of CKD. These effects include hypertension [186,187,188,198,199,200,201], kidney malformations [189], impaired renal function [190,192,194,195,197], renal hypertrophy [195], and kidney injury [196].
Several mechanisms have been implicated in environmental toxicant–induced kidney programming [5,6]. In addition to oxidative stress, key pathways include aberrant activation of the RAS, reduced nephron endowment, and dysregulation of the AhR signaling pathway, as discussed in the following sections.

5.2. Oxidative Stress

Multiple animal models have demonstrated that oxidative stress mediates kidney programming, contributing to hypertension and renal dysfunction in adult offspring [20,21]. Studies have specifically assessed the role of oxidative stress in response to prenatal exposure to toxicants such as TCDD [186,188], BPA [191], and PM2.5 [201]. Markers of oxidative DNA damage, including renal 8-OHdG, were elevated in offspring exposed in utero to TCDD [186] or BPA [191]. In a prenatal PM2.5 exposure model, offspring developed hypertension associated with oxidative stress, which was prevented by the antioxidant tempol [201]. These findings indicate that oxidative stress is a key mechanism linking early-life toxicant exposures to programmed kidney disease and hypertension. Mechanistically, oxidative stress serves as a central hub by increasing ROS through AhR activation, NADPH oxidase activity, mitochondrial dysfunction, and impaired antioxidant defenses, while interacting with pathways such as RAS activation, renal dopamine receptor signaling, and immune modulation. This oxidative milieu contributes to structural kidney alterations, nephron loss, fibrosis, and sustained elevation of BP [20,21,51].

5.3. Aberrant Activation of the RAS

The kidney is a primary target of the RAS, which is essential for regulating renal function and BP [203]. Pharmacologic inhibition of RAS remains a cornerstone for managing hypertension and CKD [204]. During nephrogenesis, RAS components are highly expressed, guiding proper renal development and physiological maturation [205]. Disruptions during this critical period—such as maternal insults—can lead to persistent RAS dysregulation, contributing to adult kidney disease and hypertension [206]. Notably, fetal exposure to RAS blockers—including ACE inhibitors, ARBs, and direct renin inhibitors—during the second and third trimesters can induce renal malformations, a phenomenon termed RAS blocker fetopathy [206].
Aberrant RAS activation has been implicated in kidney programming across multiple animal models [207,208]. Several environmental toxicants, including TCDD [188], DEHP [192], and BaP [198], appear to simultaneously affect renal development and RAS activity, resulting in programmed hypertension in adult offspring. Early-life interventions targeting the RAS have shown promise in preventing these adverse outcomes in experimental settings [208]. However, the extent to which oxidative stress mediates interactions between the RAS and environmental chemicals in the developmental origins of kidney disease and hypertension remains to be fully elucidated.

5.4. Reduced Nephron Number

Nephron endowment is a fundamental determinant of renal health across the lifespan [209]. Human kidneys typically have around 1 million nephrons, but individual counts can differ by as much as tenfold [210]. Insufficient nephron number, commonly observed in those born with low birth weight (LBW) or preterm, is strongly associated with heightened susceptibility to hypertension and CKD in later life [211,212]. Additional determinants of nephron number include age, sex, and body size. Epidemiologic studies further implicate maternal exposure to environmental toxicants—including plastics, PFOA, PAHs, and fine particulate matter (PM2.5/PM10)—in preterm birth and LBW [213,214,215,216,217], both serve as risk factors for nephron deficit. How such exposures influence nephron endowment and contribute to the developmental origins of kidney disease and hypertension remains an important unanswered question.
Within the framework of the “multi-hit” model of CKD [218], reduced nephron number can serve as an early hit, initiating compensatory hyperfiltration, elevated intraglomerular pressure, and progressive nephron attrition, thereby sensitizing the kidney to subsequent insults. Experimental evidence supports this paradigm: in a maternal DEHP exposure model, offspring developed hypertension and kidney dysfunction alongside disrupted expression of nephrogenesis-related genes [192]. These findings indicate that maternal DEHP exposure interferes with nephron formation, leading to a lasting nephron deficit and adult-onset disease. Although some heavy metals have been implicated in abnormal nephrogenesis [48], their specific role in determining nephron number is not yet established. Future studies should address whether chemical exposures affect nephron endowment in a dose- and developmental stage–dependent manner.

5.5. Dysregulated AhR Signaling Pathway

The AhR is a ligand-activated transcription factor that functions as a sensor for environmental chemicals, endogenous metabolites (such as tryptophan derivatives), and certain dietary compounds [219,220]. Once activated, AhR migrates to the nucleus, forms a complex with the aryl hydrocarbon receptor nuclear translocator (ARNT), and regulates downstream genes, including members of the cytochrome P450 enzyme family (CYP1A1, CYP1A2, CYP1B1) and the AhR repressor (AHRR). Through these actions, AhR modulates xenobiotic metabolism, oxidative balance, and immune responses. Aberrant AhR signaling, especially due to maternal exposure to exogenous ligands, has been linked to developmental programming of disease, including kidney disorders [208].
Historically, the first ligands identified for AhR were environmental pollutants such as dioxins, BPA, phthalates, PFOS, and PAHs [219]. Maternal exposure to TCDD, a dioxin, has been shown to induce hypertension in offspring through activation of the AhR/CYP1A1 axis and promotion of TH17-mediated renal inflammation [186]. BPA is likewise an AhR agonist [221], with prenatal exposure leading to offspring hypertension accompanied by increased renal expression of AhR, AHRR, CYP1A1, and ARNT [191].
Phthalates, including DEHP and DBP, also engage AhR signaling and have been associated with impaired renal outcomes in progeny, such as altered kidney function, hypertrophy, and fibrosis [192,193,194]. Gestational PFOS exposure results in offspring hypertension in both sexes by 16 weeks of age [203], though the mechanistic contribution of AhR remains to be fully defined [222]. Similarly, prenatal exposure to fine particulate matter (PM2.5) predisposes offspring to adult-onset hypertension [200,201], with emerging evidence suggesting that AhR mediates its oxidative and inflammatory effects [223].

5.6. Epigenetic Dysregulation

Pollutant-induced nephrotoxicity is increasingly recognized to involve aberrant DNA methylation, altered histone acetylation/methylation, and dysregulated non-coding RNAs. Emerging evidence suggests that these epigenetic mechanisms are central to the transgenerational inheritance of pollutant-induced kidney injury. Unlike genetic mutations, epigenetic modifications are reversible and responsive to environmental cues, making them plausible mediators of how toxicant exposures in one generation influence kidney health in unexposed descendants.
In animal models, maternal exposure to TCDD has been shown to induce persistent DNA methylation changes at specific differentially methylated regions (DMRs) within germ cells. When pregnant F0 females were exposed to TCDD, both the F1 and F3 generations were subsequently examined. Importantly, the F3 generation—without any direct exposure—showed an increased incidence of kidney disease in males. Approximately 50 sperm DMRs were identified and associated with this transgenerational kidney phenotype [224].
Emerging evidence demonstrates that Cd exerts its nephrotoxic effects in part through epigenetic modifications. Research reveals that Cd exposure enhances DNA methyltransferase (DNMT1/3a) activity leading to aberrant DNA methylation. Concurrently, Cd promotes histone modifications, including increased H3K9 mono/di-methylation via G9a and altered acetylation states through suppression of the histone deacetylase SIRT1, thereby driving maladaptive gene expression and oxidative stress in renal tubular cells [225]. Beyond DNA methylation, histone modifications play a pivotal role. Cd-induced recruitment of the chromatin reader BRD4, together with enhanced H4K16 acetylation, impairs autophagy and promotes tubular injury. Pharmacological inhibition of BRD4 (e.g., with JQ1) or knockdown approaches restored autophagy and attenuated oxidative injury in Cd-exposed mice [226]. Likewise, inhibition of G9a partially reversed Cd-induced histone methylation and improved cell survival, suggesting the reversibility of these changes [225].
Importantly, epigenetic modifications are not static. Antioxidants such as N-acetylcysteine and resveratrol (a SIRT1 activator) have been proposed to counteract Cd-induced oxidative stress and epigenetic dysregulation [227,228], while their effects in environmental toxicant-induced nephrotoxicity models remain explored. Collectively, these findings underscore the potential of epigenetic reprogramming strategies as novel and reversible approaches to mitigate pollutant-induced nephrotoxicity and reduce long-term kidney risk.

5.7. Others

Other molecular mechanisms contributing to kidney programming have been demonstrated in various animal models of developmental origins. These include gut microbiota dysbiosis and perturbations in nutrient-sensing signaling pathways [20,21]. Importantly, these mechanisms do not act in isolation. Many are influenced, directly or indirectly, by environmental toxicants, which can alter microbial composition, interfere with metabolic signaling, and induce heritable changes in gene expression through epigenetic reprogramming [229,230,231].
Such findings suggest that kidney disease and hypertension of developmental origins arise from complex interactions among these mechanisms. While much of this interplay remains to be fully clarified, accumulating evidence underscores their relevance as therapeutic targets. In this context, antioxidant interventions that attenuate oxidative stress may serve as strategies to prevent or mitigate environmental intoxicant exposure-induced kidney programming.

6. Antioxidant-Based Strategies to Preserve Kidney Health

The role of antioxidants in human CKD remains uncertain. CKD is a major risk factor for cardiovascular disease and mortality, with oxidative stress as a key contributor. A Cochrane review of 95 RCTs (10,468 patients) found that antioxidant therapy had little or no effect on all-cause or cardiovascular mortality, kidney transplant outcomes, or proteinuria, though it may modestly reduce cardiovascular events, slow CKD progression, and improve kidney function [232]. Study quality was generally low to moderate, results were heterogeneous, and antioxidant use may increase infection and heart failure risk, limiting confidence in their benefit.
Perinatal oxidative stress plays a pivotal role in environmental toxicant-induced kidney programming, leading to adult-onset kidney disease. Excess ROS or RNS may be mitigated by early-life antioxidant interventions, as suggested by preclinical animal models, even though most human trials have not confirmed protection against pollution-induced oxidative stress [233]. Antioxidants can be endogenous—enzymatic or non-enzymatic—or exogenous, including vitamins, minerals, carotenoids, flavonoids, and other dietary compounds [234], and may be classified as natural or synthetic [235].
Animal studies indicate that natural antioxidants—such as vitamins, amino acids, melatonin, and polyphenols—administered during pregnancy and lactation can protect kidney health and prevent kidney programming [20,21]. Plant-based sources, including fruits, vegetables, seeds, and nuts, are rich in vitamins, carotenoids, polyphenols, and glutathione. Synthetic antioxidants have also been tested in animal models. Nutritional programming emerges as a critical mechanism in oxidative-stress-mediated kidney programming, and certain dietary antioxidants have shown efficacy in preventing environmental toxicant-induced adult-onset kidney disease in preclinical studies, as discussed below.

6.1. Vitamins and Minerals

Although vitamins are widely recognized as dietary antioxidants, their role in preventing kidney programming induced by environmental toxicants remains uncertain. Vitamins A, C, and E, together with selenium and folate, can protect against oxidative-stress-related kidney injury [236]. Vitamin C acts as a free radical scavenger, whereas vitamin E inhibits ROS-generating enzymes [237,238]. In animal studies, perinatal supplementation with these vitamins—alone or combined with selenium and folate—reduced offspring hypertension triggered by maternal stress or inflammation, key features of kidney programming [239,240,241]. However, excessive intake of certain vitamins, particularly vitamin E at high doses (>400 IU/day) [242], as well as vitamins A and β-carotene, has been associated with increased all-cause mortality. The proposed mechanisms include pro-oxidant effects at supraphysiologic levels, interference with anticoagulation, and disruption of redox-sensitive signaling pathways [243]. Additionally, high vitamin A intake during pregnancy (more than 10,000 units per day or 25,000 units per week) is linked to birth defects [244]. Their potential as reprogramming interventions for toxicant-induced kidney programming has not been tested, and contamination of supplements with heavy metals or other toxic elements poses additional risks [245], underscoring the need for cautious perinatal use, especially under environmental pollutant exposure.

6.2. Amino Acids

Several amino acids, including cysteine, taurine, and arginine, possess antioxidant properties [246], and dietary supplementation has been shown to exert therapeutic and protective effects in kidney disease [247,248]. Amino acids play dual roles in environmental nephrotoxicity across the life course. On the one hand, they act as protective modulators: cysteine supports glutathione synthesis, taurine stabilizes membranes and mitigates oxidative stress, and arginine/citrulline sustain NO–mediated vascular and kidney function [249,250,251]. On the other hand, environmental toxicants can disrupt amino acid metabolism—for example, heavy metals deplete sulfur-containing amino acids, while pollutants alter the tryptophan–kynurenine pathway—thereby amplifying oxidative stress, inflammation, and fibrosis [252,253].
During pregnancy, toxicant-induced disturbances in maternal amino acid metabolism or placental amino acid transport can impair nephrogenesis, leading to reduced nephron endowment and greater lifelong CKD susceptibility in offspring [254,255,256]. Notably, maternal citrulline supplementation has been shown to improve NO bioavailability and protect adult rat offspring against kidney programming in a maternal adenine-induced CKD model [257]. Similarly, tryptophan and cysteine have been investigated as reprogramming interventions to prevent offspring hypertension in maternal CKD-related models [258,259]. While these antioxidant amino acids show promise in mitigating kidney programming, their specific effects in toxicant-induced models remain unclear. Given that AhR is a key target of environmental toxicants [219,220] and that tryptophan catabolites act as AhR ligands in the developmental programming of kidney disease [253], tryptophan metabolites hold potential to counteract environmental toxicant–induced kidney injury in offspring, highlighting the need for further studies to define their protective mechanisms.

6.3. Polyphenols

Polyphenols represent the most abundant class of bioactive phytochemicals and are widely recognized for their antioxidant and health-promoting properties [260,261]. Among them, resveratrol has been extensively studied; it can chelate transition metals, neutralize free radicals, activate NOS, and enhance the activity of antioxidant enzymes [262]. Because of these actions, polyphenols have attracted attention as potential interventions to support kidney health [263,264].
In the setting of environmental exposure, numerous polyphenols demonstrate nephroprotective actions in experimental models [263,264,265]. Broadly, polyphenols are divided into two categories: flavonoids and non-flavonoids [260]. Flavonoid compounds, such as quercetin, when administered during pregnancy, have been shown to protect offspring from high-fat maternal diet–induced kidney programming and subsequent hypertension [266]. Likewise, epigallocatechin gallate provided during gestation and lactation mitigated the hypertensive effects of prenatal dexamethasone exposure in rat offspring [267].
Resveratrol, a non-flavonoid polyphenol, exerts multiple antioxidant effects, including reducing levels of ROS/RNS, elevating glutathione, upregulating NOS expression, and boosting enzymatic antioxidant defenses [268,269,270]. Beyond its antioxidant profile, resveratrol also functions as an AhR antagonist [271]. In toxicant-induced models of kidney programming—such as those involving TCDD [80,188], BPA [191], and DEHP [193]—maternal resveratrol supplementation improved kidney outcomes in adult offspring. In BPA-exposed pregnancies, for example, resveratrol alleviated offspring hypertension by restoring NO bioavailability, reducing oxidative stress, and suppressing AhR signaling [191]. Emerging evidence suggests that AhR-induced oxidative stress is a key contributor to kidney disease [271]. TCDD-induced hypertension is associated with AhR activation, and resveratrol supplementation during gestation and lactation can mitigate TCDD-induced AhR signaling and oxidative stress. Similarly, perinatal resveratrol therapy reverses maternal BPA exposure–induced increases in AHR protein levels and mRNA expression of AHRR, CYP1A1, and ARNT [191]. Collectively, evidence from animal studies highlights resveratrol and other polyphenols as promising reprogramming agents against environmentally induced kidney disease and hypertension [272].

6.4. Melatonin

Melatonin, a tryptophan-derived circadian hormone, supports pregnancy and fetal development while providing antioxidant protection through ROS/RNS scavenging, activation of antioxidant enzymes, and restoration of NO signaling [273,274,275,276]. Light pollution suppresses nocturnal melatonin secretion, resulting in circadian misalignment, oxidative stress, and metabolic dysregulation [162,163,164]. During gestation, reduced maternal melatonin may impair fetal programming, decrease nephron endowment, and increase susceptibility to kidney disease in later life. Evidence from animal studies indicates that perinatal melatonin supplementation can prevent kidney programming across diverse experimental models [168,275]. Although melatonin is generally safe in children, its use during pregnancy is not currently recommended [277,278]. Further research is needed to determine whether restoring melatonin signaling can counteract kidney programming induced by light pollution as well as other environmental toxicants.

6.5. Synthetic Antioxidants

Alongside natural antioxidants, several synthetic compounds have been studied in kidney disease [232]. MitoQ, a coenzyme Q10 analog, reduces oxidative stress by suppressing superoxide and lipid peroxidation, and perinatal treatment prevented hypertension and kidney injury in offspring of smoking-exposed mice [279]. Dimethyl fumarate, an Nrf2 activator, improved renal outcomes in a dexamethasone–high-fat diet model by lowering 8-OHdG while enhancing NO [280]. Tempol, a SOD mimetic, also show protective effects in experimental hypertension [281]. NAC, a synthetic L-cysteine analog and glutathione precursor, has demonstrated perinatal protection against kidney programming in various animal models [21,282], but its role in toxicant-induced kidney programming remains untested. To date, none of these synthetic antioxidants have been applied in clinical practice during pregnancy. Figure 2 illustrates potential antioxidant-based strategies to counter kidney programming and preserve kidney health in the face of environmental pollutants.

7. Conclusions and Future Perspectives

Preventing environmental nephrotoxicity requires a life-course-oriented strategy that integrates risk reduction, early intervention, and ongoing monitoring [283,284] (Figure 3). At the earliest stage, minimizing exposure to nephrotoxic pollutants and optimizing maternal and neonatal environments—including limiting light pollution to preserve circadian rhythms—represents a critical first line of defense. Interventions during pregnancy and early life, such as maternal antioxidant supplementation (e.g., vitamins C and E) [285,286] or promotion of exclusive breastfeeding [287,288,289], may help counteract oxidative-stress-driven kidney programming. Early detection through noninvasive monitoring and the use of oxidative stress biomarkers can identify fetuses, neonates, and children at risk, enabling timely preventive or therapeutic measures [290]. Later in life, lifestyle guidance, targeted interventions, and longitudinal follow-up remain essential to protect kidney health and reduce long-term disease burden [291].
Despite promising preclinical evidence, translation into human practice remains limited. Most data on antioxidant use to prevent pollutant-induced kidney damage in pregnancy are derived from animal studies, providing mechanistic insights but limited direct applicability. While antioxidants can reduce oxidative stress in CKD patients [292,293], none are currently recommended in clinical guidelines, and careful attention to compound, dose, and timing is needed to avoid pro-oxidant effects—for example, vitamin E may increase oxidative stress if co-antioxidants like vitamin C are insufficient [294]. Candidate biomarkers (e.g., urinary 8-OHdG, F2-isoprostanes, glutathione redox ratio) may help identify individuals with elevated oxidative burden who are most likely to benefit from targeted antioxidant therapy. These limitations highlight the need for well-designed human studies to guide safe antioxidant interventions during pregnancy and early life.
Early-life oxidative stress remains a major driver of kidney programming, yet maternal and neonatal interventions have been underexplored, and human studies are sparse, particularly in fetuses and neonates. Breastfeeding, rich in natural antioxidants, represents a safe and potentially effective preventive measure that warrants further study [287,288,289].
The development and validation of reliable oxidative stress biomarkers is crucial to guide intervention. Although no single biomarker is ideal, panels that capture the pathogenic processes observed in animal studies may improve early detection of kidney injury [295,296]. Non-coding RNAs, which are increasingly recognized as key regulators of kidney disease and epigenetic modifications [297], also represent innovative candidates for biomarkers in environmental nephrotoxicity and CKD. Emerging technologies, such as liquid biopsies, may further facilitate rapid and noninvasive monitoring of oxidative-stress–related kidney damage [298].
Overall, optimizing kidney outcomes requires a comprehensive, perinatal-focused approach: reducing environmental exposures, applying evidence-based antioxidant strategies, and integrating validated biomarkers for early risk identification. This integrated framework bridges preclinical findings with clinical practice, offering a pathway to prevent environmentally induced kidney disease across the life course.

Author Contributions

Conceptualization, Writing—original draft, Y.-L.T. and C.-N.H.; funding acquisition, Y.-L.T. and C.-N.H.; data curation, C.-Y.H., Y.-L.T., Y.-W.C., G.-P.C.-C., S.-F.L. and C.-N.H.; writing—review and editing, Y.-W.C., C.-Y.H., Y.-L.T., G.-P.C.-C., S.-F.L. and C.-N.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by financial assistance from the National Science and Technology Council, Taiwan, under grant numbers 114-2314-B-182A-036-MY3 and 114-2314-B-182A-048 and CMRPG8Q0261 from Kaohsiung Chang Gung Memorial Hospital, Taiwan.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Acknowledgments

We would like to thank the Super Micro Mass Research and Technology Center, the Institute of Environmental Toxin and Emerging-Contaminant, and the Center for Environmental Toxin and Emerging Contaminant Research, Cheng Shiu University, Kaohsiung, for technical support. During the preparation of this manuscript, the authors used Napkin AI Image Generator [https://www.napkin.ai/ (accessed on 31 August 2025)] for the purpose of generating figures. The authors have reviewed and edited the output and take full responsibility for the content of this publication.

Conflicts of Interest

The authors declare no conflicts of interest with regard to the contents of this manuscript.

References

  1. Jager, K.J.; Kovesdy, C.; Langham, R.; Rosenberg, M.; Jha, V.; Zoccali, C. A single number for advocacy and communicationworldwide more than 850 million individuals have kidney diseases. Nephrol. Dial. Transplant. 2019, 34, 1803–1805. [Google Scholar] [CrossRef]
  2. World Health Organization. The top 10 Causes of Death. Available online: https://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death (accessed on 6 August 2025).
  3. Waith, F.M.; Bresolin, N.L.; Antwi, S. Detect early, protect kidney health: World Kidney Day 2025. Pediatr. Nephrol. 2025, 40, 1511–1514. [Google Scholar] [CrossRef]
  4. Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work Group. KDIGO 2024 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease. Kidney Int. 2024, 105, S117–S314. [Google Scholar] [CrossRef]
  5. Tain, Y.L. Advocacy for DOHaD research optimizing child kidney health. Pediatr, Neonatol. 2025, 66 (Suppl. 1), S18–S22. [Google Scholar] [CrossRef]
  6. Kett, M.M.; Denton, K.M. Renal programming: Cause for concern? Am. J. Physiol. Regul. Integr. Comp. Physiol. 2011, 300, R791–R803. [Google Scholar] [CrossRef]
  7. Barker, D.J.; Eriksson, J.G.; Forsén, T.; Osmond, C. Fetal origins of adult disease: Strength of effects and biological basis. Int. J. Epidemiol. 2002, 31, 1235–1239. [Google Scholar] [CrossRef] [PubMed]
  8. Gluckman, P.D.; Hanson, M.A.; Buklijas, T. A Conceptual Framework for the Developmental Origins of Health and Disease. J. Dev. Origins Orig. Health Dis. 2010, 1, 6–18. [Google Scholar] [CrossRef] [PubMed]
  9. Lao, X.Q.; Bo, Y.; Chen, D.; Zhang, K.; Szeto, C.C. Environmental pollution to kidney disease: An updated review of current knowledge and future directions. Kidney Int. 2024, 106, 214–225. [Google Scholar] [CrossRef] [PubMed]
  10. Hsu, C.N.; Tain, Y.L. Adverse Impact of Environmental Chemicals on Developmental Origins of Kidney Disease and Hypertension. Front. Endocrinol. 2021, 12, 745716. [Google Scholar] [CrossRef]
  11. Jalili, C.; Kazemi, M.; Cheng, H.; Mohammadi, H.; Babaei, A.; Taheri, E.; Moradi, S. Associations between exposure to heavy metals and the risk of chronic kidney disease: A systematic review and meta-analysis. Crit. Rev. Toxicol. 2021, 51, 165–182. [Google Scholar] [CrossRef]
  12. Wu, M.Y.; Lo, W.C.; Chao, C.T.; Wu, M.S.; Chiang, C.K. Association between air pollutants and development of chronic kidney disease: A systematic review and meta-analysis. Sci. Total Environ. 2020, 706, 135522. [Google Scholar] [CrossRef]
  13. Ramya Ranjan Nayak, S.P.; Boopathi, S.; Haridevamuthu, B.; Arockiaraj, J. Toxic ties: Unraveling the complex relationship between endocrine disrupting chemicals and chronic kidney disease. Environ. Pollut. 2023, 338, 122686. [Google Scholar] [CrossRef]
  14. Kataria, A.; Trasande, L.; Trachtman, H. The effects of environmental chemicals on renal function. Nat. Rev. Nephrol. 2015, 11, 610–625. [Google Scholar] [CrossRef] [PubMed]
  15. de Oliveira, R.B.; Pelepenko, L.E.; Masaro, D.A.; Lustosa, G.M.M.M.; de Oliveira, M.C.; Roza, N.A.V.; Marciano, M.A.; Dos Reis, L.M.; Kamel, S.; Louvet, L.; et al. Effects of microplastics on the kidneys: A narrative review. Kidney Int. 2024, 106, 400–407. [Google Scholar] [CrossRef]
  16. Wu, H.; Eckhardt, C.M.; Baccarelli, A.A. Molecular mechanisms of environmental exposures and human disease. Nat. Rev. Genet. 2023, 24, 332–344. [Google Scholar] [CrossRef]
  17. Ahmad, S. Oxidative stress from environmental pollutants. Arch. Insect. Biochem. Physiol. 1995, 29, 135–157. [Google Scholar] [CrossRef] [PubMed]
  18. Xu, W.; Wang, S.; Jiang, L.; Sun, X.; Wang, N.; Liu, X.; Yao, X.; Qiu, T.; Zhang, C.; Li, J.; et al. The influence of PM2.5 exposure on kidney diseases. Hum. Exp. Toxicol. 2022, 41, 9603271211069982. [Google Scholar] [CrossRef]
  19. Lee, S.E.; Kim, D.Y.; Jeong, T.S.; Park, Y.S. Micro- and Nano-Plastic-Induced Adverse Health Effects on Lungs and Kidneys Linked to Oxidative Stress and Inflammation. Life 2025, 15, 392. [Google Scholar] [CrossRef]
  20. Hsu, C.N.; Tain, Y.L. Developmental Origins of Kidney Disease: Why Oxidative Stress Matters? Antioxidants 2020, 10, 33. [Google Scholar] [CrossRef] [PubMed]
  21. Tain, Y.L.; Hsu, C.N. Perinatal Oxidative Stress and Kidney Health: Bridging the Gap between Animal Models and Clinical Reality. Antioxidants 2022, 12, 13. [Google Scholar] [CrossRef]
  22. Park, M.Y.; Kang, M.Y. Occupational Risk Factors for Kidney Disease: A Comprehensive Review. J. Korean Med. Sci. 2025, 40, e224. [Google Scholar] [CrossRef]
  23. Radi, R. Oxygen radicals, nitric oxide, and peroxynitrite: Redox pathways in molecular medicine. Proc. Natl. Acad. Sci. USA 2018, 115, 5839–5848. [Google Scholar] [CrossRef] [PubMed]
  24. Wilcox, C.S. Reactive oxygen species: Roles in blood pressure and kidney function. Curr. Hypertens. Rep. 2002, 4, 160–166. [Google Scholar] [CrossRef] [PubMed]
  25. Kone, B.C. Nitric oxide synthesis in the kidney: Isoforms, biosynthesis, and functions in health. Semin. Nephrol. 2004, 24, 299–315. [Google Scholar] [CrossRef] [PubMed]
  26. Wilcox, C.S. Oxidative stress and nitric oxide deficiency in the kidney: A critical link to hypertension? Am. J. Physiol. Regul. Integr. Comp. Physiol. 2005, 289, R913–R935. [Google Scholar] [CrossRef]
  27. Rochette, L.; Lorin, J.; Zeller, M.; Guilland, J.C.; Lorgis, L.; Cottin, Y.; Vergely, C. Nitric oxide synthase inhibition and oxidative stress in cardiovascular diseases: Possible therapeutic targets? Pharmacol. Ther. 2013, 140, 239–257. [Google Scholar] [CrossRef]
  28. Baylis, C.; Qiu, C. Importance of nitric oxide in the control of renal hemodynamics. Kidney Int. 1996, 49, 1727–1731. [Google Scholar] [CrossRef]
  29. Pacher, P.; Beckman, J.S.; Liaudet, L. Nitric oxide and peroxynitrite in health and disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef]
  30. Jenkins, C.; Wilson, R.; Roberts, J.; Miller, H.; McKillop, J.H.; Walker, J.J. Antioxidants: Their role in pregnancy and miscarriage. Antioxid. Redox Signal. 2000, 2, 623–628. [Google Scholar] [CrossRef]
  31. Zhang, J.J.; Du, J.; Kong, N.; Zhang, G.Y.; Liu, M.Z.; Liu, C. Mechanisms and pharmacological applications of ferroptosis: A narrative review. Ann. Transl. Med. 2021, 9, 1503. [Google Scholar] [CrossRef]
  32. Daenen, K.; Andries, A.; Mekahli, D.; Van Schepdael, A.; Jouret, F.; Bammens, B. Oxidative stress in chronic kidney disease. Pediatr. Nephrol. 2019, 34, 975–991. [Google Scholar] [CrossRef] [PubMed]
  33. Kishi, S.; Nagasu, H.; Kidokoro, K.; Kashihara, N. Oxidative stress and the role of redox signalling in chronic kidney disease. Nat. Rev. Nephrol. 2024, 20, 101–119. [Google Scholar] [CrossRef]
  34. Poulianiti, K.P.; Kaltsatou, A.; Mitrou, G.I.; Jamurtas, A.Z.; Koutedakis, Y.; Maridaki, M.; Stefanidis, I.; Sakkas, G.K.; Karatzaferi, C. Systemic Redox Imbalance in Chronic Kidney Disease: A Systematic Review. Oxid. Med. Cell. Longev. 2016, 2016, 8598253. [Google Scholar] [CrossRef]
  35. Tucker, P.S.; Dalbo, V.J.; Han, T.; Kingsley, M.I. Clinical and research markers of oxidative stress in chronic kidney disease. Biomarkers 2013, 18, 103–115. [Google Scholar] [CrossRef]
  36. Sahni, N.; Gupta, K.L.; Rana, S.V.; Prasad, R.; Bhalla, A.K. Intake of antioxidants and their status in chronic kidney disease patients. J. Ren. Nutr. 2012, 22, 389–399. [Google Scholar] [CrossRef] [PubMed]
  37. Praticò, D.; Iuliano, L.; Mauriello, A.; Spagnoli, L.; Lawson, J.A.; Rokach, J.; Maclouf, J.; Violi, F.; FitzGerald, G.A. Localization of distinct F2-isoprostanes in human atherosclerotic lesions. J. Clin. Investig. 1997, 100, 2028–2034. [Google Scholar] [CrossRef] [PubMed]
  38. Ward, R.A.; Ouseph, R.; McLeish, K.R. Effects of high-flux hemodialysis on oxidant stress. Kidney Int. 2003, 63, 353–359. [Google Scholar] [CrossRef]
  39. Dai, L.; Watanabe, M.; Qureshi, A.R.; Mukai, H.; Machowska, A.; Heimbürger, O.; Barany, P.; Lindholm, B.; Stenvinkel, P. Serum 8-hydroxydeoxyguanosine, a marker of oxidative DNA damage, is associated with mortality independent of inflammation in chronic kidney disease. Eur. J. Intern. Med. 2019, 68, 60–65. [Google Scholar] [CrossRef]
  40. Crawford, A.; Fassett, R.G.; Coombes, J.S.; Kunde, D.A.; Ahuja, K.D.; Robertson, I.K.; Ball, M.J.; Geraghty, D.P. Glutathione peroxidase, superoxide dismutase and catalase genotypes and activities and the progression of chronic kidney disease. Nephrol. Dial. Transplant. 2011, 26, 2806–2813. [Google Scholar] [CrossRef]
  41. Annuk, M.; Zilmer, M.; Lind, L.; Linde, T.; Fellström, B. Oxidative stress and endothelial function in chronic renal failure. J. Am. Soc. Nephrol. 2001, 12, 2747–2752. [Google Scholar] [CrossRef]
  42. Lee, O.Y.A.; Wong, A.N.N.; Ho, C.Y.; Tse, K.W.; Chan, A.Z.; Leung, G.P.; Kwan, Y.W.; Yeung, M.H.Y. Potentials of Natural Antioxidants in Reducing Inflammation and Oxidative Stress in Chronic Kidney Disease. Antioxidants 2024, 13, 751. [Google Scholar] [CrossRef] [PubMed]
  43. Tsinari, A.; Roumeliotis, S.; Neofytou, I.E.; Varouktsi, G.; Veljkovic, A.; Stamou, A.; Leivaditis, K.; Liakopoulos, V. The Clinical Utility and Plausibility of Oxidative and Antioxidant Variables in Chronic and End-Stage Kidney Disease: A Review of the Literature. Int. J. Mol. Sci. 2025, 26, 3376. [Google Scholar] [CrossRef]
  44. Lu, P.C.; Tain, Y.L.; Lin, Y.J.; Hsu, C.N. Oxidative Stress in Maternal and Offspring Kidney Disease and Hypertension: A Life-Course Perspective. Antioxidants 2025, 14, 387. [Google Scholar] [CrossRef]
  45. Hsu, C.N.; Tain, Y.L. Early-Life Programming and Reprogramming of Adult Kidney Disease and Hypertension: The Interplay between Maternal Nutrition and Oxidative Stress. Int. J. Mol. Sci. 2020, 21, 3572. [Google Scholar] [CrossRef]
  46. Dennery, P.A. Oxidative stress in development: Nature or nurture? Free Radic. Biol. Med. 2010, 49, 1147–1151. [Google Scholar] [CrossRef]
  47. Thompson, L.P.; Al-Hasan, Y. Impact of oxidative stress in fetal programming. J. Pregnancy 2012, 2012, 582748. [Google Scholar] [CrossRef]
  48. Luyckx, V.A.; Brenner, B.M. The clinical importance of nephron mass. J. Am. Soc. Nephrol. 2010, 21, 898–910. [Google Scholar] [CrossRef]
  49. Murugapoopathy, V.; Gupta, I.R. A Primer on Congenital Anomalies of the Kidneys and Urinary Tracts (CAKUT). Clin. J. Am. Soc. Nephrol. 2020, 15, 723–731. [Google Scholar] [CrossRef] [PubMed]
  50. Dwivedi, R.S.; Herman, J.G.; McCaffrey, T.A.; Raj, D.S. Beyond genetics: Epigenetic code in chronic kidney disease. Kidney Int. 2011, 79, 23–32. [Google Scholar] [CrossRef] [PubMed]
  51. Tain, Y.L.; Huang, L.T.; Chan, J.Y.; Lee, C.T. Transcriptome analysis in rat kidneys: Importance of genes involved in programmed hypertension. Int. J. Mol. Sci. 2015, 16, 4744–4758. [Google Scholar] [CrossRef] [PubMed]
  52. Tain, Y.L.; Lee, C.T.; Chan, J.Y.; Hsu, C.N. Maternal melatonin or N-acetylcysteine therapy regulates hydrogen sulfide generating pathway and renal transcriptome to prevent prenatal N(G)-Nitro-L-arginine methyl ester (L-NAME)-induced fetal programming of hypertension in adult male offspring. Am. J. Obstet. Gynecol. 2016, 215, 636.E1–636.E72. [Google Scholar] [CrossRef] [PubMed]
  53. Langley-Evans, S.C. Nutritional programming of disease: Unravelling the mechanism. J. Anat. 2009, 215, 36–51. [Google Scholar] [CrossRef]
  54. Martínez Gascón, L.E.; Ortiz, M.C.; Galindo, M.; Sanchez, J.M.; Sancho-Rodriguez, N.; Albaladejo Otón, M.D.; Rodriguez Mulero, M.D.; Rodriguez, F. Role of heme oxygenase in the regulation of the renal hemodynamics in a model of sex dependent programmed hypertension by maternal diabetes. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2022, 322, R181–R191. [Google Scholar] [CrossRef]
  55. Vieira, L.D.; Farias, J.S.; de Queiroz, D.B.; Cabral, E.V.; Lima-Filho, M.M.; Sant’Helena, B.R.M.; Aires, R.S.; Ribeiro, V.S.; SantosRocha, J.; Xavier, F.E.; et al. Oxidative stress induced by prenatal LPS leads to endothelial dysfunction and renal haemodynamic changes through angiotensin II/NADPH oxidase pathway: Prevention by early treatment with α-tocopherol. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 3577–3587. [Google Scholar] [CrossRef]
  56. Sukjamnong, S.; Chan, Y.L.; Zakarya, R.; Nguyen, L.T.; Anwer, A.G.; Zaky, A.A.; Santiyanont, R.; Oliver, B.G.; Goldys, E.; Pollock, C.A.; et al. MitoQ supplementation prevent long-term impact of maternal smoking on renal development, oxidative stress and mitochondrial density in male mice offspring. Sci. Rep. 2018, 8, 6631. [Google Scholar] [CrossRef]
  57. Tai, I.H.; Sheen, J.M.; Lin, Y.J.; Yu, H.R.; Tiao, M.M.; Chen, C.C.; Huang, L.T.; Tain, Y.L. Maternal N-acetylcysteine therapy regulates hydrogen sulfide-generating pathway and prevents programmed hypertension in male offspring exposed to prenatal dexamethasone and postnatal high-fat diet. Nitric Oxide 2016, 53, 6–12. [Google Scholar] [CrossRef]
  58. Larkin, B.P.; Saad, S.; Glastras, S.J.; Nguyen, L.T.; Hou, M.; Chen, H.; Wang, R.; Pollock, C.A. Low-dose hydralazine during gestation reduces renal fibrosis in rodent offspring exposed to maternal high fat diet. PLoS ONE 2021, 16, e0248854. [Google Scholar] [CrossRef] [PubMed]
  59. Svitok, P.; Okuliarova, M.; Varga, I.; Zeman, M. Renal impairment induced by prenatal exposure to angiotensin II in male rat offspring. Exp. Biol. Med. 2019, 244, 923–931. [Google Scholar] [CrossRef] [PubMed]
  60. Do Nascimento, L.C.P.; Neto, J.P.R.C.; de Andrade Braga, V.; Lagranha, C.J.; de Brito Alves, J.L. Maternal exposure to high-fat and high-cholesterol diet induces arterial hypertension and oxidative stress along the gut-kidney axis in rat offspring. Life Sci. 2020, 261, 118367. [Google Scholar] [CrossRef]
  61. Tain, Y.L.; Lee, W.C.; Hsu, C.N.; Lee, W.C.; Huang, L.T.; Lee, C.T.; Lin, C.Y. Asymmetric dimethylarginine is associated with developmental programming of adult kidney disease and hypertension in offspring of streptozotocin-treated mothers. PLoS ONE 2013, 8, e55420. [Google Scholar] [CrossRef]
  62. Tain, Y.L.; Hsu, C.N. The NOS/NO System in Renal Programming and Reprogramming. Antioxidants 2023, 12, 1629. [Google Scholar] [CrossRef]
  63. Ranjbar, A.; Ghasemi, H.; Hatami, M.; Dadras, F.; Heidary Shayesteh, T.; Khoshjou, F. Tempol effects on diabetic nephropathy in male rats. J. Renal Inj. Prev. 2016, 5, 74–78. [Google Scholar] [CrossRef]
  64. Lin, Y.J.; Lin, I.C.; Yu, H.R.; Sheen, J.M.; Huang, L.T.; Tain, Y.L. Early Postweaning Treatment with Dimethyl Fumarate Prevents Prenatal Dexamethasone- and Postnatal High-Fat Diet-Induced Programmed Hypertension in Male Rat Offspring. Oxid. Med. Cell. Longev. 2018, 2018, 5343462. [Google Scholar] [CrossRef]
  65. Kaur, H.; Satyanarayana, P.S.; Chopra, K. Effect of U-74500A, a 21-aminosteroid on renal ischemia-reperfusion injury in rats. Ren. Fail. 2003, 25, 157–171. [Google Scholar] [CrossRef] [PubMed]
  66. Koeners, M.P.; Braam, B.; Joles, J.A. Perinatal inhibition of NF-kappaB has long-term antihypertensive effects in spontaneously hypertensive rats. J. Hypertens. 2011, 29, 1160–1166. [Google Scholar] [CrossRef] [PubMed]
  67. Das, A. The emerging role of microplastics in systemic toxicity: Involvement of reactive oxygen species (ROS). Sci. Total Environ. 2023, 895, 165076. [Google Scholar] [CrossRef] [PubMed]
  68. Quasmi, M.N.; Singh, J.; Kumar, D.; Dhingra, D.; Jangra, A. Insights into the molecular mechanisms underlying phthalates-induced nephrotoxicity. Toxicology 2025, 516, 154187. [Google Scholar] [CrossRef]
  69. Ye, J.J.; Wang, S.S.; Fang, Y.; Zhang, X.J.; Hu, C.Y. Ambient air pollution exposure and risk of chronic kidney disease: A systematic review of the literature and meta-analysis. Environ. Res. 2021, 195, 110867. [Google Scholar] [CrossRef]
  70. Gowder, S.J. Nephrotoxicity of bisphenol A (BPA)—an updated review. Curr. Mol. Pharmacol. 2013, 6, 163–172. [Google Scholar] [CrossRef]
  71. Yadav, R.; Kumar, D.; Singh, J.; Jangra, A. Environmental toxicants and nephrotoxicity: Implications on mechanisms and therapeutic strategies. Toxicology 2024, 504, 153784. [Google Scholar] [CrossRef]
  72. Xu, X.; Yu, Z.; Han, B.; Li, S.; Sun, Y.; Du, Y.; Wang, Z.; Gao, D.; Zhang, Z. Luteolin alleviates inorganic mercury-induced kidney injury via activation of the AMPK/mTOR autophagy pathway. J. Inorg. Biochem. 2021, 224, 111583. [Google Scholar] [CrossRef] [PubMed]
  73. Bosch-Panadero, E.; Mas, S.; Civantos, E.; Abaigar, P.; Camarero, V.; Ruiz-Priego, A.; Ortiz, A.; Egido, J.; González-Parra, E. Bisphenol A is an exogenous toxin that promotes mitochondrial injury and death in tubular cells. Environ. Toxicol. 2018, 33, 325–332. [Google Scholar] [CrossRef] [PubMed]
  74. Dopico, M.; Gόmez, A. Review of the Current State and Main Sources of Dioxins around the World. J. Air Waste Manag. Assoc. 2015, 65, 1033–1049. [Google Scholar] [CrossRef]
  75. Milbrath, M.O.; Wenger, Y.; Chang, C.W.; Emond, C.; Garabrant, D.; Gillespie, B.W.; Jolliet, O. Apparent half-lives of dioxins, furans, and polychlorinated biphenyls as a function of age, body fat, smoking status, and breast-feeding. Environ. Health Perspect. 2009, 117, 417–425. [Google Scholar] [CrossRef]
  76. Bock, K.W. Aryl hydrocarbon or dioxin receptor: Biologic and toxic responses. Rev. Physiol. Biochem. Pharmacol. 1994, 125, 1–42. [Google Scholar]
  77. Doğan, M.F.; Başak Türkmen, N.; Taşlıdere, A.; Şahin, Y.; Çiftçi, O. The protective effects of capsaicin on oxidative damage-induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin in rats. Drug Chem. Toxicol. 2022, 45, 2463–2470. [Google Scholar] [CrossRef]
  78. Zhang, X.; Flaws, J.A.; Spinella, M.J.; Irudayaraj, J. The Relationship between Typical Environmental Endocrine Disruptors and Kidney Disease. Toxics 2022, 11, 32. [Google Scholar] [CrossRef]
  79. Viluksela, M.; Pohjanvirta, R. Multigenerational and Transgenerational Effects of Dioxins. Int. J. Mol. Sci. 2019, 20, 2947. [Google Scholar] [CrossRef]
  80. Hsu, C.N.; Hung, C.H.; Hou, C.Y.; Chang, C.I.; Tain, Y.L. Perinatal Resveratrol Therapy to Dioxin-Exposed Dams Prevents the Programming of Hypertension in Adult Rat Offspring. Antioxidants 2021, 10, 1393. [Google Scholar] [CrossRef] [PubMed]
  81. Aragon, A.C.; Goens, M.B.; Carbett, E.; Walker, M.K. Perinatal 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure sensitizes offspring to angiotensin II-induced hypertension. Cardiovasc. Toxicol. 2008, 8, 145–154. [Google Scholar] [CrossRef]
  82. Morrissey, R.E.; Harris, M.W.; Diliberto, J.J.; Birnbaum, L.S. Limited PCB antagonism of TCDD-induced malformations in mice. Toxicol. Lett. 1992, 60, 19–25. [Google Scholar] [CrossRef]
  83. Hahladakis, J.N.; Velis, C.A.; Weber, R.; Lacovidou, E.; Purnell, P. An overview of chemical additives present in plastics: Migration, release, fate and environmental impact during their use, disposal and recycling. J. Hazard. Mater. 2018, 344, 179–199. [Google Scholar] [CrossRef]
  84. Symeonides, C.; Aromataris, E.; Mulders, Y.; Dizon, J.; Stern, C.; Barker, T.H.; Whitehorn, A.; Pollock, D.; Marin, T.; Dunlop, S. An Umbrella Review of Meta-Analyses Evaluating Associations between Human Health and Exposure to Major Classes of Plastic-Associated Chemicals. Ann. Glob. Health. 2024, 90, 52. [Google Scholar] [CrossRef]
  85. Rudel, R.A.; Camann, D.E.; Spengler, J.D.; Korn, L.R.; Brody, J.G. Phthalates, alkylphenols, pesticides, polybrominated diphenyl ethers, and other endocrine-disrupting compounds in indoor air and dust. Environ. Sci. Technol. 2003, 37, 4543–4553. [Google Scholar] [CrossRef] [PubMed]
  86. Giuliani, A.; Zuccarini, M.; Cichelli, A.; Khan, H.; Reale, M. Critical review on the presence of phthalates in food and evidence of their biological impact. Int. J. Environ. Res. Public Health 2020, 17, 5655. [Google Scholar] [CrossRef]
  87. Zhang, Y.J.; Guo, J.L.; Xue, J.C.; Bai, C.L.; Guo, Y. Phthalate metabolites: Characterization, toxicities, global distribution, and exposure assessment. Environ. Pollut. 2021, 291, 118106. [Google Scholar] [CrossRef] [PubMed]
  88. UNEP. Chemicals in Plastics—A Technical Report. United Nations Environment Programme (UNEP) and Secretariat of the Basel, Rotterdam and Stockholm Conventions. 2023. Available online: http://www.unep.org/resources/report/chemicals-plastics-technical-report (accessed on 10 August 2025).
  89. Sweeney, M.R.; O’Leary, K.G.; Jeney, Z.; Braunlin, M.C.; Gibb, H.J. Systematic review and quality ranking of studies of two phthalate metabolites and anogenital distance, bone health, inflammation, and oxidative stress. Crit. Rev. Toxicol. 2019, 49, 281–301. [Google Scholar] [CrossRef] [PubMed]
  90. Kirtana, A.; Seetharaman, B. Comprehending the Role of Endocrine Disruptors in Inducing Epigenetic Toxicity. Endocr. Metab. Immune Disord. Drug Targets 2022, 22, 1059–1072. [Google Scholar] [CrossRef]
  91. Hau, A.K.; Kwan, T.H.; Li, P.K. Melamine toxicity and the kidney. J. Am. Soc Nephrol. 2009, 20, 245–250. [Google Scholar] [CrossRef]
  92. Kang, H.; Lee, J.P.; Choi, K. Exposure to phthalates and environmental phenols in association with chronic kidney disease (CKD) among the general US population participating in multi-cycle NHANES (2005-2016). Sci. Total Environ. 2021, 791, 148343. [Google Scholar] [CrossRef]
  93. Han, C.; Hong, Y.C. Bisphenol A, Hypertension, and Cardiovascular Diseases: Epidemiological, Laboratory, and Clinical Trial Evidence. Curr. Hypertens. Rep. 2016, 18, 11. [Google Scholar] [CrossRef]
  94. Bae, S.; Lim, Y.H.; Lee, Y.A.; Shin, C.H.; Oh, S.Y.; Hong, Y.C. Maternal Urinary Bisphenol A Concentration During Midterm Pregnancy and Children’s Blood Pressure at Age 4. Hypertension 2017, 69, 367–374. [Google Scholar] [CrossRef]
  95. Sol, C.M.; Santos, S.; Asimakopoulos, A.G.; Martinez-Moral, M.P.; Duijts, L.; Kannan, K.; Trasande, L.; Jaddoe, V.W.V. Associations of maternal phthalate and bisphenol urine concentrations during pregnancy with childhood blood pressure in a population-based prospective cohort study. Environ. Int. 2020, 138, 105677. [Google Scholar] [CrossRef]
  96. Ouyang, F.; Zhang, G.H.; Du, K.; Shen, L.; Ma, R.; Wang, X.; Wang, X.; Zhang, J. Maternal prenatal urinary bisphenol A level and child cardio-metabolic risk factors: A prospective cohort study. Environ. Pollut. 2020, 265, 115008. [Google Scholar] [CrossRef] [PubMed]
  97. Lee, C.C.; Bhandari, J.C.; Winston, J.M.; House, W.B.; Peters, P.J.; Dixon, R.L.; Woods, J.S. Inhalation toxicity of vinyl chloride and vinylidene chloride. Environ. Health Perspect. 1977, 21, 25–32. [Google Scholar] [CrossRef]
  98. Hsu, Y.H.; Chuang, H.C.; Lee, Y.H.; Lin, Y.F.; Chiu, Y.J.; Wang, Y.L.; Wu, M.S.; Chiu, H.W. Induction of Fibrosis and Autophagy in Kidney Cells by Vinyl Chloride. Cells 2019, 8, 601. [Google Scholar] [CrossRef]
  99. Mutti, A.; Coccini, T.; Alinovi, R.; Toubeau, G.; Broeckaert, F.; Bergamaschi, E.; Mozzoni, P.; Nonclercq, D.; Bernard, A.; Manzo, L. Exposure to hydrocarbons and renal disease: An experimental animal model. Ren. Fail. 1999, 21, 369–385. [Google Scholar] [CrossRef] [PubMed]
  100. Li, X.; Wang, Q.; Jiang, N.; Lv, H.; Liang, C.; Yang, H.; Yao, X.; Wang, J. Occurrence, source, ecological risk, and mitigation of phthalates (PAEs) in agricultural soils and the environment: A review. Environ. Res. 2023, 220, 115196. [Google Scholar] [CrossRef]
  101. Li, Z.; He, J.; Li, X.; Chen, J.; You, M.; Sun, B.; Yang, G. A narrative review of phthalates: From environmental release to kidney injury. Environ. Pollut. 2025, 376, 126380. [Google Scholar] [CrossRef] [PubMed]
  102. Jin, S.; Cui, S.; Mu, X.; Liu, Z.; Han, Y.; Cui, T.; Xiong, W.; Xi, W.; Zhang, X. Exposure to phthalates and their alternatives in relation to biomarkers of inflammation and oxidative stress in adults: Evidence from NHANES 2017–2018. Environ. Sci. Pollut. Res. Int. 2023, 30, 123770–123784. [Google Scholar] [CrossRef]
  103. Kabekkodu, S.P.; Gladwell, L.R.; Choudhury, M. The mitochondrial link: Phthalate exposure and cardiovascular disease. Biochim. Biophys. Acta Mol. Cell Res. 2024, 1871, 119708. [Google Scholar] [CrossRef]
  104. Chang, W.H.; Herianto, S.; Lee, C.C.; Hung, H.; Chen, H.L. The effects of phthalate ester exposure on human health: A review. Sci. Total Environ. 2021, 786, 147371. [Google Scholar] [CrossRef] [PubMed]
  105. Qian, Y.; Shao, H.; Ying, X.; Huang, W.; Hua, Y. The Endocrine Disruption of Prenatal Phthalate Exposure in Mother and Offspring. Front. Public Health 2020, 8, 366. [Google Scholar] [CrossRef] [PubMed]
  106. Trasande, L.; Sathyanarayana, S.; Spanier, A.J.; Trachtman, H.; Attina, T.M.; Urbina, E.M. Urinary Phthalates Are Associated With Higher Blood Pressure in Childhood. J. Pediatr. 2013, 163, 747–753. [Google Scholar] [CrossRef]
  107. Lee, I.; Park, J.Y.; Kim, S.; An, J.N.; Lee, J.; Park, H.; Jung, S.K.; Kim, S.Y.; Lee, J.P.; Choi, K. Association of Exposure to Phthalates and Environmental Phenolics With Markers of Kidney Function: Korean National Environmental Health Survey (Konehs) 2015–2017. Environ. Int. 2020, 143, 105877. [Google Scholar] [CrossRef] [PubMed]
  108. NTP (National Toxicology Program). Report on Carcinogens, 15th ed.; U.S. Department of Health and Human Services, Public Health Service: Research Triangle Park, NC, USA, 2021. [Google Scholar] [CrossRef]
  109. Xue, J.; Xiao, Q.; Zhang, M.; Li, D.; Wang, X. Toxic Effects and Mechanisms of Polybrominated Diphenyl Ethers. Int. J. Mol. Sci. 2023, 24, 13487. [Google Scholar] [CrossRef]
  110. Stubbings, W.A.; Harrad, S. Extent and mechanisms of brominated flame retardant emissions from waste soft furnishings and fabrics: A critical review. Environ. Int. 2014, 71, 164–175. [Google Scholar] [CrossRef]
  111. Albina, M.L.; Alonso, V.; Linares, V.; Bellés, M.; Sirvent, J.J.; Domingo, J.L.; Sánchez, D.J. Effects of exposure to BDE-99 on oxidative status of liver and kidney in adult rats. Toxicology 2010, 271, 51–56. [Google Scholar] [CrossRef]
  112. Zhao, X.; Chen, T.; Wang, D.; Du, Y.; Wang, Y.; Zhu, W.; Bekir, M.; Yu, D.; Shi, Z. Polybrominated diphenyl ethers and decabromodiphenyl ethane in paired hair/serum and nail/serum from corresponding chemical manufacturing workers and their correlations to thyroid hormones, liver and kidney injury markers. Sci. Total Environ. 2020, 729, 139049. [Google Scholar] [CrossRef]
  113. Milovanovic, V.; Buha, A.; Matovic, V.; Curcic, M.; Vucinic, S.; Nakano, T.; Antonijevic, B. Oxidative stress and renal toxicity after subacute exposure to decabrominated diphenyl ether in Wistar rats. Environ. Sci. Pollut. Res. Int. 2018, 25, 7223–7230. [Google Scholar] [CrossRef]
  114. Coutinho, J.V.; Freitas-Lima, L.C.; Freitas, F.F.; Freitas, F.P.; Podratz, P.L.; Magnago, R.P.; Porto, M.L.; Meyrelles, S.S.; Vasquez, E.C.; Brandão, P.A.; et al. Tributyltin chloride induces renal dysfunction by inflammation and oxidative stress in female rats. Toxicol. Lett. 2016, 260, 52–69. [Google Scholar] [CrossRef]
  115. Mustieles, V.; Balogh, R.K.; Axelstad, M.; Montazeri, P.; Márquez, S.; Vrijheid, M.; Draskau, M.K.; Taxvig, C.; Peinado, F.M.; Berman, T.; et al. Benzophenone-3: Comprehensive review of the toxicological and human evidence with meta-analysis of human biomonitoring studies. Environ. Int. 2023, 173, 107739. [Google Scholar] [CrossRef] [PubMed]
  116. Cunha Carvalho, A.E.; Rodrigues de Oliveira, A.C.; de Sousa Holanda, F.; Libanio Reis Santos, E.; de Oliveira-Lima, J. Benzophenone-3 (oxybenzone) in zebrafish: Histopathological and oxidative stress analysis. Xenobiotica 2025, 55, 71–77. [Google Scholar] [CrossRef] [PubMed]
  117. Elewa, H.S.; Salama, D.A.; Hikal, M.S.; El Hamid, M.F.A.; Eid, M.H.; Khalil, F.M.A.; Albadrani, M.S.; Abdelaal, K.; El-Tokhy, A.I. Protective effects of resveratrol and naringenin against nonylphenol-induced oxidative stress in rats. AMB Express 2025, 15, 7. [Google Scholar] [CrossRef]
  118. Alimi, O.S.; Farner Budarz, J.; Hernandez, L.M.; Tufenkji, N. Microplastics and Nanoplastics in Aquatic Environments: Aggregation, Deposition, and Enhanced Contaminant Transport. Environ. Sci. Technol. 2018, 52, 1704–1724. [Google Scholar] [CrossRef]
  119. Lin, Y.D.; Huang, P.H.; Chen, Y.W.; Hsieh, C.W.; Tain, Y.L.; Lee, B.H.; Hou, C.Y.; Shih, M.K. Sources, Degradation, Ingestion and Effects of Microplastics on Humans: A Review. Toxics 2023, 11, 747. [Google Scholar] [CrossRef]
  120. Lee, Y.; Cho, J.; Sohn, J.; Kim, C. Health Effects of Microplastic Exposures: Current Issues and Perspectives in South Korea. Yonsei. Med. J. 2023, 64, 301–308. [Google Scholar] [CrossRef]
  121. Massardo, S.; Verzola, D.; Alberti, S.; Caboni, C.; Santostefano, M.; Eugenio Verrina, E.; Angeletti, A.; Lugani, F.; Ghiggeri, G.M.; Bruschi, M.; et al. MicroRaman spectroscopy detects the presence of microplastics in human urine and kidney tissue. Environ. Int. 2024, 184, 108444. [Google Scholar] [CrossRef]
  122. Meng, X.; Yin, K.; Zhang, Y.; Wang, D.; Lu, H.; Hou, L.; Zhao, H.; Xing, M. Polystyrene microplastics induced oxidative stress, inflammation and necroptosis via NF-κB and RIP1/RIP3/MLKL pathway in chicken kidney. Toxicology 2022, 478, 153296. [Google Scholar] [CrossRef]
  123. Hong, R.; Shi, Y.; Fan, Z.; Gao, Y.; Chen, H.; Pan, C. Chronic exposure to polystyrene microplastics induces renal fibrosis via ferroptosis. Toxicology 2024, 509, 153996. [Google Scholar] [CrossRef] [PubMed]
  124. Tan, R.Y.; She, Q.Y.; Ma, Y.C.; Liu, M.H.; Li, L.J.; Huang, L.L.; Zhong, Y.W.; Bi, H.X. The threat of microplastics to human kidney health: Mechanisms of nephrotoxicity and future research directions. Environ. Res. 2025, 283, 122124. [Google Scholar] [CrossRef]
  125. Tain, Y.L.; Chang-Chien, G.P.; Lin, S.F.; Hou, C.Y.; Hsu, C.N. Protective Effect of Resveratrol on Kidney Disease and Hypertension Against Microplastics Exposure in Male Juvenile Rats. Antioxidants 2024, 13, 1457. [Google Scholar] [CrossRef]
  126. Järup, L. Hazards of heavy metal contamination. Br. Med. Bull. 2003, 68, 167–182. [Google Scholar] [CrossRef]
  127. Jomova, K.; Alomar, S.Y.; Nepovimova, E.; Kuca, K.; Valko, M. Heavy metals: Toxicity and human health effects. Arch. Toxicol. 2025, 99, 153–209. [Google Scholar] [CrossRef]
  128. Farkhondeh, T.; Naseri, K.; Esform, A.; Aramjoo, H.; Naghizadeh, A. Drinking water heavy metal toxicity and chronic kidney diseases: A systematic review. Rev. Environ. Health 2020, 36, 359–366. [Google Scholar] [CrossRef]
  129. Valko, M.; Morris, H.; Cronin, M.T. Metals, toxicity and oxidative stress. Curr. Med. Chem. 2005, 12, 1161–1208. [Google Scholar] [CrossRef] [PubMed]
  130. Jomova, K.; Jenisova, Z.; Feszterova, M.; Baros, S.; Liska, J.; Hudecova, D.; Rhodes, C.J.; Valko, M. Arsenic: Toxicity, oxidative stress and human disease. J. Appl. Toxicol. 2011, 31, 95–107. [Google Scholar] [CrossRef] [PubMed]
  131. Lopes, A.C.; Peixe, T.S.; Mesas, A.E.; Paoliello, M.M. Lead Exposure and Oxidative Stress: A Systematic Review. Rev. Environ. Contam. Toxicol. 2016, 236, 193–238. [Google Scholar] [PubMed]
  132. Turkington, R.E.; Hukriede, N.A.; Ho, J.; Jayasundara, N.; Sanders, A.P. Metal mechanisms of mitochondrial toxicity: Recent review of arsenic, cadmium, and lead-induced nephrotoxicity. Environ. Sci. Pollut. Res. Int. 2025, 32, 14439–14451. [Google Scholar] [CrossRef]
  133. Lutz, E.; Lind, B.; Herin, P.; Krakau, I.; Bui, T.H.; Vahter, M. Concentrations of Mercury, Cadmium and Lead in Brain and Kidney of Second Trimester Fetuses and Infants. J. Trace Elem. Med. Biol. 1996, 10, 61–67. [Google Scholar] [CrossRef]
  134. Wedeen, R.P.; D’Haese, P.; Van de Vyver, F.L.; Verpooten, G.A.; De Broe, M.E. Lead Nephropathy. Am. J. Kidney Dis. 1986, 8, 380–383. [Google Scholar] [CrossRef]
  135. Yang, H.; Shu, Y. Cadmium Transporters in the Kidney and Cadmium-Induced Nephrotoxicity. Int. J. Mol. Sci. 2015, 16, 1484–1494. [Google Scholar] [CrossRef]
  136. Zalups, R.K. Molecular interactions with mercury in the kidney. Pharmacol. Rev. 2000, 52, 113–143. [Google Scholar] [CrossRef]
  137. Weidemann, D.K.; Weaver, V.M.; Fadrowski, J.J. Toxic Environmental Exposures and Kidney Health in Children. Pediatr. Nephrol. 2016, 31, 2043–2054. [Google Scholar] [CrossRef] [PubMed]
  138. Solhaug, M.J.; Bolger, P.M.; Jose, P.A. The Developing Kidney and Environmental Toxins. Pediatrics 2004, 113, 1084–1091. [Google Scholar] [CrossRef] [PubMed]
  139. Mallah, M.A.; Changxing, L.; Mallah, M.A.; Noreen, S.; Liu, Y.; Saeed, M.; Xi, H.; Ahmed, B.; Feng, F.; Mirjat, A.A.; et al. Polycyclic aromatic hydrocarbon and its effects on human health: An overeview. Chemosphere 2022, 296, 133948. [Google Scholar] [CrossRef]
  140. Barbosa, F., Jr.; Rocha, B.A.; Souza, M.C.O.; Bocato, M.Z.; Azevedo, L.F.; Adeyemi, J.A.; Santana, A.; Campiglia, A.D. Polycyclic aromatic hydrocarbons (PAHs): Updated aspects of their determination, kinetics in the human body, and toxicity. J. Toxicol. Environ. Health B. Crit. Rev. 2023, 26, 28–65. [Google Scholar] [CrossRef] [PubMed]
  141. Kim, K.B.; Lee, B.M. Oxidative stress to DNA, protein, and antioxidant enzymes (superoxide dismutase and catalase) in rats treated with benzo(a)pyrene. Cancer Lett. 1997, 113, 205–212. [Google Scholar] [CrossRef]
  142. Miao, H.; Wu, X.Q.; Wang, Y.N.; Chen, D.Q.; Chen, L.; Vaziri, N.D.; Zhuang, S.; Guo, Y.; Su, W.; Ma, S.X.; et al. 1-Hydroxypyrene mediates renal fibrosis through aryl hydrocarbon receptor signalling pathway. Br. J. Pharmacol. 2022, 179, 103–124. [Google Scholar] [CrossRef]
  143. Patel, A.B.; Shaikh, S.; Jain, K.R.; Desai, C.; Madamwar, D. Polycyclic Aromatic Hydrocarbons: Sources, Toxicity, and Remediation Approaches. Front. Microbiol. 2020, 11, 562813. [Google Scholar] [CrossRef]
  144. Pulkrabova, J.; Stupak, M.; Svarcova, A.; Rossner, P.; Rossnerova, A.; Ambroz, A.; Sram, R.; Hajslova, J. Relationship Between Atmospheric Pollution in the Residential Area and Concentrations of Polycyclic Aromatic Hydrocarbons (Pahs) in Human Breast Milk. Sci. Total Environ. 2016, 562, 640–647. [Google Scholar] [CrossRef]
  145. Drwal, E.; Rak, A.; Gregoraszczuk, E.L. Review: Polycyclic Aromatic Hydrocarbons (Pahs)-Action on Placental Function and Health Risks in Future Life of Newborns. Toxicology 2019, 411, 133–142. [Google Scholar] [CrossRef]
  146. Perera, F.; Rauh, V.; Whyatt, R.; Tang, D.; Tsai, W.; Bernert, J.; Tu, Y.; Andrews, H.; Barr, D.; Camann, D.; et al. A Summary of Recent Findings on Birth Outcomes and Developmental Effects of Prenatal ETS, PAH, and Pesticide Exposures. Neurotoxicology 2005, 26, 573–587. [Google Scholar] [CrossRef]
  147. Farzan, S.F.; Chen, Y.; Trachtman, H.; Trasande, L. Urinary Polycyclic Aromatic Hydrocarbons and Measures of Oxidative Stress, Inflammation and Renal Function in Adolescents: NHANES 2003–2008. Environ. Res. 2016, 144, 149–157. [Google Scholar] [CrossRef] [PubMed]
  148. Jacobs, L.; Buczynska, A.; Walgraeve, C.; Delcloo, A.; Potgieter-Vermaak, S.; Van Grieken, R.; Demeestere, K.; Dewulf, J.; Van Langenhove, H.; De Backer, H.; et al. Acute Changes in Pulse Pressure in Relation to Constituents of Particulate Air Pollution in Elderly Persons. Environ. Res. 2012, 117, 60–67. [Google Scholar] [CrossRef] [PubMed]
  149. Li, J.; Fan, H.; Liu, K.; Li, X.; Fan, D.; Lu, X.; Xia, Y.; Cao, Y.; Xiao, C. Associations of Urinary Polycyclic Aromatic Hydrocarbons With Albuminuria in U.S. Adults, NHANES 2003–2014. Ecotoxicol. Environ. Saf. 2020, 195, 110445. [Google Scholar] [CrossRef]
  150. Sunderland, E.M.; Hu, X.C.; Dassuncao, C.; Tokranov, A.K.; Wagner, C.C.; Allen, J.G. A Review of the Pathways of Human Exposure to Poly- and Perfluoroalkyl Substances (Pfass) and Present Understanding of Health Effects. J. Expo. Sci. Environ. Epidemiol. 2019, 29, 131–147. [Google Scholar] [CrossRef]
  151. Kato, K.; Wong, L.Y.; Jia, L.T.; Kuklenyik, Z.; Calafat, A.M. Trends in Exposure to Polyfluoroalkyl Chemicals in the U.S. Population: 1999–2008. Environ. Sci. Technol. 2011, 45, 8037–8045. [Google Scholar] [CrossRef]
  152. Shankar, A.; Xiao, J.; Ducatman, A. Perfluoroalkyl Chemicals and Chronic Kidney Disease in US Adults. Am. J. Epidemiol. 2011, 174, 893–900. [Google Scholar] [CrossRef]
  153. Watkins, D.J.; Josson, J.; Elston, B.; Bartell, S.M.; Shin, H.-M.; Vieira, V.M.; Savitz, D.A.; Fletcher, T.; Wellenius, G.A. Exposure to Perfluoroalkyl Acids and Markers of Kidney Function Among Children and Adolescents Living Near a Chemical Plant. Environ. Health Perspect. 2013, 121, 625–630. [Google Scholar] [CrossRef] [PubMed]
  154. Stanifer, J.W.; Stapleton, H.M.; Souma, T.; Wittmer, A.; Zhao, X.; Boulware, L.E. Perfluorinated Chemicals as Emerging Environmental Threats to Kidney Health: A Scoping Review. Clin. J. Am. Soc. Nephrol. 2018, 13, 1479–1492. [Google Scholar] [CrossRef]
  155. Solan, M.E.; Koperski, C.P.; Senthilkumar, S.; Lavado, R. Short-chain per- and polyfluoralkyl substances (PFAS) effects on oxidative stress biomarkers in human liver, kidney, muscle, and microglia cell lines. Environ. Res. 2023, 223, 115424. [Google Scholar] [CrossRef]
  156. Afsar, B.; Elsurer Afsar, R.; Kanbay, A.; Covic, A.; Ortiz, A.; Kanbay, M. Air Pollution and Kidney Disease: Review of Current Evidence. Clin. Kidney J. 2019, 12, 19–32. [Google Scholar] [CrossRef] [PubMed]
  157. Bowe, B.; Xie, Y.; Li, T.; Yan, Y.; Xian, H.; Al-Aly, Z. Particulate Matter Air Pollution and the Risk of Incident CKD and Progression to ESRD. J. Am. Soc. Nephrol. 2018, 29, 218–230. [Google Scholar] [CrossRef] [PubMed]
  158. Bowe, B.; Xie, Y.; Li, T.; Yan, Y.; Xian, H.; Al-Aly, Z. Associations of Ambient Coarse Particulate Matter, Nitrogen Dioxide, and Carbon Monoxide With the Risk of Kidney Disease: A Cohort Study. Lancet Planet Health 2017, 1, e267–e276. [Google Scholar] [CrossRef]
  159. Xu, C.; Zhang, Q.; Huang, G.; Huang, J.; Zhang, H. The impact of PM2.5 on kidney. J. Appl. Toxicol. 2023, 43, 107–121. [Google Scholar] [CrossRef]
  160. Mudway, I.S.; Kelly, F.J.; Holgate, S.T. Oxidative stress in air pollution research. Free Radic. Biol. Med. 2020, 151, 2–6. [Google Scholar] [CrossRef]
  161. Feng, Y.; Liu, X.; Zhang, X.; Zhao, X.; Chang, H.; Ouyang, F.; Yu, Z.; Gao, Z.; Zhang, H. Global air pollution exposure and congenital anomalies: An updated systematic review and meta-analysis of epidemiological studies. Int. J. Environ. Health Res. 2024, 34, 2333–2352. [Google Scholar] [CrossRef]
  162. Cho, Y.; Ryu, S.H.; Lee, B.R.; Kim, K.H.; Lee, E.; Choi, J. Effects of artificial light at night on human health: A literature review of observational and experimental studies applied to exposure assessment. Chronobiol. Int. 2015, 32, 1294–1310. [Google Scholar] [CrossRef]
  163. Tsao, J.Y.; Han, J.; Haitz, R.H.; Pattison, P.M. The blue LED Nobel prize: Historical context, current scientific understanding, human benefit. Ann. Phys. 2015, 527, A53–A61. [Google Scholar] [CrossRef]
  164. Ishida, A.; Mutoh, T.; Ueyama, T.; Bando, H.; Masubuchi, S.; Nakahara, D.; Tsujimoto, G.; Okamura, H. Light activates the adrenal gland: Timing of gene expression and glucocorticoid release. Cell Metab. 2005, 2, 297–307. [Google Scholar] [CrossRef] [PubMed]
  165. Gumz, M.L. Molecular origin of the kidney clock. Kidney Int. 2014, 86, 873–874. [Google Scholar] [CrossRef] [PubMed]
  166. Bonny, O.; Vinciguerra, M.; Gumz, M.L.; Mazzoccoli, G. Molecular bases of circadian rhythmicity in renal physiology and pathology. Nephrol. Dial. Transpl. 2013, 28, 2421–2431. [Google Scholar] [CrossRef]
  167. Seron-Ferre, M.; Valenzuela, G.J.; Torres-Farfan, C. Circadian clocks during embryonic and fetal development. Birth Defects Res. C Embryo Today 2007, 81, 204–214. [Google Scholar] [CrossRef]
  168. Hsu, C.N.; Tain, Y.L. Light and Circadian Signaling Pathway in Pregnancy: Programming of Adult Health and Disease. Int. J. Mol. Sci. 2020, 21, 2232. [Google Scholar] [CrossRef]
  169. Simko, F.; Reiter, R.J.; Pechanova, O.; Paulis, L. Experimental models of melatonin-deficient hypertension. Front. Biosci. 2013, 18, 616–625. [Google Scholar] [CrossRef]
  170. Tain, Y.L.; Lin, Y.J.; Chan, J.Y.H.; Lee, C.T.; Hsu, C.N. Maternal melatonin or agomelatine therapy prevents programmed hypertension in male offspring of mother exposed to continuous light. Biol. Reprod. 2017, 97, 636–643. [Google Scholar] [CrossRef]
  171. Cisternas, C.D.; Compagnucci, M.V.; Conti, N.R.; Ponce, R.H.; Vermouth, N.T. Protective effect of maternal prenatal melatonin administration on rat pups born to mothers submitted to constant light during gestation. Braz. J. Med. Biol. Res. 2010, 43, 874–882. [Google Scholar] [CrossRef]
  172. Jobst, M.; Guerreiro, I.; Payros, D.; Oswald, I.P.; Fernandes, A.S.; Warth, B.; Del Favero, G. New approach methodologies for risk assessment of urinary occurring toxicants. Ecotoxicol. Environ. Saf. 2025, 302, 118649. [Google Scholar] [CrossRef] [PubMed]
  173. Khoi, C.S.; Chen, J.H.; Lin, T.Y.; Chiang, C.K.; Hung, K.Y. Ochratoxin A-Induced Nephrotoxicity: Up-to-Date Evidence. Int. J. Mol. Sci. 2021, 22, 11237. [Google Scholar] [CrossRef]
  174. Wang, Y.; Li, Y.; Wu, Y.; Wu, A.; Xiao, B.; Liu, X.; Zhang, Q.; Feng, Y.; Yuan, Z.; Yi, J.; et al. Endoplasmic reticulum stress promotes oxidative stress, inflammation, and apoptosis: A novel mechanism of citrinin-induced renal injury and dysfunction. Ecotoxicol. Environ. Saf. 2024, 284, 116946. [Google Scholar] [CrossRef]
  175. Zhou, Q.; Jiang, L.; Su, T.; Liu, G.; Yang, L. Overview of aristolochic acid nephropathy: An update. Kidney Res. Clin. Pract. 2023, 42, 579–590. [Google Scholar] [CrossRef]
  176. Chen, L.; Cheng, S.; Ying, J.; Zhang, Q.; Wang, C.; Wu, H.; Wang, Y.; Zhang, H.; Wang, J.; Ye, J.; et al. Aristolochic acid I promotes renal tubulointerstitial fibrosis by up-regulating expression of indoleamine 2,3-dioxygenase-1 (IDO1). Toxicol. Lett. 2024, 402, 44–55. [Google Scholar] [CrossRef] [PubMed]
  177. Alvito, P.; Pereira-da-Silva, L. Mycotoxin Exposure during the First 1000 Days of Life and Its Impact on Children’s Health: A Clinical Overview. Toxins 2022, 14, 189. [Google Scholar] [CrossRef] [PubMed]
  178. Ma, Y.; Hong, Y.; Gao, R.; Zhang, Y.; Geng, Y.; Yin, X.; Chen, X.; Li, F.; Mu, X.; He, J. Maternal exposure to Aristolochic Acid I affects meiotic I progression by impairing DNA damage repair in fetal oocytes. Ecotoxicol. Environ. Saf. 2025, 295, 118137. [Google Scholar] [CrossRef]
  179. Qiqi, L.; Junlin, H.; Xuemei, C.; Yi, H.; Fangfang, L.; Yanqing, G.; Yan, Z.; Lamptey, J.; Zhuxiu, C.; Fangfei, L.; et al. Fetal exposure of Aristolochic Acid I undermines ovarian reserve by disturbing primordial folliculogenesis. Ecotoxicol. Environ. Saf. 2022, 236, 113480. [Google Scholar] [CrossRef] [PubMed]
  180. Debelle, F.D.; Vanherweghem, J.L.; Nortier, J.L. Aristolochic acid nephropathy: A worldwide problem. Kidney Int. 2008, 74, 158–169. [Google Scholar] [CrossRef]
  181. Jelaković, B.; Dika, Ž.; Arlt, V.M.; Stiborova, M.; Pavlović, N.M.; Nikolić, J.; Colet, J.M.; Vanherweghem, J.L.; Nortier, J.L. Balkan Endemic Nephropathy and the Causative Role of Aristolochic Acid. Semin. Nephrol. 2019, 39, 284–296. [Google Scholar] [CrossRef]
  182. Jelaković, B.; Karanović, S.; Vuković-Lela, I.; Miller, F.; Edwards, K.L.; Nikolić, J.; Tomić, K.; Slade, N.; Brdar, B.; Turesky, R.J.; et al. Aristolactam-DNA adducts are a biomarker of environmental exposure to aristolochic acid. Kidney Int. 2012, 81, 559–567. [Google Scholar] [CrossRef]
  183. Hoang, M.L.; Chen, C.H.; Sidorenko, V.S.; He, J.; Dickman, K.G.; Yun, B.H.; Moriya, M.; Niknafs, N.; Douville, C.; Karchin, R.; et al. Mutational signature of aristolochic acid exposure as revealed by whole-exome sequencing. Sci. Transl. Med. 2013, 5, 197ra102. [Google Scholar] [CrossRef]
  184. Rajput, P.; Kumar, P.; Priya, A.K.; Kumari, S.; Shiade, S.R.G.; Rajput, V.D.; Fathi, A.; Pradhan, A.; Sarfraz, R.; Sushkova, S.; et al. Nanomaterials and biochar mediated remediation of emerging contaminants. Sci. Total Environ. 2024, 916, 170064. [Google Scholar] [CrossRef]
  185. Albrahim, T. Silver nanoparticles-induced nephrotoxicity in rats: The protective role of red beetroot (Beta vulgaris) juice. Environ. Sci. Pollut. Res. Int. 2020, 27, 38871–38880. [Google Scholar] [CrossRef]
  186. Hsu, C.N.; Chan, J.Y.H.; Yu, H.R.; Lee, W.C.; Wu, K.L.H.; Chang-Chien, G.P.; Lin, S.; Hou, C.Y.; Tain, Y.L. Targeting on Gut Microbiota-Derived Metabolite Trimethylamine to Protect Adult Male Rat Offspring Against Hypertension Programmed by Combined Maternal High-Fructose Intake and Dioxin Exposure. Int. J. Mol. Sci. 2020, 21, 5488. [Google Scholar] [CrossRef]
  187. Hsu, C.N.; Hou, C.Y.; Lee, C.T.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Maternal 3,3-Dimethyl-1-Butanol Therapy Protects Adult Male Rat Offspring against Hypertension Programmed by Perinatal TCDD Exposure. Nutrients 2021, 13, 3041. [Google Scholar] [CrossRef] [PubMed]
  188. Hsu, C.N.; Lin, Y.J.; Lu, P.C.; Tain, Y.L. Maternal Resveratrol Therapy Protects Male Rat Offspring Against Programmed Hypertension Induced by TCDD and Dexamethasone Exposures: Is it Relevant to Aryl Hydrocarbon Receptor? Int. J. Mol. Sci. 2018, 19, 2459. [Google Scholar] [CrossRef] [PubMed]
  189. Aragon, A.C.; Kopf, P.G.; Campen, M.J.; Huwe, J.K.; Walker, M.K. In Utero and Lactational 2,3,7,8-Tetrachlorodibenzo-P-Dioxin Exposure: Effects on Fetal and Adult Cardiac Gene Expression and Adult Cardiac and Renal Morphology. Toxicol. Sci. 2008, 101, 321–330. [Google Scholar] [CrossRef]
  190. Nuñez, P.; Fernandez, T.; García-Arévalo, M.; Alonso-Magdalena, P.; Nadal, A.; Perillan, C. Effects of Bisphenol a Treatment During Pregnancy on Kidney Development in Mice: A Stereological and Histopathological Study. J. Dev. Orig. Health Dis. 2018, 9, 208–214. [Google Scholar] [CrossRef]
  191. Hsu, C.N.; Lin, Y.J.; Tain, Y.L. Maternal Exposure to Bisphenol a Combined with High-Fat Diet-Induced Programmed Hypertension in Adult Male Rat Offspring: Effects of Resveratrol. Int. J. Mol. Sci. 2019, 20, 4382. [Google Scholar] [CrossRef] [PubMed]
  192. Wei, Z.; Song, L.; Wei, J.; Chen, T.; Chen, J.; Lin, Y.; Xia, W.; Xu, B.; Li, X.; Chen, X.; et al. Maternal Exposure to Di-(2-Ethylhexyl) Phthalate Alters Kidney Development Through the Renin-Angiotensin System in Offspring. Toxicol. Lett. 2012, 212, 212–221. [Google Scholar] [CrossRef]
  193. Tain, Y.L.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Hsu, C.N. Resveratrol Butyrate Ester Supplementation Blunts the Development of Offspring Hypertension in a Maternal Di-2-ethylhexyl Phthalate Exposure Rat Model. Nutrients 2023, 15, 697. [Google Scholar] [CrossRef]
  194. Ye, Q.; Zhao, S.; Zhang, Y.; Su, Y.M.; Chen, M.; Zhao, J.; Jia, G.Z.; Han, B.M.; Jiang, J.T. Activation of the Rhoa/ROCK Pathway Contributes to Renal Fibrosis in Offspring Rats Induced by Maternal Exposure to Di-N-Butyl Phthalate. Toxicology 2020, 443, 152573. [Google Scholar] [CrossRef]
  195. Chen, X.; Wang, L.; Liu, K.; Wang, Q.; Li, R.; Niu, L.; Wu, H. Maternal exposure to polystyrene nanoplastics induces sex-specific kidney injury in offspring. Ecotoxicol. Environ. Saf. 2025, 293, 118006. [Google Scholar] [CrossRef]
  196. Saillenfait, A.M.; Payan, J.P.; Brondeau, M.T.; Zissu, D.; de Ceaurriz, J. Changes in Urinary Proximal Tubule Parameters in Neonatal Rats Exposed to Cadmium Chloride During Pregnancy. J. Appl. Toxicol. 1991, 11, 23–27. [Google Scholar] [CrossRef]
  197. Jacquillet, G.; Barbier, O.; Rubera, I.; Tauc, M.; Borderie, A.; Namorado, M.C.; Martin, D.; Sierra, G.; Reyes, J.L.; Poujeol, P.; et al. Cadmium Causes Delayed Effects on Renal Function in the Offspring of Cadmium-Contaminated Pregnant Female Rats. Am. J. Physiol. Renal Physiol. 2007, 293, F1450–F1560. [Google Scholar] [CrossRef] [PubMed]
  198. Dangudubiyyam, S.V.; Mishra, J.S.; Zhao, H.; Kumar, S. Perfluorooctane sulfonic acid (PFOS) exposure during pregnancy increases blood pressure and impairs vascular relaxation mechanisms in the adult offspring. Reprod. Toxicol. 2020, 98, 165–173. [Google Scholar] [CrossRef]
  199. Jules, G.E.; Pratap, S.; Ramesh, A.; Hood, D.B. In Utero Exposure to Benzo (a) Pyrene Predisposes Offspring to Cardiovascular Dysfunction in Later-Life. Toxicology 2012, 295, 56–67. [Google Scholar] [CrossRef]
  200. Pan, K.; Jiang, S.; Du, X.; Zeng, X.; Zhang, J.; Song, L.; Lei, L.; Zhou, J.; Kan, H.; Sun, Q. Parental PM2.5 Exposure Changes Th17/Treg Cells in Offspring, Is Associated with the Elevation of Blood Pressure. Environ. Toxicol. 2021, 36, 1152–1161. [Google Scholar] [CrossRef] [PubMed]
  201. Ye, Z.; Lu, X.; Deng, Y.; Wang, X.; Zheng, S.; Ren, H.; Zhang, M.; Chen, T.; Jose, P.A.; Yang, J.; et al. In Utero Exposure to Fine Particulate Matter Causes Hypertension Due to Impaired Renal Dopamine D1 Receptor in Offspring. Cell Physiol. Biochem. 2018, 46, 148–159. [Google Scholar] [CrossRef]
  202. Sengupta, P. The Laboratory Rat: Relating Its Age with Human’s. Int. J. Prev. Med. 2013, 4, 624–630. [Google Scholar] [PubMed]
  203. Patel, S.; Rauf, A.; Khan, H.; Abu-Izneid, T. Renin-Angiotensin-aldosterone (RAAS): The ubiquitous system for homeostasis and pathologies. Biomed. Pharmacother. 2017, 94, 317–325. [Google Scholar] [CrossRef]
  204. Cravedi, P.; Ruggenenti, P.; Remuzzi, G. Which antihypertensive drugs are the most nephroprotective and why? Expert Opin. Pharmacother. 2010, 11, 2651–2663. [Google Scholar] [CrossRef]
  205. Gubler, M.C.; Antignac, C. Renin-Angiotensin system in kidney development: Renal tubular dysgenesis. Kidney Int. 2010, 77, 400–406. [Google Scholar] [CrossRef] [PubMed]
  206. Yosypiv, I.V. Renin-Angiotensin system in ureteric bud branching morphogenesis: Insights into the mechanisms. Pediatr. Nephrol. 2011, 26, 1499–1512. [Google Scholar] [CrossRef] [PubMed]
  207. Alexander, B.T.; South, A.M.; August, P.; Bertagnolli, M.; Ferranti, E.P.; Grobe, J.L.; Jones, E.J.; Loria, A.S.; Safdar, B.; SequeiraLopez, M.L.S.; et al. Appraising the Preclinical Evidence of the Role of the Renin-Angiotensin-Aldosterone System in Antenatal Programming of Maternal and Offspring Cardiovascular Health Across the Life Course: Moving the Field Forward: A Scientific Statement From the American Heart Association. Hypertension 2023, 80, e75–e89. [Google Scholar]
  208. Hsu, C.N.; Tain, Y.L. Targeting the Renin-Angiotensin-Aldosterone System to Prevent Hypertension and Kidney Disease of Developmental Origins. Int. J. Mol. Sci. 2021, 22, 2298. [Google Scholar] [CrossRef]
  209. Luyckx, V.A.; Brenner, B.M. Clinical consequences of developmental programming of low nephron number. Anat. Rec. (Hoboken) 2020, 303, 2613–2631. [Google Scholar] [CrossRef]
  210. Bertram, J.F.; Douglas-Denton, R.N.; Diouf, B.; Hughson, M.D.; Hoy, W.E. Human nephron number: Implications for health and disease. Pediatr. Nephrol. 2011, 26, 1529–1533. [Google Scholar] [CrossRef]
  211. Rosenblum, S.; Pal, A.; Reidy, K. Renal development in the fetus and premature infant. Semin. Fetal Neonatal Med. 2017, 22, 58–66. [Google Scholar] [CrossRef]
  212. Hsu, C.N.; Tain, Y.L. The First Thousand Days: Kidney Health and Beyond. Healthcare 2021, 9, 1332. [Google Scholar] [CrossRef]
  213. Gao, X.; Ni, W.; Zhu, S.; Wu, Y.; Cui, Y.; Ma, J.; Liu, Y.; Qiao, J.; Ye, Y.; Yang, P.; et al. Per- and Polyfluoroalkyl Substances Exposure During Pregnancy and Adverse Pregnancy and Birth Outcomes: A Systematic Review and Meta-Analysis. Environ. Res. 2021, 201, 111632. [Google Scholar] [CrossRef] [PubMed]
  214. Choi, H.; Wang, L.; Lin, X.; Spengler, J.D.; Perera, F.P. Fetal Window of Vulnerability to Airborne Polycyclic Aromatic Hydrocarbons on Proportional Intrauterine Growth Restriction. PLoS ONE 2012, 7, e35464. [Google Scholar] [CrossRef]
  215. Kumar, S.N.; Saxena, P.; Patel, R.; Sharma, A.; Pradhan, D.; Singh, H.; Deval, R.; Bhardwaj, S.K.; Borgohain, D.; Akhtar, N.; et al. Predicting Risk of Low Birth Weight Offspring From Maternal Features and Blood Polycyclic Aromatic Hydrocarbon Concentration. Reprod. Toxicol. 2020, 94, 92–100. [Google Scholar] [CrossRef] [PubMed]
  216. Uwak, I.; Olson, N.; Fuentes, A.; Moriarty, M.; Pulczinski, J.; Lam, J.; Xu, X.; Taylor, B.D.; Taiwo, S.; Koehler, K.; et al. Application of the navigation guide systematic review methodology to evaluate prenatal exposure to particulate matter air pollution and infant birth weight. Environ. Int. 2021, 148, 106378. [Google Scholar] [CrossRef]
  217. Belmaker, I.; Anca, E.D.; Rubin, L.P.; Magen-Molho, H.; Miodovnik, A.; van der Hal, N. Adverse health effects of exposure to plastic, microplastics and their additives: Environmental, legal and policy implications for Israel. Isr. J. Health Policy Res. 2024, 13, 44. [Google Scholar] [CrossRef] [PubMed]
  218. Nenov, V.D.; Taal, M.W.; Sakharova, O.V.; Brenner, B.M. Multi-hit nature of chronic renal disease. Curr. Opin. Nephrol. Hypertens. 2000, 9, 85–97. [Google Scholar] [CrossRef]
  219. Vogel, C.F.A.; Van Winkle, L.S.; Esser, C.; Haarmann-Stemmann, T. The aryl hydrocarbon receptor as a target of environmental stressors—Implications for pollution mediated stress and inflammatory responses. Redox Biol. 2020, 34, 101530. [Google Scholar] [CrossRef]
  220. Kou, Z.; Dai, W. Aryl hydrocarbon receptor: Its roles in physiology. Biochem. Pharmacol. 2021, 185, 114428. [Google Scholar] [CrossRef]
  221. Tain, Y.L.; Hsu, C.N. The Impact of the Aryl Hydrocarbon Receptor on Antenatal Chemical Exposure-Induced Cardiovascular-Kidney-Metabolic Programming. Int. J. Mol. Sci. 2024, 25, 4599. [Google Scholar] [CrossRef]
  222. Lin, L.; Dai, Y.; Xia, Y. An overview of aryl hydrocarbon receptor ligands in the Last two decades (2002–2022): A medicinal chemistry perspective. Eur. J. Med. Chem. 2022, 244, 114845. [Google Scholar] [CrossRef]
  223. Feng, S.; Duan, E.; Shi, X.; Zhang, H.; Li, H.; Zhao, Y.; Chao, L.; Zhong, X.; Zhang, W.; Li, R.; et al. Hydrogen ameliorates lung injury in a rat model of subacute exposure to concentrated ambient PM2.5 via Aryl hydrocarbon receptor. Int. Immunopharmacol. 2019, 77, 105939. [Google Scholar] [CrossRef] [PubMed]
  224. Manikkam, M.; Tracey, R.; Guerrero-Bosagna, C.; Skinner, M.K. Dioxin (TCDD) induces epigenetic transgenerational inheritance of adult onset disease and sperm epimutations. PLoS ONE 2012, 7, e46249. [Google Scholar] [CrossRef] [PubMed]
  225. Guo, A.H.; Kumar, S.; Lombard, D.B. Epigenetic mechanisms of cadmium-induced nephrotoxicity. Curr. Opin. Toxicol. 2022, 32, 100372. [Google Scholar] [CrossRef] [PubMed]
  226. Gong, Z.G.; Zhao, Y.; Wang, Z.Y.; Fan, R.F.; Liu, Z.P.; Wang, L. Epigenetic regulator BRD4 is involved in cadmium-induced acute kidney injury via contributing to lysosomal dysfunction, autophagy blockade and oxidative stress. J. Hazard. Mater. 2022, 423, 127110. [Google Scholar] [CrossRef]
  227. Hirao-Suzuki, M.; Takeda, S.; Sakai, G.; Waalkes, M.P.; Sugihara, N.; Takiguchi, M. Cadmium-stimulated invasion of rat liver cells during malignant transformation: Evidence of the involvement of oxidative stress/TET1-sensitive machinery. Toxicology 2021, 447, 152631. [Google Scholar] [CrossRef]
  228. Fernandes, G.F.S.; Silva, G.D.B.; Pavan, A.R.; Chiba, D.E.; Chin, C.M.; Dos Santos, J.L. Epigenetic Regulatory Mechanisms Induced by Resveratrol. Nutrients 2017, 9, 1201. [Google Scholar] [CrossRef]
  229. Rosenfeld, C.S. Gut Dysbiosis in Animals Due to Environmental Chemical Exposures. Front. Cell Infect. Microbiol. 2017, 7, 396. [Google Scholar] [CrossRef]
  230. Belkin, S. Microbial Whole-Cell Sensing Systems of Environmental Pollutants. Curr. Opin. Microbiol. 2003, 6, 206–212. [Google Scholar] [CrossRef] [PubMed]
  231. Chappell, G.; Pogribny, I.P.; Guyton, K.Z.; Rusyn, I. Epigenetic Alterations Induced by Genotoxic Occupational and Environmental Human Chemical Carcinogens: A Systematic Literature Review. Mutat. Res. Rev. Mutat. Res. 2016, 768, 27–45. [Google Scholar] [CrossRef] [PubMed]
  232. Colombijn, J.M.; Hooft, L.; Jun, M.; Webster, A.C.; Bots, M.L.; Verhaar, M.C.; Vernooij, R.W. Antioxidants for adults with chronic kidney disease. Cochrane Database Syst. Rev. 2023, 11, CD008176. [Google Scholar] [CrossRef]
  233. Poljšak, B.; Fink, R. The protective role of antioxidants in the defence against ROS/RNS-mediated environmental pollution. Oxid. Med. Cell Longev. 2014, 2014, 671539. [Google Scholar] [CrossRef]
  234. Neha, K.; Haider, M.R.; Pathak, A.; Yar, M.S. Medicinal prospects of antioxidants: A review. Eur. J. Med. Chem. 2019, 178, 687–704. [Google Scholar] [CrossRef]
  235. Wojcik, M.; Burzynska-Pedziwiatr, I.; Wozniak, L.A. A review of natural and synthetic antioxidants important for health and longevity. Curr. Med. Chem. 2010, 17, 3262–3288. [Google Scholar] [CrossRef]
  236. Rapa, S.F.; Di Iorio, B.R.; Campiglia, P.; Heidland, A.; Marzocco, S. Inflammation and Oxidative Stress in Chronic Kidney Disease-Potential Therapeutic Role of Minerals, Vitamins and Plant-Derived Metabolites. Int. J. Mol. Sci. 2019, 21, 263. [Google Scholar] [CrossRef]
  237. Said, H.M.; Nexo, E. Gastrointestinal Handling of Water-Soluble Vitamins. Compr. Physiol. 2018, 8, 1291–1311. [Google Scholar] [CrossRef]
  238. Niki, E. Role of vitamin E as a lipid-soluble peroxyl radical scavenger: In vitro and in vivo evidence. Free Radic. Biol. Med. 2014, 66, 3–12. [Google Scholar] [CrossRef]
  239. Wang, J.; Yin, N.; Deng, Y.; Wei, Y.; Huang, Y.; Pu, X.; Li, L.; Zheng, Y.; Guo, J.; Yu, J.; et al. Ascorbic Acid Protects against Hypertension through Downregulation of ACE1 Gene Expression Mediated by Histone Deacetylation in Prenatal Inflammation-Induced Offspring. Sci. Rep. 2016, 6, 39469. [Google Scholar] [CrossRef]
  240. Farias, J.S.; Santos, K.M.; Lima, N.K.S.; Cabral, E.V.; Aires, R.S.; Veras, A.C.; Paixão, A.D.; Vieira, L.D. Maternal endotoxemia induces renal collagen deposition in adult offspring: Role of NADPH oxidase/TGF-β1/MMP-2 signaling pathway. Arch. Biochem. Biophys. 2020, 684, 108306. [Google Scholar] [CrossRef] [PubMed]
  241. Franco Mdo, C.; Ponzio, B.F.; Gomes, G.N.; Gil, F.Z.; Tostes, R.; Carvalho, M.H.; Fortes, Z.B. Micronutrient prenatal supplementation prevents the development of hypertension and vascular endothelial damage induced by intrauterine malnutrition. Life Sci. 2009, 85, 327–333. [Google Scholar] [CrossRef] [PubMed]
  242. Miller, E.R., 3rd; Pastor-Barriuso, R.; Dalal, D.; Riemersma, R.A.; Appel, L.J.; Guallar, E. Meta-analysis: High-dosage vitamin E supplementation may increase all-cause mortality. Ann. Intern. Med. 2005, 142, 37–46. [Google Scholar] [CrossRef] [PubMed]
  243. Bjelakovic, G.; Nikolova, D.; Gluud, L.L.; Simonetti, R.G.; Gluud, C. Antioxidant supplements for prevention of mortality in healthy participants and patients with various diseases. Cochrane Database Syst. Rev. 2012, 3, CD007176. [Google Scholar] [CrossRef]
  244. Abadie, R.B.; Staples, A.A.; Lauck, L.V.; Dautel, A.D.; Spillers, N.J.; Klapper, R.J.; Hirsch, J.D.; Varrassi, G.; Ahmadzadeh, S.; Shekoohi, S.; et al. Vitamin A-Mediated Birth Defects: A Narrative Review. Cureus 2023, 15, e50513. [Google Scholar] [CrossRef]
  245. Schwalfenberg, G.; Rodushkin, I.; Genuis, S.J. Heavy metal contamination of prenatal vitamins. Toxicol. Rep. 2018, 5, 390–395. [Google Scholar] [CrossRef]
  246. Ali, S.S.; Ahsan, H.; Zia, M.K.; Siddiqui, T.; Khan, F.H. Understanding oxidants and antioxidants: Classical team with new players. J. Food Biochem. 2020, 44, e13145. [Google Scholar] [CrossRef] [PubMed]
  247. Li, X.; Zheng, S.; Wu, G. Amino Acid Metabolism in the Kidneys: Nutritional and Physiological Significance. Adv. Exp. Med. Biol. 2020, 1265, 71–95. [Google Scholar]
  248. Davani-Davari, D.; Karimzadeh, I.; Sagheb, M.M.; Khalili, H. The Renal Safety of L-Carnitine, L-Arginine, and Glutamine in Athletes and Bodybuilders. J. Ren. Nutr. 2019, 29, 221–234. [Google Scholar] [CrossRef] [PubMed]
  249. Correia, M.J.; Pimpão, A.B.; Fernandes, D.G.F.; Morello, J.; Sequeira, C.O.; Calado, J.; Antunes, A.M.M.; Almeida, M.S.; Branco, P.; Monteiro, E.C.; et al. Cysteine as a Multifaceted Player in Kidney, the Cysteine-Related Thiolome and Its Implications for Precision Medicine. Molecules 2022, 27, 1416. [Google Scholar] [CrossRef]
  250. Baliou, S.; Adamaki, M.; Ioannou, P.; Pappa, A.; Panayiotidis, M.I.; Spandidos, D.A.; Christodoulou, I.; Kyriakopoulos, A.M.; Zoumpourlis, V. Protective role of taurine against oxidative stress (Review). Mol. Med. Rep. 2021, 24, 605. [Google Scholar] [CrossRef]
  251. Hsu, C.N.; Tain, Y.L. Regulation of nitric oxide production in the developmental programming of hypertension and kidney disease. Int. J. Mol. Sci. 2019, 20, 681. [Google Scholar] [CrossRef]
  252. Colovic, M.B.; Vasic, V.M.; Djuric, D.M.; Krstic, D.Z. Sulphur-containing Amino Acids: Protective Role Against Free Radicals and Heavy Metals. Curr. Med. Chem. 2018, 25, 324–335. [Google Scholar] [CrossRef] [PubMed]
  253. Hsu, C.N.; Tain, Y.L. Developmental Programming and Reprogramming of Hypertension and Kidney Disease: Impact of Tryptophan Metabolism. Int. J. Mol. Sci. 2020, 21, 8705. [Google Scholar] [CrossRef]
  254. Hsu, C.N.; Tain, Y.L. Impact of Arginine Nutrition and Metabolism during Pregnancy on Offspring Outcomes. Nutrients 2019, 11, 1452. [Google Scholar] [CrossRef]
  255. Endou, H. Recent advances in molecular mechanisms of nephrotoxicity. Toxicol. Lett. 1998, 102–103, 29–33. [Google Scholar] [CrossRef]
  256. Wagner, C.A. Beyond SGLT2: Proximal tubule transporters as potential drug targets for chronic kidney disease. Nephrol. Dial. Transplant. 2025, 40, i18–i28. [Google Scholar] [CrossRef] [PubMed]
  257. Hsu, C.N.; Yang, H.W.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Maternal Adenine-Induced Chronic Kidney Disease Programs Hypertension in Adult Male Rat Offspring: Implications of Nitric Oxide and Gut Microbiome Derived Metabolites. Int. J. Mol. Sci. 2020, 21, 7237. [Google Scholar] [CrossRef] [PubMed]
  258. Hsu, C.N.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Dietary Supplementation with Cysteine during Pregnancy Rescues Maternal Chronic Kidney Disease-Induced Hypertension in Male Rat Offspring: The Impact of Hydrogen Sulfide and Microbiota-Derived Tryptophan Metabolites. Antioxidants 2022, 11, 483. [Google Scholar] [CrossRef]
  259. Tain, Y.L.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Hsu, C.N. Protective Role of Taurine on Rat Offspring Hypertension in the Setting of Maternal Chronic Kidney Disease. Antioxidants 2023, 12, 2059. [Google Scholar] [CrossRef]
  260. Durazzo, A.; Lucarini, M.; Souto, E.B.; Cicala, C.; Caiazzo, E.; Izzo, A.A.; Novellino, E.; Santini, A. Polyphenols: A concise over view on the chemistry, occurrence, and human health. Phytother. Res. 2019, 33, 2221–2243. [Google Scholar] [CrossRef]
  261. Bao, H.; Peng, A. The Green Tea Polyphenol (—)-epigallocatechin-3-gallate and its beneficial roles in chronic kidney disease. J. Transl. Int. Med. 2016, 4, 99–103. [Google Scholar] [CrossRef]
  262. Guerreiro, Í.; Ferreira-Pêgo, C.; Carregosa, D.; Santos, C.N.; Menezes, R.; Fernandes, A.S.; Costa, J.G. Polyphenols and Their Metabolites in Renal Diseases: An Overview. Foods 2022, 11, 1060. [Google Scholar] [CrossRef] [PubMed]
  263. Chen, L.; Mo, H.; Zhao, L.; Gao, W.; Wang, S.; Cromie, M.M.; Lu, C.; Wang, J.S.; Shen, C.L. Therapeutic properties of green tea against environmental insults. J. Nutr. Biochem. 2017, 40, 1–13. [Google Scholar] [CrossRef]
  264. Clementino, M.; Shi, X.; Zhang, Z. Prevention of Polyphenols Against Carcinogenesis Induced by Environmental Carcinogens. J. Environ. Pathol. Toxicol. Oncol. 2017, 36, 87–98. [Google Scholar] [CrossRef]
  265. Lagoa, R.; Marques-da-Silva, D.; Diniz, M.; Daglia, M.; Bishayee, A. Molecular mechanisms linking environmental toxicants to cancer development: Significance for protective interventions with polyphenols. Semin. Cancer Biol. 2022, 80, 118–144. [Google Scholar] [CrossRef] [PubMed]
  266. Wu, Z.; Zhao, J.; Xu, H.; Lyv, Y.; Feng, X.; Fang, Y.; Xu, Y. Maternal quercetin administration during gestation and lactation decrease endoplasmic reticulum stress and related inflammation in the adult offspring of obese female rats. Eur. J. Nutr. 2014, 53, 1669–1683. [Google Scholar] [CrossRef]
  267. Lamothe, J.; Khurana, S.; Tharmalingam, S.; Williamson, C.; Byrne, C.J.; Lees, S.J.; Khaper, N.; Kumar, A.; Tai, T.C. Oxidative Stress Mediates the Fetal Programming of Hypertension by Glucocorticoids. Antioxidants 2021, 10, 531. [Google Scholar] [CrossRef]
  268. Singh, A.P.; Singh, R.; Verma, S.S.; Rai, V.; Kaschula, C.H.; Maiti, P.; Gupta, S.C. Health benefits of resveratrol: Evidence from 926 clinical studies. Med. Res. Rev. 2019, 39, 1851–1891. [Google Scholar] [CrossRef]
  269. Truong, V.L.; Jun, M.; Jeong, W.S. Role of resveratrol in regulation of cellular defense systems against oxidative stress. Biofactors 2018, 44, 36–49. [Google Scholar] [CrossRef]
  270. Casper, R.F.; Quesne, M.; Rogers, I.M.; Shirota, T.; Jolivet, A.; Milgrom, E.; Savouret, J.F. Resveratrol has antagonist activity on the aryl hydrocarbon receptor: Implications for prevention of dioxin toxicity. Mol. Pharmacol. 1999, 56, 784–790. [Google Scholar] [CrossRef] [PubMed]
  271. Tan, Y.Q.; Wang, Y.N.; Feng, H.Y.; Guo, Z.Y.; Li, X.; Nie, X.L.; Zhao, Y.Y. Host/microbiota interactions-derived tryptophan metabolites modulate oxidative stress and inflammation via aryl hydrocarbon receptor signaling. Free Radic. Biol. Med. 2022, 184, 30–41. [Google Scholar] [CrossRef] [PubMed]
  272. Hsu, C.N.; Hou, C.Y.; Tain, Y.L. Preventive Aspects of Early Resveratrol Supplementation in Cardiovascular and Kidney Disease of Developmental Origins. Int. J. Mol. Sci. 2021, 22, 4210. [Google Scholar] [CrossRef]
  273. Tamura, H.; Nakamura, Y.; Terron, M.P.; Flores, L.J.; Manchester, L.C.; Tan, D.X.; Sugino, N.; Reiter, R.J. Melatonin and pregnancy in the human. Reprod. Toxicol. 2008, 25, 291–303. [Google Scholar] [CrossRef]
  274. Reiter, R.J.; Tan, D.X.; Terron, M.P.; Flores, L.J.; Czarnocki, Z. Melatonin and its metabolites: New findings regarding their production and their radical scavenging actions. Acta Biochim. Pol. 2007, 54, 1–9. [Google Scholar] [CrossRef]
  275. Tain, Y.L.; Huang, L.T.; Hsu, C.N. Developmental Programming of Adult Disease: Reprogramming by Melatonin? Int. J. Mol. Sci. 2017, 18, 426. [Google Scholar] [CrossRef]
  276. Raza, Z.; Naureen, Z. Melatonin ameliorates the drug induced nephrotoxicity: Molecular insights. Nefrologia 2020, 40, 12–25. [Google Scholar] [CrossRef]
  277. Aversa, S.; Pellegrino, S.; Barberi, I.; Reiter, R.J.; Gitto, E. Potential utility of melatonin as an antioxidant during pregnancy and in the perinatal period. J. Matern. Fetal Neonatal Med. 2012, 25, 207–221. [Google Scholar] [CrossRef] [PubMed]
  278. Marseglia, L.; D’Angelo, G.; Manti, S.; Reiter, R.J.; Gitto, E. Potential utility of melatonin in preeclampsia, intrauterine fetal growth retardation, and perinatal asphyxia. Reprod. Sci. 2016, 23, 970–977. [Google Scholar] [CrossRef]
  279. James, A.M.; Smith, R.A.; Murphy, M.P. Antioxidant and prooxidant properties of mitochondrial Coenzyme Q. Arch. Biochem. Biophys. 2004, 423, 47–56. [Google Scholar] [CrossRef]
  280. Hsu, C.N.; Lin, Y.J.; Yu, H.R.; Lin, I.C.; Sheen, J.M.; Huang, L.T.; Tain, Y.L. Protection of Male Rat Offspring against Hypertension Programmed by Prenatal Dexamethasone Administration and Postnatal High-Fat Diet with the Nrf2 Activator Dimethyl Fumarate during Pregnancy. Int. J. Mol. Sci. 2019, 20, 3957. [Google Scholar] [CrossRef] [PubMed]
  281. Rosa, A.C.; Corsi, D.; Cavi, N.; Bruni, N.; Dosio, F. Superoxide Dismutase Administration: A Review of Proposed Human Uses. Molecules 2021, 26, 1844. [Google Scholar] [CrossRef] [PubMed]
  282. Salamon, S.; Kramar, B.; Marolt, T.P.; Poljšak, B.; Milisav, I. Medical and Dietary Uses of N-Acetylcysteine. Antioxidants 2019, 8, 111. [Google Scholar] [CrossRef]
  283. Gunjyal, N.; Rani, S.; Asgari Lajayer, B.; Senapathi, V.; Astatkie, T. A review of the effects of environmental hazards on humans, their remediation for sustainable development, and risk assessment. Environ. Monit. Assess. 2023, 195, 795. [Google Scholar] [CrossRef]
  284. Falck, A.J.; Mooney, S.; Kapoor, S.S.; White, K.M.; Bearer, C.; El Metwally, D. Developmental Exposure to Environmental Toxicants. Pediatr. Clin. 2015, 62, 1173–1197. [Google Scholar] [CrossRef]
  285. Conde-Agudelo, A.; Romero, R.; Kusanovic, J.P.; Hassan, S.S. Supplementation with vitamins C and E during pregnancy for the prevention of preeclampsia and other adverse maternal and perinatal outcomes: A systematic review and metaanalysis. Am. J. Obstet. Gynecol. 2011, 204, 503.e1–503.e12. [Google Scholar] [CrossRef]
  286. Sen, S.; Cherkerzian, S.; Herlihy, M.; Hacker, M.R.; McElrath, T.F.; Cantonwine, D.E.; Fichorova, R.; Oken, E.; Meydani, S.N. Supplementation with antioxidant micronutrients in pregnant women with obesity: A randomized controlled trial. Int. J. Obes. 2024, 48, 796–807. [Google Scholar] [CrossRef]
  287. Yuksel, S.; Yigit, A.A.; Cinar, M.; Atmaca, N.; Onaran, Y. Oxidant and Antioxidant Status of Human Breast Milk during Lactation Period. Dairy Sci. Technol. 2015, 95, 295–302. [Google Scholar] [CrossRef]
  288. Section on Breastfeeding. Breastfeeding and the use of human milk. Pediatrics 2012, 129, e827–e841. [CrossRef] [PubMed]
  289. Tain, Y.L.; Lin, Y.J.; Hsu, C.N. Breastfeeding and Future Cardiovascular, Kidney, and Metabolic Health—A Narrative Review. Nutrients 2025, 17, 995. [Google Scholar] [CrossRef] [PubMed]
  290. Domínguez-Perles, R.; Gil-Izquierdo, A.; Ferreres, F.; Medina, S. Update on oxidative stress and inflammation in pregnant women, unborn children (nasciturus), and newborns-Nutritional and dietary effects. Free Radic. Biol. Med. 2019, 142, 38–51. [Google Scholar] [CrossRef] [PubMed]
  291. Madero, M.; Levin, A.; Ahmed, S.B.; Carrero, J.J.; Foster, B.; Francis, A.; Hall, R.K.; Herrington, W.G.; Hill, G.; Inker, L.A.; et al. Evaluation and Management of Chronic Kidney Disease: Synopsis of the Kidney Disease: Improving Global Outcomes 2024 Clinical Practice Guideline. Ann. Intern. Med. 2025, 178, 705–713. [Google Scholar] [CrossRef]
  292. Liakopoulos, V.; Roumeliotis, S.; Bozikas, A.; Eleftheriadis, T.; Dounousi, E. Antioxidant Supplementation in Renal Replacement Therapy Patients: Is There Evidence? Oxidative Med. Cell. Longev. 2019, 2019, 9109473. [Google Scholar] [CrossRef]
  293. Roumeliotis, S.; Roumeliotis, A.; Dounousi, E.; Eleftheriadis, T.; Liakopoulos, V. Dietary Antioxidant Supplements and Uric Acid in Chronic Kidney Disease: A Review. Nutrients 2019, 11, 1911. [Google Scholar] [CrossRef]
  294. Sotler, R.; Poljšak, B.; Dahmane, R.; Jukić, T.; Pavan Jukić, D.; Rotim, C.; Trebše, P.; Starc, A. Prooxidant activities of antioxidants and their impact on health. Acta Clin. Croat. 2019, 58, 726–736. [Google Scholar] [CrossRef] [PubMed]
  295. Okamura, D.M.; Himmelfarb, J. Tipping the redox balance of oxidative stress in fibrogenic pathways in chronic kidney disease. Pediatr. Nephrol. 2009, 24, 2309–2319. [Google Scholar] [CrossRef]
  296. Schena, F.P.; Cox, S.N. Biomarkers and Precision Medicine in IgA Nephropathy. Semin. Nephrol. 2018, 38, 521–530. [Google Scholar] [CrossRef]
  297. Metzinger, L.; Moreno, J.A.; Metzinger-Le Meuth, V. The Role of Non-Coding RNAs in Kidney Diseases. Int. J. Mol. Sci. 2022, 23, 6624. [Google Scholar] [CrossRef] [PubMed]
  298. Jin, C.; Wu, P.; Li, L.; Xu, W.; Qian, H. Exosomes: Emerging Therapy Delivery Tools and Biomarkers for Kidney Diseases. Stem Cells Int. 2021, 2021, 7844455. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic illustrating mechanisms through which environmental pollutants impair kidney health. Figure created using Napkin AI Image Generator [https://www.napkin.ai/ (accessed on 31 August 2025)].
Figure 1. Schematic illustrating mechanisms through which environmental pollutants impair kidney health. Figure created using Napkin AI Image Generator [https://www.napkin.ai/ (accessed on 31 August 2025)].
Antioxidants 14 01205 g001
Figure 2. Antioxidant-based approaches for preserving kidney health against environmental pollutants across the life course. Figure created using Napkin AI Image Generator [https://www.napkin.ai/ (accessed on 31 August 2025)].
Figure 2. Antioxidant-based approaches for preserving kidney health against environmental pollutants across the life course. Figure created using Napkin AI Image Generator [https://www.napkin.ai/ (accessed on 31 August 2025)].
Antioxidants 14 01205 g002
Figure 3. A life-course approach to the prevention of environmental nephrotoxicity. Figure created using Napkin AI Image Generator [https://www.napkin.ai/ (accessed on 31 August 2025)].
Figure 3. A life-course approach to the prevention of environmental nephrotoxicity. Figure created using Napkin AI Image Generator [https://www.napkin.ai/ (accessed on 31 August 2025)].
Antioxidants 14 01205 g003
Table 1. Summary of Animal Models Linking Early-Life Toxicant Exposure and Kidney Programming.
Table 1. Summary of Animal Models Linking Early-Life Toxicant Exposure and Kidney Programming.
ToxicantExposure Period and DoseSpeciesAge at Evaluation (Weeks)Kidney ProgrammingRef
TCDD200 ng/kg orally at GD14/21 and PND7/14SD rats/M12Hypertension[186]
TCDD200 ng/kg, 4 oral doses across gestation and lactationSD rats/M12Hypertension[80,187]
TCDD200 ng/kg, 4 oral doses across gestation and lactationSD rats/M16Hypertension[188]
TCDD6 µg/g orally at GD 14.5C57BL/6N
mice/M
12Kidney malformation[189]
BPA10/100 mg/kg/day during GD9–16OF1 mice/M
& F
5Disturbed kidney function[190]
BPA50 mg/kg/day across gestation and
Lactation
SD rats/M16Hypertension[191]
DEHP0.25/6.25 mg/kg/day during pregnancyWistar rats/
M & F
21Disturbed kidney function and renal hypertrophy[192]
DEHP10 mg/kg/day across pregnancy and
lactation
SD rats/M12Hypertension[193]
DBP850 mg/kg/day during GD14–18SD rats/M8Disturbed kidney function and renal fibrosis[194]
NPs1 mg/L polystyrene-NPs in water across pregnancy and lactationC57BL/6 J mice/M & F4Disturbed kidney function and renal hypertrophy [195]
CdCd chloride 2.0/2.5 mg/kg/day at GD8, 10, 12 & 14SD rats/M7Kidney injury[196]
CdCd chloride 0.5 mg/kg/day during pregnancyWistar rats/
M & F
8Disturbed kidney function[197]
BaP600/1200 mg/kg/day during GD14-17LEH rats/M
& F
8Hypertension[198]
PFOS50 μg/mL from GD4 to deliverySD rats/M & F16Hypertension[199]
PM2.5PM2.5, 16 wk prior to deliveryC57BL/6N
mice/M & F
12Hypertension[200]
PM2.5Oropharyngeal PM2.5, 1.0 mg/kg, GD8, 10, 12SD rats/M14Hypertension[201]
GD, gestational day; PND, postnatal day; TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin; BPA, bisphenol A; DEHP, di-2-ethylhexylphthalate; DBP, di-n-butyl phthalate; PFOS, perfluorooctane sulfonic acid; BaP, benzo(a)pyrene; NPs, nanoparticles; Cd, cadmium; PM2.5 (particulate matter < 2.5 mm); SD, Sprague-Dawley rat; OF1, Oncins France 1; LEH, Long Evans Hooded; M, male; F, female.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hsu, C.-N.; Hou, C.-Y.; Chen, Y.-W.; Chang-Chien, G.-P.; Lin, S.-F.; Tain, Y.-L. Environmental Nephrotoxicity Across the Life Course: Oxidative Stress Mechanisms and Opportunities for Early Intervention. Antioxidants 2025, 14, 1205. https://doi.org/10.3390/antiox14101205

AMA Style

Hsu C-N, Hou C-Y, Chen Y-W, Chang-Chien G-P, Lin S-F, Tain Y-L. Environmental Nephrotoxicity Across the Life Course: Oxidative Stress Mechanisms and Opportunities for Early Intervention. Antioxidants. 2025; 14(10):1205. https://doi.org/10.3390/antiox14101205

Chicago/Turabian Style

Hsu, Chien-Ning, Chih-Yao Hou, Yu-Wei Chen, Guo-Ping Chang-Chien, Shu-Fen Lin, and You-Lin Tain. 2025. "Environmental Nephrotoxicity Across the Life Course: Oxidative Stress Mechanisms and Opportunities for Early Intervention" Antioxidants 14, no. 10: 1205. https://doi.org/10.3390/antiox14101205

APA Style

Hsu, C.-N., Hou, C.-Y., Chen, Y.-W., Chang-Chien, G.-P., Lin, S.-F., & Tain, Y.-L. (2025). Environmental Nephrotoxicity Across the Life Course: Oxidative Stress Mechanisms and Opportunities for Early Intervention. Antioxidants, 14(10), 1205. https://doi.org/10.3390/antiox14101205

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop