Cadmium (Cd) is a naturally occurring toxic heavy metal found ubiquitously throughout the environment. Anthropogenic activities since the onset of industrialization have led to widespread environmental contamination that has substantially increased human exposure and associated health risks. As one of the top ten chemicals of major public health concern of the World Health Organization, Cd poses significant risks to human health, particularly when exposure occurs during the critical stages of development. Cd accumulates in the placenta and can be detected in cord blood and fetal and neonatal tissues, so it is crucial to understand the consequences of early-life Cd exposure and the underlying molecular mechanisms. In this Review, we provide an overview of the models currently used to study developmental Cd exposure and integrate the findings from epidemiological, animal and in vitro studies. We explore the impacts and mechanisms of early-life Cd exposure on the placenta, growth and development, and organ systems, identifying common themes across diverse model systems. Finally, we pinpoint knowledge gaps and propose key research priorities that will advance our understanding and inform mitigation strategies for reducing the developmental risks of Cd exposure.

Cadmium (Cd) is a heavy metal and non-essential trace element that is toxic to nearly all species (Andresen and Küpper, 2013; Dutta et al., 2020; Jacobson et al., 1981; Thévenod and Lee, 2013; Trevors et al., 1986; Xu and Morel, 2013). Cd naturally occurs in the Earth's crust at low levels [0.1-0.5 parts per million (ppm)] and is released into the environment through both natural processes (volcanic eruptions, fires, etc.) and human activities (mining, manufacturing, etc.) [Agency for Toxic Substances and Disease Registry (ATSDR), 2012; Cullen and Maldonado, 2013]. Over the past century, there has been a surge in industrial Cd consumption, the majority of which is driven by the manufacturing of nickel-Cd rechargeable batteries (Cullen and Maldonado, 2013).

Humans are predominantly exposed to Cd through two routes: inhalation and ingestion (Fig. 1) (ATSDR, 2012). Around 6% of ingested Cd is absorbed through the intestines, while 10-50% of inhaled Cd is absorbed through the lungs (Genchi et al., 2020; Järup, 2002; Mikolić et al., 2015). Tobacco plants efficiently uptake Cd from the soil, and, consequently, the Cd body burden of smokers is approximately double that of non-smokers (ATSDR, 2012; Cullen and Maldonado, 2013; Satarug et al., 2003). Cd can also be inhaled through contaminated dust or fume particles present in occupational settings (Cullen and Maldonado, 2013). In non-smokers, ingestion is the primary source of exposure and occurs predominantly through Cd-contaminated food (Kim et al., 2019) and water from unregulated private wells (Idrees et al., 2018; Kubier et al., 2019; Sanders et al., 2014).

Fig. 1.

An overview of the common sources and routes of cadmium (Cd) exposure. (A) Cd is released into the environment through human activity, including industrial processes required for the production of nickel-Cd batteries. Cd can enter air and water from factory exhaust and accidental spills. Natural processes, including volcanic eruptions and wildfires, also emit Cd, which contaminates air through direct aerosolization and enters the water cycle. (B) Common sources of maternal Cd exposure include dust and fumes in occupational settings, and tobacco products, which are absorbed through the inhalation route. Cd is also ingested from contaminated drinking water, typically from unregulated private wells, and foods like leafy greens, which accumulate Cd. Both inhaled and ingested Cd can be absorbed and affect placental and fetal development.

Fig. 1.

An overview of the common sources and routes of cadmium (Cd) exposure. (A) Cd is released into the environment through human activity, including industrial processes required for the production of nickel-Cd batteries. Cd can enter air and water from factory exhaust and accidental spills. Natural processes, including volcanic eruptions and wildfires, also emit Cd, which contaminates air through direct aerosolization and enters the water cycle. (B) Common sources of maternal Cd exposure include dust and fumes in occupational settings, and tobacco products, which are absorbed through the inhalation route. Cd is also ingested from contaminated drinking water, typically from unregulated private wells, and foods like leafy greens, which accumulate Cd. Both inhaled and ingested Cd can be absorbed and affect placental and fetal development.

Close modal

Regardless of the exposure route, Cd is rapidly removed from the bloodstream and stored in tissues, with a biological half-life of ∼26 years (Genchi et al., 2020; Waalkes, 2003). The liver and kidneys are the main targets for Cd accumulation, with the latter storing upwards of 50% of the total body burden (Genchi et al., 2020). Pregnancy is a particularly vulnerable state because the programmed increase in essential nutritive metal uptake to support fetal demands leads to a concurrent increase in absorption of Cd (Delaney et al., 2020; Olsson et al., 2002; Satarug et al., 2010; Vahter et al., 2002). Interestingly, cord blood levels of Cd are much lower than those seen in maternal blood or the placenta, suggesting that there is limited Cd transport into fetal tissues (Dong et al., 2023).

In this Review, we summarize the utility of epidemiological studies, animal models and in vitro approaches (Box 1) and discuss findings that assess the effects of developmental Cd exposure. We present the current evidence linking Cd exposure to gestational and birth outcomes, including placental insufficiency, fetal growth restriction (FGR) and congenital anomalies, followed by an exploration of dysregulation and dysfunction in major organs and organ systems: the cardiovascular system, nervous system, reproductive system, liver and kidneys. Finally, we address the gaps in our current knowledge and propose areas that warrant further study.

Box 1. Approaches used for investigating the effects of developmental cadmium (Cd) exposure

Perinatal epidemiological studies aim to understand the maternal, prenatal and postnatal health outcomes of events that occur during pregnancy and are often used to investigate the health effects of developmental Cd exposure (Buck Louis and Platt, 2011; Dunne et al., 2021; Giannakou, 2021). Given that these studies are observational, they typically use a cohort design in which pregnant women, child-mother pairs and/or children are recruited based on known or possible Cd exposure. Alternatively, they employ a case-control design in which recruitment is based on the occurrence of a specific health impairment (Chandravanshi et al., 2021; Song and Chung, 2010). Cd can be non-invasively quantified in maternal or fetal biological specimens (e.g. blood, urine, hair, cord blood, placenta, breast milk) to determine whether an observed phenotype can be attributed to Cd exposure (Vilahur et al., 2015).

Experimental animal studies enable specific hypothesis testing of the mechanisms underlying developmental Cd toxicity (Parasuraman, 2011). Rodents are most commonly used in developmental studies to model human exposure as they share similar anatomical structures (e.g. placenta), physiological processes and molecular pathways during development (Alberts et al., 2002; Carter, 2020). In developmental Cd studies, female rodents are administered Cd before and/or during pregnancy which permits investigation of the combined effects of Cd exposure on maternal physiology and direct transfer of Cd to the fetus (King-Herbert and Vasbinder, 2020; Leroy and Allais, 2013; Marsden and Leroy, 2013; Parasuraman, 2011). Fish, chickens and frogs are also used in studies to determine the direct effects of Cd exposure on developing offspring, as their ex utero development and semi-transparent embryos allow for easier exposure, visualization and manipulation (Liu et al., 2016b; Stark and Ross, 2019; Veldman and Lin, 2008).

In vitro models have been used in toxicology research for decades as they are cost effective, use few to no live animals, and require less time and physical space. Common in vitro models include cultured cells, micromass, organs, tissues and whole embryos (Winn, 2019; Lee et al., 2012; Pamies et al., 2011). The simplified nature of in vitro models allows direct investigation of the effects of Cd and facilitates precise control of physical, chemical and genetic parameters (Allen et al., 2008; Brannen et al., 2016; Winn, 2019; Il'yasova et al., 2015; Lee et al., 2012; Pamies et al., 2011). However, mimicking the gestational environment in placental animals is challenging when using in vitro methods; therefore, in vitro models can complement animal models to further explore the mechanisms driving the response to developmental Cd exposure.

In mammals, the placenta acts as a semi-permeable barrier to the maternal-fetal exchange of Cd and is where most Cd accumulates (Barrett et al., 2023). Only a small amount of Cd enters fetal circulation compared to other metals like lead (Pb) and mercury (Hg) (Kantola et al., 2000; Plöckinger et al., 1993; Sakamoto et al., 2013). The amount of Cd that crosses the placental barrier can vary based on the dose and route of exposure, gestational age, fetal sex, maternal health, diet, lifestyle and other factors (Ahokas and Dilts, 1979; Jackson et al., 2022; Sonawane et al., 1975; Dong et al., 2023). Notably, early rodent studies found that fetuses could tolerate direct Cd injections at concentrations that were embryonic lethal when administered to the mother (Levin et al., 1981), indicating that the impacts of maternal Cd exposure are more likely due to impaired placental function and/or poor gestational environment rather than direct effects of Cd within fetal tissues.

Cd is sequestered in the placenta owing to a high affinity for metal-binding metallothionein (MT) (see Glossary, Box 2) proteins (Coyle et al., 2002) (Fig. 2). Placental Cd-MT accumulation triggers a positive feedback loop, whereby MT synthesis is upregulated. Because of the non-specific affinity of MT for metal binding, zinc (Zn), copper (Cu), iron (Fe) and selenium (Se) also become sequestered in the placenta, reducing transfer of these essential nutrients to the fetus (Coyle et al., 2002; Waalkes and Goering, 1990). Higher placental Cd levels are associated with reduced Zn transport to the fetus, regulated by placental zinc transporter (Znt)1 (also known as Slc30a1) and Znt2 (also known as Slc30a2) (Wang et al., 2016b). Cd may also interfere with placental transporters for Fe [e.g. divalent metal transporter 1 (DMT1; also known as SLC11A2)] (Leazer et al., 2002; Somsuan et al., 2019) and calcium (Ca) [e.g. transient receptor potential cation channel subfamily V member 6 (TRPV6) and voltage-dependent L-type calcium channel subunit alpha-1C (CACNA1C)] (Phuapittayalert et al., 2016). In addition, Cd exposure has been shown to reduce the abundance of placental transporters ATP-binding cassette super-family G member 2 (ABCG2) and ATP-binding cassette, subfamily B, member 4 (ABCB4), which are responsible for translocating heavy metals from the placental cell membranes back into maternal circulation (Liu et al., 2016a). Recent in vitro, in vivo and epidemiological studies have further bolstered the role of placental ABCG2 in regulating maternal Cd toxicity; reduced efflux efficiency conferred by the Q141K variant was found to be less protective against the Cd-induced activation of stress genes and cytotoxicity in vitro (Wen et al., 2021) and to increase the strength of the inverse relationship between placental Cd concentration and placental weight in newborns (Barrett et al., 2023). Cd-exposed Abcg2 knockout mice had significantly higher renal Cd accumulation than wild-type mice owing to reduced urinary excretion (Wen et al., 2021).

Box 2. Glossary

11β-hydroxysteroid dehydrogenase 2 (11β-HSD2): an enzyme responsible for limiting the transfer of the glucocorticoid cortisol from maternal circulation to the fetus by catalyzing the inactivating conversion of cortisol to cortisone.

AKT signaling: a signaling pathway that functions as a key regulator of cell growth, survival and metabolism.

Cardiomegaly: a condition characterized by enlargement of the heart, usually symptomatic of an underlying pathology.

Ectrodactyly: a rare birth defect that causes one or more fingers or toes to be missing or malformed.

Glomerulus: a cluster of capillaries in the kidneys responsible for filtering cells and proteins from the blood to create a glomerular filtrate, which ultimately becomes excreted as waste in the form of urine.

Gonadotropin-releasing hormone: a hormone released from the hypothalamus that triggers the production and secretion of luteinizing hormone and follicle-stimulating hormone from the pituitary gland. These produced hormones are important for regulating sex-specific hormone production in the gonads.

Granulosa cells: a follicular cell type located in the ovaries that clusters around the developing egg and provides estrogen and progesterone at key points in the menstrual cycle.

Implantation: the first stage of gestation when a newly fertilized egg attaches to the uterine lining, establishing a pregnancy.

Intraperitoneal: within the membrane (peritoneum) that lines the abdominal cavity and covers the abdominal organs.

Leptin: a hormone predominantly secreted by adipose tissue that regulates energy balance through influencing appetite, feeding behaviors and satiety.

Leydig cells: an interstitial cell type located in the testes, responsible for providing testosterone and other androgens to support sperm production, sexual maturation and development of secondary sex characteristics.

Luteinizing hormone: a hormone that stimulates ovulation in females and the development of secondary sexual characteristics in men.

Metabolic dysfunction-associated steatotic liver disease (MASLD): a chronic and progressive liver condition that occurs when fat accumulates in the liver. It was previously known as nonalcoholic fatty liver disease (NAFLD).

Metallohormone: a description given to metals that appear to display steroid hormone-like activity, despite otherwise having no known biological function.

Metallothionein: a family of proteins that bind metals, including metals that are essential (e.g. zinc) and toxic (e.g. cadmium). Metallothionein protects against oxidative stress and metal toxicity.

Orofacial cleft: a birth defect that causes tissues of the lip and mouth to not form properly.

Placenta previa: a condition whereby the placenta attaches to the lower part of the uterus and partially or completely covers the cervix, often leading to preterm birth and maternal hemorrhage.

Placentation: the location and process of placenta formation following implantation of the embryo in the uterus, allowing for exchange of nutrients and oxygen from maternal to fetal circulation.

Preeclampsia: a serious high-blood pressure condition that can occur during pregnancy or after giving birth.

Preneoplastic lesions: abnormal cells that have a higher risk of developing into non-cancerous or cancerous tumors.

Proximal tubule cells: a major epithelial cell type in the kidneys that make up the proximal tubules, a substructure of the nephron responsible for reabsorbing fluid that has been filtered through the glomerulus back into circulation.

Sertoli cells: a structural cell type located in the seminiferous tubules of the testes, characterized by a columnar shape and very large nuclei, responsible for supporting sperm production.

Striatal: related to the striatum, a coordinated group of structures in the forebrain with critical roles in cognition, motor function and learning.

Teratogenic: a substance or exposure that has the potential to cause abnormal development in an embryo or fetus.

Transferrin: a family of glycoproteins involved in the transfer and homeostasis of iron.

Ubiquitination: the process by which ubiquitin proteins are attached to target proteins, marking them for degradation. This process is important for maintaining proper transcriptional regulation, cell surface signaling and regeneration.

WNT/β-catenin pathway: a signaling pathway that plays critical roles in embryonic development and adult tissue homeostasis.

Fig. 2.

A summary of the impacts of developmental cadmium (Cd) exposure on the placenta and multiple organ systems. (A) Maternal Cd exposure is linked to fetal growth restriction and affects multiple organ systems, including the nervous, cardiovascular and reproductive systems, as well as the liver and kidneys. (B) Cd (orange circles) disrupts homeostasis of essential trace metals, including zinc (Zn), selenium (Se), iron (Fe) and copper (Cu) (blue triangles). The presence of Cd in the placenta induces upregulation of metallothionein (MT), which non-specifically sequesters metals. Additionally, developmental Cd exposure has been shown to interfere with specific transporters, including zinc transporter (ZNT)1 (also known as SLC30A1), ZNT2 (also known as SLC30A2), divalent metal transporter 1 (DMT1; also known as SLC11A2), transient receptor potential cation channel subfamily V member 6 (TRPV6), voltage-dependent L-type calcium channel subunit alpha-1C (CACNA1C), ATP-binding cassette super-family G member 2 (ABCG2) and ATP-binding cassette, subfamily B, member 4 (ABCB4). Together, these perturbations limit the transfer of both Cd and essential trace metals to the developing fetus.

Fig. 2.

A summary of the impacts of developmental cadmium (Cd) exposure on the placenta and multiple organ systems. (A) Maternal Cd exposure is linked to fetal growth restriction and affects multiple organ systems, including the nervous, cardiovascular and reproductive systems, as well as the liver and kidneys. (B) Cd (orange circles) disrupts homeostasis of essential trace metals, including zinc (Zn), selenium (Se), iron (Fe) and copper (Cu) (blue triangles). The presence of Cd in the placenta induces upregulation of metallothionein (MT), which non-specifically sequesters metals. Additionally, developmental Cd exposure has been shown to interfere with specific transporters, including zinc transporter (ZNT)1 (also known as SLC30A1), ZNT2 (also known as SLC30A2), divalent metal transporter 1 (DMT1; also known as SLC11A2), transient receptor potential cation channel subfamily V member 6 (TRPV6), voltage-dependent L-type calcium channel subunit alpha-1C (CACNA1C), ATP-binding cassette super-family G member 2 (ABCG2) and ATP-binding cassette, subfamily B, member 4 (ABCB4). Together, these perturbations limit the transfer of both Cd and essential trace metals to the developing fetus.

Close modal

Early-life Cd exposure has been consistently linked with FGR and low birth weight (LBW) in epidemiological studies (Table S1) (Röllin et al., 2015; Tian et al., 2009; Vidal et al., 2015; Zhang et al., 2004) and animal models (Table S2) (Castillo et al., 2012; Enli et al., 2010; Hudson et al., 2021; Ronco et al., 2011; Thompson and Bannigan, 2001; Zhang et al., 2012, 2015), in both placental and non-placental species. FGR is typically concurrent with placental insufficiency, wherein the placenta does not adequately support the growing fetus (Nardozza et al., 2017) and increases the risk of premature birth, sudden infant death syndrome, metabolic disorders, impaired neurodevelopment and heart disease (Sabra et al., 2017), highlighting the importance of mitigating exposure to Cd and other potential FGR risk factors during pregnancy (Fig. 2). Regardless of dose, maternal intraperitoneal (Box 2) injection during gestation more consistently leads to FGR and LBW (Fernandez et al., 2003; Ji et al., 2011; Zhang et al., 2016). However, growth restriction is generally only observed following maternal ingestion of water containing ≥30 ppm Cd (Ronco et al., 2009, 2011; Zhao et al., 2018; Hudson et al., 2019), suggesting a dose-dependent threshold to induce FGR and LBW. Female offspring appear to be more susceptible to Cd-impaired fetal growth, particularly in studies with large cohorts or those conducted in areas with notable Cd contamination (Kippler et al., 2012; Cheng et al., 2017; Taylor et al., 2016; Zhang et al., 2018a), indicating that there may be subtle sex-specific differences that become more pronounced at higher doses. Co-administration of Cu or Zn can prevent FGR/LBW (Enli et al., 2010; Fernandez et al., 2003), and, considering that Cd is able to bind essential metal transporters, it is likely that FGR and LBW are consequences of altered essential trace metal homeostasis.

Understanding the pathological impacts of Cd exposure on FGR requires a comprehensive examination of its impact on the placenta. Numerous studies have shown impacts of maternal Cd exposure on placental structure and function, including implantation (Box 2), nutrient transport, hormone signaling, oxidative stress, and genomic and epigenomic effects, which are discussed in detail below and summarized in Tables S3-S5.

Impaired placentation and implantation

Cd reportedly impacts the earliest stages of fetal development by affecting the process and site of placentation (Box 2) and implantation during early development. In support of this, women with placenta previa (Box 2) had higher blood Cd levels (Tsuji et al., 2019). Furthermore, activities of the implantation enzymes cathepsin-D and alkaline phosphatase were downregulated or upregulated, respectively, by maternal Cd exposure in rats (Nampoothiri and Gupta, 2008). Cd accumulates in the labyrinth zone of the mature placenta of rodents, which is important for maternal-fetal nutrient and gas exchange (Yamagishi et al., 2016). Trophoblasts in the placental labyrinth zone display reduced proliferation, increased apoptosis and a muted response to differentiation induction, leading to placental insufficiency and FGR (Erboga and Kanter, 2016; Lee et al., 2009; Wang et al., 2012; Wier et al., 1990; Yamagishi et al., 2016; Zhou et al., 2016).

Fetomaternal endocrine disruption

Cd exposure during pregnancy changes the abundance of maternal and fetal hormones, including glucocorticoids and steroid hormones that drive differentiation in favor of proliferation, leading some to assign Cd the classification of metallohormone (Box 2) (Wyrwoll and Holmes, 2012). Fetal exposure to elevated glucocorticoids is associated with LBW, and Cd was found in rodents to increase glucocorticoid concentrations in the fetus, placenta and maternal plasma (Ronco et al., 2009). Cd may lead to increased glucocorticoids through reduced expression, protein abundance and enzymatic activity of 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2; also known as HSD11B2; Box 2), a negative regulator of maternal-fetal glucocorticoid transfer, as this has been observed both in vitro in human placental trophoblast cells and in vivo in rats (Wang et al., 2014; Yang et al., 2006). The role of 11β-HSD2 in Cd-induced FGR might depend on gestational timing, route and duration of maternal exposure, as well as the functionality of other mechanisms that mitigate Cd toxicity in the placenta. For example, one study found that placental 11β-HSD2 was seemingly unaffected by midgestational maternal Cd exposure in both MT-null and wild-type mice despite FGR occurring in the highest-dosed MT-null pups (Selvaratnam et al., 2013). An explanation offered for the lack of observed effects on 11β-HSD2 during Cd-induced FGR was that the effects are more pronounced in late gestation after 11β-HSD2 expression has peaked. Other Cd-induced impairments to placental glucocorticoid signaling may occur through altered glucocorticoid receptor expression, as one study identified Cd-associated changes to placental DNA methylation of nuclear receptor subfamily 3 group C member 1 (NR3C1), a glucocorticoid receptor gene (Appleton et al., 2017). Cd exposure is also associated with preeclampsia (Box 2), which may be linked to abnormal glucocorticoid homeostasis in the placenta, although a causative mechanism has not been established (Brooks et al., 2016; Wang et al., 2014, 2018; Zhang et al., 2016).

Cd exposure disrupts the synthesis and regulation of other key placental hormones that are critical for fetal development and maternal health, such as placental lactogens (Lee et al., 2009) and leptin (Box 2). Synthesis of progesterone, a hormone required for pregnancy maintenance and fetal development, is also disrupted by Cd exposure in vitro and in vivo (Jolibois et al., 1999a; Kawai et al., 2002). Accordingly, placentae from smokers have higher levels of Cd and lower levels of progesterone (Piasek et al., 2002). The underlying mechanisms leading to impaired placental progesterone synthesis may involve a Cd-induced reduction in placental cholesterol levels, altered expression of the low-density lipoprotein receptor or reduction of steroidogenic enzymes (Jolibois et al., 1999b; Nampoothiri and Gupta, 2008). Progesterone supplementation in pregnant rats can alleviate Cd-induced placental abnormalities and preeclampsia-related symptoms (Zhang et al., 2018b). Placental estrogen synthesis is similarly impaired and can also be ameliorated by supplementation with synthetic estrogen (Liu et al., 2022).

Oxidative stress

Increased placental oxidative stress (OS) is often seen in the pathology of pregnancy-related disorders like preeclampsia and FGR. Elevated OS impairs trophoblast invasion and differentiation, triggers increased apoptosis of trophoblasts and induces placental endoplasmic reticulum (ER) stress (Wang et al., 2012; Wu et al., 2015). Gestational exposure to Cd has been shown to exacerbate placental biomarkers of OS, including free radicals, oxidative DNA damage, and DNA damage repair proteins apurinic/apyrimidinic endonuclease 1 (APE1; also known as APEX1) and p53 (also known as TP53), along with a reduction in total antioxidant capacity (Zhang et al., 2016). Additionally, the key antioxidant glutathione is reduced in Cd-exposed rodent placentae (Enli et al., 2010; Wang et al., 2012). Disruption of placental glutathione metabolism may contribute to preeclampsia (Jin et al., 2017). In vivo work suggests that supplementation with N-acetylcysteine, a glutathione precursor, can rescue the negative effects of Cd on placental development, gene expression, ER stress and FGR (Guo et al., 2018).

Cd-induced reactive oxygen species (ROS) further disrupt placental development by activating hypoxia-inducible factor 1 (HIF-1; also known as HIF1A), a key regulator of oxygen homeostasis, and associated HIF-1 targets, and by altering expression of transforming growth factor beta (TGF-β) pathway members that are required for trophoblast differentiation and invasion (Adebambo et al., 2018). In addition, Cd exposure activates the OS-sensitive transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), which plays a central role in placental inflammatory responses (Ronco et al., 2011). Mechanistically, Cd promotes placental inflammation at least in part through activating the AKT serine/threonine kinase (AKT) signaling (Box 2) pathway both in vivo and in vitro; inhibition of this pathway has been shown to alleviate Cd-induced upregulation of pro-inflammatory cytokines (Hu et al., 2018). Antioxidant supplementation during Cd exposure has been shown to alleviate Cd-induced OS and related phenotypes in both the placenta and fetus, supporting the notion that increased OS contributes to the pathophysiology of maternal Cd exposure and providing promising support for antioxidant therapies (Adebambo et al., 2018; Enli et al., 2010; Guo et al., 2018; Wang et al., 2012).

Disrupted carbohydrate metabolism

Glycogen, the storage form of glucose, is abundant in the placenta during early pregnancy, although its exact role remains debated. However, elevated placental glycogen levels are often observed in poorly functioning placentae associated with FGR and preeclampsia (Akison et al., 2017; Arkwright et al., 1993). Maternal Cd exposure has been associated with elevated placental glycogen levels during late rodent gestation when levels would typically decline (Roelfzema et al., 1987, 1988a,b, 1989; Yoruk et al., 2003). Impaired enzymatic activity of glycogen phosphorylase, which breaks down glycogen into glucose, might be expected to contribute to glycogen accumulation; however, one study in rats found increased placental glycogen phosphorylase activity following maternal Cd exposure, indicating higher glycogen turnover and disrupted downstream carbohydrate metabolism (Roelfzema et al., 1989). In support of this idea, glucose transporter 3 (Glut3; also known as Slc2a3) expression was found to be significantly reduced in the murine placenta owing to maternal Cd exposure (Xu et al., 2016). Additionally, Cd-induced maternal and fetal Fe deficiencies during gestation have been associated with hypoxia in newborn mouse tissues, and a hypoxic environment during development may impair aerobic respiration and shift energy production towards inefficient anaerobic pathways to meet the energy demands of developing fetal tissues (Hudson et al., 2019, 2021). In contrast, another study reported decreased placental glycogen levels after subcutaneous Cd administration throughout pregnancy (Nampoothiri and Gupta, 2008); however, this finding was accompanied by significant reductions in all other measured biomarkers, suggesting severe placental damage and a pre-apoptotic state.

Altered regulation and stability of the genome and epigenome

Maternal Cd exposure also exerts effects on the placental genome and epigenome. Elevated placental Cd is associated with shortened telomere length, a hallmark of cancer and aging (Lin et al., 2013). Similarly, an in vitro model of embryonic exposure to cigarette smoke also demonstrated reduced telomere length (Huang et al., 2010). Cd has also been shown to damage DNA in vitro through OS (Nagaraju et al., 2022; Skipper et al., 2016). Female rats treated with Cd during pregnancy exhibited a significant reduction in placental DNA and RNA content (Lee et al., 2009; Nampoothiri and Gupta, 2008; Zhang et al., 2016), consistent with Cd-induced placental DNA damage.

Developmental Cd exposure is also associated with altered fetal DNA methylation in humans and animal models (Kippler et al., 2013; Sanders et al., 2014; Zhu et al., 2011). In humans, higher maternal Cd exposure is associated with altered fetal DNA methylation at genomic loci implicated in body weight and at imprinted genes that contribute to fetal growth (Cowley et al., 2018). In fact, altered DNA methylation at specific loci has been linked to maternal Cd burden and birth weight in the same population (Kippler et al., 2013). In the placenta, Cd-associated differentially methylated regions (DMRs) have been found in or near genes with roles in inflammatory signaling, cell growth, birth metrics, hormone receptors and fetal growth (Appleton et al., 2017; Everson et al., 2016, 2018). Some placental DMRs are sex specific; in female offspring, Cd-associated DMRs were identified in promoters of genes associated with cell damage response, whereas in males the DMRs were associated with genes involved in cell differentiation, angiogenesis and organ development (Mohanty et al., 2015). However, a genome-wide analysis of DNA methylation in the mouse placenta did not identify any DMRs associated with Cd exposure, suggesting that Cd exerts species-specific effects on the placental epigenome, possibly reflecting differences between murine and human placental structure (Simmers et al., 2023). The aforementioned approach does not rule out Cd-induced differential methylation in mice; additional studies are needed to determine whether this was inherent to the study design or whether there are truly species-specific differences in how Cd affects the placental epigenome. Differences between murine and human placental structure and gene expression patterns have been documented, so novel techniques like three-dimensional placenta organoids and embryoid bodies provide promising research platforms to accurately study the impact of Cd exposure on the placenta (Hemberger et al., 2020).

The preceding sections highlight the critical role of the placenta in the early pathogenesis of adverse health outcomes owing to maternal Cd exposure; the following sections will explore the effects on non-placental tissues and organ systems.

Congenital anomalies

Evidence from experimental animal models suggests that Cd is teratogenic (Box 2); however, this is not well substantiated in humans (Calle et al., 2015). One recent study in humans connected placental Cd levels to newborn orofacial clefts (Box 2), but the simultaneous presence of Pb and Hg made it unclear whether Cd alone was responsible (Pi et al., 2018). In rodent studies, teratogenic effects depend on the dose, route and gestational timing of Cd administration and genetic background (Table S6). When pregnant female rodents are exposed orally to Cd, there do not appear to be teratogenic effects (Barański et al., 1982; Saxena et al., 1986; Wardell et al., 1982). However, when Cd is administered to pregnant female rodents through intravenous or intraperitoneal injection, congenital anomalies are nearly always observed (Chen et al., 2008; Elsaid et al., 2010; Fernandez et al., 2003; Holt and Webb, 1987; Murata et al., 1993; Padmanabhan, 1987; Samarawickrama and Webb, 1979; Webb et al., 1988; Webster and Messerle, 1980). Teratogenic effects of Cd have also been reported in zebrafish and chick embryos, likely owing to the direct route of Cd exposure in an aqueous medium (Bosch et al., 2023; Chouchene et al., 2022; Di Paola et al., 2022; Hen Chow et al., 2009; Manigandan et al., 2015; Thompson and Bannigan, 2008; Zhang et al., 2015). Cd-induced defects in non-mammalian models appear to depend on the dose and timing of exposure; low concentrations cause no observable abnormalities (Di Paola et al., 2022), whereas higher doses consistently induce malformations (Chow et al., 2008; Zhang et al., 2015). Cd telluride (CdTe) dots, nanoparticles that have many applications in science and technology, are also linked to spinal defects, although the role of Cd remains unclear because the severity of the effect is dependent on the selected functionalization compound (Bosch et al., 2023).

Multiple pathways underlie the teratogenic effects of Cd. In mouse embryos, maternal intraperitoneal injection of Cd during neurulation altered the expression of genes involved in metabolism, transcription, translation, mitochondrial organization, cell cycle and stress response; genes that were significantly differentially expressed depended on the Cd dose and post-injection latency (Robinson et al., 2011). In rodents, Cd-induced forelimb ectrodactyly (Box 2) may be caused by disruption to fibroblast growth factor (FGF) and sonic hedgehog (SHH) signaling pathways (Elsaid et al., 2007; Schreiner et al., 2009). Disruption of SHH-directed anterior-posterior patterning of vertebrate limbs could explain why increasing doses of Cd lead to increasingly anterior forelimb ectrodactyly (Scott et al., 2005). SHH signaling may be further inhibited by a Cd-induced perturbation of Zn, a required structural component of the SHH protein (Schreiner et al., 2009). Abnormal levels of retinoic acid during development is a well-established cause of congenital malformation, and Cd has been shown to alter its signaling in cultured cells and in mice (Cui and Freedman, 2009; Hudson et al., 2021). Cd-induced developmental defects in zebrafish parallel those seen with retinoic acid depletion, and retinoic acid supplementation can rescue some of the Cd-induced phenotypes (Zhang et al., 2015).

Cardiovascular system

Developmental exposure to Cd can impair cardiovascular development and influence susceptibility to cardiovascular disease (CVD) (Table S7), although findings vary between animal models and human studies. In mice, maternal Cd exposure results in cardiomegaly (Box 2) and reduced myocyte numbers, alongside altered expression of genes linked to WNT/β-catenin signaling and hypertension (Hudson et al., 2019; Omidi et al., 2019). In contrast, human cardiac organoids derived from embryonic stem cells display reduced differentiation and suppression of the WNT/β-catenin pathway (Box 2) (Wu et al., 2022).

Evidence from rodent models supports the long-term cardiovascular consequences of developmental Cd exposure. Rodents born to Cd-exposed mothers have altered heart morphology, vascular reactivity and susceptibility to hypertension in adulthood (Hudson et al., 2019; Ronco et al., 2011). Female mice exposed to Cd from conception to adulthood, in combination with a high-fat diet after weaning, develop cardiac hypertrophy and fibrosis, although the contribution of prenatal Cd exposure to the extent of this phenotype is unclear (Liang et al., 2019). Paradoxically, one study suggested that maternal Cd exposure may aid in resistance to ischemic injury during myocardial infarction in adult rats (Zepeda et al., 2012).

Non-placental experimental models further highlight the detrimental effects of Cd on cardiovascular development (Table S7). Cd-exposed zebrafish embryos display abnormal vasculature, reduced complexity of craniofacial vasculature and reduced heart rates (Cheng et al., 2001; Zhang et al., 2012). Embryonic Japanese medaka fish exposed to Cd-contaminated sediment developed elevated heart rates during development and cardiovascular abnormalities at hatching (Barjhoux et al., 2012). In frog tadpoles, exposure to a low dose of Cd considered in Brazil to be environmentally safe depressed cardiac functions (Dal-Medico et al., 2014).

Although there is strong experimental evidence that Cd negatively impacts cardiovascular development in animals, there are limited findings from epidemiological studies. One study found no association between maternal urinary Cd during pregnancy and blood pressure in 4.5-year-olds (Hawkesworth et al., 2013); another found no association between blood Cd in 2-year-olds and blood pressure throughout childhood (Cao et al., 2009). In humans, Cd may have additive or synergistic effects with other heavy metals that affect cardiovascular development and function. One study reported that elevated maternal blood Cd, in combination with Pb, was associated with increased offspring risk for congenital heart defects, although no significant association was observed for Cd alone (Ou et al., 2017). Another study found that high maternal Cd exposure, as measured in maternal hair samples, was associated with an increased risk of congenital heart defects that was more pronounced when combined with arsenic exposure (Jin et al., 2016). It is also possible that the cardiotoxic effects of developmental Cd exposure are unique to rodent models at high doses that are rarely encountered in human populations, although additional epidemiological studies are required to support this assertion.

Malnutrition during development is associated with increased CVD risk (Wadhwa et al., 2009), and changes to essential trace element homeostasis during prenatal development may be a contributing factor to Cd-associated CVD (Hudson et al., 2019; Mikolić et al., 2015). For example, fetal Fe deficiency induced by maternal Cd exposure may impair fetal oxygen homeostasis (Hudson et al., 2019, 2021) and indirectly lead to OS, a proposed direct mechanism of Cd-induced cardiotoxicity (Almenara et al., 2013). Exposure to Cd during early postnatal life may further impair Fe metabolism in cardiovascular tissues. In postnatally developing rats, Cd increased cardiac transferrin (Box 2) uptake independent of dietary Fe supply (Crowe and Morgan, 1997). Early postnatal Cd exposure may also affect Zn and Cu homeostasis in the heart; Cd-exposed chick embryos displayed delayed vasculogenesis and lacked clearly defined vessels, a phenotype that could explain Cd-associated malnutrition in the embryo (Gheorghescu and Thompson, 2016).

Epigenetic alterations offer another possible mechanism. At the epigenetic level, Cd-associated DMRs were identified in newborn human cord blood at loci involved in cardiometabolic functions, indicating a potential mechanism through which maternal Cd exposure may program long-term susceptibility to CVD (Cowley et al., 2018). However, mechanistic knowledge is lacking, and additional work is needed to identify causal mechanisms linking early-life Cd exposure with impaired cardiovascular development and disease susceptibility.

Nervous system

Extensive epidemiological research links developmental Cd exposure to neurodevelopmental delays and disorders (Table S8). Higher maternal Cd levels correlate with a greater risk of emotional problems and diminished performance in cognition, perception, quantitation, locomotion, intelligence and social development tests (Bonithon-Kopp et al., 1986; Jeong et al., 2015; Kippler et al., 2012, 2016; Sioen et al., 2013; Tian et al., 2009; Wang et al., 2016a). Sex-specific differences have been reported, although it is unclear whether males or females are more sensitive (Ciesielski et al., 2012; Kippler et al., 2012; Sioen et al., 2013). Conversely, others report no significant relationship between maternal, newborn or juvenile Cd levels and impaired neurodevelopment or behavior, possibly owing to differences in behavioral testing and Cd sampling methodologies (de Assis Araujo et al., 2022; Freire et al., 2018; Garí et al., 2022).

Numerous animal studies have described the neurodevelopmental effects of early-life Cd exposure (Table S9). Maternal Cd has been linked with altered brain sizes when measured at birth or during postnatal development (Antonio et al., 1998; Gulati et al., 1986; Gupta et al., 1991; Hudson et al., 2021; Mikolić et al., 2016; Mimouna et al., 2018; Xu et al., 1993; Zhang et al., 2009), as well as altered behaviors, e.g. activity levels, exploration, sensorimotor skills, aversion or avoidance, grooming, cognitive development, learning, memory, stress, anxiety, aggression and sensitivity to sound (Ali et al., 1986; Allam et al., 2016; Barański, 1986; Barański et al., 1983; Dési et al., 1998; Feng et al., 2019; Gulati et al., 1986; Halder et al., 2016, 2019; Hudson et al., 2021; Lehotzky et al., 1990; Mimouna et al., 2018; Minetti and Reale, 2006; Nagymajtényi et al., 1997; Newland et al., 1986; Petersson Grawé et al., 2004; Rai et al., 2010; Rastogi et al., 1977; Zhao et al., 2018). There is also evidence in rodents that paternal Cd exposure may affect offspring behavior and neurodevelopment (Zhao et al., 2015), although the underlying mechanisms are unclear.

Evidence for Cd-impaired neurodevelopment in animal studies is often highly spatiotemporally dependent. In one study, gestational exposure alone was sufficient to induce neuronal degeneration in the cortex and hippocampus in gestational day (GD)20 fetuses (Allam et al., 2016), while a separate cross-fostering study found that both gestational and lactational exposure was required to elicit morphological changes to the hippocampus (Zhao et al., 2018). Some studies suggest that exposure cessation at birth mitigates adverse effects compared to exposure continuing through lactation (Liapi et al., 2013; Mikolić et al., 2016; Yargiçoğlu et al., 1997; Zhao et al., 2018). Multiple rodent studies found that maternal Cd exposure damaged subpopulations of neuronal cells and altered the architecture of multiple regions of the brain (Allam et al., 2016; Bekheet, 2011; Ben Mimouna et al., 2018b; Feng et al., 2019; Zhao et al., 2018). Hydrocephaly has also been linked to late-gestation Cd injection in rat fetuses (White et al., 1990).

Early-life Cd exposure alters lipid metabolism in ways that increase brain OS (Gupta et al., 1995, 1996; Gutiérrez-Reyes et al., 1998; Xu et al., 1993; Zhang et al., 2009; Zhao et al., 2015) and are more prominent when exposure continues into the postnatal period (Agar et al., 2000; Gulati et al., 1986; Gupta and Shukla, 1996; Yargiçoğlu et al., 1997). Increased ROS and impaired antioxidant response linked to lipid peroxidation are implicated in brain injury, inflammation and cell death (Anthonymuthu et al., 2016) and could be one mechanism through which developmental Cd exposure exerts neurotoxic effects. Concordantly, supplementation of early-life Cd exposure with antioxidants has been neuroprotective (Allam et al., 2016; Bekheet, 2011; Ben Mimouna et al., 2018a,b; Halder et al., 2016, 2019; Mimouna et al., 2018; Mukherjee et al., 2010). Myelination is a unique form of lipid metabolism that is affected by Cd, although it is unclear whether exposure has a stimulatory or suppressive effect (Gupta and Shukla, 1996; Hudson et al., 2021).

Early-life exposure to Cd can affect essential trace metal homeostasis in the brain. Decreases in brain Zn levels in response to Cd may vary with Cd dose, route of exposure and, particularly, age of the individual, as some studies have noted a significant Cd-induced decrease in brain Zn in only a subset of the timepoints measured (Barański, 1986; Gupta et al., 1993; Mikolić et al., 2016). Meanwhile, other studies have observed no significant Cd-induced changes in brain Zn when measured at a single timepoint (Gulati et al., 1986; Sowa and Steibert, 1985). Regardless, supplemental Zn has a neuroprotective effect during developmental Cd exposure (Ben Mimouna et al., 2018a,b; Mimouna et al., 2018). Cu is also essential for brain development and function (Scheiber et al., 2014). Fewer studies have quantified brain Cu in response to developmental Cd exposure; but, in general, a decrease in brain Cu is observed (Barański, 1987; Gulati et al., 1986; Gupta and Shukla, 1996; Mukherjee et al., 2010; Sowa and Steibert, 1985; Thomas and Mushak, 1986). Fe is involved in numerous essential processes, including oxygen homeostasis, myelin production, neurotransmitter synthesis and cellular metabolism (Ward et al., 2014). Fe dysregulation is evident but complex, as animal models have found increases (Mukherjee et al., 2010), decreases (Mikolić et al., 2016) or no change (Gupta and Shukla, 1996; Sowa and Steibert, 1985) in brain Fe following Cd exposure. The brain is the body's most resource-demanding organ and requires Fe to support its high energy and metabolic needs (Ward et al., 2014). Maternal Cd exposure can lead to systemic Fe deficiency at birth (Hudson et al., 2019). In Fe deficiency, oxygen transport and cellular energy production are impaired, potentially leading to hypoxic environments within brain tissue. To compensate, the body may prioritize the brain's Fe, oxygen and nutrient supply through a phenomenon known as ‘brain-sparing’, which frequently coincides with FGR (Cohen et al., 2015).

In concert with perturbed metal homeostasis, developmental Cd exposure increases MT levels in the brain (Ben Mimouna et al., 2018a,b; Gutiérrez-Reyes et al., 1998; Mimouna et al., 2018). Artificially increasing MT levels during Cd exposure in early postnatal rats alleviated Cd-induced increases in striatal (Box 2) lipid peroxidation and dopamine release, possibly by increasing the available MT to bind Cd or Zn (Gutiérrez-Reyes et al., 1998). In support of a protective role for MT, MT-null mice were more sensitive to thyroid enzyme activation in the brain following early-life Cd exposure (Mori et al., 2006).

Rodent models have shown that early-life exposure to Cd can affect the levels of multiple neurotransmitters and hormones. The dopaminergic pathways – which are involved in motivation, reward-seeking behaviors and addiction (Volkow et al., 2017) – are perturbed by developmental Cd exposure, but the effects vary by area of the brain or exposure parameters (Allam et al., 2016; Antonio et al., 1998; Gutiérrez-Reyes et al., 1998; Rastogi et al., 1977). Serotonin signaling is implicated in depression and anxiety disorders, may influence reward and aversion behavior, and is affected by developmental Cd exposure in rodents (Hu, 2016; Szeitz and Bandiera, 2018). Rat pups orally dosed with Cd early in life had increased norepinephrine in multiple regions of the brain (Rastogi et al., 1977). Another study found that mice born to mothers who received Cd injections during gestation had low brain acetylcholine after weaning, which could be partially rescued by antioxidant-rich parsley juice supplementation (Allam et al., 2016). The activity of acetylcholinesterase, the enzyme that breaks down acetylcholine to terminate its neurotransmission, has also been found to be altered by maternal Cd exposure, although studies are inconsistent on whether the enzyme activity is increased or decreased (Liapi et al., 2013; Mukherjee et al., 2010; Stolakis et al., 2013).

Consistent with the classification of Cd as a metallohormone, developmental exposure has been shown to affect sex hormone signaling in the brain (Byrne et al., 2009). Mice born to mothers who ingested a moderate dose of Cd during pregnancy and through the first 10 days of postnatal life had reduced sex hormone receptors in their brains that were receptor and sex specific (Ishitobi et al., 2007). Synthesis of placenta-derived estrogen is impaired by Cd and linked to delays in neuronal development and escape behavior (Liu et al., 2022). Excess glucocorticoids can affect neurodevelopment and behaviors, and Cd increases fetal glucocorticoids and methylation of placental glucocorticoid receptor genes (Appleton et al., 2017). Perinatal Cd exposure has also been found to interfere with thyroid hormone metabolism in neonatal mice with MT deficiency (Mori et al., 2006).

Although less commonly used, non-mammalian and in vitro models provide valuable insight into Cd-induced dysneurogenesis (Chow et al., 2008; Gulisano et al., 2009; Hen Chow et al., 2009; Ohtani-Kaneko et al., 2008). Zebrafish embryos exposed to Cd exhibit reduced motor activity and hypersensitivity to auditory stimuli, and their brains appear smaller than those of zebrafish embryos without Cd exposure, with poorly defined regions (Chow et al., 2008). There is also evidence from zebrafish that implicates the WNT/β-catenin pathway and Ca signaling in impaired neurodevelopment (Green et al., 2023; Xu et al., 2022a,b). Proliferation of primary human fetal neuroblasts is altered in a dose-dependent manner, whereby a low dose of Cd increases growth, but a higher dose reduces proliferation and increases cell death (Gulisano et al., 2009). Meanwhile, primary fetal rat cortical cells exposed to Cd exhibit decreased dendritic and synaptic development, and decreased expression of proteins involved in neuronal development, with no change in cell proliferation (Ohtani-Kaneko et al., 2008). Thus, the effects of Cd on the nervous system are wide ranging and incompletely understood.

Reproductive system

Knowledge of the impacts of developmental Cd exposure on the reproductive system is primarily derived from rodent studies (Table S10). Despite inefficient transfer across the placenta, Cd is detectable in testicular and ovarian tissues following developmental exposure (Bekheet, 2011; Chemek et al., 2016; Li et al., 2018a,b; Zhao et al., 2015). In general, Cd causes a reduction in gonad weight (Chemek et al., 2016; Ji et al., 2011; Liu et al., 2021; Luo et al., 2015; Samuel et al., 2011; Tam and Liu, 1985; Tian et al., 2018), although a handful of studies have shown little to no change (Li et al., 2018b; Luo et al., 2015; Pillai et al., 2012). The direct interaction of Cd with the gonads manifests in histological, morphological or structural changes in several cell types including germ cells, Sertoli cells (Box 2), Leydig cells (Box 2), granulosa cells (Box 2) and spermatogenic cells. One of the more notable effects in male offspring is altered diameter, structure and number of seminiferous tubules (Bekheet, 2011; Chemek et al., 2016; Corpas and Antonio, 1998; Huang et al., 2020; Liu et al., 2021). Production of sperm in the testes occurs in the seminiferous tubules, where disorganization or dysfunction can lead to reduced sperm counts, decreased motility or other sperm abnormalities (Banzato et al., 2012; Huang et al., 2020; Luo et al., 2015; Pillai et al., 2012; Zhang et al., 2019). Interestingly, maternal Zn supplementation during gestation and lactation has been shown to rescue testicular morphology and mitigate sperm abnormalities caused by maternal Cd exposure (Chemek et al., 2018). In female offspring, maternal Cd exposure alters ovarian follicles. Although it is unclear whether folliculogenesis is primarily promoted or impaired (Li et al., 2018b; Samuel et al., 2011), any dysregulation of folliculogenesis or normal follicular function can impact fertility (Chang and Cook-Andersen, 2013).

Developmental Cd exposure also affects reproductive hormone levels in blood serum and gonadal tissues. In males, exposure consistently reduces levels of reproductive hormones, including serum and testicular testosterone, serum gonadotropin-releasing hormone (Box 2), luteinizing hormone (Box 2) and progesterone (Chemek et al., 2016; Ji et al., 2011; Pillai et al., 2012). In females, findings are more variable; serum levels of testosterone, progesterone, estradiol and ovarian progesterone generally decrease following Cd exposure, although some studies report elevated serum estradiol and progesterone (Li et al., 2018b). Differences in serum and tissue hormone levels may be partially explained by changes in the expression of genes and enzymes involved in regulating steroidogenesis (Huang et al., 2020; Ji et al., 2011; Kariyazono et al., 2015; Liu et al., 2020, 2021; Tian et al., 2018), the process by which cholesterol is converted to steroid hormones in the gonads and other endocrine organs (Miller and Auchus, 2011). Notably, a consistent finding across studies is the co-downregulation of steroidogenic acute regulatory protein (StAR), the rate-limiting enzyme in the biosynthesis of steroid hormones, with testosterone and other steroids in both male and female gonads (Manna et al., 2009). One study reporting increased serum steroid hormones in females found a concurrent increase in ovarian StAR enzyme expression (Li et al., 2018b). These results suggest that Cd disrupts reproductive function by altering steroid hormone production.

Some studies showed that F1 females exposed to Cd during development had alterations in the day of vaginal opening: two studies found an earlier onset (Li et al., 2018b; Parodi et al., 2017), and two studies found delayed onset (Salvatori et al., 2004; Samuel et al., 2011), while others found no effect (Ishitobi and Watanabe, 2005; Luo et al., 2015). Despite these variations in the age of vaginal opening, fertility was unaffected. One study even found that exposed female F1 mice had larger litter sizes (Li et al., 2018b). A handful of studies have also shown that Cd exposure in F1 mice has detrimental effects on the reproductive systems of F2 and F3 generations (see Table S10) (Huang et al., 2020; Li et al., 2018b; Liu et al., 2020, 2021), highlighting the persistent intergenerational and transgenerational effects of Cd.

Although reproductive studies typically have focused on maternal exposures, paternal Cd exposure can also affect offspring reproductive health. A recent study in mice found that paternal exposure affected sex hormone synthesis and lipid metabolism in female offspring (Zhang et al., 2023). Whereas one rat study found that paternal Cd exposure negatively affected sperm quality in the male offspring (Zhao et al., 2015), a separate rat study found no changes in paternally exposed testes (Nan et al., 2020), suggesting potential species-, sex- or strain-specific sensitivities to paternal Cd exposure.

Liver

Although animal studies have provided a wealth of evidence supporting a link between developmental Cd exposure and later-onset liver disease, including metabolic dysfunction-associated steatotic liver disease (MASLD; Box 2) (Table S11), few epidemiological studies have directly linked maternal Cd burden with offspring liver function. A handful of studies have examined the effect of maternal tobacco smoking on fetal liver and found molecular changes, but did not implicate Cd itself as a contributing factor (Drake et al., 2015; Filis et al., 2015; O'Shaughnessy et al., 2011).

Numerous rodent studies have shown that mice born to Cd-exposed mothers are deficient in hepatic Zn and Fe (Barański, 1986; Roelfzema et al., 1989; Hudson et al., 2019; Kuriwaki et al., 2005; Mikolić et al., 2016; Pillai and Gupta, 2005; Roelfzema et al., 1987; Sasser et al., 1985; Sorell and Graziano, 1990; Sowa and Steibert, 1985). Studies have also found perturbed homeostasis in other hepatic essential metal ion homeostasis (Hudson et al., 2019) that may be more pronounced in females (Jackson et al., 2022).

Maternal Cd exposure has been shown to disrupt metabolic pathways and functions in the developing liver, affecting insulin regulation (Jackson et al., 2020; Jacquet et al., 2019; Men et al., 2021; Mimouna et al., 2018), metabolism of sugars and glycogen (Roelfzema et al., 1987; Jackson et al., 2020; Pillai and Gupta, 2005; Stewart et al., 1984; Yi et al., 2021; Yoruk et al., 2003), hormone production and regulation (Castillo et al., 2012), and lipid peroxidation and deposition (Jackson et al., 2020; Xu et al., 1993). One study found that maternally exposed male mice had hyperglycemia and enhanced hepatic gluconeogenesis during puberty, the latter of which persisted into adulthood (Yi et al., 2021). Additionally, fetal livers displayed increased OS-related proteins [including NADPH oxidase (NOX)2 (also known as CYBB), NOX4, heme oxygenase-1 (HO-1; also known as HMOX1) and superoxide dismutase (SOD)2)] and SOD activity, and a decrease in glutathione content (Yi et al., 2021). Because OS is a hallmark of metabolic diseases, the markers of OS detected in this study provide evidence that maternal Cd exposure perturbs metabolism during critical windows of liver development. Perinatal Cd exposure has been linked to histological signs of steatosis and fibrosis and increased expression of MASLD-related and imprinted genes, indicating that early-life exposure is sufficient to program MASLD in young animals (Riegl et al., 2023). Another study revealed a sex-specific difference in the effects of maternal Cd exposure on offspring later in life. Although both males and females had dysregulated glucose homeostasis, females, overall, had more severe outcomes, including dyslipidemia, hepatic steatosis, perturbed expression of MASLD-related genes, preneoplastic lesions (Box 2), disrupted retinoic acid signaling, mitochondrial dysfunction, and oxidative and ER stress (Jackson et al., 2020). Finally, the impact of developmental Cd exposure on hepatic glycogen regulation is less clear; one study found that maternally exposed pups had decreased glycogen content in their livers (Pillai and Gupta, 2005), whereas other studies found that liver glycogen content was unaffected by maternal Cd exposure (Roelfzema et al., 1987; Jackson et al., 2020; Yoruk et al., 2003).

Gestational Cd exposure also impacts protein degradation by inducing ubiquitination (Box 2) (Kurita et al., 2016, 2018). One study demonstrated increased abundance of polyubiquitinated protein and decreased monoubiquitinated protein in fetal livers (Kurita et al., 2018). Another showed a decrease in the transcript level of ubiquitin C (Ubc), which encodes a protein that maintains stress-related responses and removes damaged or misfolded proteins (Kurita et al., 2016). Because disturbed expression of Ubc can lead to defective hepatocyte proliferation (Park et al., 2013), these findings present a possible mechanism through which Cd impacts liver development.

Non-mammalian studies have also shown that developmental Cd exposure adversely impacts liver function. In ovo Cd exposure causes significant changes in the size, number and structure of the mitochondria in chicken embryo hepatocytes, which may alter their function and induce OS (Dżugan et al., 2018). Meanwhile, in ovo Cd exposure during organogenesis leads to sinusoidal dilation and necrosis of the liver and causes ruptured cellular membranes, irregular chromatin condensation, and damaged or missing organelles in hepatocytes (Venter et al., 2015).

Maternal Cd exposure may have lasting effects on offspring liver through altered hepatic DNA methylation and expression of DNA methyltransferases (Dnmt genes). In maternally exposed males, livers were found to have hypermethylated glucocorticoid receptor gene promoters and increased transcript abundance of Dnmt3a in the liver, whereas female mice displayed hypomethylation at the hepatic glucocorticoid receptor gene promoter and decreased Dnmt3a expression (Castillo et al., 2012).

Kidney

In humans, the relationship between developmental Cd exposure and kidney health remains poorly understood, and the few studies that do exist examined childhood and adolescent exposure but not the perinatal period. Once Cd enters the body, the kidneys are the primary organs of Cd accumulation, which persists for decades (Genchi et al., 2020). Small amounts of Cd can be detected in fetal kidney tissue (Lutz et al., 1996); therefore, it is plausible that Cd could exert adverse effects on the developing kidney during gestation and early life. One of the few relevant epidemiological studies estimated dietary Cd intake from 1 to 9 years of age, and, at 9 years, a panel of kidney function biomarkers revealed no association between Cd consumption and kidney function (Rodríguez-López et al., 2020).

Rodent studies provide stronger evidence that perinatal Cd exposure disrupts kidney development and function (Table S12). Notably, a majority of these studies do not extensively examine the mechanisms of developmental Cd-induced kidney toxicity but rather measure markers of renal damage or dysfunction. One study found elevated gene expression of kidney injury molecule-1 (Kim-1; also known as Havcr1), a marker of damaged proximal tubule cells (Box 2) (Blum et al., 2015). Another found disorganized expression of tight junction proteins claudin-5 and claudin-2 in the glomerulus (Box 2) and proximal tubule (Jacquillet et al., 2007). This disorganization was associated with increased fractional excretion of Na+, K+, Ca2+, Mg2+ and PO43−, indicating dysfunctional glomerular filtration (Jacquillet et al., 2007). HIF-1 provides a possible mechanism of Cd-induced kidney toxicity, as one study found that gestational Cd exposure reduced the DNA-binding ability of HIF-1 protein in GD21 kidneys, which, if sustained, could harm renal function (Jacobo-Estrada et al., 2018).

There is limited evidence that developmental Cd exposure is able to perturb essential metal homeostasis in the kidneys. Fe and Zn levels were decreased and increased, respectively, in kidneys at postnatal day 21, and the Na/K ratio was decreased in fetal kidneys at GD19 (Mikolić et al., 2016). Patients with chronic kidney disease have essential metal imbalances and are at a greater risk of disease progression if the imbalances are not resolved (Xie et al., 2022), suggesting that Cd-induced changes could exacerbate underlying renal conditions.

Parental Cd burden before, during and after conception renders offspring more vulnerable to poor birth outcomes, organ dysfunction and disease. Epidemiological studies, in which existing Cd burden is quantified in maternal biofluid or tissue, cord blood or placenta, implicate Cd in the etiology of small neonatal size and childhood neurobehavioral delays, including poor cognitive performance, social impairments and emotional challenges (Kippler et al., 2012; Röllin et al., 2015; Sioen et al., 2013; Vidal et al., 2015; Zhang et al., 2004). However, epidemiological studies that link developmental Cd exposure with congenital anomalies, cardiovascular and reproductive outcomes, or liver and kidney problems in humans, are rare and have consistently failed to find evidence of teratogenicity (Calle et al., 2015; Pi et al., 2018). It is still uncertain whether humans are vulnerable to developmental Cd-induced changes in the heart, liver, kidneys or gonads, simply because very few studies have incorporated measurement of both Cd and parameters related to these organ systems. Studies of maternal smoking provide opportunities to examine populations with high Cd burden and have been performed in maternal-child cohorts (Drake et al., 2015; Filis et al., 2015; O'Shaughnessy et al., 2011), but these studies cannot distinguish the impact of Cd alone from other harmful substances in tobacco smoke. Additional constraints and variability in epidemiological study findings stem from differences in methods and biological samples used to quantify Cd burden. For example, animal models suggest that hair Cd levels do not accurately reflect internal concentrations of Cd (Combs et al., 1983; Tete et al., 2014). Quantifying Cd levels in newborn tissues may also not accurately reflect maternal exposure owing to placental accumulation of Cd. Opportunities remain to widen the breadth of epidemiological studies that include measurement of maternal Cd burden.

Findings from experimental studies align with neurodevelopmental and growth impairment observations in humans. In contrast, rodent models have yielded evidence that developmental Cd exposure induces congenital anomalies and changes in the cardiovascular and reproductive systems, despite a lack of such evidence from epidemiological studies. Overall, cardiomegaly is the most commonly observed cardiovascular outcome. Although the reproductive organs exhibit cellular and molecular changes, there is little evidence of infertility linked to developmental Cd exposure. Disparate findings between humans and rodents may be explained by differences in anatomy and physiology, route of administration, form of Cd salt or genetic backgrounds. Rats and mice are the only placental species among the commonly used animal models of developmental exposure; however, their placental structures differ from those of humans and may be more susceptible to Cd damage or transport, which could explain why there are more severe outcomes in rodent offspring. Additionally, Cd doses that are much higher than typically encountered in the environment are often used in toxicological studies to elucidate a mechanism of action and may lead to exaggerated or unexpected consequences, a phenomenon particularly noticeable in the context of congenital anomalies (Adami et al., 2011). Humans are exposed to Cd primarily through ingestion or inhalation (WHO, 2019); therefore, Cd administered to animals through injections may not result in similar mechanisms of absorption and action. However, precisely timed injections might be most suitable when investigating specific developmental stages. In aqueous solution, Cd is in the form of a soluble salt that is converted to free ionic Cd, whereas humans ingest ligand-bound Cd. Additionally, gastrointestinal absorption rates differ between species and depend on factors such as Fe intake, making it difficult to convert Cd intake levels in animals to the amount of equivalent human exposure.

In conclusion, the findings of this Review encourage mitigation of parental Cd exposure during the perinatal period. Owing to overwhelming evidence from both controlled experiments and human populations, we conclude that FGR and neurobehavioral impairments are the outcomes of greatest concern. Further studies of the efficacy of intervention with antioxidant and essential trace metals are warranted, and additional recommendations include more robust investigation of the effects of developmental Cd exposure on reproductive, hepatic and renal function through epidemiological studies with longer routine monitoring.

We thank members of the Cowley Laboratory for helpful discussion on the manuscript.

Funding

This work was supported by the National Institutes of Health under the following grants: R01ES031596, K22ES027510, P30ES025128 and T32ES007046.

Adami
,
H.-O.
,
Berry
,
S. C. L.
,
Breckenridge
,
C. B.
,
Smith
,
L. L.
,
Swenberg
,
J. A.
,
Trichopoulos
,
D.
,
Weiss
,
N. S.
and
Pastoor
,
T. P.
(
2011
).
Toxicology and epidemiology: improving the science with a framework for combining toxicological and epidemiological evidence to establish causal inference
.
Toxicol. Sci.
122
,
223
-
234
.
Adebambo
,
O. A.
,
Shea
,
D.
and
Fry
,
R. C.
(
2018
).
Cadmium disrupts signaling of the hypoxia-inducible (HIF) and transforming growth factor (TGF-beta) pathways in placental JEG-3 trophoblast cells via reactive oxygen species
.
Toxicol. Appl. Pharmacol.
342
,
108
-
115
.
Agar
,
A.
,
Yargiçoğlu
,
P.
,
Izgüt-Uysal
,
V. N.
,
Sentürk
,
U. K.
and
Aktekin
,
B.
(
2000
).
The effect of pre and postnatal cadmium exposure on somatosensory evoked potentials: relation to lipid peroxidation
.
Int. J. Neurosci.
101
,
45
-
56
.
Ahokas
,
R. A.
and
Dilts
,
P. V. J.
(
1979
).
Cadmium uptake by the rat embryo as a function of gestational age
.
Am. J. Obstet. Gynecol.
135
,
219
-
222
.
Akison
,
L. K.
,
Nitert
,
M. D.
,
Clifton
,
V. L.
,
Moritz
,
K. M.
and
Simmons
,
D. G.
(
2017
).
Review: Alterations in placental glycogen deposition in complicated pregnancies: current preclinical and clinical evidence
.
Placenta
54
,
52
-
58
.
Alberts
,
B.
,
Johnson
,
A.
and
Lewis
,
J.
(
2002
).
Molecular Biology of the Cell
, 4th edn.
New York
:
Garland Science
.
Ali
,
M. M.
,
Murthy
,
R. C.
and
Chandra
,
S. V.
(
1986
).
Developmental and longterm neurobehavioral toxicity of low level in-utero cadmium exposure in rats
.
Neurobehav. Toxicol. Teratol.
8
,
463
-
468
.
Allam
,
A. A.
,
Maodaa
,
S. N.
,
Abo-Eleneen
,
R.
and
Ajarem
,
J.
(
2016
).
Protective effect of parsley juice (Petroselinum crispum, Apiaceae) against cadmium deleterious changes in the developed albino mice newborns (Mus musculus) brain
.
Oxid. Med. Cell Longev.
2016
,
2646840
.
Allen
,
D. D.
,
Caviedes
,
R.
,
Cárdenas
,
A. M.
,
Shimahara
,
T.
,
Segura-Aguilar
,
J.
and
Caviedes
,
P. A.
(
2008
).
Cell lines as in vitro models for drug screening and toxicity studies
.
31
,
757
-
768
.
Almenara
,
C. C. P.
,
Broseghini-Filho
,
G. B.
,
Vescovi
,
M. V. A.
,
Angeli
,
J. K.
,
Faria
,
T. O.
,
Stefanon
,
I.
,
Vassallo
,
D. V.
and
Padilha
,
A. S.
(
2013
).
Chronic cadmium treatment promotes oxidative stress and endothelial damage in isolated rat aorta
.
PLoS ONE
8
,
e68418
.
Andresen
,
E.
and
Küpper
,
H.
(
2013
).
Cadmium toxicity in plants
. In
Metal Ions in Life Sciences
.
11
, pp.
395
-
413
.
Anthonymuthu
,
T. S.
,
Kenny
,
E. M.
and
Bayir
,
H.
(
2016
).
Therapies targeting lipid peroxidation in traumatic brain injury
.
Brain Res.
1640
,
57
-
76
.
Antonio
,
M. T.
,
Benito
,
M. J.
,
Leret
,
M. L.
and
Corpas
,
I.
(
1998
).
Gestational administration of cadmium alters the neurotransmitter levels in newborn rat brains
.
J. Appl. Toxicol.
18
,
83
-
88
.
Appleton
,
A. A.
,
Jackson
,
B. P.
,
Karagas
,
M.
and
Marsit
,
C. J.
(
2017
).
Prenatal exposure to neurotoxic metals is associated with increased placental glucocorticoid receptor DNA methylation
.
Epigenetics
12
,
607
-
615
.
Arkwright
,
P. D.
,
Rademacher
,
T. W.
,
Dwek
,
R. A.
and
Redman
,
C. W.
(
1993
).
Pre-eclampsia is associated with an increase in trophoblast glycogen content and glycogen synthase activity, similar to that found in hydatidiform moles
.
J. Clin. Investig.
91
,
2744
-
2753
.
Banzato
,
T. P.
,
Godinho
,
A. F.
,
Da Silva Zacarin
,
E. C. M.
,
Perobelli
,
J. E.
,
Fernandez
,
C. D. B.
,
Favareto
,
A. P. A.
and
Kempinas
,
W. D. G.
(
2012
).
Sperm quality in adult male rats exposed to cadmium in utero and lactation
.
J. Toxicol. Environ. Health A Curr. Issues
75
,
1047
-
1058
.
Barański
,
B.
(
1986
).
Effect of maternal cadmium exposure on postnatal development and tissue cadmium, copper and zinc concentrations in rats
.
Arch. Toxicol.
58
,
255
-
260
.
Barański
,
B.
(
1987
).
Effect of cadmium on prenatal development and on tissue cadmium, copper, and zinc concentrations in rats
.
Environ. Res.
42
,
54
-
62
.
Barański
,
B.
,
Stetkiewicz
,
I.
,
Trzcinka-Ochocka
,
M.
,
Sitarek
,
K.
and
Szymczak
,
W.
(
1982
).
Teratogenicity, fetal toxicity and tissue concentration of cadmium administered to female rats during organogenesis
.
J. Appl. Toxicol.
2
,
255
-
259
.
Barański
,
B.
,
Stetkiewicz
,
I.
,
Sitarek
,
K.
and
Szymczak
,
W.
(
1983
).
Effects of oral, subchronic cadmium administration on fertility, prenatal and postnatal progeny development in rats
.
Arch. Toxicol.
54
,
297
-
302
.
Barjhoux
,
I.
,
Baudrimont
,
M.
,
Morin
,
B.
,
Landi
,
L.
,
Gonzalez
,
P.
and
Cachot
,
J.
(
2012
).
Effects of copper and cadmium spiked-sediments on embryonic development of Japanese medaka (Oryzias latipes)
.
Ecotoxicol. Environ. Saf.
79
,
272
-
282
.
Barrett
,
E. S.
,
Rivera-Núñez
,
Z.
,
Getz
,
K.
,
Ohman-Strickland
,
P.
,
Zhang
,
R.
,
Kozlosky
,
D.
,
Doherty
,
C. L.
,
Buckley
,
B. T.
,
Brunner
,
J.
,
Miller
,
R. K.
et al.
(
2023
).
Protective role of the placental efflux transporter BCRP/ABCG2 in the relationship between prenatal cadmium exposure, placenta weight, and size at birth
.
Environ. Res.
225
,
115597
.
Bekheet
,
S. H. M.
(
2011
).
Comparative effects of repeated administration of cadmium chloride during pregnancy and lactation and selenium protection against cadmium toxicity on some organs in immature rats’ offsprings
.
Biol. Trace Elem. Res.
144
,
1008
-
1023
.
Ben Mimouna
,
S.
,
Boughammoura
,
S.
,
Chemek
,
M.
,
Haouas
,
Z.
,
Banni
,
M.
and
Messaoudi
,
I.
(
2018a
).
Disruption of the zinc metabolism in rat foetal brain after prenatal exposure to cadmium
.
Chem. Biol. Interact.
286
,
88
-
95
.
Ben Mimouna
,
S.
,
Chemek
,
M.
,
Boughammoura
,
S.
,
Haouas
,
Z.
and
Messaoudi
,
I.
(
2018b
).
Protective role of zinc against the neurotoxicity induced by exposure to cadmium during gestation and lactation periods on hippocampal volume of pups tested in early adulthood
.
Drug Chem. Toxicol.
41
,
424
-
433
.
Blum
,
J. L.
,
Edwards
,
J. R.
,
Prozialeck
,
W. C.
,
Xiong
,
J. Q.
and
Zelikoff
,
J. T.
(
2015
).
Effects of maternal exposure to cadmium oxide nanoparticles during pregnancy on maternal and offspring kidney injury markers using a murine model
.
J. Toxicol. Environ. Health A
78
,
711
.
Bonithon-Kopp
,
C.
,
Huel
,
G.
,
Moreau
,
T.
and
Wendling
,
R.
(
1986
).
Prenatal exposure to lead and cadmium and psychomotor development of the child at 6 years
.
Neurobehav. Toxicol. Teratol.
8
,
307
-
310
.
Bosch
,
S.
,
Botha
,
T. L.
and
Wepener
,
V.
(
2023
).
Influence of different functionalized CdTe quantum dots on the accumulation of metals, developmental toxicity and respiration in different development stages of the zebrafish (Danio rerio)
.
Front. Toxicol.
5
,
1176172
.
Brannen
,
K. C.
,
Chapin
,
R. E.
,
Jacobs
,
A. C.
and
Green
,
M. L.
(
2016
).
Alternative models of developmental and reproductive toxicity in pharmaceutical risk assessment and the 3Rs
.
ILAR J.
57
,
144
-
156
.
Brooks
,
S. A.
,
Martin
,
E.
,
Smeester
,
L.
,
Grace
,
M. R.
,
Boggess
,
K.
and
Fry
,
R. C.
(
2016
).
miRNAs as common regulators of the transforming growth factor (TGF)-β pathway in the preeclamptic placenta and cadmium-treated trophoblasts: Links between the environment, the epigenome and preeclampsia
.
Food Chem. Toxicol.
98
,
50
-
57
.
Buck Louis
,
G. M.
and
Platt
,
R. W.
(
2011
).
Introduction to reproductive and perinatal epidemiology
. In
Reproductive and Perinatal Epidemiology
, pp.
3
-
15
.
Oxford University Press
.
Byrne
,
C.
,
Divekar
,
S. D.
,
Storchan
,
G. B.
,
Parodi
,
D. A.
and
Martin
,
M. B.
(
2009
).
Cadmium - a metallohormone
?
Toxicol. Appl. Pharmacol.
238
,
266
-
271
.
Calle
,
J. S.
,
Pachajoa
,
H.
,
Charry
,
J. D.
and
Pachecho
,
R.
(
2015
).
Teratogenic effect of cadmium: from the developing embryo to the fetus
.
Rev. Colombiana Salud Ocup.
5
,
21
-
26
.
Cao
,
Y.
,
Chen
,
A.
,
Radcliffe
,
J.
,
Dietrich
,
K. N.
,
Jones
,
R. L.
,
Caldwell
,
K.
and
Rogan
,
W. J.
(
2009
).
Postnatal cadmium exposure, neurodevelopment, and blood pressure in children at 2, 5, and 7 years of age
.
Environ. Health Perspect.
117
,
1580
-
1586
.
Carter
,
A. M.
(
2020
).
Animal models of human pregnancy and placentation: alternatives to the mouse
.
Reproduction
160
,
R129
-
R143
.
Castillo
,
P.
,
Ibáñez
,
F.
,
Guajardo
,
A.
,
Llanos
,
M. N.
and
Ronco
,
A. M.
(
2012
).
Impact of cadmium exposure during pregnancy on hepatic glucocorticoid receptor methylation and expression in rat fetus
.
PLoS ONE
7
,
e44139
.
Chandravanshi
,
L.
,
Shiv
,
K.
and
Kumar
,
S.
(
2021
).
Developmental toxicity of cadmium in infants and children: a review
.
Environ. Anal. Health Toxicol.
36
,
e2021003-e0
.
Chang
,
R. J.
and
Cook-Andersen
,
H.
(
2013
).
Disordered follicle development
.
Mol. Cell. Endocrinol.
373
,
51
.
Chemek
,
M.
,
Mimouna
,
S. B.
,
Boughammoura
,
S.
,
Delbès
,
G.
and
Messaoudi
,
I.
(
2016
).
Protective role of zinc against the toxicity induced by exposure to cadmium during gestation and lactation on testis development
.
Reprod. Toxicol.
63
,
151
-
160
.
Chemek
,
M.
,
Venditti
,
M.
,
Boughamoura
,
S.
,
Mimouna
,
S. B.
,
Messaoudi
,
I.
and
Minucci
,
S.
(
2018
).
Involvement of testicular DAAM1 expression in zinc protection against cadmium-induced male rat reproductive toxicity
.
J. Cell. Physiol.
233
,
630
-
640
.
Chen
,
H.
,
Boontheung
,
P.
,
Ogorzalek Loo
,
R. R.
,
Xie
,
Y.
,
Loo
,
J. A.
,
Rao
,
J.
and
Collins
,
M. D.
(
2008
).
Proteomic analysis to characterize differential mouse strain sensitivity to cadmium-induced forelimb teratogenesis
.
Birth Defects Res. A Clin. Mol. Teratol
82
,
187
-
199
.
Cheng
,
S. H.
,
Chan
,
P. K.
and
Wu
,
R. S.
(
2001
).
The use of microangiography in detecting aberrant vasculature in zebrafish embryos exposed to cadmium
.
Aquat. Toxicol.
52
,
61
-
71
.
Cheng
,
L.
,
Zhang
,
B.
,
Zheng
,
T.
,
Hu
,
J.
,
Zhou
,
A.
,
Bassig
,
B. A.
,
Xia
,
W.
,
Savitz
,
D. A.
,
Buka
,
S.
,
Xiong
,
C.
et al.
(
2017
).
Critical windows of prenatal exposure to cadmium and size at birth
.
Int. J. Environ. Res. Public Health
14
,
58
.
Chouchene
,
L.
,
Kessabi
,
K.
,
Gueguen
,
M.-M.
,
Kah
,
O.
,
Pakdel
,
F.
and
Messaoudi
,
I.
(
2022
).
Interference with zinc homeostasis and oxidative stress induction as probable mechanisms for cadmium-induced embryo-toxicity in zebrafish
.
Environ. Sci. Pollut. Res.
29
,
39578
-
39592
.
Chow
,
E. S. H.
,
Hui
,
M. N. Y.
,
Lin
,
C. C.
and
Cheng
,
S. H.
(
2008
).
Cadmium inhibits neurogenesis in zebrafish embryonic brain development
.
Aquat. Toxicol.
87
,
157
-
169
.
Ciesielski
,
T.
,
Weuve
,
J.
,
Bellinger
,
D. C.
,
Schwartz
,
J.
,
Lanphear
,
B.
and
Wright
,
R. O.
(
2012
).
Cadmium exposure and neurodevelopmental outcomes in U.S. children
.
Environ. Health Perspect.
120
,
758
-
763
.
Cohen
,
E.
,
Baerts
,
W.
and
Van Bel
,
F.
(
2015
).
Brain-sparing in intrauterine growth restriction: considerations for the neonatologist
.
Neonatology
108
,
269
-
276
.
Combs
,
D. K.
,
Goodrich
,
R. D.
and
Meiske
,
J. C.
(
1983
).
Influence of dietary zinc or cadmium on hair and tissue mineral concentrations in rats and goats
.
J. Anim. Sci.
56
,
184
-
193
.
Corpas
,
I.
and
Antonio
,
M. T.
(
1998
).
Study of alterations produced by cadmium and cadmium/lead administration during gestational and early lactation periods in the reproductive organs of the rat
.
Ecotoxicol. Environ. Saf.
41
,
180
-
188
.
Cowley
,
M.
,
Skaar
,
D. A.
,
Jima
,
D. D.
,
Maguire
,
R. L.
,
Hudson
,
K. M.
,
Park
,
S. S.
,
Sorrow
,
P.
and
Hoyo
,
C.
(
2018
).
Effects of cadmium exposure on DNA methylation at imprinting control regions and genome-wide in mothers and newborn children
.
Environ. Health Perspect.
126
,
037003
.
Coyle
,
P.
,
Philcox
,
J. C.
,
Carey
,
L. C.
and
Rofe
,
A. M.
(
2002
).
Metallothionein: the multipurpose protein
.
Cell. Mol. Life Sci.
59
,
627
-
647
.
Crowe
,
A.
and
Morgan
,
E. H.
(
1997
).
Effect of dietary cadmium on iron metabolism in growing rats
.
Toxicol. Appl. Pharmacol.
145
,
136
-
146
.
Cui
,
Y.
and
Freedman
,
J. H.
(
2009
).
Cadmium induces retinoic acid signaling by regulating retinoic acid metabolic gene expression
.
J. Biol. Chem.
284
,
24925
-
24932
.
Cullen
,
J. T.
and
Maldonado
,
M. T.
(
2013
).
Biogeochemistry of cadmium and its release to the environment
. In
Cadmium: From Toxicity to Essentiality
(ed.
A.
Sigel
,
H.
Sigel
and
R. K.
Sigel
), pp.
31
-
62
.
Dordrecht
:
Springer Netherlands
.
Dal-Medico
,
S. E.
,
Rissoli
,
R. Z.
,
Gamero
,
F. U.
,
Victório
,
J. A.
,
Salla
,
R. F.
,
Abdalla
,
F. C.
,
Silva-Zacarin
,
E. C. M.
,
Carvalho
,
C. S.
and
Costa
,
M. J.
(
2014
).
Negative impact of a cadmium concentration considered environmentally safe in Brazil on the cardiac performance of bullfrog tadpoles
.
Ecotoxicol. Environ. Saf.
104
,
168
-
174
.
de Assis Araujo
,
M. S.
,
Froes-Asmus
,
C. I. R.
,
De Figueiredo
,
N. D.
,
Camara
,
V. M.
,
Luiz
,
R. R.
,
Prata-Barbosa
,
A.
,
Martins
,
M. M.
,
Jacob
,
S. C.
,
Santos
,
L. M. G. D.
,
Vicentini Neto
,
S. A.
et al.
(
2022
).
Prenatal exposure to metals and neurodevelopment in infants at six months: rio birth cohort study of environmental exposure and childhood development (PIPA Project)
.
Int. J. Environ. Res. Public Health
19
,
4295
.
Delaney
,
K. M.
,
Guillet
,
R.
,
Pressman
,
E. K.
,
Caulfield
,
L. E.
,
Zavaleta
,
N.
,
Abrams
,
S. A.
and
O'Brien
,
K. O.
(
2020
).
Iron absorption during pregnancy is underestimated when iron utilization by the placenta and fetus is ignored
.
Am. J. Clin. Nutr.
112
,
576
-
585
.
Dési
,
I.
,
Nagymajtényi
,
L.
and
Schulz
,
H.
(
1998
).
Behavioural and neurotoxicological changes caused by cadmium treatment of rats during development
.
J. Appl. Toxicol.
18
,
63
-
70
.
Di Paola
,
D.
,
Natale
,
S.
,
Iaria
,
C.
,
Crupi
,
R.
,
Cuzzocrea
,
S.
,
Spanò
,
N.
,
Gugliandolo
,
E.
and
Peritore
,
A. F.
(
2022
).
Environmental co-exposure to potassium perchlorate and Cd caused toxicity and thyroid endocrine disruption in zebrafish embryos and larvae (Danio rerio)
.
Toxics
10
,
198
.
Dong
,
X.
,
Ding
,
A.
,
Hu
,
H.
,
Xu
,
F.
,
Liu
,
L.
and
Wu
,
M.
(
2023
).
Placental barrier on cadmium transfer from mother to fetus in related to pregnancy complications
.
Int. J. Womens Health Volume
15
,
179
-
190
.
Drake
,
A. J.
,
O'Shaughnessy
,
P. J.
,
Bhattacharya
,
S.
,
Monteiro
,
A.
,
Kerrigan
,
D.
,
Goetz
,
S.
,
Raab
,
A.
,
Rhind
,
S. M.
,
Sinclair
,
K. D.
,
Meharg
,
A. A.
et al.
(
2015
).
In utero exposure to cigarette chemicals induces sex-specific disruption of one-carbon metabolism and DNA methylation in the human fetal liver
.
BMC Med.
13
,
18
.
Dunne
,
J.
,
Tessema
,
G. A.
,
Ognjenovic
,
M.
and
Pereira
,
G.
(
2021
).
Quantifying the influence of bias in reproductive and perinatal epidemiology through simulation
.
Ann. Epidemiol.
63
,
86
-
101
.
Dutta
,
A.
,
Patra
,
A.
,
Jatav
,
H. S.
,
Jatav
,
S. S.
,
Singh
,
S. K.
,
Sathyanarayana
,
E.
,
Verma
,
S.
,
Singh
,
P.
,
Dutta
,
A.
,
Patra
,
A.
et al.
(
2020
).
Toxicity of cadmium in soil-plant-human continuum and its bioremediation techniques
. In
Soil Contamination - Threats and Sustainable Solutions
(ed.
M. L.
Larramendy
and
S.
Soloneski
), pp.
59
-
80
.
IntechOpen
.
Dżugan
,
M.
,
Trybus
,
W.
,
Lis
,
M.
,
Wesołowska
,
M.
,
Trybus
,
E.
,
Kopacz-Bednarska
,
A.
and
Król
,
T.
(
2018
).
Cadmium-induced ultrastructural changes in primary target organs of developing chicken embryos (Gallus domesticus)
.
J. Trace Elem. Med. Biol.
50
,
167
-
174
.
Elsaid
,
A. F.
,
Délot
,
E. C.
and
Collins
,
M. D.
(
2007
).
Differential perturbation of the Fgf/Erk1/2 and Shh pathways in the C57BL/6N and SWV embryonic limb buds after mid-gestational cadmium chloride administration
.
Mol. Genet. Metab.
92
,
258
-
270
.
Elsaid
,
A. F.
,
Koriem
,
K. M. M.
and
Collins
,
M. D.
(
2010
).
Sensitivity to cadmium-chloride-induced forelimb ectrodactyly is independent of the p53 gene-dosage in the C57BL/6J mouse
.
Birth Defects Res. A
88
,
223
-
227
.
Enli
,
Y.
,
Turgut
,
S.
,
Oztekin
,
O.
,
Demir
,
S.
,
Enli
,
H.
and
Turgut
,
G.
(
2010
).
Cadmium intoxication of pregnant rats and fetuses: interactions of copper supplementation
.
Arch. Med. Res.
41
,
7
-
13
.
Erboga
,
M.
and
Kanter
,
M.
(
2016
).
Effect of cadmium on trophoblast cell proliferation and apoptosis in different gestation periods of rat placenta
.
Biol. Trace Elem. Res.
169
,
285
-
293
.
Everson
,
T. M.
,
Armstrong
,
D. A.
,
Jackson
,
B. P.
,
Green
,
B. B.
,
Karagas
,
M. R.
and
Marsit
,
C. J.
(
2016
).
Maternal cadmium, placental PCDHAC1, and fetal development
.
Reprod. Toxicol.
65
,
263
-
271
.
Everson
,
T. M.
,
Punshon
,
T.
,
Jackson
,
B. P.
,
Hao
,
K.
,
Lambertini
,
L.
,
Chen
,
J.
,
Karagas
,
M. R.
and
Marsit
,
C. J.
(
2018
).
Cadmium-associated differential methylation throughout the placental genome: epigenome-wide association study of two U.S. birth cohorts
.
Environ. Health Perspect.
126
,
17010
.
Feng
,
J.
,
Chen
,
S.
,
Wang
,
Y.
,
Liu
,
Q.
,
Yang
,
M.
,
Li
,
X.
,
Nie
,
C.
,
Qin
,
J.
,
Chen
,
H.
,
Yuan
,
X.
et al.
(
2019
).
Maternal exposure to cadmium impairs cognitive development of male offspring by targeting the Coronin-1a signaling pathway
.
Chemosphere
225
,
765
-
774
.
Fernandez
,
E. L.
,
Gustafson
,
A.-L.
,
Andersson
,
M.
,
Hellman
,
B.
and
Dencker
,
L.
(
2003
).
Cadmium-induced changes in apoptotic gene expression levels and DNA damage in mouse embryos are blocked by zinc
.
Toxicol. Sci.
76
,
162
-
170
.
Filis
,
P.
,
Nagrath
,
N.
,
Fraser
,
M.
,
Hay
,
D. C.
,
Iredale
,
J. P.
,
O'Shaughnessy
,
P.
and
Fowler
,
P. A.
(
2015
).
Maternal smoking dysregulates protein expression in second trimester human fetal livers in a sex-specific manner
.
J. Clin. Endocrinol. Metab.
100
,
E861
-
E870
.
Freire
,
C.
,
Amaya
,
E.
,
Gil
,
F.
,
Fernández
,
M. F.
,
Murcia
,
M.
,
Llop
,
S.
,
Andiarena
,
A.
,
Aurrekoetxea
,
J.
,
Bustamante
,
M.
,
Guxens
,
M.
et al.
(
2018
).
Prenatal co-exposure to neurotoxic metals and neurodevelopment in preschool children: The Environment and Childhood (INMA) Project
.
Sci. Total Environ.
621
,
340
-
351
.
Garí
,
M.
,
Grzesiak
,
M.
,
Krekora
,
M.
,
Kaczmarek
,
P.
,
Jankowska
,
A.
,
Król
,
A.
,
Kaleta
,
D.
,
Jerzyńska
,
J.
,
Janasik
,
B.
,
Kuraś
,
R.
et al.
(
2022
).
Prenatal exposure to neurotoxic metals and micronutrients and neurodevelopmental outcomes in early school age children from Poland
.
Environ. Res.
204
,
112049
.
Genchi
,
G.
,
Sinicropi
,
M. S.
,
Lauria
,
G.
,
Carocci
,
A.
and
Catalano
,
A.
(
2020
).
The effects of cadmium toxicity
.
Int. J. Environ. Res. Public Health
17
,
3782
.
Gheorghescu
,
A.
and
Thompson
,
J.
(
2016
).
Delayed vasculogenesis and impaired angiogenesis due to altered Ang-2 and VE-cadherin levels in the chick embryo model following exposure to cadmium
.
Pediatr. Surg. Int.
32
,
175
-
186
.
Giannakou
,
K.
(
2021
).
Perinatal epidemiology: Issues, challenges, and potential solutions
.
Obstet. Med.
14
,
77
.
Green
,
A. J.
,
Wall
,
A. R.
,
Weeks
,
R. D.
,
Mattingly
,
C. J.
,
Marsden
,
K. C.
and
Planchart
,
A.
(
2023
).
Developmental cadmium exposure disrupts zebrafish vestibular calcium channels interfering with otolith formation and inner ear function
.
Neurotoxicology
96
,
129
-
139
.
Gulati
,
S.
,
Gill
,
K. D.
and
Nath
,
R.
(
1986
).
Effect of cadmium on lipid composition of the weanling rat brain
.
Acta Pharmacol. Toxicol.
59
,
89
-
93
.
Gulisano
,
M.
,
Pacini
,
S.
,
Punzi
,
T.
,
Morucci
,
G.
,
Quagliata
,
S.
,
Delfino
,
G.
,
Sarchielli
,
E.
,
Marini
,
M.
and
Vannelli
,
G. B.
(
2009
).
Cadmium modulates proliferation and differentiation of human neuroblasts
.
J. Neurosci. Res.
87
,
228
-
237
.
Guo
,
M.-Y.
,
Wang
,
H.
,
Chen
,
Y.-H.
,
Xia
,
M.-Z.
,
Zhang
,
C.
and
Xu
,
D.-X.
(
2018
).
N-acetylcysteine alleviates cadmium-induced placental endoplasmic reticulum stress and fetal growth restriction in mice
.
PLoS ONE
13
,
e0191667
.
Gupta
,
A.
and
Shukla
,
G. S.
(
1996
).
Ontogenic profile of brain lipids following perinatal exposure to cadmium
.
J. Appl. Toxicol.
16
,
227
-
233
.
Gupta
,
A. A.
,
Gupta
,
A. A.
and
Chandra
,
S. V.
(
1991
).
Gestational cadmium exposure and brain development: a biochemical study
.
Ind. Health
29
,
65
-
71
.
Gupta
,
A.
,
Gupta
,
A.
,
Murthy
,
R. C.
and
Chandra
,
S. V.
(
1993
).
Neurochemical changes in developing rat brain after pre- and postnatal cadmium exposure
.
Bull. Environ. Contam. Toxicol.
51
,
12
-
17
.
Gupta
,
A.
,
Gupta
,
A.
and
Shukla
,
G. S.
(
1995
).
Development of brain free radical scavenging system and lipid peroxidation under the influence of gestational and lactational cadmium exposure
.
Hum. Exp. Toxicol.
14
,
428
-
433
.
Gupta
,
A.
,
Gupta
,
A.
and
Shukla
,
G. S.
(
1996
).
Maternal cadmium intoxication: effects on fetal brain antioxidant defense parameters
.
Redox Rep.
2
,
199
-
204
.
Gutiérrez-Reyes
,
E. Y.
,
Albores
,
A.
and
Ríos
,
C.
(
1998
).
Increase of striatal dopamine release by cadmium in nursing rats and its prevention by dexamethasone-induced metallothionein
.
Toxicology
131
,
145
-
154
.
Halder
,
S.
,
Kar
,
R.
,
Galav
,
V.
,
Mehta
,
A. K.
,
Bhattacharya
,
S. K.
,
Mediratta
,
P. K.
and
Banerjee
,
B. D.
(
2016
).
Cadmium exposure during lactation causes learning and memory-impairment in F1 generation mice: amelioration by quercetin
.
Drug Chem. Toxicol.
39
,
272
-
278
.
Halder
,
S.
,
Kar
,
R.
,
Chakraborty
,
S.
,
Bhattacharya
,
S. K.
,
Mediratta
,
P. K.
and
Banerjee
,
B. D.
(
2019
).
Cadmium level in brain correlates with memory impairment in F1 and F2 generation mice: improvement with quercetin
.
Environ. Sci. Pollut. Res.
26
,
9632
-
9639
.
Hawkesworth
,
S.
,
Wagatsuma
,
Y.
,
Kippler
,
M.
,
Fulford
,
A. J. C.
,
Arifeen
,
S. E.
,
Persson
,
L.-A.
,
Moore
,
S. E.
and
Vahter
,
M.
(
2013
).
Early exposure to toxic metals has a limited effect on blood pressure or kidney function in later childhood, rural Bangladesh
.
Int. J. Epidemiol.
42
,
176
-
185
.
Hemberger
,
M.
,
Hanna
,
C. W.
and
Dean
,
W.
(
2020
).
Mechanisms of early placental development in mouse and humans
.
Nat. Rev. Genet.
21
,
27
-
43
.
Hen Chow
,
E. S.
,
Yu Hui
,
M. N.
,
Cheng
,
C. W.
and
Cheng
,
S. H.
(
2009
).
Cadmium affects retinogenesis during zebrafish embryonic development
.
Toxicol. Appl. Pharmacol.
235
,
68
-
76
.
Holt
,
D.
and
Webb
,
M.
(
1987
).
Teratogenicity of ionic cadmium in the Wistar rat
.
Arch. Toxicol.
59
,
443
-
447
.
Hu
,
H.
(
2016
).
Reward and aversion
.
Annu. Rev. Neurosci.
39
,
297
-
324
.
Hu
,
J.
,
Wang
,
H.
,
Hu
,
Y.-F.
,
Xu
,
X.-F.
,
Chen
,
Y.-H.
,
Xia
,
M.-Z.
,
Zhang
,
C.
and
Xu
,
D.-X.
(
2018
).
Cadmium induces inflammatory cytokines through activating Akt signaling in mouse placenta and human trophoblast cells
.
Placenta
65
,
7
-
14
.
Huang
,
J.
,
Okuka
,
M.
,
McLean
,
M.
,
Keefe
,
D. L.
and
Liu
,
L.
(
2010
).
Telomere susceptibility to cigarette smoke-induced oxidative damage and chromosomal instability of mouse embryos in vitro
.
Free Radic. Biol. Med.
48
,
1663
-
1676
.
Huang
,
Y.
,
Zhu
,
J.
,
Li
,
H.
,
Wang
,
W.
,
Li
,
Y.
,
Yang
,
X.
,
Zheng
,
N.
,
Liu
,
Q.
,
Zhang
,
Q.
,
Zhang
,
W.
et al.
(
2020
).
Cadmium exposure during prenatal development causes testosterone disruption in multigeneration via SF-1 signaling in rats
.
Food Chem. Toxicol.
135
,
110897
.
Hudson
,
K. M.
,
Belcher
,
S. M.
and
Cowley
,
M.
(
2019
).
Maternal cadmium exposure in the mouse leads to increased heart weight at birth and programs susceptibility to hypertension in adulthood
.
Sci. Rep.
9
,
13553
.
Hudson
,
K. M.
,
Shiver
,
E.
,
Yu
,
J.
,
Mehta
,
S.
,
Jima
,
D. D.
,
Kane
,
M. A.
,
Patisaul
,
H. B.
and
Cowley
,
M.
(
2021
).
Transcriptomic, proteomic, and metabolomic analyses identify candidate pathways linking maternal cadmium exposure to altered neurodevelopment and behavior
.
Sci. Rep.
11
,
16302
.
Idrees
,
N.
,
Tabassum
,
B.
,
Abd_Allah
,
E. F.
,
Hashem
,
A.
,
Sarah
,
R.
and
Hashim
,
M.
(
2018
).
Groundwater contamination with cadmium concentrations in some West U.P. Regions, India
.
Saudi J. Biol. Sci.
25
,
1365
-
1368
.
Il'yasova
,
D.
,
Kloc
,
N.
and
Kinev
,
A.
(
2015
).
Cord blood cells for developmental toxicology and environmental health
.
Front. Public Health
3
,
265
.
Ishitobi
,
H.
and
Watanabe
,
C.
(
2005
).
Effects of low-dose perinatal cadmium exposure on tissue zinc and copper concentrations in neonatal mice and on the reproductive development of female offspring
.
Toxicol. Lett.
159
,
38
-
46
.
Ishitobi
,
H.
,
Mori
,
K.
,
Yoshida
,
K.
and
Watanabe
,
C.
(
2007
).
Effects of perinatal exposure to low-dose cadmium on thyroid hormone-related and sex hormone receptor gene expressions in brain of offspring
.
Neurotoxicology
28
,
790
-
797
.
Jackson
,
T. W.
,
Ryherd
,
G. L.
,
Scheibly
,
C. M.
,
Sasser
,
A. L.
,
Guillette
,
T. C.
and
Belcher
,
S. M.
(
2020
).
Gestational Cd exposure in the CD-1 mouse induces sex-specific hepatic insulin insensitivity, obesity, and metabolic syndrome in adult female offspring
.
Toxicol. Sci.
178
,
264
-
280
.
Jackson
,
T. W.
,
Baars
,
O.
and
Belcher
,
S. M.
(
2022
).
Gestational Cd exposure in the CD-1 mouse sex-specifically disrupts essential metal ion homeostasis
.
Toxicol. Sci.
187
,
254
-
266
.
Jacobo-Estrada
,
T.
,
Cardenas-Gonzalez
,
M.
,
Santoyo-Sánchez
,
M. P.
,
Thevenod
,
F.
and
Barbier
,
O.
(
2018
).
Intrauterine exposure to cadmium reduces HIF-1 DNA-binding ability in rat fetal kidneys
.
Toxics
6
,
53
.
Jacobson
,
K. B.
,
Opresko
,
L.
,
Owenby
,
R. K.
and
Christie
,
N. T.
(
1981
).
Effects of cadmium on drosophila: toxicity, proteins, and transfer RNAs
.
Toxicol. Appl. Pharmacol.
60
,
368
-
378
.
Jacquet
,
A.
,
Barbeau
,
D.
,
Arnaud
,
J.
,
Hijazi
,
S.
,
Hazane-Puch
,
F.
,
Lamarche
,
F.
,
Quiclet
,
C.
,
Couturier
,
K.
,
Fontaine
,
E.
,
Moulis
,
J.-M.
et al.
(
2019
).
Impact of maternal low-level cadmium exposure on glucose and lipid metabolism of the litter at different ages after weaning
.
Chemosphere
219
,
109
-
121
.
Jacquillet
,
G.
,
Barbier
,
O.
,
Rubera
,
I.
,
Tauc
,
M.
,
Borderie
,
A.
,
Namorado
,
M. C.
,
Martin
,
D.
,
Sierra
,
G.
,
Reyes
,
J. L.
,
Poujeol
,
P.
et al.
(
2007
).
Cadmium causes delayed effects on renal function in the offspring of cadmium-contaminated pregnant female rats
.
Am. J. Physiol. Renal Physiol.
293
,
F1450
-
F1460
.
Järup
,
L.
(
2002
).
Cadmium overload and toxicity
.
Nephrol. Dial. Transplant.
17
Suppl. 2
,
35
-
39
.
Jeong
,
K. S.
,
Park
,
H.
,
Ha
,
E.
,
Hong
,
Y.-C.
,
Ha
,
M.
,
Park
,
H.
,
Kim
,
B.-N.
,
Lee
,
B.-E.
,
Lee
,
S.-J.
,
Lee
,
K. Y.
et al.
(
2015
).
Performance IQ in children is associated with blood cadmium concentration in early pregnancy
.
J. Trace Elem. Med. Biol.
30
,
107
-
111
.
Ji
,
Y.-L.
,
Wang
,
H.
,
Liu
,
P.
,
Zhao
,
X.-F.
,
Zhang
,
Y.
,
Wang
,
Q.
,
Zhang
,
H.
,
Zhang
,
C.
,
Duan
,
Z.-H.
,
Meng
,
C.
et al.
(
2011
).
Effects of maternal cadmium exposure during late pregnant period on testicular steroidogenesis in male offspring
.
Toxicol. Lett.
205
,
69
-
78
.
Jin
,
X.
,
Tian
,
X.
,
Liu
,
Z.
,
Hu
,
H.
,
Li
,
X.
,
Deng
,
Y.
,
Li
,
N.
and
Zhu
,
J.
(
2016
).
Maternal exposure to arsenic and cadmium and the risk of congenital heart defects in offspring
.
Reprod. Toxicol.
59
,
109
-
116
.
Jin
,
X.
,
Xu
,
Z.
,
Cao
,
J.
,
Shao
,
P.
,
Zhou
,
M.
,
Qin
,
Z.
,
Liu
,
Y.
,
Yu
,
F.
,
Zhou
,
X.
,
Ji
,
W.
et al.
(
2017
).
Proteomics analysis of human placenta reveals glutathione metabolism dysfunction as the underlying pathogenesis for preeclampsia
.
Biochim. Biophys. Acta Proteins Proteomics
1865
,
1207
-
1214
.
Jolibois
,
L. S. J.
,
Shi
,
W.
,
George
,
W. J.
,
Henson
,
M. C.
and
Anderson
,
M. B.
(
1999a
).
Cadmium accumulation and effects on progesterone release by cultured human trophoblast cells
.
Reprod. Toxicol.
13
,
215
-
221
.
Jolibois
,
L. S. J.
,
Burow
,
M. E.
,
Swan
,
K. F.
,
George
,
W. J.
,
Anderson
,
M. B.
and
Henson
,
M. C.
(
1999b
).
Effects of cadmium cell viability, trophoblastic development, and expression of low density lipoprotein receptor transcripts in cultured human placental cells
.
Reprod. Toxicol.
13
,
473
-
480
.
Kantola
,
M.
,
Purkunen
,
R.
,
Kröger
,
P.
,
Tooming
,
A.
,
Juravskaja
,
J.
,
Pasanen
,
M.
,
Saarikoski
,
S.
and
Vartiainen
,
T.
(
2000
).
Accumulation of cadmium, zinc, and copper in maternal blood and developmental placental tissue: differences between Finland, Estonia, and St. Petersburg
.
Environ. Res.
83
,
54
-
66
.
Kariyazono
,
Y.
,
Taura
,
J.
,
Hattori
,
Y.
,
Ishii
,
Y.
,
Narimatsu
,
S.
,
Fujimura
,
M.
,
Takeda
,
T.
and
Yamada
,
H.
(
2015
).
Effect of in utero exposure to endocrine disruptors on fetal steroidogenesis governed by the pituitary-gonad axis: a study in rats using different ways of administration
.
J. Toxicol. Sci.
40
,
909
-
916
.
Kawai
,
M.
,
Swan
,
K. F.
,
Green
,
A. E.
,
Edwards
,
D. E.
,
Anderson
,
M. B.
and
Henson
,
M. C.
(
2002
).
Placental endocrine disruption induced by cadmium: effects on P450 cholesterol side-chain cleavage and 3beta-hydroxysteroid dehydrogenase enzymes in cultured human trophoblasts
.
Biol. Reprod.
67
,
178
-
183
.
Kim
,
K.
,
Melough
,
M. M.
,
Vance
,
T. M.
,
Noh
,
H.
,
Koo
,
S. I.
and
Chun
,
O. K.
(
2019
).
Dietary cadmium intake and sources in the US
.
Nutrients
11
,
2
.
King-Herbert
,
A. P.
and
Vasbinder
,
M. A.
(
2020
).
Toxicology
. In
The Laboratory Rat
, 3rd edn (ed. M. A. Suckow, F. C. Hankenson, R. P. Wilson and P. L. Foley), pp.
849
-
862
. Academic Press.
Kippler
,
M.
,
Tofail
,
F.
,
Hamadani
,
J. D.
,
Gardner
,
R. M.
,
Grantham-McGregor
,
S. M.
,
Bottai
,
M.
and
Vahter
,
M.
(
2012
).
Early-life cadmium exposure and child development in 5-year-old girls and boys: a cohort study in rural Bangladesh
.
Environ. Health Perspect.
120
,
1462
-
1468
.
Kippler
,
M.
,
Engström
,
K.
,
Mlakar
,
S. J.
,
Bottai
,
M.
,
Ahmed
,
S.
,
Hossain
,
M. B.
,
Raqib
,
R.
,
Vahter
,
M.
and
Broberg
,
K.
(
2013
).
Sex-specific effects of early life cadmium exposure on DNA methylation and implications for birth weight
.
Epigenetics
8
,
494
-
503
.
Kippler
,
M.
,
Bottai
,
M.
,
Georgiou
,
V.
,
Koutra
,
K.
,
Chalkiadaki
,
G.
,
Kampouri
,
M.
,
Kyriklaki
,
A.
,
Vafeiadi
,
M.
,
Fthenou
,
E.
,
Vassilaki
,
M.
et al.
(
2016
).
Impact of prenatal exposure to cadmium on cognitive development at preschool age and the importance of selenium and iodine
.
Eur. J. Epidemiol.
31
,
1123
-
1134
.
Kubier
,
A.
,
Wilkin
,
R. T.
and
Pichler
,
T.
(
2019
).
Cadmium in soils and groundwater: a review
.
Appl. Geochem.
108
,
104388
.
Kurita
,
H.
,
Nagase
,
H.
,
Tokumoto
,
M.
,
Lee
,
J.-Y.
and
Satoh
,
M.
(
2016
).
DNA microarray analysis of fetal liver of C57BL/6J mice exposed to cadmium during gestation
.
Fundam. Toxicol. Sci.
3
,
257
-
280
.
Kurita
,
H.
,
Hasegawa
,
T.
,
Seko
,
Y.
,
Nagase
,
H.
,
Tokumoto
,
M.
,
Lee
,
J.-Y.
and
Satoh
,
M.
(
2018
).
Effect of gestational cadmium exposure on fetal growth, polyubiquitinated protein and monoubiqutin levels in the fetal liver of mice
.
J. Toxicol. Sci.
43
,
19
-
24
.
Kuriwaki
,
J.
,
Nishijo
,
M.
,
Honda
,
R.
,
Tawara
,
K.
,
Nakagawa
,
H.
,
Hori
,
E.
and
Nishijo
,
H.
(
2005
).
Effects of cadmium exposure during pregnancy on trace elements in fetal rat liver and kidney
.
Toxicol. Lett.
156
,
369
-
376
.
Leazer
,
T. M.
,
Liu
,
Y.
and
Klaassen
,
C. D.
(
2002
).
Cadmium absorption and its relationship to divalent metal transporter-1 in the pregnant rat
.
Toxicol. Appl. Pharmacol.
185
,
18
-
24
.
Lee
,
C. K.
,
Lee
,
J. T.
,
Yu
,
S. J.
,
Kang
,
S. G.
,
Moon
,
C. S.
,
Choi
,
Y. H.
,
Kim
,
J. H.
,
Kim
,
D. H.
,
Son
,
B. C.
,
Lee
,
C. H.
et al.
(
2009
).
Effects of cadmium on the expression of placental lactogens and Pit-1 genes in the rat placental trophoblast cells
.
Mol. Cell. Endocrinol.
298
,
11
-
18
.
Lee
,
H. Y.
,
Inselman
,
A. L.
,
Kanungo
,
J.
and
Hansen
,
D. K.
(
2012
).
Alternative models in developmental toxicology
.
58
,
10
-
22
.
Lehotzky
,
K.
,
Ungváry
,
G.
,
Polinák
,
D.
and
Kiss
,
A.
(
1990
).
Behavioral deficits due to prenatal exposure to cadmium chloride in CFY rat pups
.
Neurotoxicol. Teratol.
12
,
169
-
172
.
Leroy
,
M.
and
Allais
,
L.
(
2013
).
Teratology studies in the rat
.
Methods Mol. Biol.
947
,
95
-
109
.
Levin
,
A. A.
,
Plautz
,
J. R.
,
Di Sant'Agnese
,
P. A.
and
Miller
,
R. K.
(
1981
).
Cadmium: placental mechanisms of fetal toxicity
.
Placenta Suppl.
3
,
303
-
318
.
Li
,
X.
,
Liu
,
J.
,
Wu
,
S.
,
Zheng
,
W.
,
Li
,
H.
,
Bao
,
S.
,
Chen
,
Y.
,
Guo
,
X.
,
Zhang
,
L.
and
Ge
,
R.-S.
(
2018a
).
In utero single low-dose exposure of cadmium induces rat fetal Leydig cell dysfunction
.
Chemosphere
194
,
57
-
66
.
Li
,
Z.
,
Li
,
T.
,
Leng
,
Y.
,
Chen
,
S.
,
Liu
,
Q.
,
Feng
,
J.
,
Chen
,
H.
,
Huang
,
Y.
and
Zhang
,
Q.
(
2018b
).
Hormonal changes and folliculogenesis in female offspring of rats exposed to cadmium during gestation and lactation
.
Environ. Pollut.
238
,
336
-
347
.
Liang
,
Y.
,
Young
,
J. L.
,
Kong
,
M.
,
Tong
,
Y.
,
Qian
,
Y.
,
Freedman
,
J. H.
and
Cai
,
L.
(
2019
).
Gender differences in cardiac remodeling induced by a high-fat diet and lifelong, low-dose cadmium exposure
.
Chem. Res. Toxicol.
32
,
1070
-
1081
.
Liapi
,
C.
,
Stolakis
,
V.
,
Zarros
,
A.
,
Zissis
,
K. M.
,
Botis
,
J.
,
Al-Humadi
,
H.
and
Tsakiris
,
S.
(
2013
).
Gestational exposure to cadmium alters crucial offspring rat brain enzyme activities: the role of cadmium-free lactation
.
Environ. Toxicol. Pharmacol.
36
,
835
-
839
.
Lin
,
S.
,
Huo
,
X.
,
Zhang
,
Q.
,
Fan
,
X.
,
Du
,
L.
,
Xu
,
X.
,
Qiu
,
S.
,
Zhang
,
Y.
,
Wang
,
Y.
and
Gu
,
J.
(
2013
).
Short placental telomere was associated with cadmium pollution in an electronic waste recycling town in China
.
PLoS ONE
8
,
e60815
.
Liu
,
L.
,
Zhou
,
L.
,
Hu
,
S.
,
Zhou
,
S.
,
Deng
,
Y.
,
Dong
,
M.
,
Huang
,
J.
,
Zeng
,
Y.
,
Chen
,
X.
,
Zhao
,
N.
et al.
(
2016a
).
Down-regulation of ABCG2 and ABCB4 transporters in the placenta of rats exposed to cadmium
.
Oncotarget
7
,
38154
-
38163
.
Liu
,
L. S.
,
Zhao
,
L. Y.
,
Wang
,
S. H.
and
Jiang
,
J. P.
(
2016b
).
Research proceedings on amphibian model organisms
.
Zool. Res.
37
,
237
-
245
.
Liu
,
J.
,
Zeng
,
L.
,
Zhuang
,
S.
,
Zhang
,
C.
,
Li
,
Y.
,
Zhu
,
J.
and
Zhang
,
W.
(
2020
).
Cadmium exposure during prenatal development causes progesterone disruptors in multiple generations via steroidogenic enzymes in rat ovarian granulosa cells
.
Ecotoxicol. Environ. Saf.
201
,
110765
.
Liu
,
Y.
,
Wen
,
X.
,
Wang
,
D.
and
Liao
,
X.
(
2021
).
Maternal exposure to trace cadmium affects gonadal differentiation and development in male offspring rats though a star pathway
.
Food Sci. Technol.
41
,
635
-
642
.
Liu
,
W.-B.
,
Zhu
,
H.-L.
,
Xiong
,
Y.-W.
,
Lv
,
J.
,
Huang
,
Y.-C.
and
Wang
,
H.
(
2022
).
Environmental cadmium exposure during gestation impairs fetal brain and cognitive function of adult offspring via reducing placenta-derived E2 level
.
Chemosphere
307
,
135668
.
Luo
,
X.
,
Li
,
L.
,
Ma
,
M.
and
Li
,
R.
(
2015
).
Effects of low-dose cadmium exposure during gestation and lactation on development and reproduction in rats
.
Environ. Sci. Pollut. Res.
22
,
10569
-
10579
.
Lutz
,
E.
,
Lind
,
B.
,
Herin
,
P.
,
Krakau
,
I.
,
Bui
,
T. H.
and
Vahter
,
M.
(
1996
).
Concentrations of mercury, cadmium and lead in brain and kidney of second trimester fetuses and infants
.
J. Trace Elem. Med. Biol.
10
,
61
-
67
.
Manigandan
,
K.
,
Jayaraj
,
R. L.
,
Jagatheesh
,
K.
and
Elangovan
,
N.
(
2015
).
Taxifolin mitigates oxidative DNA damage in vitro and protects zebrafish (Danio rerio) embryos against cadmium toxicity
.
Environ. Toxicol. Pharmacol.
39
,
1252
-
1261
.
Manna
,
P. R.
,
Dyson
,
M. T.
and
Stocco
,
D. M.
(
2009
).
Regulation of the steroidogenic acute regulatory protein gene expression: present and future perspectives
.
Mol. Hum. Reprod.
15
,
321
.
Marsden
,
E.
and
Leroy
,
M.
(
2013
).
Teratology studies in the mouse
.
Methods Mol. Biol.
947
,
111
-
123
.
Men
,
H.
,
Young
,
J. L.
,
Zhou
,
W.
,
Zhang
,
H.
,
Wang
,
X.
,
Xu
,
J.
,
Lin
,
Q.
,
Tan
,
Y.
,
Zheng
,
Y.
and
Cai
,
L.
(
2021
).
Early-life exposure to low-dose cadmium accelerates diethylnitrosamine and diet-induced liver cancer
.
Oxid. Med. Cell Longev.
2021
,
1
-
15
.
Mikolić
,
A.
,
Piasek
,
M.
,
Sulimanec Grgec
,
A.
,
Varnai
,
V. M.
,
Stasenko
,
S.
and
Kralik Oguić
,
S.
(
2015
).
Oral cadmium exposure during rat pregnancy: assessment of transplacental micronutrient transport and steroidogenesis at term
.
J. Appl. Toxicol.
35
,
508
-
519
.
Mikolić
,
A.
,
Schönwald
,
N.
and
Piasek
,
M.
(
2016
).
Cadmium, iron and zinc interaction and hematological parameters in rat dams and their offspring
.
J. Trace Elem. Med. Biol.
38
,
108
-
116
.
Miller
,
W. L.
and
Auchus
,
R. J.
(
2011
).
The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders
.
Endocr. Rev.
32
,
81
.
Mimouna
,
S. B.
,
Chemek
,
M.
,
Boughammoura
,
S.
,
Banni
,
M.
and
Messaoudi
,
I.
(
2018
).
Early-life exposure to cadmium triggers distinct Zn-dependent protein expression patterns and impairs brain development
.
Biol. Trace Elem. Res.
184
,
409
-
421
.
Minetti
,
A.
and
Reale
,
C. A.
(
2006
).
Sensorimotor developmental delays and lower anxiety in rats prenatally exposed to cadmium
.
J. Appl. Toxicol.
26
,
35
-
41
.
Mohanty
,
A. F.
,
Farin
,
F. M.
,
Bammler
,
T. K.
,
MacDonald
,
J. W.
,
Afsharinejad
,
Z.
,
Burbacher
,
T. M.
,
Siscovick
,
D. S.
,
Williams
,
M. A.
and
Enquobahrie
,
D. A.
(
2015
).
Infant sex-specific placental cadmium and DNA methylation associations
.
Environ. Res.
138
,
74
-
81
.
Mori
,
K.
,
Yoshida
,
K.
,
Hoshikawa
,
S.
,
Ito
,
S.
,
Yoshida
,
M.
,
Satoh
,
M.
and
Watanabe
,
C.
(
2006
).
Effects of perinatal exposure to low doses of cadmium or methylmercury on thyroid hormone metabolism in metallothionein-deficient mouse neonates
.
Toxicology
228
,
77
-
84
.
Mukherjee
,
R.
,
Desai
,
F.
,
Singh
,
S.
,
Gajaria
,
T.
,
Singh
,
P. K.
,
Baxi
,
D. B.
,
Sharma
,
D.
,
Bhatnagar
,
M.
and
Ramachandran
,
A. V.
(
2010
).
Melatonin protects against alterations in hippocampal cholinergic system, trace metals and oxidative stress induced by gestational and lactational exposure to cadmium
.
EXCLI J.
9
,
119
-
132
.
Murata
,
M.
,
Takigawa
,
H.
and
Sakamoto
,
H.
(
1993
).
Teratogenic effects of noise and cadmium in mice: does noise have teratogenic potential?
J. Toxicol. Environ. Health
39
,
237
-
245
.
Nagaraju
,
R.
,
Kalahasthi
,
R.
,
Balachandar
,
R.
and
Bagepally
,
B. S.
(
2022
).
Cadmium exposure and DNA damage (genotoxicity): a systematic review and meta-analysis
.
Crit. Rev. Toxicol.
52
,
786
-
798
.
Nagymajtényi
,
L.
,
Schulz
,
H.
and
Dési
,
I.
(
1997
).
Behavioural and functional neurotoxicological changes caused by cadmium in a three-generational study in rats
.
Hum. Exp. Toxicol.
16
,
691
-
699
.
Nampoothiri
,
L. P.
and
Gupta
,
S.
(
2008
).
Biochemical effects of gestational coexposure to lead and cadmium on reproductive performance, placenta, and ovary
.
J. Biochem. Mol. Toxicol.
22
,
337
-
344
.
Nan
,
Y.
,
Yi
,
S.-J.
,
Zhu
,
H.-L.
,
Xiong
,
Y.-W.
,
Shi
,
X.-T.
,
Cao
,
X.-L.
,
Zhang
,
C.
,
Gao
,
L.
,
Zhao
,
L.-L.
,
Zhang
,
J.
et al.
(
2020
).
Paternal cadmium exposure increases the susceptibility to diet-induced testicular injury and spermatogenic disorders in mouse offspring
.
Chemosphere
246
,
125776
.
Nardozza
,
L. M. M.
,
Caetano
,
A. C. R.
,
Zamarian
,
A. C. P.
,
Mazzola
,
J. B.
,
Silva
,
C. P.
,
Marçal
,
V. M. G.
,
Lobo
,
T. F.
,
Peixoto
,
A. B.
and
Araujo Júnior
,
E.
(
2017
).
Fetal growth restriction: current knowledge
.
Arch. Gynecol. Obstet.
295
,
1061
-
1077
.
Newland
,
M. C.
,
Ng
,
W. W.
,
Baggs
,
R. B.
,
Gentry
,
G. D.
,
Weiss
,
B.
and
Miller
,
R. K.
(
1986
).
Operant behavior in transition reflects neonatal exposure to cadmium
.
Teratology
34
,
231
-
241
.
Ohtani-Kaneko
,
R.
,
Tazawa
,
H.
,
Yokosuka
,
M.
,
Yoshida
,
M.
,
Satoh
,
M.
and
Watanabe
,
C.
(
2008
).
Suppressive effects of cadmium on neurons and affected proteins in cultured developing cortical cells
.
Toxicology
253
,
110
-
116
.
Olsson
,
I. M.
,
Bensryd
,
I.
,
Lundh
,
T.
,
Ottosson
,
H.
,
Skerfving
,
S.
and
Oskarsson
,
A.
(
2002
).
Cadmium in blood and urine--impact of sex, age, dietary intake, iron status, and former smoking--association of renal effects
.
Environ. Health Perspect.
110
,
1185
.
Omidi
,
M.
,
Niknahad
,
H.
,
Noorafshan
,
A.
,
Fardid
,
R.
,
Nadimi
,
E.
,
Naderi
,
S.
,
Bakhtari
,
A.
and
Mohammadi-Bardbori
,
A.
(
2019
).
Co-exposure to an aryl hydrocarbon receptor endogenous ligand, 6-Formylindolo[3,2-b]carbazole (FICZ), and cadmium induces cardiovascular developmental abnormalities in mice
.
Biol. Trace Elem. Res.
187
,
442
-
451
.
O'Shaughnessy
,
P. J.
,
Monteiro
,
A.
,
Bhattacharya
,
S.
and
Fowler
,
P. A.
(
2011
).
Maternal smoking and fetal sex significantly affect metabolic enzyme expression in the human fetal liver
.
J. Clin. Endocrinol. Metab.
96
,
2851
-
2860
.
Ou
,
Y.
,
Bloom
,
M. S.
,
Nie
,
Z.
,
Han
,
F.
,
Mai
,
J.
,
Chen
,
J.
,
Lin
,
S.
,
Liu
,
X.
and
Zhuang
,
J.
(
2017
).
Associations between toxic and essential trace elements in maternal blood and fetal congenital heart defects
.
Environ. Int.
106
,
127
-
134
.
Padmanabhan
,
R.
(
1987
).
Abnormalities of the ear associated with exencephaly in mouse fetuses induced by maternal exposure to cadmium
.
Teratology
35
,
9
-
18
.
Pamies
,
D.
,
Martínez
,
C. E.
,
Sogorb
,
M. A.
and
Vilanova
,
E.
(
2011
).
Mechanism-based models in reproductive and developmental toxicology
. In
Reproductive and Developmental Toxicology
(ed. R. C. Gupta), pp.
135
-
146
. Academic Press.
Parasuraman
,
S.
(
2011
).
Toxicological screening
.
J. Pharmacol. Pharmacother.
2
,
74
-
79
.
Park
,
H.
,
Yoon
,
M. S.
and
Ryu
,
K. Y.
(
2013
).
Disruption of polyubiquitin gene Ubc leads to defective proliferation of hepatocytes and bipotent fetal liver epithelial progenitor cells
.
Biochem. Biophys. Res. Commun.
435
,
434
-
440
.
Parodi
,
D.
,
Greenfield
,
M.
,
Evans
,
C.
,
Chichura
,
A.
,
Alpaugh
,
A.
,
Williams
,
J.
,
Cyrus
,
K.
and
Martin
,
M.
(
2017
).
Alteration of mammary gland development and gene expression by in utero exposure to cadmium
.
Int. J. Mol. Sci.
18
,
1939
.
Petersson Grawé
,
K.
,
Teiling-Gårdlund
,
A.
,
Jalkesten
,
E.
and
Oskarsson
,
A.
(
2004
).
Increased spontaneous motor activity in offspring after maternal cadmium exposure during lactation
.
Environ. Toxicol. Pharmacol.
17
,
35
-
43
.
Phuapittayalert
,
L.
,
Saenganantakarn
,
P.
,
Supanpaiboon
,
W.
,
Cheunchoojit
,
S.
,
Hipkaeo
,
W.
and
Sakulsak
,
N.
(
2016
).
Increasing CACNA1C expression in placenta containing high Cd level: an implication of Cd toxicity
.
Environ. Sci. Pollut. Res.
23
,
24592
-
24600
.
Pi
,
X.
,
Qiao
,
Y.
,
Wei
,
Y.
,
Jin
,
L.
,
Li
,
Z.
,
Liu
,
J.
,
Zhang
,
Y.
,
Wang
,
L.
,
Liu
,
Y.
,
Xie
,
Q.
et al.
(
2018
).
Concentrations of selected heavy metals in placental tissues and risk for neonatal orofacial clefts
.
Environ. Pollut.
242
,
1652
-
1658
.
Piasek
,
M.
,
Laskey
,
J. W.
,
Kostial
,
K.
and
Blanuša
,
M.
(
2002
).
Assessment of steroid disruption using cultures of whole ovary and/or placenta in rat and in human placental tissue
.
Int. Arch. Occup. Environ. Health
75
Suppl. 1
,
S36
-
S44
.
Pillai
,
A.
and
Gupta
,
S.
(
2005
).
Effect of gestational and lactational exposure to lead and/or cadmium on reproductive performance and hepatic oestradiol metabolising enzymes
.
Toxicol. Lett.
155
,
179
-
186
.
Pillai
,
P.
,
Pandya
,
C.
,
Bhatt
,
N.
and
Gupta
,
S. S.
(
2012
).
Biochemical and reproductive effects of gestational/lactational exposure to lead and cadmium with respect to testicular steroidogenesis, antioxidant system, endogenous sex steroid and cauda-epididymal functions
.
Andrologia
44
,
92
-
101
.
Plöckinger
,
B.
,
Dadak
,
C.
and
Meisinger
,
V.
(
1993
).
[Lead, mercury and cadmium in newborn infants and their mothers]
.
Z. Geburtshilfe Perinatol.
197
,
104
-
107
.
Rai
,
A.
,
Maurya
,
S. K.
,
Khare
,
P.
,
Srivastava
,
A.
and
Bandyopadhyay
,
S.
(
2010
).
Characterization of developmental neurotoxicity of As, Cd, and Pb mixture: synergistic action of metal mixture in glial and neuronal functions
.
Toxicol. Sci.
118
,
586
-
601
.
Rastogi
,
R. B.
,
Merali
,
Z.
and
Singhal
,
R. L.
(
1977
).
Cadmium alters behaviour and the biosynthetic capacity for catecholamines and serotonin in neonatal rat brain
.
J. Neurochem.
28
,
789
-
794
.
Riegl
,
S. D.
,
Starnes
,
C.
,
Jima
,
D. D.
,
Baptissart
,
M.
,
Diehl
,
A. M.
,
Belcher
,
S. M.
and
Cowley
,
M.
(
2023
).
The imprinted gene Zac1 regulates steatosis in developmental cadmium-induced nonalcoholic fatty liver disease
.
Toxicol. Sci.
191
,
34
-
46
.
Robinson
,
J. F.
,
Yu
,
X.
,
Moreira
,
E. G.
,
Hong
,
S.
and
Faustman
,
E. M.
(
2011
).
Arsenic- and cadmium-induced toxicogenomic response in mouse embryos undergoing neurulation
.
Toxicol. Appl. Pharmacol.
250
,
117
-
129
.
Rodríguez-López
,
E.
,
Tamayo-Ortiz
,
M.
,
Ariza
,
A. C.
,
Ortiz-Panozo
,
E.
,
Deierlein
,
A. L.
,
Pantic
,
I.
,
Tolentino
,
M. C.
,
Estrada-Gutiérrez
,
G.
,
Parra-Hernández
,
S.
,
Espejel-Núñez
,
A.
et al.
(
2020
).
Early-life dietary cadmium exposure and kidney function in 9-year-old children from the PROGRESS cohort
.
Toxics
8
,
1
-
11
.
Roelfzema
,
W. H.
,
Roelofsen
,
A. M.
,
Herber
,
R. F.
,
Leene
,
W.
and
Copius Peereboom-Stegeman
,
J. H.
(
1987
).
Effects of chronic cadmium administration on placental and fetal development
.
J. Trace Elem. Electrolytes Health Dis.
1
,
49
-
53
.
Roelfzema
,
W. H.
,
Zahn-Breidenbach
,
U.
and
Copius Peereboom-Stegeman
,
J. H.
(
1988a
).
Light and electron microscopic investigation of the rat placenta after cadmium administration during pregnancy
.
Anat. Embryol.
178
,
345
-
351
.
Roelfzema
,
W. H.
,
Roelofsen
,
A. M.
,
Copius Peereboom-Stegeman
,
J. H.
and
Van Noorden
,
C. J.
(
1988b
).
Glycogen content of placenta and of fetal and maternal liver of cadmium-exposed rats. II: a quantitative histochemical study
.
Placenta
9
,
39
-
45
.
Roelfzema
,
W. H.
,
Hacker
,
H. J.
and
Van Noorden
,
C. J.
(
1989
).
Effects of cadmium exposure on glycogen phosphorylase activity in rat placenta as demonstrated by histochemical means
.
Histochemistry
91
,
305
-
308
.
Roelfzema
,
W. H.
,
Roelofsen
,
A. M.
and
Peereboom-Stegeman
,
J. H. J. C.
(
1987
).
Glycogen content of placenta and of fetal and maternal liver in cadmium-exposed rats. I: a descriptive light microscopic study
.
Placenta
8
,
27
-
36
.
Röllin
,
H. B.
,
Kootbodien
,
T.
,
Channa
,
K.
and
Odland
,
J. Ø.
(
2015
).
Prenatal exposure to cadmium, placental permeability and birth outcomes in coastal populations of South Africa
.
PLoS ONE
10
,
e0142455
.
Ronco
,
A. M.
,
Urrutia
,
M.
,
Montenegro
,
M.
and
Llanos
,
M. N.
(
2009
).
Cadmium exposure during pregnancy reduces birth weight and increases maternal and foetal glucocorticoids
.
Toxicol. Lett.
188
,
186
-
191
.
Ronco
,
A. M.
,
Montenegro
,
M.
,
Castillo
,
P.
,
Urrutia
,
M.
,
Saez
,
D.
,
Hirsch
,
S.
,
Zepeda
,
R.
and
Llanos
,
M. N.
(
2011
).
Maternal exposure to cadmium during gestation perturbs the vascular system of the adult rat offspring
.
Toxicol. Appl. Pharmacol.
251
,
137
-
145
.
Sabra
,
S.
,
Malmqvist
,
E.
,
Saborit
,
A.
,
Gratacós
,
E.
and
Gomez Roig
,
M. D.
(
2017
).
Heavy metals exposure levels and their correlation with different clinical forms of fetal growth restriction
.
PLoS ONE
12
,
e0185645
.
Sakamoto
,
M.
,
Yasutake
,
A.
,
Domingo
,
J. L.
,
Chan
,
H. M.
,
Kubota
,
M.
and
Murata
,
K.
(
2013
).
Relationships between trace element concentrations in chorionic tissue of placenta and umbilical cord tissue: potential use as indicators for prenatal exposure
.
Environ. Int.
60
,
106
-
111
.
Salvatori
,
F.
,
Talassi
,
C. B.
,
Salzgeber
,
S. A.
,
Spinosa
,
H. S.
and
Bernardi
,
M. M.
(
2004
).
Embryotoxic and long-term effects of cadmium exposure during embryogenesis in rats
.
Neurotoxicol. Teratol.
26
,
673
-
680
.
Samarawickrama
,
G. P.
and
Webb
,
M.
(
1979
).
Acute effects of cadmium on the pregnant rat and embryo-fetal development
.
Environ. Health Perspect.
28
,
245
-
249
.
Samuel
,
J. B.
,
Stanley
,
J. A.
,
Princess
,
R. A.
,
Shanthi
,
P.
and
Sebastian
,
M. S.
(
2011
).
Gestational cadmium exposure-induced ovotoxicity delays puberty through oxidative stress and impaired steroid hormone levels
.
J. Med. Toxicol.
7
,
195
-
204
.
Sanders
,
A. P.
,
Desrosiers
,
T. A.
,
Warren
,
J. L.
,
Herring
,
A. H.
,
Enright
,
D.
,
Olshan
,
A. F.
,
Meyer
,
R. E.
and
Fry
,
R. C.
(
2014
).
Association between arsenic, cadmium, manganese, and lead levels in private wells and birth defects prevalence in North Carolina: a semi-ecologic study
.
BMC Public Health
14
,
955
.
Sasser
,
L. B.
,
Kelman
,
B. J.
,
Levin
,
A. A.
and
Miller
,
R. K.
(
1985
).
The influence of maternal cadmium exposure or fetal cadmium injection on hepatic metallothionein concentrations in the fetal rat
.
Toxicol. Appl. Pharmacol.
80
,
299
-
307
.
Satarug
,
S.
,
Baker
,
J. R.
,
Urbenjapol
,
S.
,
Haswell-Elkins
,
M.
,
Reilly
,
P. E. B.
,
Williams
,
D. J.
and
Moore
,
M. R.
(
2003
).
A global perspective on cadmium pollution and toxicity in non-occupationally exposed population
.
Toxicol. Lett.
137
,
65
-
83
.
Satarug
,
S.
,
Garrett
,
S. H.
,
Sens
,
M. A.
and
Sens
,
D. A.
(
2010
).
Cadmium, environmental exposure, and health outcomes
.
Environ. Health Perspect.
118
,
182
-
190
.
Saxena
,
D. K.
,
Murthy
,
R. C.
and
Chandra
,
S. V.
(
1986
).
Embryotoxic and teratogenic effects of interaction of cadmium and lindane in rats
.
Acta Pharmacol. Toxicol.
59
,
175
-
178
.
Scheiber
,
I. F.
,
Mercer
,
J. F. B.
and
Dringen
,
R.
(
2014
).
Metabolism and functions of copper in brain
.
Prog. Neurobiol.
116
,
33
-
57
.
Schreiner
,
C. M.
,
Bell
,
S. M.
and
Scott
,
W. J. J.
(
2009
).
Microarray analysis of murine limb bud ectoderm and mesoderm after exposure to cadmium or acetazolamide
.
Birth Defects Res. A
85
,
588
-
598
.
Scott
,
W. J. J.
,
Schreiner
,
C. M.
,
Goetz
,
J. A.
,
Robbins
,
D.
and
Bell
,
S. M.
(
2005
).
Cadmium-induced postaxial forelimb ectrodactyly: association with altered sonic hedgehog signaling
.
Reprod. Toxicol.
19
,
479
-
485
.
Selvaratnam
,
J.
,
Guan
,
H.
,
Koropatnick
,
J.
and
Yang
,
K.
(
2013
).
Metallothionein-I- and -II-deficient mice display increased susceptibility to cadmium-induced fetal growth restriction
.
Am. J. Physiol. Endocrinol. Metab.
305
,
E727
-
E735
.
Simmers
,
M. D.
,
Hudson
,
K. M.
,
Baptissart
,
M.
and
Cowley
,
M.
(
2023
).
Epigenetic control of the imprinted growth regulator Cdkn1c in cadmium-induced placental dysfunction
.
Epigenetics
18
,
2088173
.
Sioen
,
I.
,
Den Hond
,
E.
,
Nelen
,
V.
,
Van De Mieroop
,
E.
,
Croes
,
K.
,
Van Larebeke
,
N.
,
Nawrot
,
T. S.
and
Schoeters
,
G.
(
2013
).
Prenatal exposure to environmental contaminants and behavioural problems at age 7–8 years
.
Environ. Int.
59
,
225
-
231
.
Skipper
,
A.
,
Sims
,
J. N.
,
Yedjou
,
C. G.
and
Tchounwou
,
P. B.
(
2016
).
Cadmium chloride induces DNA damage and apoptosis of human liver carcinoma cells via oxidative stress
.
Int. J. Environ. Res. Public Health
13
,
88
.
Somsuan
,
K.
,
Phuapittayalert
,
L.
,
Srithongchai
,
Y.
,
Sonthi
,
P.
,
Supanpaiboon
,
W.
,
Hipkaeo
,
W.
and
Sakulsak
,
N.
(
2019
).
Increased DMT-1 expression in placentas of women living in high-Cd-contaminated areas of Thailand
.
Environ. Sci. Pollut. Res. Int.
26
,
141
-
151
.
Sonawane
,
B. R.
,
Nordberg
,
M.
,
Nordberg
,
G. F.
and
Lucier
,
G. W.
(
1975
).
Placental transfer of cadmium in rats: influence of dose and gestational age
.
Environ. Health Perspect.
12
,
97
-
102
.
Song
,
J. W.
and
Chung
,
K. C.
(
2010
).
Observational studies: cohort and case-control studies
.
Plast. Reconstr. Surg.
126
,
2234
.
Sorell
,
T. L.
and
Graziano
,
J. H.
(
1990
).
Effect of oral cadmium exposure during pregnancy on maternal and fetal zinc metabolism in the rat
.
Toxicol. Appl. Pharmacol.
102
,
537
-
545
.
Sowa
,
B.
and
Steibert
,
E.
(
1985
).
Effect of oral cadmium administration to female rats during pregnancy on zinc, copper, and iron content in placenta, foetal liver, kidney, intestine, and brain
.
Arch. Toxicol.
56
,
256
-
262
.
Stark
,
M. R.
and
Ross
,
M. M.
(
2019
).
The chicken embryo as a model in developmental toxicology
.
Methods Mol. Biol.
1965
,
155
-
171
.
Stewart
,
J. L.
,
Newhouse
,
C. L.
,
Wagner
,
M. V.
and
Bradshaw
,
W. S.
(
1984
).
The effects of prenatal exposure to structurally diverse chemicals on the ontogeny of rat dehydrogenases
.
Neonatology
46
,
69
-
79
.
Stolakis
,
V.
,
Tsakiris
,
S.
,
Kalafatakis
,
K.
,
Zarros
,
A.
,
Skandali
,
N.
,
Gkanti
,
V.
,
Kyriakaki
,
A.
and
Liapi
,
C.
(
2013
).
Developmental neurotoxicity of cadmium on enzyme activities of crucial offspring rat brain regions
.
Biometals
26
,
1013
-
1021
.
Szeitz
,
A.
and
Bandiera
,
S. M.
(
2018
).
Analysis and measurement of serotonin
.
Biomed. Chromatogr.
32
.
Tam
,
P. P. L.
and
Liu
,
W. K.
(
1985
).
Gonadal development and fertility of mice treated prenatally with cadmium during the early organogenesis stages
.
Teratology
32
,
453
-
462
.
Taylor
,
C. M.
,
Golding
,
J.
and
Emond
,
A. M.
(
2016
).
Moderate prenatal cadmium exposure and adverse birth outcomes: a role for sex-specific differences?
Paediatr. Perinat. Epidemiol.
30
,
603
-
611
.
Tete
,
N.
,
Afonso
,
E.
,
Crini
,
N.
,
Drouhot
,
S.
,
Prudent
,
A.-S.
and
Scheifler
,
R.
(
2014
).
Hair as a noninvasive tool for risk assessment: do the concentrations of cadmium and lead in the hair of wood mice (Apodemus sylvaticus) reflect internal concentrations?
Ecotoxicol. Environ. Saf.
108
,
233
-
241
.
Thévenod
,
F.
and
Lee
,
W.-K.
(
2013
).
Toxicology of cadmium and its damage to mammalian organs
.
Met. Ions Life Sci.
11
,
415
-
490
.
Thomas
,
D. J.
and
Mushak
,
P.
(
1986
).
Effects of cadmium exposure on zinc and copper distribution in neonatal rats
.
Arch. Toxicol.
58
,
130
-
135
.
Thompson
,
J.
and
Bannigan
,
J.
(
2001
).
Effects of cadmium on formation of the ventral body wall in chick embryos and their prevention by zinc pretreatment
.
Teratology
64
,
87
-
97
.
Thompson
,
J.
and
Bannigan
,
J.
(
2008
).
Cadmium: toxic effects on the reproductive system and the embryo
.
Reprod. Toxicol.
25
,
304
-
315
.
Tian
,
L. L.
,
Zhao
,
Y. C.
,
Wang
,
X. C.
,
Gu
,
J. L.
,
Sun
,
Z. J.
,
Zhang
,
Y. L.
and
Wang
,
J. X.
(
2009
).
Effects of gestational cadmium exposure on pregnancy outcome and development in the offspring at age 4.5 years
.
Biol. Trace Elem. Res.
132
,
51
-
59
.
Tian
,
H.
,
Chen
,
S.
,
Leng
,
Y.
,
Li
,
T.
,
Li
,
Z.
,
Chen
,
H.
and
Zhang
,
Q.
(
2018
).
Exposure to cadmium during gestation and lactation affects development and function of Leydig cells in male offspring
.
Environ. Toxicol.
33
,
351
-
360
.
Trevors
,
J. T.
,
Stratton
,
G. W.
and
Gadd
,
G. M.
(
1986
).
Cadmium transport, resistance, and toxicity in bacteria, algae, and fungi
.
Can. J. Microbiol.
32
,
447
-
464
.
Tsuji
,
M.
,
Shibata
,
E.
,
Askew
,
D. J.
,
Morokuma
,
S.
,
Aiko
,
Y.
,
Senju
,
A.
,
Araki
,
S.
,
Sanefuji
,
M.
,
Ishihara
,
Y.
,
Tanaka
,
R.
et al.
(
2019
).
Associations between metal concentrations in whole blood and placenta previa and placenta accreta: the Japan Environment and Children's Study (JECS)
.
Environ. Health Prev. Med.
24
,
1
-
10
.
Vahter
,
M.
,
Berglund
,
M.
,
Åkesson
,
A.
and
Lidén
,
C.
(
2002
).
Metals and women's health
.
Environ. Res.
88
,
145
-
155
.
Veldman
,
M. B.
and
Lin
,
S.
(
2008
).
Zebrafish as a developmental model organism for pediatric research
.
Pediatr. Res.
64
,
470
-
476
.
Venter
,
C.
,
Oberholzer
,
H. M.
,
Taute
,
H.
,
Cummings
,
F. R.
and
Bester
,
M. J.
(
2015
).
An in ovo investigation into the hepatotoxicity of cadmium and chromium evaluated with light- and transmission electron microscopy and electron energy-loss spectroscopy
.
J. Environ. Sci. Health A Tox. Hazard. Subst. Environ. Eng.
50
,
830
-
838
.
Vidal
,
A. C.
,
Semenova
,
V.
,
Darrah
,
T.
,
Vengosh
,
A.
,
Huang
,
Z.
,
King
,
K.
,
Nye
,
M. D.
,
Fry
,
R.
,
Skaar
,
D.
,
Maguire
,
R.
et al.
(
2015
).
Maternal cadmium, iron and zinc levels, DNA methylation and birth weight
.
BMC Pharmacol. Toxicol.
16
,
20
.
Vilahur
,
N.
,
Vahter
,
M.
and
Broberg
,
K.
(
2015
).
The epigenetic effects of prenatal cadmium exposure
.
Curr. Environ. Health Rep.
2
,
195
-
203
.
Volkow
,
N. D.
,
Wise
,
R. A.
and
Baler
,
R.
(
2017
).
The dopamine motive system: implications for drug and food addiction
.
Nat. Rev. Neurosci.
18
,
741
-
752
.
Waalkes
,
M. P.
(
2003
).
Cadmium carcinogenesis
.
Mutat. Res. Fund. Mol. Mech. Mutagen.
533
,
107
-
120
.
Waalkes
,
M. P.
and
Goering
,
P. L.
(
1990
).
Metallothionein and other cadmium-binding proteins: recent developments
.
Res. Toxicol.
3
,
281
-
288
.
Wadhwa
,
P.
,
Buss
,
C.
,
Entringer
,
S.
and
Swanson
,
J.
(
2009
).
Developmental origins of health and disease: brief history of the approach and current focus on epigenetic mechanisms
.
Semin. Reprod. Med.
27
,
358
-
368
.
Wang
,
Z.
,
Wang
,
H.
,
Xu
,
Z. M.
,
Ji
,
Y.-L.
,
Chen
,
Y.-H.
,
Zhang
,
Z.-H.
,
Zhang
,
C.
,
Meng
,
X.-H.
,
Zhao
,
M.
and
Xu
,
D.-X.
(
2012
).
Cadmium-induced teratogenicity: association with ROS-mediated endoplasmic reticulum stress in placenta
.
Toxicol. Appl. Pharmacol.
259
,
236
-
247
.
Wang
,
F.
,
Zhang
,
Q.
,
Zhang
,
X.
,
Luo
,
S.
,
Ye
,
D.
,
Guo
,
Y.
,
Chen
,
S.
and
Huang
,
Y.
(
2014
).
Preeclampsia induced by cadmium in rats is related to abnormal local glucocorticoid synthesis in placenta
.
Reprod. Biol. Endocrinol.
12
,
77
.
Wang
,
Y.
,
Chen
,
L.
,
Gao
,
Y.
,
Zhang
,
Y.
,
Wang
,
C.
,
Zhou
,
Y.
,
Hu
,
Y.
,
Shi
,
R.
and
Tian
,
Y.
(
2016a
).
Effects of prenatal exposure to cadmium on neurodevelopment of infants in Shandong, China
.
Environ. Pollut.
211
,
67
-
73
.
Wang
,
H.
,
Wang
,
Y.
,
Bo
,
Q.-L.
,
Ji
,
Y.-L.
,
Liu
,
L.
,
Hu
,
Y.-F.
,
Chen
,
Y.-H.
,
Zhang
,
J.
,
Zhao
,
L.-L.
and
Xu
,
D.-X.
(
2016b
).
Maternal cadmium exposure reduces placental zinc transport and induces fetal growth restriction in mice
.
Reprod. Toxicol.
63
,
174
-
182
.
Wang
,
F.
,
Fan
,
F.
,
Wang
,
L.
,
Ye
,
W.
,
Zhang
,
Q.
and
Xie
,
S.
(
2018
).
Maternal cadmium levels during pregnancy and the relationship with preeclampsia and fetal biometric parameters
.
Biol. Trace Elem. Res.
186
,
322
-
329
.
Ward
,
R. J.
,
Zucca
,
F. A.
,
Duyn
,
J. H.
,
Crichton
,
R. R.
and
Zecca
,
L.
(
2014
).
The role of iron in brain ageing and neurodegenerative disorders
.
Lancet Neurol.
13
,
1045
-
1060
.
Wardell
,
R. E.
,
Seegmiller
,
R. E.
and
Bradshaw
,
W. S.
(
1982
).
Induction of prenatal toxicity in the rat by diethylstilbestrol, zeranol, 3,4,3′,4′,–tetrachlorobiphenyl, cadmium, and lead
.
Teratology
26
,
229
-
237
.
Webb
,
M.
,
Holt
,
D.
,
Brown
,
N.
and
Hard
,
G. C.
(
1988
).
The teratogenicity of cadmium-metallothionein in the rat
.
Arch. Toxicol.
61
,
457
-
467
.
Webster
,
W. S.
and
Messerle
,
K.
(
1980
).
Changes in the mouse neuroepithelium associated with cadmium-induced neural tube defects
.
Teratology
21
,
79
-
88
.
Wen
,
X.
,
Kozlosky
,
D.
,
Zhang
,
R.
,
Doherty
,
C.
,
Buckley
,
B.
,
Barrett
,
E.
and
Aleksunes
,
L. M.
(
2021
).
BCRP/ABCG2 transporter regulates accumulation of cadmium in kidney cells: role of the Q141K variant in modulating nephrotoxicity
.
Drug Metab. Dispos.
49
,
629
-
637
.
White
,
T. E. K.
,
Baggs
,
R. B.
and
Miller
,
R. K.
(
1990
).
Central nervous system lesions in the wistar rat fetus following direct fetal injections of cadmium
.
Teratology
42
,
7
-
13
.
Wier
,
P. J.
,
Miller
,
R. K.
,
Maulik
,
D.
and
Disant'agnese
,
P. A.
(
1990
).
Toxicity of cadmium in the perfused human placenta
.
Toxicol. Appl. Pharmacol.
105
,
156
-
171
.
Winn
,
L. M.
(
2019
).
In vitro models in developmental toxicology
.
Methods Mol. Biol.
1965
,
1
-
6
.
Wu
,
F.
,
Tian
,
F.-J.
and
Lin
,
Y.
(
2015
).
Oxidative stress in placenta: health and diseases
.
Biomed. Res. Int.
2015
,
293271
.
Wu
,
X.
,
Chen
,
Y.
,
Luz
,
A.
,
Hu
,
G.
and
Tokar
,
E. J.
(
2022
).
Cardiac development in the presence of cadmium: an in vitro study using human embryonic stem cells and cardiac organoids
.
Environ. Health Perspect.
130
,
117002
.
Wyrwoll
,
C. S.
and
Holmes
,
M. C.
(
2012
).
Prenatal excess glucocorticoid exposure and adult affective disorders: a role for serotonergic and catecholamine pathways
.
Neuroendocrinology
95
,
47
-
55
.
Xie
,
Y.
,
Liu
,
F.
,
Zhang
,
X.
,
Jin
,
Y.
,
Li
,
Q.
,
Shen
,
H.
,
Fu
,
H.
and
Mao
,
J.
(
2022
).
Benefits and risks of essential trace elements in chronic kidney disease: a narrative review
.
Ann. Transl. Med.
10
,
1400
-
1400
.
Xu
,
Y.
and
Morel
,
F. M. M.
(
2013
).
Cadmium in marine phytoplankton
. In
Cadmium: From Toxicity to Essentiality. Metal Ions in Life Sciences
(ed. A. Sigel, H. Sigel and R. Sigel),
11
, pp.
509
-
528
.
Xu
,
B.
,
Jin
,
Y.
,
Feng
,
Z.
,
Xu
,
Z.
and
Matsushita
,
T.
(
1993
).
Lipid peroxidation induced by maternal cadmium exposure in mouse pups
.
Bull. Environ. Contam. Toxicol.
51
,
772
-
779
.
Xu
,
P.
,
Wu
,
Z.
,
Xi
,
Y.
and
Wang
,
L.
(
2016
).
Epigenetic regulation of placental glucose transporters mediates maternal cadmium-induced fetal growth restriction
.
Toxicology
372
,
34
-
41
.
Xu
,
Y.
,
Zhao
,
H.
,
Wang
,
Z.
,
Gao
,
H.
,
Liu
,
J.
,
Li
,
K.
,
Song
,
Z.
,
Yuan
,
C.
,
Lan
,
X.
,
Pan
,
C.
et al.
(
2022a
).
Developmental exposure to environmental levels of cadmium induces neurotoxicity and activates microglia in zebrafish larvae: From the perspectives of neurobehavior and neuroimaging
.
Chemosphere
291
,
132802
.
Xu
,
Y.
,
Liu
,
J.
,
Tian
,
Y.
,
Wang
,
Z.
,
Song
,
Z.
,
Li
,
K.
,
Zhang
,
S.
and
Zhao
,
H.
(
2022b
).
Wnt/β-catenin signaling pathway is strongly implicated in cadmium-induced developmental neurotoxicity and neuroinflammation: clues from zebrafish neurobehavior and in vivo neuroimaging
.
Int. J. Mol. Sci.
23
,
11434
.
Yamagishi
,
Y.
,
Furukawa
,
S.
,
Tanaka
,
A.
,
Kobayashi
,
Y.
and
Sugiyama
,
A.
(
2016
).
Histopathological localization of cadmium in rat placenta by LA-ICP-MS analysis
.
J. Toxicol. Pathol.
29
,
279
-
283
.
Yang
,
K.
,
Julan
,
L.
,
Rubio
,
F.
,
Sharma
,
A.
and
Guan
,
H.
(
2006
).
Cadmium reduces 11β-hydroxysteroid dehydrogenase type 2 activity and expression in human placental trophoblast cells
.
Am. J. Physiol. Endocrinol. Metab.
290
,
E135
-
E142
.
Yargiçoğlu
,
P.
,
Ağar
,
A.
,
Oğuz
,
Y.
,
Izgüt-Uysal
,
V. N.
,
Sentürk
,
U. K.
and
Oner
,
G.
(
1997
).
The effect of developmental exposure to cadmium (Cd) on visual evoked potentials (VEPs) and lipid peroxidation
.
Neurotoxicol. Teratol.
19
,
213
-
219
.
Yi
,
S.-J.
,
Xiong
,
Y.-W.
,
Zhu
,
H.-L.
,
Dai
,
L.-M.
,
Cao
,
X.-L.
,
Liu
,
W.-B.
,
Shi
,
X.-T.
,
Zhou
,
G.-X.
,
Liu
,
A.-Y.
,
Zhao
,
L.-L.
et al.
(
2021
).
Environmental cadmium exposure during pregnancy causes diabetes-like phenotypes in mouse offspring: association with oxidative stress in the fetal liver
.
Sci. Total Environ.
777
,
146006
.
Yoruk
,
M.
,
Kanter
,
M.
,
Meral
,
I.
and
Agaoglu
,
Z.
(
2003
).
Localization of glycogen in the placenta and fetal and maternal livers of cadmium-exposed diabetic pregnant rats
.
Biol. Trace Elem. Res.
96
,
217
-
226
.
Zepeda
,
R.
,
Castillo
,
P.
,
Sáez
,
D.
,
Llanos
,
M. N.
and
Ronco
,
A. M.
(
2012
).
Cardiac tissue injury resistance during myocardial infarction at adulthood by developmental exposure to cadmium
.
Cardiovasc. Toxicol.
12
,
64
-
72
.
Zhang
,
Y.-L.
,
Zhao
,
Y.-C.
,
Wang
,
J.-X.
,
Zhu
,
H.-D.
,
Liu
,
Q.-F.
,
Fan
,
Y.-G.
,
Wang
,
N.-F.
,
Zhao
,
J.-H.
,
Liu
,
H.-S.
,
Ou-Yang
,
L.
et al.
(
2004
).
Effect of environmental exposure to cadmium on pregnancy outcome and fetal growth: a study on healthy pregnant women in China
.
J. Environ. Sci. Health A Tox. Hazard. Subst. Environ. Eng.
39
,
2507
-
2515
.
Zhang
,
Y.-M.
,
Liu
,
X.-Z.
,
Lu
,
H.
,
Mei
,
L.
and
Liu
,
Z.-P.
(
2009
).
Lipid peroxidation and ultrastructural modifications in brain after perinatal exposure to lead and/or cadmium in rat pups
.
Biomed. Environ. Sci.
22
,
423
-
429
.
Zhang
,
W.
,
Lin
,
K.
,
Miao
,
Y.
,
Dong
,
Q.
,
Huang
,
C.
,
Wang
,
H.
,
Guo
,
M.
and
Cui
,
X.
(
2012
).
Toxicity assessment of zebrafish following exposure to CdTe QDs
.
J. Hazard. Mater.
213-214
,
413
-
420
.
Zhang
,
T.
,
Zhou
,
X.-Y.
,
Ma
,
X.-F.
and
Liu
,
J.-X.
(
2015
).
Mechanisms of cadmium-caused eye hypoplasia and hypopigmentation in zebrafish embryos
.
Aquat. Toxicol.
167
,
68
-
76
.
Zhang
,
X.
,
Xu
,
Z.
,
Lin
,
F.
,
Wang
,
F.
,
Ye
,
D.
and
Huang
,
Y.
(
2016
).
Increased oxidative DNA damage in placenta contributes to cadmium-induced preeclamptic conditions in rat
.
Biol. Trace Elem. Res.
170
,
119
-
127
.
Zhang
,
Y.
,
Xu
,
X.
,
Chen
,
A.
,
Davuljigari
,
C. B.
,
Zheng
,
X.
,
Kim
,
S. S.
,
Dietrich
,
K. N.
,
Ho
,
S.-M.
,
Reponen
,
T.
and
Huo
,
X.
(
2018a
).
Maternal urinary cadmium levels during pregnancy associated with risk of sex-dependent birth outcomes from an e-waste pollution site in China
.
Reprod. Toxicol.
75
,
49
-
55
.
Zhang
,
Q.
,
Huang
,
Y.
,
Zhang
,
K.
,
Yan
,
Y.
,
Wu
,
J.
,
Wang
,
F.
,
Zhao
,
Y.
,
Xu
,
H.
,
Jiang
,
W.
,
Yu
,
D.
et al.
(
2018b
).
Progesterone attenuates hypertension and autoantibody levels to the angiotensin II type 1 receptor in response to elevated cadmium during pregnancy
.
Placenta
62
,
16
-
24
.
Zhang
,
T.
,
Gao
,
X.
,
Luo
,
X.
,
Li
,
L.
,
Ma
,
M.
,
Zhu
,
Y.
,
Zhao
,
L.
and
Li
,
R.
(
2019
).
The effects of long-term exposure to low doses of cadmium on the health of the next generation of mice
.
Chem. Biol. Interact.
312
,
108792
.
Zhang
,
Y.
,
Zhou
,
J.
,
Zeng
,
L.
,
Xiong
,
Y.
,
Wang
,
X.
,
Xiang
,
W.
and
Su
,
P.
(
2023
).
Paternal cadmium exposure affects estradiol synthesis by impairing intracellular cholesterol homeostasis and mitochondrial function in offspring female mice
.
Ecotoxicol. Environ. Saf.
263
,
115280
.
Zhao
,
X.
,
Cheng
,
Z.
,
Zhu
,
Y. I.
,
Li
,
S.
,
Zhang
,
L.
and
Luo
,
Y.
(
2015
).
Effects of paternal cadmium exposure on the sperm quality of male rats and the neurobehavioral system of their offspring
.
Exp Ther. Med.
10
,
2356
-
2360
.
Zhao
,
Q.
,
Gao
,
L.
,
Liu
,
Q.
,
Cao
,
Y.
,
He
,
Y.
,
Hu
,
A.
,
Chen
,
W.
,
Cao
,
J.
,
Hu
,
C.
,
Li
,
L.
et al.
(
2018
).
Impairment of learning and memory of mice offspring at puberty, young adulthood, and adulthood by low-dose Cd exposure during pregnancy and lactation via GABAAR α5 and δ subunits
.
Ecotoxicol. Environ. Saf.
166
,
336
-
344
.
Zhou
,
T.
,
Wang
,
H.
,
Zhang
,
S.
,
Jiang
,
X.
and
Wei
,
X.
(
2016
).
S100P is a potential molecular target of cadmium-induced inhibition of human placental trophoblast cell proliferation
.
Exp. Toxicol. Pathol.
68
,
565
-
570
.
Zhu
,
H.
,
Li
,
K.
,
Liang
,
J.
,
Zhang
,
J.
and
Wu
,
Q.
(
2011
).
Changes in the levels of DNA methylation in testis and liver of SD rats neonatally exposed to 5-aza-2′-deoxycytidine and cadmium
.
J. Appl. Toxicol.
31
,
484
-
495
.

Competing interests

The authors declare no competing or financial interests.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.

Supplementary information