Abstract
Metal ions play crucial roles in numerous biological processes, facilitating biochemical reactions by binding to various proteins. An increasing body of evidence suggests that neurotoxicity associated with exposure to nonessential metals (e.g., Pb2+) involves disruption of synaptic activity, and these observed effects are associated with the ability of Pb2+ to interfere with Zn2+ and Ca2+-dependent functions. However, the molecular mechanism behind Pb2+ toxicity remains a topic of debate. In this review, we first discuss potential neuronal Ca2+ binding protein (CaBP) targets for Pb2+ such as calmodulin (CaM), synaptotagmin, neuronal calcium sensor-1 (NCS-1), N-methyl-D-aspartate receptor (NMDAR) and family C of G-protein coupled receptors (cGPCRs), and their involvement in Ca2+-signalling pathways. We then compare metal binding properties between Ca2+ and Pb2+ to understand the structural implications of Pb2+ binding to CaBPs. Statistical and biophysical studies (e.g., NMR and fluorescence spectroscopy) of Pb2+ binding are discussed to investigate the molecular mechanism behind Pb2+ toxicity. These studies identify an opportunistic, allosteric binding of Pb2+ with CaM that is distinct from ionic displacement. Together, this data suggests three potential modes of Pb2+ activity related to molecular and/or neural toxicity: (i) Pb2+ can occupy Ca2+-binding sites, inhibiting the activity of the protein by structural modulation, (ii) Pb2+ can mimic Ca2+ in the binding sites, falsely activating the protein and perturbing downstream activities, or (iii) Pb2+ binds outside of the Ca2+-binding sites, resulting in allosteric modulation of the proteins activity. Moreover, the data further suggest that even low concentrations of Pb2+ can interfere at multiple points within the neuronal Ca2+ signalling pathways to cause neurotoxicity.
Introduction
Given that approximately 40% of all proteins have some interaction with physiologically relevant metal cations, the role of non-essential metals on cellular toxicity and neurotoxicity remains a relevant area of research. Of all the known toxic metals, Pb2+ has likely received the most attention, as it has been a persistent anthropogenic toxicant for much of recorded human history, and continues to be a health concern (1, 2) in urban areas and countries with emerging industrial production (3, 4). Toxicity associated with Pb2+ occurs even at very low concentrations. The current exposure threshold, as recommended by the CDC, is <5 μg/dL (http://www.cdc.gov/nceh/lead/acclpp/blood_lead_levels.htm). About 20% of Pb2+ absorbed by the body, primarily through inhalation and consumption, enters the blood stream where it is redistributed to different tissues and bone (5). The toxicity associated with Pb2+ and its relationship to anaemia was documented well over a century ago (6), although it was only in the latter half of the 20th century that Pb2+ toxicity was investigated at the molecular level. Exposure to Pb2+ produces a variety of systemic effects including anaemia, hypertension, kidney damage, and decrease in male fertility (7–9). Studies have reported that Pb2+ ions can pass cross through the blood-brain barrier (BBB) and the placenta in pregnant women (10, 11). More significantly, exposure to Pb2+ at even low concentrations produces a number of neurotoxic effects that induce severe cognitive deficiencies such as irreversible IQ loss in children (12), which can in turn lead to subsequent behavioral disorders (13) that persist through adolescence and adulthood.
An increasing body of evidence suggests that the neurotoxicity associated with Pb2+ exposure involves disruption of synaptic activity (14, 15), and these observed neurotoxic effects are associated with the ability of Pb2+ to interfere with Zn2+ and Ca2+-dependent functions, as recently summarized by Neal and Guilarte (16). However, the molecular mechanism for Pb2+ has yet to be clearly defined and is a topic of some controversy. In this review, cellular Pb2+ toxicity will be discussed in the context of calcium binding proteins (CaBPs) and the relationship between Ca2+, Pb2+, and Ca2+-regulated pathways. The metal binding properties of CaBPs with respect to both Ca2+ and Pb2+ will be examined, and structural analysis of CaBPs and proteins known to bind Pb2+ will be discussed with possible proposed mechanisms for molecular toxicity.
Calcium Signalling and CaBPs
Calcium, one of the most universal and versatile signal indicators, acts as an extracellular first messenger, and as an intracellular second messenger within mammalian cells to regulate almost all aspects of cellular processes. This begins at the inception of life during fertilization, and ends with cell death or apoptosis. Outside of the cell, available Ca2+ can reach mM concentrations, whereas cytosolic Ca2+ concentration ([Ca2+]CYT) at a resting state is maintained at about 100 nM, with the endoplasmic reticulum (ER) Ca2+ concentration ([Ca2+]ER) at several hundred μM (Fig. 1). Thus, cells have to maintain a >10,000-fold Ca2+ gradient across the plasma membrane and maintain intracellular Ca2+ homeostasis to avoid acute, massive fluctuations of Ca2+ within cells (17). A specific spatiotemporal pattern of cytosolic Ca2+ signalling is made possible by Ca2+ from two major sources: the internal Ca2+ store (mainly ER or sarcoplasmic reticulum) and the extracellular medium (Fig. 1). The entry of Ca2+ across the plasma membrane as mediated by specific receptors and Ca2+ channels is usually triggered by stimuli, including membrane depolarization, mechanical stretch, external agonists, depletion of internal stores and intracellular messengers. The mobilization of Ca2+ from the internal stores in response to Ca2+ itself or an intracellular messenger is primarily mediated by the IP3 receptors (IP3R) (18, 19) and the ryanodine receptors (RyR) (20). Several distinct mechanisms such as the plasma membrane Ca2+-ATPase (PMCA), the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA), the secretory pathway Ca2+-ATPase (SPCA), the Na+/Ca2+ exchanger (NCX), and the mitochondrial uniporter, are responsible for sequestering Ca2+ from the cytosol by transporting Ca2+ either to an external medium or into different cellular compartments (17, 21, 22). The temporal and spatial changes in Ca2+ concentration, and the interaction between Ca2+ and different classes of CaBPs in different cellular environments subsequently stabilizes the CaBPs and regulates numerous biological processes.
Fig. 1.

Ca2+ and Pb2+ targeted proteins involved directly and indirectly in neurotoxicity and calcium homeostasis are illustrated. The Ca2+ gradient is maintained at nM range intracellularly and at a mM range extracellularly in neurons. The black arrows represent the influx and the yellow arrows represent the outflux of divalent metal ions in the system. A general activity profile of receptor proteins is shown by a sigmoid and a bell curve as a result of Ca2+ and Pb2+ ions, respectively. Pb2+ is known to activate Ca2+ associated neuronal proteins at low concentrations and deactivate at higher concentrations, as characterized by a bell curve.
Potential Molecular Pb2+ Targets in Neurons
Neurons, like other cells, contain numerous proteins that interact either directly or indirectly with Ca2+ as part of a broader Ca2+-dependent signalling system (23). Changes in the free Ca2+ concentration regulates signalling in neural pathways along the CNS by triggering neurotransmitter release in synapses. Therefore, the well-established relationship between Ca2+ and Pb2+ makes the intracellular compartment a target-rich environment where Pb2+ may compete with Ca2+ to gain entrance to the cell and further compete with Ca2+ to erroneously activate or inhibit Ca2+-regulated processes via interactions with CABPs (Fig. 1). This section will focus on several classes of neuronal CaBPs as potential Pb2+ targets in the intracellular environment, including CaM (Fig. 2A and 2B), NCS (Fig. 2C), the transmembrane protein synaptotagmin, and extracellular proteins such as family C of GPCR.
Fig. 2.

(A) Proposed allosteric binding site for Pb2+ in human Ca2+-bound CaM (PDB ID 4bw8) in carboxyl-rich region of central helix comprising residues 78-84 (DTDSEEE). (B) Pb2+-bound human CaM (PDB ID 4bw8) (C) Human Ca2+-bound NCS-1 (PDB ID 4guk) with four EF-Hand sites similar to those observed in CaM (D) Pb2+ bound in site EF-IV of CaM (PDB ID 2v01). (E) Comparison of Ca2+- and Pb2+-binding characteristics with respect to the pentagonal plane. DLig-Ca and DLig-Pb, indicate distances between oxygen ligands and ions. C-Lig-Ca and C-Lig-Pb represent angles ϕ formed between the carbon atoms, ligands and the ions. DCa and DPb indicate ion distance above the pentagonal plane formed by Cγ, Oδ1 and Oδ2. (F) Ca2+ bound in site EF-IV of CaM (PDB ID 3cln). Figs. 2A, B, C, D, and F rendered with Chimera (24).
Calmodulin
In the neural cell, the intracellular protein CaM, upon being activated by binding with Ca2+ (Fig. 2A), regulates a variety of processes (Fig. 1). These processes include: gene expression and modifications to synaptic plasticity (25, 26); inhibition of Ca2+ release from intracellular stores through binding with inositol 1,4,5-trisphosphate receptor type 1 (IP3R1) (27, 28); binding with calcineurin (which also binds Ca2+ directly), which activates T cells and may play a role in cognitive function (29, 30); and activation of short transient receptor potential channel 5 (TrpC5) proteins, which participate in the formation of Ca2+-permeable ion channels, and can either inhibit or potentiate neuron growth cone formation (31). There is a preponderance of evidence indicating that Pb2+ interacts with CaM (Fig. 2B), and these interactions likely play a prominent role in Pb2+ neurotoxicity. Consequently, the ability of CaM to be activated by low concentration of Pb2+ suggests that each of these downstream protein interactions represents a potential avenue for neurotoxicity (Fig. 1), and research has already demonstrated that CaM activated at low concentrations of Pb2+ can subsequently activate phosphodiesterase (PDE) (32), and it can release norepinephrine (NE) in bovine neuroendocrine chromaffin cells (33).
Additionally, studies have reported that Trcp5 may be directly activated by low micromolar concentrations of Pb2+ through interaction with a glutamate residue (E543) (34), and another neurotoxic metal, Hg2+, through interaction with extracellular cysteine residues (C553 and C558) (35). As these interactions are not dependent on CaM, they represent an allosteric effect where toxic metals are binding opportunistically on the protein surface. These results also suggest that blocking of Trcp5 may present an approach to preventing neurotoxic effects of these metals (35).
Synaptotagmin
Synaptotagmin I, one of at least 15 isoforms of synaptotagmin, is a C2 domain-containing protein that is localized to synaptic vesicles (Fig. 1) (36) and plays an important regulatory role in neurotransmitter exocytosis by responding to changes in Ca2+ concentration (37). Synaptotagmin I contains a transmembrane domain and a short intravesicular N-terminus domain. The two C2 domains (C2A and C2B), homologous to the C2 domain of protein kinase C (PKC) are a part of their cytoplasmic domain (36), which is also known to bind Pb2+. Synaptotagmin I interacts with the plasma membrane protein syntaxin (38) and negatively charged phospholipids such as phosphatidylserine and phosphatidylcholine (39). These interactions occur in a Ca2+ dependent manner via the C2A domain of synaptotagmin I (36). A study by Bouton et al in 2001 reported that synaptotagmin binds Pb2+ with higher affinity than Ca2+, and that Pb2+ at nanomolar concentrations can mimic the effects of Ca2+ on synaptotagmin I, producing some of the same effects observed with Ca2+ (40). For example, Pb2+ was found to protect synaptotagmin I from proteolytic cleavage like Ca2+, but the Pb2+-synaptotagmin complex could not interact with syntaxin. This suggests a direct competitive interaction of Pb2+ Vs Ca2+ at low Pb2+ concentration, and that Pb2+-binding may stabilize the protein by displacing Ca2+, while simultaneously inhibiting the proteins function through the direct interaction with the Ca2+-binding sites, thus interfering with Ca2+-mediated release of neurotransmitters.
Additionally, a study by Garcia and Godwin reported that synaptotagmin II, which shares high sequence identity with synaptotagmin I, will self-associate to form a dimer in the presence of low concentrations of Pb2+ (~10 μM), and a multimer at 1.1 mM, while self-association due to Ca2+ occurs only at a higher concentration (5 mM) (41). Thus, the availability of Pb2+ may lead to inactivation of synaptotagmin through a self-association mechanism.
Family C of GPCRs
The family C of G-protein-coupled receptors (GPCRs) represents one of the largest families of cell surface receptors, consisting of 14 members including the calcium sensing receptor (CaSR), metabotropic glutamate receptors (mGluR), ɣ-aminobutyric acid (GABA)B receptors, amino acid taste receptors, pheromone receptors, odorant receptors in fish and several orphan receptor (42). GPCRs have been implicated in neuronal and glial signalling, and function to protect neurons from pathological stresses (43). The cGPCRs are potential targets for Pb2+ due to their direct activation by binding to Ca2+, or indirect modulation by interactions with other CABPs, as seen in the case of CaM. Consequently, Pb2+ can interfere with downstream Ca2+ signalling pathways through displacement of Ca2+, or through opportunistic binding of Pb2+ to regions besides the Ca2+ binding sites.
ɣ-aminobutyric acid (GABA)B receptors
Glutamatergic and GABAergic neurotransmitters within the amygdala are linked with neurons and are related to processes associated with emotion, fear regulation, anxiety, learning and memory (44, 45). GABA is the primary inhibitory neurotransmitter in the adult central nervous system (CNS). GABA functions as an excitatory neurotrophic factor (46) during early CNS development, and therefore plays a crucial role in cell proliferation, neuronal migration, neurite growth, axonal growth, and synaptogenesis (46).
Rats chronically exposed to low levels of Pb2+ have been observed to exhibit reduced Ca2+-dependent glutamate and γ-aminobutyric acid release in the hippocampus (47) indicating presynaptic neuron dysfunction during Pb2+ exposure mediated through glutamatergic and GABAergic systems. Studies of hippocampal neurons (48) and brain slices (47) exhibited a deficit in neurotransmission in both the glutamatergic and GABAergic systems, following exposure to Pb2+, as indicated by the impaired excitatory postsynaptic currents (EPSCs) and inhibitory postsynaptic currents (IPSCs). Similarly, Neal et al reported that Pb2+ exposure resulted in reduced expression of the presynaptic proteins synaptophysin (Syn) and synaptobrevin (Syb), and deficits in vesicular neurotransmitter release which were associated with both glutamatergic and GABAergic synapses (49). Additionally, a study by Wirbisky et al analysing tissue up-take, gross morphological alterations, gene expression, and neurotransmitter levels in zebrafish, indicated that Pb2+ exposure at lower doses affected the GABAergic system of the developing CNS in a manner that was dependent both on the dose and development stage (50). For example, the GABAergic system exhibited reduced expression of GABA genes gad2, gabra1, gabbr1a, and vgat at 48 hours post-fertilization (hpf) in the presence of 50 and 100 ppb doses of Pb, while conversely, increased expression of these genes was observed at 60 hpf, but only following treatment with 50 ppb. This may have been due to the lower concentration of Pb2+ mimicking the presence of Ca2+ in the protein Ca2+-binding site, thereby falsely activating the protein.
Metabotropic glutamate receptor 1 (mGluR1)
Metabotropic glutamate receptor 1 (mGluR1) modulates excitatory neurotransmission, neurotransmitter release, and synaptic plasticity, and is emerging as a potential drug target for various disorders, including a range of psychiatric and chronic neuronal degenerative diseases (51). Structural studies have shown that the receptors are activated when the endogenous agonist L-glutamate (L-Glu), the major excitatory neurotransmitter in the central nervous system, binds at the hinge region of the extracellular domain (ECD) of the receptor. This subsequently stimulates phospholipase C, resulting in accumulation of inositol trisphosphate (ITP) and an increase of intracellular calcium concentration ([Ca2+]i) (52–54), and activation of PKC (55). In addition to being activated by glutamate, mGluR1 is also modulated by extracellular Ca2+, and Jiang et al recently identified a Ca2+-binding site within the hinge region of the ECD of mGluR1 comprised of residues D318, E325 and D322 (51), adjacent to the reported L-Glu-binding site (56, 57).
Several studies have identified a crucial relationship between Pb2+ neurotoxicity, CaM, mGLuRs, and their signalling pathways. The proteins mGluR1, mGluR5, and mGluR7 are known to interact with CaM, which plays a role in mGLuR regulation. Picomolar concentrations of Pb2+ have been shown to replace micromolar concentrations of Ca2+ in a PKC enzyme assay (58), and a relationship between Pb2+ and increased PKC activity was reported in a study of lead workers who were observed to exhibit higher tibia bone Pb (BPb) levels and an apparent associated loss of cognitive skills (59). Studies have also reported that PKC phosphorylation of mGluR5 (60) and mGLuR7 (61) inhibits CaM binding and vice-versa. Additionally, mRNA expression of both PKC and CaM was observed to decrease in the hippocampus as a result of chronic lead exposure (62), while conversely, a study by Xu et al reported that Pb2+ exposure produced no obvious effect on hippocampal mGluR3 and mGluR7 mRNA expression, suggesting that these isoforms might not be associated with Pb2+-induced neurotoxicity (63). Moreover, a study by Nakajima reported that CaBPI (also known to bind Pb2+ (64)) competes with CaM for binding with presynaptic group III mGluRs 4, 7, and 8, in a Ca2+-dependent manner, and this binding can be blocked by PKC (61). These results suggest that Pb2+ may act on mGluR indirectly, either through binding with PKC or CaM, or by interfering with the expression levels of the two proteins, which may be indicative of another route of neurotoxicity not reported with the GABAergic system and other cited proteins.
N-methyl-D-aspartate receptor (NMDAR)
The N-methyl-D-aspartate receptor (NMDAR) is important for presynaptic plasticity through the generation and/or release of trans-synaptic retrograde signalling molecules, such as brain-derived neurotrophic factor (BDNF) (65–68). Pb2+ is a potent NMDAR antagonist (69, 70). Both exposure to Pb2+, and inhibition of NMDAR through its antagonist aminophosphonovaleric acid (APV), exhibit similar effects, including the disruption of developing synapses by reducing selective synaptic proteins, Syn and Syb, and this activity can be rescued by BDNF (71). This indicated that the effect is mediated via NMDAR inhibition and disruption of BDNF signalling (14). NMDAR consists of an obligatory NR1 subunit along with one or more accessory subunits from the NR2 and NR3 families (16). Pb2+ has been shown to bind the Zn2+ regulatory site in NMDAR in a voltage-independent manner with greater binding affinity to its NR2A subunit (72). This result is supported by electrophysiological studies where NR2A-NMDARs are inhibited by Pb2+ more than its other forms (73, 74). Excessive Pb2+ exposure has also been shown to correlate with reduced NR2A in the hippocampus (75–78), altered expression of NR1 splice variants (79), and slightly increased or unchanged NR2B mRNA levels (49, 75–78, 80). Altered expressions of NR2 family members consequently could interfere with NR2 related downstream pathways such as MAPK signalling (81), calcium/calmodulin kinase II (CaMKII) activity (82) and cyclic AMP response element binding protein (CREB) phosphorylation and binding affinity (80, 83).
Neuronal calcium sensor protein-1 (NCS-1)
Neuronal calcium sensor protein 1 (NCS-1) is a Ca2+-sensing EF-Hand protein involved in the regulation of neurotransmission (84), neuron development (85, 86), and cognitive functions (87). NCS-1, when activated by Ca2+, interacts with a wide variety of downstream proteins (23), including dopamine receptor D2R (88), Ca2+-dependent activator protein for secretion (CAPS), phosphatidylinositol-4 kinase-IIIβ (PI4KIIIβ) (89, 90), ARFI (23, 90), AP1, AP2, TGFBR1, and calcineurin, among others. Despite having a low sequence identity to CaM (20%), NCS-1 possess four EF-Hand motifs similar to CaM (23). Likewise, NCS-1 is very sensitive to small increases in intracellular Ca2+ concentrations. As seen in the structure of NCS-1 (Fig. 2C), Ca2+ binds in only three of the four EF-Hand sites, while the fourth site can accommodate Na+. Although a review of the literature failed to identify a direct link between Pb2+ neurotoxicity and NCS-1, the fact that NCS-1 is an EF-Hand protein with high affinity for Ca2+ strongly suggests that it could bind Pb2+ in one or more of the EF-Hand sites, including the site that is unable to bind Ca2+, and that interaction with Pb2+ could alter downstream activity when NCS-1 interacts with other proteins.
Moreover, while NCS-1 interacts with various proteins that are not directly involved in neurotransmission, these interactions can involve other cellular regulatory and assembly activities that are critical support functions that may be indirectly affected by Pb2+. For example, ADP-ribosylation factor 1 (ARF1) is a GTPase that interacts with NCS-1 and PI4KIIIβ, and regulates vesicular trafficking in cells (23, 91). ARF1 also performs several important functions at the Golgi complex related to vesicle coat assembly and trafficking from the trans-Golgi-network to the plasma membrane and endosomes (92). Although no evidence to date suggests that ARF1 interacts directly with Pb2+, it does interact with Ca2+-binding NCS-1 and may have other functions regulated by Ca2+ availability.
Conversely, CAPS does play an important role in neural signalling. CAPS exists in two primary isoforms in neuroendocrine cells and the brain, CAPS1 and CAPS2. CAPS1 is a cytosolic protein that plays a role in large dense-core vesicle (LDCV) exocytosis and neurotransmitter release, while CAPS2 is also expressed in the lung, liver, and testis (93). The two isoforms are expressed differently during development, with low expression of CAPS1 prior to birth, followed by increased expression until postnatal day 21, while expression of CAPS2 remains constant from embryonic day 10 until postnatal day 60. Due to differences in the permeability of the BBB during fetal development as compared with adults, the developing brain may be subject to increased accumulation of Pb2+ (94), and Pb2+ may act either directly or indirectly (through Pb2+-modulated NCS-1) with CAPS proteins during embryonic or early childhood development. This would represent another potential route for neurotoxicity that could contribute to the observed problems associated with neural development and cognitive deficiencies observed in children.
Stim and Orai
A recent study by Moccia et al reported that Stim1-2 and Orai1-2 are expressed in neurons (95). Stim1 is a Ca2+-sensing EF-Hand protein that responds to decreasing Ca2+ concentration in the ER by activating ORAI1 to refill the ER store of Ca2+. Results of this study suggested that store operated Ca2+ entry (SOCE) may play a role in the excitation of neuron and the regulation of synaptic activity, and may further contribute to neurological disorders (95). As these are all Ca2+-modulated activities, they also represent potential targets for disruption by Pb2+.
Pb2+ effects on functional activity of proteins associated with Ca2+
The effects of Pb2+ on the functional activities associated with Ca2+ vary in a concentration dependent manner, with protein activation observed at low concentrations and inhibition observed with increasing Pb2+ concentration (Fig. 1). At initially low concentrations, CABPs may not be activated by competing essential metals, but instead may be activated by toxic metals mimicking their native metals. For example, the intracellular protein CaM may be activated by either Pb2+ (32) or Cd2+ (96), and Richardt et al reported that Pb2+ will bind with CaM, Parvalbumin, TnC, CaBPI and CaBPII, with the highest Pb2+ affinities being observed with CaM and CaBPI (64). Several other studies have reported that Pb2+ may initially activate and then deactivate CABPs with increasing concentration. Habermann et al (32) found that Pb2+ could replace Ca2+ in CaM, and the Pb2+-CaM complex subsequently activated cyclic nucleotide phosphodiesterase (PDE) to phosphorylate several membrane proteins. This behavior was observed up to a Pb2+ concentration of near 10−4 M, which corresponds to the minimum Ca2+ concentration required for activation. Phosphorylation was subsequently inhibited with increasing concentration of Pb2+. This suggests that Pb2+ may activate CaM at lower concentrations by mimicking Ca2+ in the EF-Hand binding sites (Fig. 2D and Fig. 2F), with the reported loss of activity at increasing Pb2+ concentration suggesting that Pb2+ can bind to another region of the protein, resulting in allosteric inhibition. Additionally, Chao et al reported that Pb2+ produces a biphasic activation and inactivation of MLCK with increasing Pb2+ concentration, but only in the presence of CaM, indicating that Pb2+ interacts directly with CaM, not MLCK (97). Chao et al reported in a later study that Pb2+ activated TnC, which subsequently stimulated myofibrillar ATPase (98). Therefore, the effects of Pb2+ on different processes may occur indirectly via interactions with upstream proteins.
Additionally, there is little evidence indicating that Pb2+ occupancy of the Ca2+ sites inhibits the function of the protein. Instead, the evidence suggests that it impersonates Ca2+ in these sites, thereby falsely activating the protein and altering downstream activities at inappropriate times. However, the subsequent inhibition of these proteins at higher concentrations suggests a second mode of toxicity where inactivation or inhibition of the protein is induced as result of Pb2+ binding to the proteins in regions outside of the normal Ca2+-binding sites (Fig. 3).
Fig. 3.
Proposed mechanism for Pb2+ toxicity in CaM. Path A illustrates normal activity associated with binding of Ca2+. Ca2+ first cooperatively binds the two higher affinity sites EF-III and EF-IV, followed by cooperative binding in sites EF-I and EF-II. The Ca2+ loaded protein then interacts with ligand molecules. In Path B, Pb2+ binds in a single higher affinity site in the C-terminal domain, while the remaining sites may bind Pb2+ with equivalent affinity. Binding may not occur cooperatively. At lower concentration of Pb2+, CaM is activated and functions as if bound with Ca2+. In either the Ca2+ or Pb2+ loaded states, an increase in the concentration of Pb2+ results in Pb2+ binding to a secondary site in the central helix, altering the conformation of the protein and allosterically inhibiting the activity of the protein.
Pb2+ toxicity and ionic displacement in proteins
Under normal cellular conditions, proteins are generally not activated by competing essential metal ions (e.g., Ca2+, Zn2+, Mn2+ or Fe2+). As example, Mg2+ ions can occupy intracellular Ca2+-binding sites in CaM, when the concentration of Ca2+ is low, but doing so occurs without activation of CaM. The Mg2+ ions are then displaced with an influx of the higher affinity intracellular Ca2+ ions to activate the protein. A study by Ouyang et al reported that Mg2+ binding in the EF-Hand sites of CaM did not produce conformational changes (99), explaining its inability to activate the protein. However, it is possible that Mg2+ (and/or Zn2+) allosterically regulates binding of Ca2+ to some extent, via binding at auxiliary sites on the protein (100).
The ability of ions to occupy metal binding sites and interact with ligands is related to a number of variables, including: the ionic radius (101); charge and electronegativity of the metal ion; the types of ligands in the binding sites, as well as the structure and coordination geometry of the site; affinity; and ion concentration. Despite the differences in binding geometries and ligand types between Ca2+ (6–8 oxygen ligands) and Zn2+ (4–6 sulfur and other ligands), Pb2+ can also replace Zn2+ in proteins (102–107), which points at the versatility of Pb2+ binding and interestingly, several studies have identified this as one route through which Pb2+ induces neurotoxicity by disrupting the Zn2+-dependent modulation of NMDAR activation as mentioned above (16, 72). For example, while both Ca2+ and Zn2+ have identical charges, the ionic radius of Zn2+ is about 0.75 Å compared with approximately 1.0 Å for Ca2+, and Zn2+ utilizes different binding ligands than Ca2+ (more sulfur than oxygen) with a tetrahedral coordination geometry (108), so that the two ions are generally not competitive for their respective target proteins. The stronger binding of Zn2+ (1018 -1010 M−1) (109, 110) compared to Ca2+ (107 M−1) (109) to the binding sites in proteins, along with the smaller off rate of Zn2+ (10−9 s−1) (110) as compared to that of Ca2+ (103 s−1) (109, 111) should also be considered. Therefore, the exchange of Zn2+ with Pb2+ is likely much slower and less probable than the exchange of Ca2+ with Pb2+, even when the binding constant of Pb2+ to Zn2+ sites are higher in magnitude than the binding of Pb2+ to Ca2+.
Classification of an ion as being a hard, soft, or borderline Lewis acid also appears to play a role in the interaction affinity between ions and proteins. This is particularly relevant with respect to toxic metal ions, which can occupy metal binding sites in proteins, thereby replacing or displacing the targeted essential metal ions. The divalent Ca2+ and Pb2+ ions are identical in charge and can be similar in size (0.99 – 1.12 Å vs. 1.12 – 1.19 Å, respectively) depending on the coordination geometry (112). However, Pb2+ is approximately 2.3 times as electronegative as Ca2+, and is classified as a borderline Lewis acid, whereas Ca2+ is classified as a hard Lewis acid. The Pb2+ ion therefore tends to be more polarizable with lower energy requirements to achieve its highest occupied molecular orbitals (HOMOs), likely determining its ability to occupy metal binding sites with higher affinities (Fig. 2E).
Structural analysis of Pb2+ vs. Ca2+ binding sites
Previous work in our laboratory has included a comprehensive statistical analysis of CaBP structural data from the Protein Data Bank (PDB) (113). This study evaluated 1,605 Ca2+-binding sites from 558 PDB files with higher resolutions (R > 2.0 Å). The Ca2+-binding sites were further sub-divided into EF-Hand (EF) sites with well-defined pentagonal bipyramidal geometries, and a more general group of structurally-diverse non-EF-Hand (NEF) sites. Structural parameters used to evaluate the sites included binding ligand type (atom type and sidechain), coordination number, and ligand/ion distances and angles. Subsequently, a similar analysis was conducted for proteins reported to bind Pb2+. To better understand how Pb2+ displaces Ca2+ in proteins, data from both data sets were then compared (112), and the results of these studies (Table 1) indicated that Pb2+ sites typically utilized fewer total ligands (CN = 2 – 5) and less mean negative charge by site (charge = −2) than canonical EF-Hand Ca2+ sites (CN = 4 – 8, charge = −3). Differences in ligand selections were also observed. While Ca2+ utilizes oxygen atoms almost exclusively in the EF-Hand sites, the Pb2+ sites were more likely to contain either nitrogen or sulfur atoms in close enough proximity to represent potential ligands in addition to oxygen, making its behavior similar to both Ca2+ and Zn2+. Additionally, while the EF sites utilized approximately the same ratio of oxygen from Glu and Asp residues (26.6% and 29.7%), Pb2+ was observed to favor Glu over Asp (38.4% vs. 20.3%), presumably due to the longer sidechain of Glu providing more flexibility to accommodate the larger ionic radius of the Pb2+ ion (Fig. 2E). This is consistent with observations that when Pb2+ occupies EF-Hand sites, the ion is observed to sit slightly higher above the pentagonal plane than Ca2+ (Fig. 2E).
Table 1.
Summary of statistics comparing Pb2+ and Ca2+ binding sites in proteins.
| Pb |
Ca |
|||
|---|---|---|---|---|
| DS HR | DS Final | NEF | EF | |
| Total PDB proteins in study | 7 | 21 | 525 | 33 |
| Total binding sites evaluated | 27 | 48 | 1468 | 137 |
| Total target ligand atoms | 105 | 177 | 8549 | 958 |
| Total Oaa ligands | 86 | 118 | 5626 | 829 |
| Total OHOH ligands | 16 | 36 | 2788 | 127 |
| Total N ligands | 3 | 10 | 135 | 2 |
| Total S ligands | 0 | 13 | 0 | 0 |
| Total sites with N ligands | 3 | 9 | 106 | 2 |
| Mean CN, PLW | 4 ± 2 | 4 ± 2 | 6 ± 2 | 7 ± 1 |
| % CN 2–5 | 78 | 77 | 31 | 4 |
| % CN 6–9 | 22 | 17 | 47 | 96 |
| Mean CN, PL | 3 ± 2 | 3 ± 2 | 4 ± 2 | 6 ± 1 |
| % CN 2–5 | 70 | 73 | 58 | 9 |
| % CN 6–9 | 15 | 8 | 27 | 91 |
| Mean charge by site | −2 | −2 | −1 | −3 |
| Total identified bidentate pairs | 24 | 36 | 777 | 129 |
| Total sites with bidentate ligands | 21 | 30 | 664 | 125 |
| % Sites with bidentate ligands | 78 | 63 | 45 | 91 |
| Mean distance, Pb-Oaa (Å) | 2.7 ± 0.4 | 2.7 ± 0.4 | --- | --- |
| Mean distance, Pb-N (Å) | 2.7 ± 0.3 | 2.6 ± 0.4 | --- | --- |
| Mean distance, Pb-S (Å) | --- | 3.2 ± 0.3 | --- | --- |
| Mean distance, Ion-OHOH (Å) | 2.8 ± 0.3 | 2.8 ± 0.4 | 2.5 ± 0.3 | 2.1 ± 0.1 |
| Mean distance, Ca-Ocarbonyl (Å) | --- | --- | 2.4 ± 0.2 | 2.3 ± 0.1 |
| Mean distance, Ca-OSC (Å) | --- | --- | 2.4 ± 0.2 | 2.4 ± 0.2 |
| Mean distance, Ca-OBidentate (Å) | --- | --- | 2.6 ± 0.3 | 2.5 ± 0.2 |
CN=Coordination Number. PL=Protein Ligands only. PLW=Protein Ligands and Water. Oaa=Amino acid oxygen ligands. OHOH=Water oxygen ligands. Ocarbonyl=Carbonyl oxygen. OSC=Sidechain oxygen. Obidentate=Bidentate oxygen ligands. DS HR=High resolution dataset (R less than 1.76 Å). DS Final=Total dataset from PDB. NEF=Non-EF-Hand calcium binding sites. EF=EF-Hand calcium binding sites.
From a structural perspective, the NEF sites exhibited much greater diversity with respect to coordination number than EF sites, and generally could be classified as having a spherical (holo-directed), semi-spherical (hemi-directed), or planar geometry surrounding the Ca2+ ion (114), which was also previously reported for binding of Pb2+ (108). The holo-directed geometry is consistent with higher CN values observed with EF-Hand Ca2+-binding sites, while the hemi-directed geometry was more prevalent with Non-EF-Hand Ca2+-binding sites, and with Pb2+-binding sites (Table 1). Based on the similarity of these binding sites, and despite variances in the number of coordinating ligands, it is possible that the NEF sites may represent incomplete pentagonal-bipyramidal structures that evolved in response to different needs within the Ca2+-signalling pathways. Comparatively, the hemispheric geometry observed with C2 domain Ca2+-binding sites, as seen with PKC (114), is also observed with Pb2+-binding, and from the perspective of our previous analyses (Table 1), when EF-hand sites were excluded, the differences between binding of Pb2+ and Ca2+ diminished significantly (112).
Additionally, a detailed comparison of CaM structures bound with Ca2+ (PDB ID 1EXR) and Pb2+ (PDB ID 1N0Y) revealed only minor deviations in coordination geometry, distances between ligands and ions, bond angles, and RMSD values, when comparing the two protein backbones for the four EF-Hand sites (112). These data therefore suggested only minimal disruption to the structure based on ionic displacement, which was consistent with results reported by Kursula and Majava (115). However, the crystal structure of the Pb2+-bound CaM (Fig. 2B) exhibits binding of several ions outside of the EF-Hand sites, including at the central helix connecting the N- and C-terminal domains of the protein, where significant folding of the protein was observed. While this may potentially represent an artefact of crystallization, the region in question frequently appears as an extended helix in crystalized structures of the Ca2+-loaded protein (116, 117), and as a partially unwound coil in solution (118, 119), and these conformational changes may differentiate the Ca2+-free and Ca2+-loaded states. The flexibility of this region is believed to be necessary for conformational changes associated with peptide binding (Fig. 3), and a recent study has provided evidence that disruption within this region contributes to folding of a more compact structure associated with the Ca2+-bound state (120), as has been reported for osteocalcin (121), while binding of Ca2+ in the four EF-Hand sites may result in less flexibility (122). The compact folding observed in the crystalline structure of Pb2+-bound CaM suggested a potential allosteric binding site for Pb2+ in this region, and results of structural analyses identified the carboxyl-rich region from residues 78–84 as a strong candidate for binding of Pb2+ (Fig. 2A) (123).
Analyses of structural data from the PDB indicate that Pb2+ can occupy the binding sites of various metals with a broad range of coordination values, and may also bind opportunistically outside of known binding sites, suggesting multiple binding modes and a promiscuous interaction with proteins, which would be consistent with the systemic effects produced as a result of elevated Pb2+ concentrations, including neurotoxicity.
NMR/Binding studies
Although various studies have demonstrated that Pb2+ can physically occupy Ca2+-binding sites, displacement of Ca2+ by Pb2+ would depend on other factors, including concentrations of the ions and their relative binding affinities for a given binding site. Unfortunately, determining accurate binding affinities for Ca2+ and Pb2+ remains a challenge for several reasons. A general problem is that available analytical instrumentation often lacks sufficient sensitivity to detect very low concentrations of analytes that would correspond to high affinity binding (124). Additionally, for many EF-Hand Ca2+-binding proteins, where binding sites are frequently paired and appear to bind cooperatively, methods to determine Kd values for individual sites, such as deactivating one of the paired sites through site-directed mutagenesis (125), or grafting an individual EF-site into a scaffold protein (126), may not provide reliable affinity data as the process eliminates the effects of cooperativity. Moreover, cooperativity may occur both within and between domains. For example, each of the paired EF-hands in the respective N- and C-terminal domains of CaM appear to bind Ca2+ ions cooperatively (127), but further evidence suggests that the domains bind cooperatively, as well, so that Ca2+ occupies both EF-hand sites in the higher affinity C-terminal domain sites first, which produces minor conformational changes that enhance binding in the EF-hand sites in the N-terminal domain (128). These cooperative effects appear to be of critical importance for producing conformational changes in responses to fluctuations in Ca2+ concentrations (129), which would directly affect Ca2+ signal transduction.
As these approaches have provided very limited or incomplete data on affinity, various alternative methods of approximation have been employed to investigate Ca2+-binding affinity at the molecular or domain level, rather than the individual sites. These approaches may involve calculated upper and lower limits (130), relative affinities determined by the order in which the domains bind Ca2+, or spectrofluorometric approaches that monitor fluorescence changes in Phe and Tyr side chains that result from conformational changes associated with metal binding. This latter approach has been used to determine domain-level binding affinities for Ca2+ with CaM, as Phe produces strong signals in the N-terminal domain, while Tyr produces similar results in the C-terminal domain (131, 132). In another approach, Richardt et al utilized flow dialysis methods to evaluate competitive binding between Ca2+ and Pb2+ or other toxic metals, with a variety of intracellular Ca2+-binding proteins, and calculations of IC50 values for the various protein/metal complexes (i.e., interaction at the molecular level), indicated that the relative binding affinities were: for CaM, Pb2+ > Ca2+ > Cd2+; for troponin C, Ca2+ > Cd2+ > Pb2+; for CaBP I, Ca2+ ~ Pb2+; for CaBP II, Ca2+ > Pb2+ > Cd2+; and for parvalbumin, Cd2+ ~ Ca2+ > Pb2+ (64). Thus, while different CABPs may bind Pb2+, the affinities can vary significantly, indicating that ionic displacement is not universal, and this study verified that CaM represents an important target for Pb2+ toxicity at the molecular level.
Nuclear magnetic resonance (NMR) is another powerful tool that can be used to study protein structure and monitor dynamic properties, conformational changes and binding affinity trends. A recent study used NMR and fluorescence spectroscopy to study different aspects of Pb2+ binding in CaM, including calculation of dissociation constants (Kd) for binding of Pb2+ with CaM, and to determine whether Pb2+ displaces Ca2+ in the EF-hand binding sites (123). In the NMR study, data were collected using 2D 15N-HSQC. A baseline of chemical shift data for Ca2+ was collected at different concentrations of Ca2+, starting with a Ca2+-free structure. The magnitude of the differences in chemical shifts for residues in the EF-Hand sites was used to monitor binding of Ca2+. Results of this part of the study indicated that Ca2+ was first bound to the two EF-Hand sites (III and IV) in the higher affinity C-terminal domain, and these events further altered the structure of the N-terminal domain prior to binding of Ca2+ in EF-Hand sites I and II, which would be consistent with interdomain cooperativity. Repeating the experiment, but substituting Pb2+ for Ca2+, failed to identify an order of domain occupancy, as was observed with Ca2+, suggesting that the affinities of the four EF-Hand sites for Pb2+ were similar, which was consistent with results of other studies (99). However, the addition of Pb2+ to Ca2+-saturated CaM produced interesting results which suggested that Pb2+ displaced Ca2+ only in the N-terminal domain, where sites EF-I and EF-II bind Ca2+ with lower affinity, but not the C-terminal domain sites. Moreover, further addition of Pb2+ produced conformational changes that were consistent with binding of Pb2+ outside of the EF-Hand sites, which led us to hypothesize that increasing concentrations of Pb2+ result in opportunistic binding of Pb2+ in one or more auxiliary sites on the protein, resulting in significant conformational changes to the protein capable of inhibiting its activity, which would be consistent with the biphasic results observed with the interaction between Pb2+-CaM and PDE (32).
In the same study, Kd values for binding of Ca2+ and Pb2+ were calculated by monitoring fluorescence changes from Phe in the N-terminal domain and Tyr in the C-terminal domain. While this approach does not allow for separation of the individual binding sites, affinity values for the domains were determined, indicating that Kd values for Ca2+ for both the N-terminal (11.50 ± 0.68 μM) and C-terminal (2.04 ± 0.02 μM) were consistent with known intracellular concentrations and previously reported values (133). For Pb2+, the Kd values reported for the N- and C-terminal domains were 1.40 ± 0.30 μM and 0.73 ± 0.10 μM, respectively, which indicated that Pb2+ had slightly higher affinity for the EF-Hand sites than Ca2+. Interestingly, the tyrosine fluorescence exhibited a biphasic response: an increase in fluorescence similar to the Ca2+ response followed by a decrease in intensity with increasing Pb2+ concentration. This response was interpreted as either the presence of a single higher affinity binding site (Fig. 3), or a conformational change associated with opportunistic binding of Pb2+ in an auxiliary site, and the associated Kd was calculated to be 1.93 ± 0.32 μM, which would indicate greater affinity than that observed for Pb2+ in the N-terminal domain. If this latter hypothesis were correct, it would help to explain the biphasic response of CaM activation to Pb2+, where trace concentrations can activate the protein, which is subsequently deactivated with increasing concentration. The study further noted that Pb2+ did not produce conformational changes when Pb2+ was introduced to Ca2+-loaded CaM, despite differences in binding affinity values, which suggests that Pb2+ either displaces Ca2+ without disrupting the structure, or Pb2+ fails to displace Ca2+, but can deactivate the protein through opportunistic binding in a secondary site (123). In the latter case, it is possible that Pb2+ may bind opportunistically to regions of the protein outside of the normal metal binding sites, such as the carboxyl-rich central helix of CaM (Fig. 3), thereby allosterically inhibiting protein function through structural alteration. As previously noted, studies have identified potential secondary sites in CaM that may bind other physiologically relevant ions. Consistent with this, Kursula and Majava identified a 5th Ca2+-binding site in CaM, located in the central helix (115), and Bertini et al reported that Yb3+ may bind in this region as well (134). Results from these studies suggest that this region might represent a potential target for binding of Pb2+, and structural studies of Pb2+-CaM complexes in our lab further support this hypothesis (123).
A new perspective for the molecular basis of Pb2+ toxicity
Historically, molecular toxicity was believed to be caused by the displacement of Zn2+ or Ca2+ by Pb2+ions, and various studies have since demonstrated that Pb2+ can occupy binding sites for many physiologically essential metals, including: Mg2+ in pyrimidine 5'-nucleotidase type 1 (135); Zn2+ in 5-aminolevulinic acid dehydratase (ALAD) (136, 137), resulting in iron deficiency associated with anaemia; and Fe2+ in divalent metal transporter-1 (DMT1), which appears to be involved in transport and uptake of Pb2+ (138, 139). The Pb2+ ion can also occupy Ca2+-binding sites, and/or displace Ca2+ in many proteins, including voltage-gated calcium channels (VGCCs) (140), troponin C (98), CaM (32), PKC (58), parvalbumin (64), and synaptotagmin (40). Moreover, CABPs have also been observed to bind a number of different non-essential metals in addition to Pb2+, including Sr2+, Hg2+, Cd2+ and many lanthanides (141–145). Similar results have been reported in studies involving other toxic metals (e.g., Hg2+ and Cd2+) and in different proteins, and these data were generally consistent with a displacement model where toxic metals replace essential metals in binding sites (146).
However, studies have also reported that CABPs may be activated by binding of toxic metals in the primary metal binding sites. As previously noted, the intracellular signalling protein CaM may be activated by either Pb2+ (32, 64, 101, 147, 148) or Cd2+ (96). Moreover, evidence suggests that Pb2+ may initially activate and then deactivate CABPs with increasing concentration, as seen with CaM (147). This in turn affects CaM's subsequent interaction with phosphodiesterase (PDE) (32) and myosin light chain kinase (MLCK) (97). A biphasic response of this nature suggests that toxic metal ions like Pb2+ may effectively mimic the Ca2+ ion in the Ca2+-binding site in certain proteins by inappropriately activating the protein, rather than directly inhibiting the protein function, and increasing concentration subsequently inactivates the protein via a different mechanism or site. Research on CaM suggested that Pb2+ may alter the functional activity of the protein by binding opportunistically in regions of the protein outside of the EF-hand Ca2+ binding sites in a concentration-dependent manner, and in the presence of Ca2+ (112, 123), which would indicate an allosteric inhibitory effect. Similar results were reported by Shirran and Barran (149), and Mills and Johnson (142). Additionally, other studies have identified secondary sites in CaM capable of binding Ni2+ (in the Ca2+-bound state) (150), or Zn2+ (100), which, in the latter study, appears to inhibit binding of Ca2+ in the EF-Hand sites. The ability of Pb2+ to mimic Ca2+ may also produce neurotoxic effects indirectly. Exposure to Pb2+ has been observed to inhibit Ca2+-dependent glutamate and γ-aminobutyric acid (GABA) release in the hippocampus (47, 151, 152), while results of a study by Ferguson et al reported that Pb2+ produced an increase in Ca2+ efflux from the neural cell by activating CaM, which then stimulates Ca2+ ATPase, an enzyme that regulates Ca2+ concentration in the cell (153). Similarly, Pb2+ ions may inhibit voltage-gated calcium channels (VGCCs) that regulate synaptic transmission, and subsequently utilize the channels for entry into the cell where they presumably can interact with other proteins (140, 154). Studies also suggest that Pb2+ interferes with Ca2+ in PKC, a protein that plays a significant role in cognitive functions (155). Additionally, Pb2+ may disrupt the Zn2+-dependent modulation of N-methyl-D-aspartate receptor (NMDAR) activation (16, 72), which regulates the flow of Ca2+ and Na+ into the cell.
Collectively, these data indicate that Pb2+ may interact with multiple proteins involved in neural pathways, and while Pb2+ can interact with proteins that are not specifically CaBPs, the information presented in this review has only focused on the interactions between Pb2+ and CaBPs or proteins involved in Ca2+-signalling pathways, and how they relate to neurotoxicity, based on three potential modes of Pb2+ activity as follows (Fig. 3):
Pb2+ may occupy Ca2+-binding sites, altering the structure of the protein and inhibiting the activity of the protein.
Pb2+ may occupy Ca2+-binding sites and mimic Ca2+, thereby falsely activating the protein and perturbing downstream activities.
Pb2+ may bind with proteins in sites other than Ca2+-binding sites, thereby altering the conformation and function of the protein.
The picture that is emerging from this research suggests that there are multiple points within the Ca2+ signalling pathways in neurons and other cells that may be disrupted by the presence of even low concentrations of Pb2+. All of the available evidence strongly indicates that Pb2+ interacts with CaM, either through ionic displacement, or through opportunistic binding, which likely disrupts a wide range of downstream activities, and it is possible that Pb2+ could interact with NCS-1 in a similar manner, given the functions of NCS-1 summarized in this review, and their relationships with known effects of neurotoxicity.
The mechanisms discussed in this review can further provide insights into other systemic physiological problems associated with Pb2+ exposure. For example, Pb2+ may induce nephrotoxicity through high-affinity interaction at low concentrations with CaSR, (156–159), and long term activation of the CaSR can lead to renal injury by inhibiting growth of epithelial cells and stimulating growth of fibroblasts (160–162). Similarly, Pb2+ exposure may be a contributing factor in cardiotoxicity, producing structural changes in cardiac tissues (163, 164), while a direct correlation between hypertension and Pb+2 exposure has been reported (165).
The above associations however, do not have firmly-established mechanistic explanations, and while Pb2+ ions may occupy the Ca2+-binding sites in different CABPs, it cannot be assumed that they will replace Ca2+ ions, as the affinity for Pb2+ may vary differently than the affinities for Ca2+. In the case of the neuron, variations in affinities for Ca2+ and Pb2+ could influence the activation or inhibition of different CABPs and alter the Ca2+-mediated pathways. For example, while both Ca2+ and Pb2+ promote exocytosis of vesicular catecholamine, the process is regulated by PKC and calcineurin in the presence of Ca2+, and by CaM kinase II in the presence of Pb2+ (166). Because Ca2+-regulated activity within the neuron is complex due to different types of Ca2+ signals that can translate into subtle physiological differences, or produce opposing effects depending on their timing and magnitude (23), it is reasonable to assume that extensive research would be required to decipher the particular variations that might occur as a result of Pb2+ introduction into the cell.
Finally, the seemingly incongruent reported dose-response on the loss of IQ of children, where low concentrations of Pb2+ appear to produce greater IQ loss than higher exposure to Pb2+ (12, 167, 168), may be better understood in the context of a biphasic response of CABPs such as CaM to Pb2+, where the protein is falsely activated at lower Pb2+ concentrations, thus initiating deleterious Ca2+-modulated responses, which are subsequently inhibited by further allosteric interaction with Pb2+ at higher concentrations. Because anthropogenic Pb2+ in the environment represents a continuing problem, as seen recently with the contaminated water supply in Flint, Michigan (http://www.motherjones.com/environment/2016/02/flint-lead-poisoning-america-toxic-crisis), understanding the mechanisms associated with Pb2+ neurotoxicity is an important step towards understanding the impact of exposure on human health, and as a basis for environmental efforts and the enactment of policies directed towards elimination of this threat.
Acknowledgements
This work was supported in part by National Institutes of Health Grants GM081749 and EB007268 (to J. J. Y.), and a Center for Diagnostics and Therapeutics fellowship (to R.G.) from Georgia State University.
Biographies

Jenny J. Yang is a Distinguished University Professor in Chemistry at Georgia State University. The research in her lab focuses on predicting and understanding the role of metal ions, especially calcium , in biological, chemical and environmental systems, to design novel tools for diagnostics and research, and to create drugs for better health and overall environment.

Michael Kirberger is a Lecturer of Chemistry at Clayton State University. His diverse research interests include metalloproteins, metal toxicity, structural biology, and computational methods.

Rakshya Gorkhali is a PhD candidate and a University fellow at Georgia State University. Her research interests include the biomolecular and biophysical study of calcium regulated proteins, such as calcium sensing receptors and gap junctions, to understand the molecular basis of human diseases associated with calcium signaling.

Kenneth Huang is a PhD candidate at Georgia State University in the Department of Chemistry. His research interests include the use of HPC in the sciences, computational chemistry, and molecular dynamics.
References
- 1.Callan AC, Hinwood AL. Exposures to lead. Rev Environ Health. 2011;26(1):13–5. doi: 10.1515/reveh.2011.003. [DOI] [PubMed] [Google Scholar]
- 2.Mielke HW, Reagan PL. Soil is an important pathway of human lead exposure. Environ Health Perspect. 1998;106(Suppl 1):217–29. doi: 10.1289/ehp.98106s1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Gong M, Wu L, Bi XY, Ren LM, Wang L, Ma ZD, et al. Assessing heavy-metal contamination and sources by GIS-based approach and multivariate analysis of urban-rural topsoils in Wuhan, central China. Environ Geochem Health. 2010;32(1):59–72. doi: 10.1007/s10653-009-9265-2. [DOI] [PubMed] [Google Scholar]
- 4.Luo XS, Yu S, Zhu YG, Li XD. Trace metal contamination in urban soils of China. Sci Total Environ. 2012;421–422:17–30. doi: 10.1016/j.scitotenv.2011.04.020. [DOI] [PubMed] [Google Scholar]
- 5.Marchetti C. Interaction of metal ions with neurotransmitter receptors and potential role in neurodiseases. Biometals. 2014;27(6):1097–113. doi: 10.1007/s10534-014-9791-y. [DOI] [PubMed] [Google Scholar]
- 6.Aub JC, Reznikoff P. Lead Studies : Iii. The Effects of Lead on Red Blood Cells. Part 3. A Chemical Explanation of the Reaction of Lead with Red Blood Cells. J Exp Med. 1924;40(2):189–208. doi: 10.1084/jem.40.2.189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Apostoli P, Bellini A, Porru S, Bisanti L. The effect of lead on male fertility: a time to pregnancy (TTP) study. Am J Ind Med. 2000;38(3):310–5. doi: 10.1002/1097-0274(200009)38:3<310::aid-ajim10>3.0.co;2-9. [DOI] [PubMed] [Google Scholar]
- 8.Chetty CS, Reddy GR, Murthy KS, Johnson J, Sajwan K, Desaiah D. Perinatal lead exposure alters the expression of neuronal nitric oxide synthase in rat brain. Int J Toxicol. 2001;20(3):113–20. doi: 10.1080/109158101317097692. [DOI] [PubMed] [Google Scholar]
- 9.Moore MR, Goldberg A, Yeung-Laiwah AA. Lead effects on the heme biosynthetic pathway. Relationship to toxicity. Ann N Y Acad Sci. 1987;514:191–203. doi: 10.1111/j.1749-6632.1987.tb48774.x. [DOI] [PubMed] [Google Scholar]
- 10.Dey PM, Burger J, Gochfeld M, Reuhl KR. Developmental lead exposure disturbs expression of synaptic neural cell adhesion molecules in herring gull brains. Toxicology. 2000;146(2–3):137–47. doi: 10.1016/s0300-483x(00)00171-2. [DOI] [PubMed] [Google Scholar]
- 11.Finkelstein Y, Markowitz ME, Rosen JF. Low-level lead-induced neurotoxicity in children: an update on central nervous system effects. Brain Res Brain Res Rev. 1998;27(2):168–76. doi: 10.1016/s0165-0173(98)00011-3. [DOI] [PubMed] [Google Scholar]
- 12.Canfield RL, Henderson CR, Jr., Cory-Slechta DA, Cox C, Jusko TA, Lanphear BP. Intellectual impairment in children with blood lead concentrations below 10 microg per deciliter. N Engl J Med. 2003;348(16):1517–26. doi: 10.1056/NEJMoa022848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bellinger D, Leviton A, Allred E, Rabinowitz M. Pre- and postnatal lead exposure and behavior problems in school-aged children. Environ Res. 1994;66(1):12–30. doi: 10.1006/enrs.1994.1041. [DOI] [PubMed] [Google Scholar]
- 14.Neal AP, Guilarte TR. Molecular neurobiology of lead (Pb(2+)): effects on synaptic function. Mol Neurobiol. 2010;42(3):151–60. doi: 10.1007/s12035-010-8146-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhang XL, Guariglia SR, McGlothan JL, Stansfield KH, Stanton PK, Guilarte TR. Presynaptic mechanisms of lead neurotoxicity: effects on vesicular release, vesicle clustering and mitochondria number. PLoS One. 2015;10(5):e0127461. doi: 10.1371/journal.pone.0127461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Neal AP, Guilarte TR. Mechanisms of lead and manganese neurotoxicity. Toxicol Res (Camb) 2013;2(2):99–114. doi: 10.1039/C2TX20064C. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Berridge MJ, Bootman MD, Roderick HL. Calcium signalling: dynamics, homeostasis and remodelling. Nature reviews. 2003;4(7):517–29. doi: 10.1038/nrm1155. [DOI] [PubMed] [Google Scholar]
- 18.Mikoshiba K, Hattori M. IP3 receptor-operated calcium entry. Sci STKE. 2000;2000(51):PE1. doi: 10.1126/stke.2000.51.pe1. [DOI] [PubMed] [Google Scholar]
- 19.Zeng W, Mak DO, Li Q, Shin DM, Foskett JK, Muallem S. A new mode of Ca2+ signaling by G protein-coupled receptors: gating of IP3 receptor Ca2+ release channels by Gbetagamma. Curr Biol. 2003;13(10):872–6. doi: 10.1016/s0960-9822(03)00330-0. [DOI] [PubMed] [Google Scholar]
- 20.Hamilton SL. Ryanodine receptors. Cell calcium. 2005;38(3–4):253–60. doi: 10.1016/j.ceca.2005.06.037. [DOI] [PubMed] [Google Scholar]
- 21.Putney JW, Jr., Ribeiro CM. Signaling pathways between the plasma membrane and endoplasmic reticulum calcium stores. Cell Mol Life Sci. 2000;57(8–9):1272–86. doi: 10.1007/PL00000765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Putney JW., Jr. Calcium signaling: up, down, up, down…what's the point? Science (New York, NY. 1998;279(5348):191–2. doi: 10.1126/science.279.5348.191. [DOI] [PubMed] [Google Scholar]
- 23.Haynes LP, Sherwood MW, Dolman NJ, Burgoyne RD. Specificity, promiscuity and localization of ARF protein interactions with NCS-1 and phosphatidylinositol-4 kinase-III beta. Traffic. 2007;8(8):1080–92. doi: 10.1111/j.1600-0854.2007.00594.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, et al. UCSF Chimera--a visualization system for exploratory research and analysis. J Comput Chem. 2004;25(13):1605–12. doi: 10.1002/jcc.20084. [DOI] [PubMed] [Google Scholar]
- 25.Deisseroth K, Heist EK, Tsien RW. Translocation of calmodulin to the nucleus supports CREB phosphorylation in hippocampal neurons. Nature. 1998;392(6672):198–202. doi: 10.1038/32448. [DOI] [PubMed] [Google Scholar]
- 26.Pang ZP, Cao P, Xu W, Sudhof TC. Calmodulin controls synaptic strength via presynaptic activation of calmodulin kinase II. J Neurosci. 2010;30(11):4132–42. doi: 10.1523/JNEUROSCI.3129-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hirota J, Michikawa T, Natsume T, Furuichi T, Mikoshiba K. Calmodulin inhibits inositol 1,4,5-trisphosphate-induced calcium release through the purified and reconstituted inositol 1,4,5-trisphosphate receptor type 1. FEBS Lett. 1999;456(2):322–6. doi: 10.1016/s0014-5793(99)00973-4. [DOI] [PubMed] [Google Scholar]
- 28.Iino M, Endo M. Calcium-dependent immediate feedback control of inositol 1,4,5-triphosphate-induced Ca2+ release. Nature. 1992;360(6399):76–8. doi: 10.1038/360076a0. [DOI] [PubMed] [Google Scholar]
- 29.Rusnak F, Mertz P. Calcineurin: form and function. Physiol Rev. 2000;80(4):1483–521. doi: 10.1152/physrev.2000.80.4.1483. [DOI] [PubMed] [Google Scholar]
- 30.Zhuo M, Zhang W, Son H, Mansuy I, Sobel RA, Seidman J, et al. A selective role of calcineurin aalpha in synaptic depotentiation in hippocampus. Proc Natl Acad Sci U S A. 1999;96(8):4650–5. doi: 10.1073/pnas.96.8.4650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ordaz B, Tang J, Xiao R, Salgado A, Sampieri A, Zhu MX, et al. Calmodulin and calcium interplay in the modulation of TRPC5 channel activity. Identification of a novel C-terminal domain for calcium/calmodulin-mediated facilitation. J Biol Chem. 2005;280(35):30788–96. doi: 10.1074/jbc.M504745200. [DOI] [PubMed] [Google Scholar]
- 32.Habermann E, Crowell K, Janicki P. Lead and other metals can substitute for Ca2+ in calmodulin. Arch Toxicol. 1983;54(1):61–70. doi: 10.1007/BF00277816. [DOI] [PubMed] [Google Scholar]
- 33.Tomsig JL, Suszkiw JB. Intracellular mechanism of Pb(2+)-induced norepinephrine release from bovine chromaffin cells. Am J Physiol. 1993;265(6 Pt 1):C1630–6. doi: 10.1152/ajpcell.1993.265.6.C1630. [DOI] [PubMed] [Google Scholar]
- 34.Sukumar P, Beech DJ. Stimulation of TRPC5 cationic channels by low micromolar concentrations of lead ions (Pb2+) Biochem Biophys Res Commun. 2010;393(1):50–4. doi: 10.1016/j.bbrc.2010.01.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Xu SZ, Zeng B, Daskoulidou N, Chen GL, Atkin SL, Lukhele B. Activation of TRPC cationic channels by mercurial compounds confers the cytotoxicity of mercury exposure. Toxicol Sci. 2012;125(1):56–68. doi: 10.1093/toxsci/kfr268. [DOI] [PubMed] [Google Scholar]
- 36.Sudhof TC, Rizo J. Synaptotagmins: C2-domain proteins that regulate membrane traffic. Neuron. 1996;17(3):379–88. doi: 10.1016/s0896-6273(00)80171-3. [DOI] [PubMed] [Google Scholar]
- 37.Fernandez-Chacon R, Konigstorfer A, Gerber SH, Garcia J, Matos MF, Stevens CF, et al. Synaptotagmin I functions as a calcium regulator of release probability. Nature. 2001;410(6824):41–9. doi: 10.1038/35065004. [DOI] [PubMed] [Google Scholar]
- 38.Kee Y, Scheller RH. Localization of synaptotagmin-binding domains on syntaxin. J Neurosci. 1996;16(6):1975–81. doi: 10.1523/JNEUROSCI.16-06-01975.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Chapman ER, Jahn R. Calcium-dependent interaction of the cytoplasmic region of synaptotagmin with membranes. Autonomous function of a single C2-homologous domain. J Biol Chem. 1994;269(8):5735–41. [PubMed] [Google Scholar]
- 40.Bouton CM, Frelin LP, Forde CE, Arnold Godwin H, Pevsner J. Synaptotagmin I is a molecular target for lead. J Neurochem. 2001;76(6):1724–35. doi: 10.1046/j.1471-4159.2001.00168.x. [DOI] [PubMed] [Google Scholar]
- 41.Garcia RA, Godwin HA. High metal concentrations are required for self-association of synaptotagmin II. Biophys J. 2004;86(4):2455–66. doi: 10.1016/S0006-3495(04)74302-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Pin JP, Galvez T, Prezeau L. Evolution, structure, and activation mechanism of family 3/C G-protein-coupled receptors. Pharmacology & therapeutics. 2003;98(3):325–54. doi: 10.1016/s0163-7258(03)00038-x. [DOI] [PubMed] [Google Scholar]
- 43.Gamo K, Kiryu-Seo S, Konishi H, Aoki S, Matsushima K, Wada K, et al. G-protein-coupled receptor screen reveals a role for chemokine receptor CCR5 in suppressing microglial neurotoxicity. J Neurosci. 2008;28(46):11980–8. doi: 10.1523/JNEUROSCI.2920-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.LeDoux JE. Emotion circuits in the brain. Annu Rev Neurosci. 2000;23:155–84. doi: 10.1146/annurev.neuro.23.1.155. [DOI] [PubMed] [Google Scholar]
- 45.Roozendaal B, McEwen BS, Chattarji S. Stress, memory and the amygdala. Nat Rev Neurosci. 2009;10(6):423–33. doi: 10.1038/nrn2651. [DOI] [PubMed] [Google Scholar]
- 46.Zhang RW, Wei HP, Xia YM, Du JL. Development of light response and GABAergic excitation-to-inhibition switch in zebrafish retinal ganglion cells. J Physiol. 2010;588(Pt 14):2557–69. doi: 10.1113/jphysiol.2010.187088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Xiao C, Gu Y, Zhou CY, Wang L, Zhang MM, Ruan DY. Pb2+ impairs GABAergic synaptic transmission in rat hippocampal slices: a possible involvement of presynaptic calcium channels. Brain Res. 2006;1088(1):93–100. doi: 10.1016/j.brainres.2006.03.005. [DOI] [PubMed] [Google Scholar]
- 48.Braga MF, Pereira EF, Albuquerque EX. Nanomolar concentrations of lead inhibit glutamatergic and GABAergic transmission in hippocampal neurons. Brain Res. 1999;826(1):22–34. doi: 10.1016/s0006-8993(99)01194-4. [DOI] [PubMed] [Google Scholar]
- 49.Neal AP, Worley PF, Guilarte TR. Lead exposure during synaptogenesis alters NMDA receptor targeting via NMDA receptor inhibition. Neurotoxicology. 2011;32(2):281–9. doi: 10.1016/j.neuro.2010.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Wirbisky SE, Weber GJ, Lee JW, Cannon JR, Freeman JL. Novel dose-dependent alterations in excitatory GABA during embryonic development associated with lead (Pb) neurotoxicity. Toxicol Lett. 2014;229(1):1–8. doi: 10.1016/j.toxlet.2014.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Jiang JY, Nagaraju M, Meyer RC, Zhang L, Hamelberg D, Hall RA, et al. Extracellular calcium modulates actions of orthosteric and allosteric ligands on metabotropic glutamate receptor 1alpha. J Biol Chem. 2014;289(3):1649–61. doi: 10.1074/jbc.M113.507665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kubo Y, Miyashita T, Murata Y. Structural basis for a Ca2+-sensing function of the metabotropic glutamate receptors. Science (New York, NY. 1998;279(5357):1722–5. doi: 10.1126/science.279.5357.1722. [DOI] [PubMed] [Google Scholar]
- 53.Lavreysen H, Janssen C, Bischoff F, Langlois X, Leysen JE, Lesage AS. [3H]R214127: a novel high-affinity radioligand for the mGlu1 receptor reveals a common binding site shared by multiple allosteric antagonists. Mol Pharmacol. 2003;63(5):1082–93. doi: 10.1124/mol.63.5.1082. [DOI] [PubMed] [Google Scholar]
- 54.Lindsley CW, Wisnoski DD, Leister WH, O'Brien JA, Lemaire W, Williams DL, Jr., et al. Discovery of positive allosteric modulators for the metabotropic glutamate receptor subtype 5 from a series of N-(1,3-diphenyl-1H- pyrazol-5-yl)benzamides that potentiate receptor function in vivo. J Med Chem. 2004;47(24):5825–8. doi: 10.1021/jm049400d. [DOI] [PubMed] [Google Scholar]
- 55.Conn PJ, Pin JP. Pharmacology and functions of metabotropic glutamate receptors. Annu Rev Pharmacol Toxicol. 1997;37:205–37. doi: 10.1146/annurev.pharmtox.37.1.205. [DOI] [PubMed] [Google Scholar]
- 56.Wang X, Kirberger M, Qiu F, Chen G, Yang JJ. Towards predicting Ca(2+)-binding sites with different coordination numbers in proteins with atomic resolution. Proteins. 2008;75(4):787–98. doi: 10.1002/prot.22285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wang X, Zhao K, Kirberger M, Wong H, Chen G, Yang J. Analysis and prediction of calcium-binding pockets from apo protein structures exhibiting calcium-induced localized conformational changes. Protein Sci. 2010 doi: 10.1002/pro.394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Markovac J, Goldstein GW. Picomolar concentrations of lead stimulate brain protein kinase C. Nature. 1988;334(6177):71–3. doi: 10.1038/334071a0. [DOI] [PubMed] [Google Scholar]
- 59.Hwang KY, Lee BK, Bressler JP, Bolla KI, Stewart WF, Schwartz BS. Protein kinase C activity and the relations between blood lead and neurobehavioral function in lead workers. Environ Health Perspect. 2002;110(2):133–8. doi: 10.1289/ehp.02110133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lee JH, Lee J, Choi KY, Hepp R, Lee JY, Lim MK, et al. Calmodulin dynamically regulates the trafficking of the metabotropic glutamate receptor mGluR5. Proc Natl Acad Sci U S A. 2008;105(34):12575–80. doi: 10.1073/pnas.0712033105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Nakajima Y, Yamamoto T, Nakayama T, Nakanishi S. A relationship between protein kinase C phosphorylation and calmodulin binding to the metabotropic glutamate receptor subtype 7. J Biol Chem. 1999;274(39):27573–7. doi: 10.1074/jbc.274.39.27573. [DOI] [PubMed] [Google Scholar]
- 62.Wang FS, Liu ZH, Zhang JS, Di JR. Effects of lead exposure on protein kinase C and calmodulin expression in hippocampus and neurobehavioral function of baby rats. Zhonghua lao dong wei sheng zhi ye bing za zhi = Zhonghua laodong weisheng zhiyebing zazhi = Chinese journal of industrial hygiene and occupational diseases. 2008;26(3):139–42. [PubMed] [Google Scholar]
- 63.Xu J, Yan HC, Yang B, Tong LS, Zou YX, Tian Y. Effects of lead exposure on hippocampal metabotropic glutamate receptor subtype 3 and 7 in developmental rats. Journal of negative results in biomedicine. 2009;8:5. doi: 10.1186/1477-5751-8-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Richardt G, Federolf G, Habermann E. Affinity of heavy metal ions to intracellular Ca2+-binding proteins. Biochem Pharmacol. 1986;35(8):1331–5. doi: 10.1016/0006-2952(86)90278-9. [DOI] [PubMed] [Google Scholar]
- 65.Hartmann M, Heumann R, Lessmann V. Synaptic secretion of BDNF after high-frequency stimulation of glutamatergic synapses. The EMBO journal. 2001;20(21):5887–97. doi: 10.1093/emboj/20.21.5887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Jiang X, Tian F, Mearow K, Okagaki P, Lipsky RH, Marini AM. The excitoprotective effect of N-methyl-D-aspartate receptors is mediated by a brain-derived neurotrophic factor autocrine loop in cultured hippocampal neurons. Journal of neurochemistry. 2005;94(3):713–22. doi: 10.1111/j.1471-4159.2005.03200.x. [DOI] [PubMed] [Google Scholar]
- 67.Vanhoutte P, Bading H. Opposing roles of synaptic and extrasynaptic NMDA receptors in neuronal calcium signalling and BDNF gene regulation. Current opinion in neurobiology. 2003;13(3):366–71. doi: 10.1016/s0959-4388(03)00073-4. [DOI] [PubMed] [Google Scholar]
- 68.Walz C, Jungling K, Lessmann V, Gottmann K. Presynaptic plasticity in an immature neocortical network requires NMDA receptor activation and BDNF release. Journal of neurophysiology. 2006;96(6):3512–6. doi: 10.1152/jn.00018.2006. [DOI] [PubMed] [Google Scholar]
- 69.Alkondon M, Costa AC, Radhakrishnan V, Aronstam RS, Albuquerque EX. Selective blockade of NMDA-activated channel currents may be implicated in learning deficits caused by lead. FEBS Lett. 1990;261(1):124–30. doi: 10.1016/0014-5793(90)80652-y. [DOI] [PubMed] [Google Scholar]
- 70.Guilarte TR, Miceli RC, Jett DA. Neurochemical aspects of hippocampal and cortical Pb2+ neurotoxicity. Neurotoxicology. 1994;15(3):459–66. [PubMed] [Google Scholar]
- 71.Hu B, Nikolakopoulou AM, Cohen-Cory S. BDNF stabilizes synapses and maintains the structural complexity of optic axons in vivo. Development. 2005;132(19):4285–98. doi: 10.1242/dev.02017. [DOI] [PubMed] [Google Scholar]
- 72.Gavazzo P, Zanardi I, Baranowska-Bosiacka I, Marchetti C. Molecular determinants of Pb2+ interaction with NMDA receptor channels. Neurochemistry international. 2008;52(1–2):329–37. doi: 10.1016/j.neuint.2007.07.003. [DOI] [PubMed] [Google Scholar]
- 73.Yamada Y, Ujihara H, Sada H, Ban T. Pb2+ reduces the current from NMDA receptors expressed in Xenopus oocytes. FEBS Lett. 1995;377(3):390–2. doi: 10.1016/0014-5793(95)01402-0. [DOI] [PubMed] [Google Scholar]
- 74.Omelchenko IA, Nelson CS, Marino JL, Allen CN. The sensitivity of N-methyl-D-aspartate receptors to lead inhibition is dependent on the receptor subunit composition. The Journal of pharmacology and experimental therapeutics. 1996;278(1):15–20. [PubMed] [Google Scholar]
- 75.Guilarte TR, McGlothan JL. Hippocampal NMDA receptor mRNA undergoes subunit specific changes during developmental lead exposure. Brain Res. 1998;790(1–2):98–107. doi: 10.1016/s0006-8993(98)00054-7. [DOI] [PubMed] [Google Scholar]
- 76.Nihei MK, Desmond NL, McGlothan JL, Kuhlmann AC, Guilarte TR. N-methyl-D-aspartate receptor subunit changes are associated with lead-induced deficits of long-term potentiation and spatial learning. Neuroscience. 2000;99(2):233–42. doi: 10.1016/s0306-4522(00)00192-5. [DOI] [PubMed] [Google Scholar]
- 77.Nihei MK, Guilarte TR. NMDAR-2A subunit protein expression is reduced in the hippocampus of rats exposed to Pb2+ during development. Brain research Molecular brain research. 1999;66(1–2):42–9. doi: 10.1016/s0169-328x(99)00005-4. [DOI] [PubMed] [Google Scholar]
- 78.Zhang XY, Liu AP, Ruan DY, Liu J. Effect of developmental lead exposure on the expression of specific NMDA receptor subunit mRNAs in the hippocampus of neonatal rats by digoxigenin-labeled in situ hybridization histochemistry. Neurotoxicology and teratology. 2002;24(2):149–60. doi: 10.1016/s0892-0362(01)00210-0. [DOI] [PubMed] [Google Scholar]
- 79.Guilarte TR, McGlothan JL. Selective decrease in NR1 subunit splice variant mRNA in the hippocampus of Pb2+-exposed rats: implications for synaptic targeting and cell surface expression of NMDAR complexes. Brain research Molecular brain research. 2003;113(1–2):37–43. doi: 10.1016/s0169-328x(03)00083-4. [DOI] [PubMed] [Google Scholar]
- 80.Toscano CD, Hashemzadeh-Gargari H, McGlothan JL, Guilarte TR. Developmental Pb2+ exposure alters NMDAR subtypes and reduces CREB phosphorylation in the rat brain. Brain research Developmental brain research. 2002;139(2):217–26. doi: 10.1016/s0165-3806(02)00569-2. [DOI] [PubMed] [Google Scholar]
- 81.Cordova FM, Rodrigues AL, Giacomelli MB, Oliveira CS, Posser T, Dunkley PR, et al. Lead stimulates ERK1/2 and p38MAPK phosphorylation in the hippocampus of immature rats. Brain Res. 2004;998(1):65–72. doi: 10.1016/j.brainres.2003.11.012. [DOI] [PubMed] [Google Scholar]
- 82.Toscano CD, O'Callaghan JP, Guilarte TR. Calcium/calmodulin-dependent protein kinase II activity and expression are altered in the hippocampus of Pb2+-exposed rats. Brain Res. 2005;1044(1):51–8. doi: 10.1016/j.brainres.2005.02.076. [DOI] [PubMed] [Google Scholar]
- 83.Toscano CD, McGlothan JL, Guilarte TR. Lead exposure alters cyclic-AMP response element binding protein phosphorylation and binding activity in the developing rat brain. Brain research Developmental brain research. 2003;145(2):219–28. doi: 10.1016/j.devbrainres.2003.08.004. [DOI] [PubMed] [Google Scholar]
- 84.Dason JS, Romero-Pozuelo J, Marin L, Iyengar BG, Klose MK, Ferrus A, et al. Frequenin/NCS-1 and the Ca2+-channel alpha1-subunit co-regulate synaptic transmission and nerve-terminal growth. J Cell Sci. 2009;122(Pt 22):4109–21. doi: 10.1242/jcs.055095. [DOI] [PubMed] [Google Scholar]
- 85.Hui H, McHugh D, Hannan M, Zeng F, Xu SZ, Khan SU, et al. Calcium-sensing mechanism in TRPC5 channels contributing to retardation of neurite outgrowth. The Journal of physiology. 2006;572(Pt 1):165–72. doi: 10.1113/jphysiol.2005.102889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Nakamura TY, Jeromin A, Smith G, Kurushima H, Koga H, Nakabeppu Y, et al. Novel role of neuronal Ca2+ sensor-1 as a survival factor up-regulated in injured neurons. J Cell Biol. 2006;172(7):1081–91. doi: 10.1083/jcb.200508156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Gomez M, De Castro E, Guarin E, Sasakura H, Kuhara A, Mori I, et al. Ca2+ signaling via the neuronal calcium sensor-1 regulates associative learning and memory in C. elegans. Neuron. 2001;30(1):241–8. doi: 10.1016/s0896-6273(01)00276-8. [DOI] [PubMed] [Google Scholar]
- 88.Pandalaneni S, Karuppiah V, Saleem M, Haynes LP, Burgoyne RD, Mayans O, et al. Neuronal Calcium Sensor-1 Binds the D2 Dopamine Receptor and G-protein-coupled Receptor Kinase 1 (GRK1) Peptides Using Different Modes of Interactions. J Biol Chem. 2015;290(30):18744–56. doi: 10.1074/jbc.M114.627059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.de Barry J, Janoshazi A, Dupont JL, Procksch O, Chasserot-Golaz S, Jeromin A, et al. Functional implication of neuronal calcium sensor-1 and phosphoinositol 4-kinase-beta interaction in regulated exocytosis of PC12 cells. J Biol Chem. 2006;281(26):18098–111. doi: 10.1074/jbc.M509842200. [DOI] [PubMed] [Google Scholar]
- 90.Haynes LP, Thomas GM, Burgoyne RD. Interaction of neuronal calcium sensor-1 and ADP-ribosylation factor 1 allows bidirectional control of phosphatidylinositol 4-kinase beta and trans-Golgi network-plasma membrane traffic. J Biol Chem. 2005;280(7):6047–54. doi: 10.1074/jbc.M413090200. [DOI] [PubMed] [Google Scholar]
- 91.Pasqualato S, Renault L, Cherfils J. Arf, Arl, Arp and Sar proteins: a family of GTP-binding proteins with a structural device for 'front-back' communication. EMBO Rep. 2002;3(11):1035–41. doi: 10.1093/embo-reports/kvf221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Donaldson JG, Honda A, Weigert R. Multiple activities for Arf1 at the Golgi complex. Biochim Biophys Acta. 2005;1744(3):364–73. doi: 10.1016/j.bbamcr.2005.03.001. [DOI] [PubMed] [Google Scholar]
- 93.Speidel D, Varoqueaux F, Enk C, Nojiri M, Grishanin RN, Martin TF, et al. A family of Ca2+-dependent activator proteins for secretion: comparative analysis of structure, expression, localization, and function. J Biol Chem. 2003;278(52):52802–9. doi: 10.1074/jbc.M304727200. [DOI] [PubMed] [Google Scholar]
- 94.Johansson PA, Dziegielewska KM, Liddelow SA, Saunders NR. The blood-CSF barrier explained: when development is not immaturity. Bioessays. 2008;30(3):237–48. doi: 10.1002/bies.20718. [DOI] [PubMed] [Google Scholar]
- 95.Moccia F, Zuccolo E, Soda T, Tanzi F, Guerra G, Mapelli L, et al. Stim and Orai proteins in neuronal Ca(2+) signaling and excitability. Front Cell Neurosci. 2015;9:153. doi: 10.3389/fncel.2015.00153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Suzuki Y, Chao SH, Zysk JR, Cheung WY. Stimulation of calmodulin by cadmium ion. Arch Toxicol. 1985;57(3):205–11. doi: 10.1007/BF00290889. [DOI] [PubMed] [Google Scholar]
- 97.Chao SH, Bu CH, Cheung WY. Stimulation of myosin light-chain kinase by Cd2+ and Pb2+ Arch Toxicol. 1995;69(3):197–203. doi: 10.1007/s002040050158. [DOI] [PubMed] [Google Scholar]
- 98.Chao SH, Bu CH, Cheung WY. Activation of troponin C by Cd2+ and Pb2+ Arch Toxicol. 1990;64(6):490–6. doi: 10.1007/BF01977632. [DOI] [PubMed] [Google Scholar]
- 99.Ouyang H, Vogel HJ. Metal ion binding to calmodulin: NMR and fluorescence studies. Biometals. 1998;11(3):213–22. doi: 10.1023/a:1009226215543. [DOI] [PubMed] [Google Scholar]
- 100.Milos M, Comte M, Schaer JJ, Cox JA. Evidence for four capital and six auxiliary cation-binding sites on calmodulin: divalent cation interactions monitored by direct binding and microcalorimetry. J Inorg Biochem. 1989;36(1):11–25. doi: 10.1016/0162-0134(89)80009-1. [DOI] [PubMed] [Google Scholar]
- 101.Chao SH, Suzuki Y, Zysk JR, Cheung WY. Activation of calmodulin by various metal cations as a function of ionic radius. Mol Pharmacol. 1984;26(1):75–82. [PubMed] [Google Scholar]
- 102.Andersen RJ, diTargiani RC, Hancock RD, Stern CL, Goldberg DP, Godwin HA. Characterization of the first N2S(alkylthiolate)lead compound: a model for three-coordinate lead in biological systems. Inorg Chem. 2006;45(17):6574–6. doi: 10.1021/ic060497z. [DOI] [PubMed] [Google Scholar]
- 103.Erskine PT, Senior N, Awan S, Lambert R, Lewis G, Tickle IJ, et al. X-ray structure of 5-aminolaevulinate dehydratase, a hybrid aldolase. Nat Struct Biol. 1997;4(12):1025–31. doi: 10.1038/nsb1297-1025. [DOI] [PubMed] [Google Scholar]
- 104.Harding MM. The architecture of metal coordination groups in proteins. Acta Crystallogr D Biol Crystallogr. 2004;60(Pt 5):849–59. doi: 10.1107/S0907444904004081. [DOI] [PubMed] [Google Scholar]
- 105.Jaffe EK, Martins J, Li J, Kervinen J, Dunbrack RL., Jr The molecular mechanism of lead inhibition of human porphobilinogen synthase. J Biol Chem. 2001;276(2):1531–7. doi: 10.1074/jbc.M007663200. [DOI] [PubMed] [Google Scholar]
- 106.Koca J, Zhan CG, Rittenhouse RC, Ornstein RL. Coordination number of zinc ions in the phosphotriesterase active site by molecular dynamics and quantum mechanics. J Comput Chem. 2003;24(3):368–78. doi: 10.1002/jcc.10217. [DOI] [PubMed] [Google Scholar]
- 107.Magyar JS, Weng TC, Stern CM, Dye DF, Rous BW, Payne JC, et al. Reexamination of lead(II) coordination preferences in sulfur-rich sites: implications for a critical mechanism of lead poisoning. J Am Chem Soc. 2005;127(26):9495–505. doi: 10.1021/ja0424530. [DOI] [PubMed] [Google Scholar]
- 108.Shimoni-Livny L, Glusker JP, Bock CW. Lone pair functionality in divalent lead compounds. Inorg Chem. 1998;37(8):1853–67. [Google Scholar]
- 109.Silva JJRFd, Williams RJP. The biological chemistry of the elements : the inorganic chemistry of life. 2nd Oxford University Press; Oxford; New York: 2001. p. xvii.p. 575. [Google Scholar]
- 110.Heinz U, Kiefer M, Tholey A, Adolph HW. On the competition for available zinc. J Biol Chem. 2005;280(5):3197–207. doi: 10.1074/jbc.M409425200. [DOI] [PubMed] [Google Scholar]
- 111.Tang S, Reddish F, Zhuo Y, Yang JJ. Fast kinetics of calcium signaling and sensor design. Current opinion in chemical biology. 2015;27:90–7. doi: 10.1016/j.cbpa.2015.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kirberger M, Yang JJ. Structural differences between Pb(2+)- and Ca(2+)-binding sites in proteins: Implications with respect to toxicity. J Inorg Biochem. 2008 doi: 10.1016/j.jinorgbio.2008.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Kirberger M, Wang X, Deng H, Yang W, Chen G, Yang JJ. Statistical analysis of structural characteristics of protein Ca(2+)-binding sites. J Biol Inorg Chem. 2008 doi: 10.1007/s00775-008-0402-7. [DOI] [PubMed] [Google Scholar]
- 114.Kirberger M, Yang J. In: Calcium-Binding Sites in Proteins: Types. Kretsinger R, Uversky V, Permyakove E, editors. Encyclopedia of Metalloproteins: Springer; 2011. [Google Scholar]
- 115.Kursula P, Majava V. A structural insight into lead neurotoxicity and calmodulin activation by heavy metals. Acta crystallographica. 2007;63(Pt 8):653–6. doi: 10.1107/S1744309107034525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Chattopadhyaya R, Meador WE, Means AR, Quiocho FA. Calmodulin structure refined at 1.7 A resolution. Journal of molecular biology. 1992;228(4):1177–92. doi: 10.1016/0022-2836(92)90324-d. [DOI] [PubMed] [Google Scholar]
- 117.Wilson MA, Brunger AT. The 1.0 A crystal structure of Ca(2+)-bound calmodulin: an analysis of disorder and implications for functionally relevant plasticity. Journal of molecular biology. 2000;301(5):1237–56. doi: 10.1006/jmbi.2000.4029. [DOI] [PubMed] [Google Scholar]
- 118.Jaren OR, Kranz JK, Sorensen BR, Wand AJ, Shea MA. Calcium-induced conformational switching of Paramecium calmodulin provides evidence for domain coupling. Biochemistry. 2002;41(48):14158–66. doi: 10.1021/bi026340+. [DOI] [PubMed] [Google Scholar]
- 119.Vogel HJ. The Merck Frosst Award Lecture 1994. Calmodulin: a versatile calcium mediator protein. Biochemistry and cell biology = Biochimie et biologie cellulaire. 1994;72(9–10):357–76. [PubMed] [Google Scholar]
- 120.Kursula P. Crystallographic snapshots of initial steps in the collapse of the calmodulin central helix. Acta Crystallogr D Biol Crystallogr. 2014;70(Pt 1):24–30. doi: 10.1107/S1399004713024437. [DOI] [PubMed] [Google Scholar]
- 121.Dowd TL, Li L, Gundberg CM. The (1)H NMR structure of bovine Pb(2+)-osteocalcin and implications for lead toxicity. Biochimica et biophysica acta. 2008;1784(11):1534–45. doi: 10.1016/j.bbapap.2008.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Astegno A, La Verde V, Marino V, Dell'Orco D, Dominici P. Biochemical and biophysical characterization of a plant calmodulin: Role of the N- and C-lobes in calcium binding, conformational change, and target interaction. Biochim Biophys Acta. 2016;1864(3):297–307. doi: 10.1016/j.bbapap.2015.12.003. [DOI] [PubMed] [Google Scholar]
- 123.Kirberger M, Wong HC, Jiang J, Yang JJ. Metal toxicity and opportunistic binding of Pb(2+) in proteins. J Inorg Biochem. 2013;125:40–9. doi: 10.1016/j.jinorgbio.2013.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Perm*i*akov EA, Kretsinger RH. Calcium binding proteins. Wiley; Hoboken, N.J: 2010. [Google Scholar]
- 125.Wimberly B, Thulin E, Chazin WJ. Characterization of the N-terminal half-saturated state of calbindin D9k: NMR studies of the N56A mutant. Protein Sci. 1995;4(6):1045–55. doi: 10.1002/pro.5560040603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Ye Y, Lee HW, Yang W, Shealy S, Yang JJ. Probing site-specific calmodulin calcium and lanthanide affinity by grafting. J Am Chem Soc. 2005;127(11):3743–50. doi: 10.1021/ja042786x. [DOI] [PubMed] [Google Scholar]
- 127.Forsen S, Linse S, Drakenberg T, Kordel J, Akke M, Sellers P, et al. Ca2+ binding in proteins of the calmodulin superfamily: cooperativity, electrostatic contributions and molecular mechanisms. Ciba Found Symp. 1991;161:222–36. doi: 10.1002/9780470514146.ch14. [DOI] [PubMed] [Google Scholar]
- 128.Evans TI, Shea MA. Energetics of calmodulin domain interactions with the calmodulin binding domain of CaMKII. Proteins. 2009;76(1):47–61. doi: 10.1002/prot.22317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Chazin WJ. Relating form and function of EF-hand calcium binding proteins. Acc Chem Res. 2011;44(3):171–9. doi: 10.1021/ar100110d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Linse S, Helmersson A, Forsen S. Calcium binding to calmodulin and its globular domains. J Biol Chem. 1991;266(13):8050–4. [PubMed] [Google Scholar]
- 131.Sorensen BR, Shea MA. Interactions between domains of apo calmodulin alter calcium binding and stability. Biochemistry. 1998;37(12):4244–53. doi: 10.1021/bi9718200. [DOI] [PubMed] [Google Scholar]
- 132.VanScyoc WS, Sorensen BR, Rusinova E, Laws WR, Ross JB, Shea MA. Calcium binding to calmodulin mutants monitored by domain-specific intrinsic phenylalanine and tyrosine fluorescence. Biophys J. 2002;83(5):2767–80. doi: 10.1016/S0006-3495(02)75286-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Jiang J, Zhou Y, Zou J, Chen Y, Patel P, Yang JJ, et al. Site-specific modification of calmodulin Ca(2)(+) affinity tunes the skeletal muscle ryanodine receptor activation profile. Biochem J. 2010;432(1):89–99. doi: 10.1042/BJ20100505. [DOI] [PubMed] [Google Scholar]
- 134.Bertini I, Gelis I, Katsaros N, Luchinat C, Provenzani A. Tuning the affinity for lanthanides of calcium binding proteins. Biochemistry. 2003;42(26):8011–21. doi: 10.1021/bi034494z. [DOI] [PubMed] [Google Scholar]
- 135.Kim Y, Yoo CI, Lee CR, Lee JH, Lee H, Kim SR, et al. Evaluation of activity of erythrocyte pyrimidine 5'-nucleotidase (P5N) in lead exposed workers: with focus on the effect on hemoglobin. Ind Health. 2002;40(1):23–7. doi: 10.2486/indhealth.40.23. [DOI] [PubMed] [Google Scholar]
- 136.Bergdahl IA, Grubb A, Schutz A, Desnick RJ, Wetmur JG, Sassa S, et al. Lead binding to delta-aminolevulinic acid dehydratase (ALAD) in human erythrocytes. Pharmacol Toxicol. 1997;81(4):153–8. doi: 10.1111/j.1600-0773.1997.tb02061.x. [DOI] [PubMed] [Google Scholar]
- 137.Kelada SN, Shelton E, Kaufmann RB, Khoury MJ. Delta-aminolevulinic acid dehydratase genotype and lead toxicity: a HuGE review. American journal of epidemiology. 2001;154(1):1–13. doi: 10.1093/aje/154.1.1. [DOI] [PubMed] [Google Scholar]
- 138.Ballatori N. Transport of toxic metals by molecular mimicry. Environmental health perspectives. 2002;110(Suppl 5):689–94. doi: 10.1289/ehp.02110s5689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Bannon DI, Abounader R, Lees PS, Bressler JP. Effect of DMT1 knockdown on iron, cadmium, and lead uptake in Caco-2 cells. Am J Physiol Cell Physiol. 2003;284(1):C44–50. doi: 10.1152/ajpcell.00184.2002. [DOI] [PubMed] [Google Scholar]
- 140.Atchison WD. Effects of toxic environmental contaminants on voltage-gated calcium channel function: from past to present. J Bioenerg Biomembr. 2003;35(6):507–32. doi: 10.1023/b:jobb.0000008023.11211.13. [DOI] [PubMed] [Google Scholar]
- 141.Forsen S, Thulin E, Lilja H. 113Cd NMR in the study of calcium binding proteins: troponin C. FEBS Lett. 1979;104(1):123–6. doi: 10.1016/0014-5793(79)81097-2. [DOI] [PubMed] [Google Scholar]
- 147.Mills JS, Johnson JD. Metal ions as allosteric regulators of calmodulin. The Journal of biological chemistry. 1985;260(28):15100–5. [PubMed] [Google Scholar]
- 143.Pidcock E, Moore GR. Structural characteristics of protein binding sites for calcium and lanthanide ions. J Biol Inorg Chem. 2001;6(5–6):479–89. doi: 10.1007/s007750100214. [DOI] [PubMed] [Google Scholar]
- 144.Wang CL, Leavis PC, Gergely J. Kinetic studies show that Ca2+ and Tb3+ have different binding preferences toward the four Ca2+-binding sites of calmodulin. Biochemistry. 1984;23(26):6410–5. doi: 10.1021/bi00321a020. [DOI] [PubMed] [Google Scholar]
- 145.Yang W, Wilkins AL, Ye Y, Liu ZR, Li SY, Urbauer JL, et al. Design of a calcium-binding protein with desired structure in a cell adhesion molecule. J Am Chem Soc. 2005;127(7):2085–93. doi: 10.1021/ja0431307. [DOI] [PubMed] [Google Scholar]
- 146.Bridges CC, Zalups RK. Molecular and ionic mimicry and the transport of toxic metals. Toxicol Appl Pharmacol. 2005;204(3):274–308. doi: 10.1016/j.taap.2004.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Goldstein GW, Ar D. Lead activates calmodulin sensitive processes. Life Sci. 1983;33(10):1001–6. doi: 10.1016/0024-3205(83)90757-9. [DOI] [PubMed] [Google Scholar]
- 148.Kern M, Wisniewski M, Cabell L, Audesirk G. Inorganic lead and calcium interact positively in activation of calmodulin. Neurotoxicology. 2000;21(3):353–63. [PubMed] [Google Scholar]
- 149.Shirran SL, Barran PE. The use of ESI-MS to probe the binding of divalent cations to calmodulin. Journal of the American Society for Mass Spectrometry. 2009;20(6):1159–71. doi: 10.1016/j.jasms.2009.02.008. [DOI] [PubMed] [Google Scholar]
- 150.Raos N, Kasprzak KS. Allosteric binding of nickel(II) to calmodulin. Fundam Appl Toxicol. 1989;13(4):816–22. doi: 10.1016/0272-0590(89)90336-9. [DOI] [PubMed] [Google Scholar]
- 151.Lasley SM, Gilbert ME. Presynaptic glutamatergic function in dentate gyrus in vivo is diminished by chronic exposure to inorganic lead. Brain Res. 1996;736(1–2):125–34. doi: 10.1016/0006-8993(96)00666-x. [DOI] [PubMed] [Google Scholar]
- 152.Lasley SM, Gilbert ME. Rat hippocampal glutamate and GABA release exhibit biphasic effects as a function of chronic lead exposure level. Toxicol Sci. 2002;66(1):139–47. doi: 10.1093/toxsci/66.1.139. [DOI] [PubMed] [Google Scholar]
- 153.Ferguson C, Kern M, Audesirk G. Nanomolar concentrations of inorganic lead increase Ca2+ efflux and decrease intracellular free Ca2+ ion concentrations in cultured rat hippocampal neurons by a calmodulin-dependent mechanism. Neurotoxicology. 2000;21(3):365–78. [PubMed] [Google Scholar]
- 154.Peng S, Hajela RK, Atchison WD. Characteristics of block by Pb2+ of function of human neuronal L-, N-, and R-type Ca2+ channels transiently expressed in human embryonic kidney 293 cells. Mol Pharmacol. 2002;62(6):1418–30. doi: 10.1124/mol.62.6.1418. [DOI] [PubMed] [Google Scholar]
- 155.Marchetti C. Molecular targets of lead in brain neurotoxicity. Neurotox Res. 2003;5(3):221–36. doi: 10.1007/BF03033142. [DOI] [PubMed] [Google Scholar]
- 156.Brown EM, Gamba G, Riccardi D, Lombardi M, Butters R, Kifor O, et al. Cloning and characterization of an extracellular Ca(2+)-sensing receptor from bovine parathyroid. Nature. 1993;366(6455):575–80. doi: 10.1038/366575a0. [DOI] [PubMed] [Google Scholar]
- 157.Fullmer CS, Edelstein S, Wasserman RH. Lead-binding properties of intestinal calcium-binding proteins. J Biol Chem. 1985;260(11):6816–9. [PubMed] [Google Scholar]
- 158.Handlogten ME, Shiraishi N, Awata H, Huang C, Miller RT. Extracellular Ca(2+)-sensing receptor is a promiscuous divalent cation sensor that responds to lead. Am J Physiol Renal Physiol. 2000;279(6):F1083–91. doi: 10.1152/ajprenal.2000.279.6.F1083. [DOI] [PubMed] [Google Scholar]
- 159.Ruat M, Snowman AM, Hester LD, Snyder SH. Cloned and expressed rat Ca2+-sensing receptor. J Biol Chem. 1996;271(11):5972–5. doi: 10.1074/jbc.271.11.5972. [DOI] [PubMed] [Google Scholar]
- 160.Buras RR, Shabahang M, Davoodi F, Schumaker LM, Cullen KJ, Byers S, et al. The effect of extracellular calcium on colonocytes: evidence for differential responsiveness based upon degree of cell differentiation. Cell proliferation. 1995;28(4):245–62. doi: 10.1111/j.1365-2184.1995.tb00067.x. [DOI] [PubMed] [Google Scholar]
- 161.Chattopadhyay N, Ye CP, Yamaguchi T, Kifor O, Vassilev PM, Nishimura R, et al. Extracellular calcium-sensing receptor in rat oligodendrocytes: expression and potential role in regulation of cellular proliferation and an outward K+ channel. Glia. 1998;24(4):449–58. [PubMed] [Google Scholar]
- 162.Wada M, Furuya Y, Sakiyama J, Kobayashi N, Miyata S, Ishii H, et al. The calcimimetic compound NPS R-568 suppresses parathyroid cell proliferation in rats with renal insufficiency. Control of parathyroid cell growth via a calcium receptor. The Journal of clinical investigation. 1997;100(12):2977–83. doi: 10.1172/JCI119851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Asokan SK. Experimental lead cardiomyopathy: myocardial structural changes in rats given small amounts of lead. The Journal of laboratory and clinical medicine. 1974;84(1):20–5. [PubMed] [Google Scholar]
- 164.Khan MY, Buse M, Louria DB. Lead cardiomyopathy in mice: a correlative ultrastructural and blood level study. Archives of pathology & laboratory medicine. 1977;101(2):89–94. [PubMed] [Google Scholar]
- 165.Navas-Acien A, Guallar E, Silbergeld EK, Rothenberg SJ. Lead exposure and cardiovascular disease--a systematic review. Environ Health Perspect. 2007;115(3):472–82. doi: 10.1289/ehp.9785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Westerink RH, Klompmakers AA, Westenberg HG, Vijverberg HP. Signaling pathways involved in Ca2+- and Pb2+-induced vesicular catecholamine release from rat PC12 cells. Brain Res. 2002;957(1):25–36. doi: 10.1016/s0006-8993(02)03580-1. [DOI] [PubMed] [Google Scholar]
- 167.Jusko TA, Henderson CR, Lanphear BP, Cory-Slechta DA, Parsons PJ, Canfield RL. Blood lead concentrations < 10 microg/dL and child intelligence at 6 years of age. Environ Health Perspect. 2008;116(2):243–8. doi: 10.1289/ehp.10424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Lanphear BP, Hornung R, Khoury J, Yolton K, Baghurst P, Bellinger DC, et al. Low-level environmental lead exposure and children's intellectual function: an international pooled analysis. Environ Health Perspect. 2005;113(7):894–9. doi: 10.1289/ehp.7688. [DOI] [PMC free article] [PubMed] [Google Scholar]

