Comprehensive Physiology Wiley Online Library

The Cardiac Na+‐Ca2+ Exchanger: From Structure to Function

Full Article on Wiley Online Library



Abstract

Ca2+ homeostasis is essential for cell function and survival. As such, the cytosolic Ca2+ concentration is tightly controlled by a wide number of specialized Ca2+ handling proteins. One among them is the Na+‐Ca2+ exchanger (NCX), a ubiquitous plasma membrane transporter that exploits the electrochemical gradient of Na+ to drive Ca2+ out of the cell, against its concentration gradient. In this critical role, this secondary transporter guides vital physiological processes such as Ca2+ homeostasis, muscle contraction, bone formation, and memory to name a few.

Herein, we review the progress made in recent years about the structure of the mammalian NCX and how it relates to function. Particular emphasis will be given to the mammalian cardiac isoform, NCX1.1, due to the extensive studies conducted on this protein. Given the degree of conservation among the eukaryotic exchangers, the information highlighted herein will provide a foundation for our understanding of this transporter family. We will discuss gene structure, alternative splicing, topology, regulatory mechanisms, and NCX's functional role on cardiac physiology. Throughout this article, we will attempt to highlight important milestones in the field and controversial topics where future studies are required. © 2021 American Physiological Society. Compr Physiol 12:1‐37, 2021.

Figure 1. Figure 1. Modes of operation of the Na+‐Ca2+ exchanger. (A) In the forward mode (top) NCX transports one Ca2+ ion out of the cell in exchange for three Na+ ions entering. One net charge is moved across the membrane resulting in a measurable ionic current. This is the most physiologically relevant cycle. In the reverse mode (bottom), one Ca2+ ions enter the cell and three Na+ ions exit. The direction in which Ca2+ is moved is governed by both the ionic gradients of Na+ and Ca2+ and membrane potential 164. (B) The scheme shows NCX transport cycle. This electrogenic transporter transitions between the outward and inward‐facing states via intermediate occluding states, leading to the coupled but opposite movements of Na+ and Ca2+ ions across the plasma membrane 164.
Figure 2. Figure 2. Na+‐Ca2+ exchanger gene organization. Each panel depicts the chromosomal location, number of exons, size, mRNA example, and corresponding protein of the three human isoforms. Gene exon structures are derived from Ensembl. Exons are indicated as grey bars and include both coding and noncoding exons. Exons undergoing alternative splicing are shown in color and are named in accordance with Quednau and colleagues 220. 5′ and 3′ regions of the gene are not included so that the gene size given extends from the beginning of the first exon and ends at the 3′ end of the last exon. Of special note, NCX2 does not undergo alternative splicing and all exons encode for protein. NCX1 and NCX3 have one noncoding exon, both undergo alternative splicing. An example of mRNA profile of the indicated exchangers is shown under each corresponding gene. Graphs are to scale. The alternative spliced exons (in colors), which at the protein level result in changes in the calcium‐binding domain 2 (CBD2) 220, are depicted in blue.
Figure 3. Figure 3. Tissue distribution of the exchanger isoforms. The three exchanger isoforms and their corresponding splice variants are differentially distributed within tissues 133,136,143,220,221. Examples are given for heart, brain, kidney, and skeletal muscle. The tissue distribution has been assessed using reverse transcriptase‐polymerase chain reaction as described in Ref. 220. Numerous evidence indicates that the isoform NCX1.1 136,143,220 is the only exchanger expressed in cardiomyocytes, although a study has identified isoforms NCX1.3 and NCX1.4 133. The splice variants NCX1.6 (ACD) and NCX3.3 (BC) are absent from the table. These exchangers have been localized in the brain 136,220, but their cell specific expression is undetermined.
Figure 4. Figure 4. Progression of Na+‐Ca2+ exchanger topology. The cartoons show how the topology of the mammalian exchanger has evolved through the years since its cloning in 1990 185. (A) Initially, NCX1.1 was predicted to be organized into 12 transmembrane segments (TMS). The two highly conserved regions among the exchangers which are known as α‐repeats 245 are highlighted. These regions are essential for ion transport 183,184,195. (B) Subsequent studies 50,60,106,239 revealed that the first helix was a signal peptide not necessary for activity, restricting the mature protein to 11 TMSs with an extracellular N terminus. (C) Cysteine scanning mutagenesis and epitope tagging were then applied to investigate NCX1.1 architecture. Based on these studies the topology of NCX was reorganized into nine transmembrane segments with an intracellular C‐terminus and two reentrant loops between TMSs 2‐3 and 7‐8 35,112,114,186,187,212. (D) The crystal structure of the archaebacterial exchanger (NCX_Mj) 150 has revealed a protein of 10 TMSs, contrasting with the predicted 9 TMSs for the mammalian exchanger as shown in (C). (E) Topology of the mammalian NCX based on NCX_Mj organization. The new architecture does not include the previously modeled reentrant loops, which are now predicted to be a short extracellular loop between TMSs 2‐3, within the α1‐repeat, and a new transmembrane segment (TMS 8) in the α2‐repeat. Future studies should be conducted to confirm the absence of either or both of these two reentrant structures and to validate that the mammalian exchanger and the archaebacterial are had a similar organization. The position of the cysteine residues involved in the intramolecular disulfide bond 241 is also shown. Amino acids are numbered based on the protein lacking the signal sequence.
Figure 5. Figure 5. Inconsistencies between the topology of the Na+‐Ca2+ exchanger and the archaebacterial exchanger. (A) Cysteine scanning mutagenesis and epitope mapping studies have been instrumental in deciphering the topology of the cardiac exchanger 35,50,112,114,186,187,212. The figure shows the amino acids that were replaced with cysteine and found accessible to membrane‐impermeable sulfhydryl agents either from the cytoplasm (blue circles) or the external side of the membrane (orange circles). Based on these investigations, NCX was modeled to have an extracellular N‐terminus, and a cytoplasmic C‐terminus. Moreover, residues 120 and 125 within the α1‐repeat were found to be exposed to the intracellular environment as well as several residues within the α2‐repeat, constraining these regions into reentrant loops. This organization corresponds to the one shown in Figure 4C. (B) With the atomic structure of the archaebacterial exchanger at hand 150, NCX1.1 is now assumed to be organized in 10 transmembrane segments 228,265, as shown in Figure 4E. However, this new arrangement places several residues previously found accessible to the cytoplasmic application of sulfhydryl reagents (blue circles), now facing the extracellular environment, including the C‐terminus. Note that the new architecture does not include reentrant loops. More investigations are needed to fully elucidate the topology of the mammalian exchanger.
Figure 6. Figure 6. Sequence alignment of the α‐repeats. (A) The sequence alignment illustrates the conservation of the α repeat regions within the Na+‐Ca2+ exchanger family. This is consistent with a broad range of evidence underlying their essential role in ion transport 125,183,184,195. The α1‐repeat encompasses residues 96 to 150 of NCX1.1, while the α2‐repeat stretches from 797 to 849 (amino acid numbering of NCX1.1 without the signal sequence). These boundaries were selected as previously described in Ref. 245. The residues that coordinate Na+ and Ca2+ ions, as revealed by the atomic structure of the archaebacterial exchanger 150, are indicated with a red arrow. (B) In addition to the conservation between species, the α‐repeats show intramolecular homology. This forms the foundation for the idea that exchangers are a result of gene duplication.
Figure 7. Figure 7. Domains and residues relevant for NCX1.1 regulation. (A) Highlighted are the residues of the canine cardiac Na+‐Ca2+ exchanger found important for its regulation. For a detailed role of these regions in controlling NCX transport activity please see the main text. Briefly, the residues shown in pink constitute the XIP region, which is intimately involved with the Na+‐dependent inactivation 166. The two helix bundle domain (THB) follows in yellow and its structure is shown in (B) (PDB# 6BV7) 295. The role of this domain remains to be determined. The residues encompassing the two Ca2+ regulatory domains are shown in light blue (Ca2+‐binding domain 1, CBD1) and dark blue (Ca2+‐binding domain 2, CBD2), their atomic structure is shown in (C) and (D), respectively [CBD1 PDB# 2PDK 189 and CBD2 PDB# 2QVM 16]. At the distal end of the large cytoplasmic loop, there is a stretch of residues (in dark grey) forming an amphipathic helix, required for palmitoylation of cysteine at position 739 207. Single point mutations that affect Na+, Ca2+, and pH regulation are indicated.
Figure 8. Figure 8. The XIP region is essential for Na+‐dependent inactivation. (A) Sequence alignment of the XIP region (amino acids 219 to 238, note NCX_Mj has no XIP region). (B) Mutations within this region either accelerate or abrogate the time course of current decay due to raised intracellular Na+, so‐called “Na+‐dependent inactivation”. Modified, with permission, from Matsuoka S, et al., 1997 166.
Figure 9. Figure 9. Properties of the exchanger Ca2+‐binding domain 1 (CBD1). (A) Protein sequence alignment of the CBD1 region among human exchanger isoforms. Numbering is reported according to NCX1.1 sequence, excluding the signal peptide. Residues involved in ion coordination are highlighted with red arrows. (B) This domain coordinates 4 Ca2+ ions (PDB code 2DPK, Ca2+ ions are shown in yellow) 189 but only the ion coordinating sites Ca3 and Ca4 have been shown to be relevant for regulation of the cardiac exchanger 196. (C) The ionic current generated by the cardiac exchanger can be evaluated using the giant patch technique 100. Shown is the Ca2+ dependency of WT NCX1.1 ionic currents and the indicated mutants 196. Mutations E385A and D446A‐D447A disrupt the binding of Ca2+ from sites 3 and 4 respectively and decreases the sensitivity of NCX1.1 to cytoplasmic Ca2+ of approximately 10‐fold.
Figure 10. Figure 10. Properties of the exchanger Ca2+‐binding domain 2 (CBD2). (A) Protein sequence alignment of CBD2 regions among human exchanger isoforms. The protein regions generated by alternative splicing (see Figure 2) are shown. Key residues for Ca2+ coordination are indicated. The lysine at position 585 does not directly coordinate Ca2+ ions but maintains CBD2 structural integrity by coordinating Asp 552 and Glu 648 in the absence of Ca2+ 16,93. Residue 683 (Glu) corresponds to Glu 648 in isoforms expressing exons AD and to Glu 647 in exon BD expressing exchangers. Numbering is reported according to NCX1.1 sequence, excluding the signal peptide. (B) The alternative splicing within CBD2 affects Ca2+ coordination. NMR and X‐ray structures show that isoforms expressing exon A allows coordination of two Ca2+ ions (PDB code 2QVM) 16,93,95, while those expressing exon B (PDB code 2KLT) 93 are unable to coordinate Ca2+. The presence of a positive charge at position 578 and a cysteine at position 585 in exon B expressing CBD2s prevents Ca2+ coordination 16,93,95. (C) Example of ionic currents recorded from exchangers expressing either exon A or B is shown. While the steady state current of the brain isoform NCX1.4 (AD exon) augmented in the presence of 10 μM cytoplasmic Ca2+, the kidney NCX1.3 (BD exon) steady state ionic current was insensitive to changes in intracellular Ca2+ concentration. The binding of Ca2+ to CBD2 enables exon A expressing isoforms to overcome the Na+‐dependent inactivation with elevation in intracellular Ca2+ 49. In contrast, exon B expressing exchangers lack this important regulatory feature. Modified, with permission, from Dunn J, et al., 2002 49.
Figure 11. Figure 11. The ion coordinating residues are conserved between the archaebacterial and mammalian exchangers. (A) The atomic structure of the archaebacterial exchanger revealed 12 amino acids within the α‐repeats (TMS 2 yellow, TMS 3 green, TMS 7 pink, and TMS 8 cyan) that coordinate Na+ and Ca2+ ions (PDB code 3V5U) 150. (B) The residues involved in ion coordination in NCX_Mj are shown. In parenthesis are reported the homologous residues in the mammalian NCX1.1 (numbered without taking into account the signal peptide). The three sodium coordinating sites are identified as SExt, SMid, and SInt. The Ca2+ ion binding site is referred to as SCa 150. New evidence indicates that SMid coordinates water instead of Na+ 151,162.
Figure 12. Figure 12. Residues relevant for NCX transport activity. The location of residues whose replacement perturbed NCX1.1 transport activity is shown. Amino acid numbering is based on the sequence of the canine NCX1.1 exchanger, excluding the signal peptide. The indicated residues were mutated one at a time and the activity of the resulting exchanger was analyzed either electrophysiologically or as Na‐dependent 45Ca2+ uptake 47,112,114,125,183,184,195. Mutations within the α‐repeats drastically affect transport activity. This is consistent with these regions harboring the residues involved in ion coordination, as revealed by the crystal structure of NCX_Mj 150 (see Figure 11). The amino acids coordinating the transported ions (marked with a red circle) are well conserved between NCX_Mj and the mammalian exchanger (see Figure 11) and their replacement impairs activity (grey circle).
Figure 13. Figure 13. Molecular mechanisms of ion transport. (A) Superimposed structures of NCX_Mj obtained in the presence of high Na+ (100 mM, gold, PDB code 5HXE) or low Na+ (10 mM, blue, PDB code 5HWY), Ca2+ ions were absent 151. Binding of Na+ (red spheres) elicits conformational changes throughout the protein. The movements of the external half of TMS 7 govern the transition from outward facing state to the occluded configuration 151. In the absence of Na+, TMS 7 is a contiguous “straight” helix. Upon binding of Na+ to SExt, TMS 7 bends (forming two helices) resulting in its movement away from TMS 6. This allows TMSs 1 and 6 to slide perpendicular to the membrane and transition the protein through occluded state (not shown) to the inward state (not shown). (B) Cysteines scanning mutagenesis allowed the mapping of the residues of the canine cardiac exchanger (NCX1.1) accessible from the cytoplasmic side. Shown are the homologous NCX_Mj residues mapped onto the inward model 125,150. Cysteine introduced at positions highlighted in cyan were accessible to cytoplasmically applied membrane‐impermeable sulfhydryl agent independently of NCX1.1 conformational state of. Residues labeled in orange were accessible to intracellular sulfhydryl agents only when the exchanger was constrained to the inward state. Residues labeled in grey were unaffected by sulfhydryl agents as assessed by ionic current inhibition. (C) Surface representation of NCX_Mj viewed from the cytoplasm. The inward‐facing model predicts two separate cytoplasmic pathways for ions to reach their binding sites 150 and cysteine scanning mutagenesis conducted in the mammalian exchanger support this hypothesis 125. Residues are color coded as described in (B). (D) NCX_Mj residues corresponding to those of NCX1.1 tested for reactivity to sulfhydryl reagents are highlighted and indicated in the table. (B–D) Modified, with permission, from John SA, et al., 2013 125, National Academy of Sciences.
Figure 14. Figure 14. pH regulation of NCX1.1. (A) Cytoplasmic acidification inhibits NCX activity; conversely, alkaline pH enhances NCX ionic currents and renders NCX insensitive to both Ca2+ and Na+ (not shown) 45,46,124. Outward exchanger currents were recorded from oocytes expressing the canine cardiac exchanger NCX1.1 using the giant patch technique. (B) Detailed electrophysiological studies show that mutations that disrupt binding of Ca2+ either to CBD1 (Δ446–499) or CBD2 (E516L) do not decrease NCX pH sensitivity 124. This result suggests that displacement of Ca2+ from the regulatory sites is not essential to confer pH sensitivity to the full‐length exchanger. This contrasts with experiments conducted with the isolated Ca2+‐binding domains (CBD1 and CBD2) showing that protons can displace Ca2+ from their binding sites 22. (C) Replacement of histidine 125 and 165 with alanine drastically decreases the sensitivity of the cardiac exchanger to cytosolic protons and in the case of position 165 also affected the cooperativity of the binding. Measurements were conducted on the canine cardiac exchanger expressed in Xenopus laevis oocytes as detailed in 124. (A,C) Modified, with permission, from John S, et al., 2018 124, © 2018, Rockfeller University Press.
Figure 15. Figure 15. NCX1.1 distribution in cardiac cells. The cartoon summarizes the distribution of NCX within adult cardiomyocytes and its coupling to other proteins essential for excitation‐contraction coupling. (A) The exchanger has been localized in proximity to voltage‐dependent Na+ (Nav,). The influx of Na+ via voltage‐dependent Na+ channels drives NCX to function in reverse mode and therefore prime the dyadic cleft with Ca2+ 140,271. (B) Changes in NCX1.1 expression (overexpression or knockout) regulate the extent of the voltage‐dependent Ca2+ channels (LTCCs) Ca2+‐dependent inactivation process, suggesting that these two proteins are in close proximity within the dyadic cleft. (C) NCX forms a macromolecular complex with the Na+/K+ ATPase (NKA), inositol triphosphate receptors (IP3R) and ankyrin (ANKB) 172. Genetic disorders due to defective expression of ankyrin result in disruption of this macromolecular complex leading to cardiac arrhythmia 173. (D) NCX appears to be in tight connection with the transient receptor potential channel (TRPC3) channels. The current model predicts that the influx of Na+ via TRPC3 tunes NCX transport activity by changing the Na+ levels in its proximity. If elevated enough, Na+ may reverse NCX augmenting intracellular Ca2+ levels 53,54.
Figure 16. Figure 16. NCX1 genetically modified mouse lines. (A) The cartoon shows a timeline of the mouse lines generated with altered expression levels of NCX. (B,C) Overexpression or ablation of NCX affects the cardiac action potential duration. The effects on the action potential waveforms have been connected to changes in K+ currents density 219, although both Ca2+ channels and NCX may contribute to this phenotype. Additional studies are needed to fully elucidate the impact of NCX on both the contractile and electrical properties of the heart. (B,C) Modified, with permission, from Bögeholz N, et al., 2016 20, © 2016, Elsevier.


Figure 1. Modes of operation of the Na+‐Ca2+ exchanger. (A) In the forward mode (top) NCX transports one Ca2+ ion out of the cell in exchange for three Na+ ions entering. One net charge is moved across the membrane resulting in a measurable ionic current. This is the most physiologically relevant cycle. In the reverse mode (bottom), one Ca2+ ions enter the cell and three Na+ ions exit. The direction in which Ca2+ is moved is governed by both the ionic gradients of Na+ and Ca2+ and membrane potential 164. (B) The scheme shows NCX transport cycle. This electrogenic transporter transitions between the outward and inward‐facing states via intermediate occluding states, leading to the coupled but opposite movements of Na+ and Ca2+ ions across the plasma membrane 164.


Figure 2. Na+‐Ca2+ exchanger gene organization. Each panel depicts the chromosomal location, number of exons, size, mRNA example, and corresponding protein of the three human isoforms. Gene exon structures are derived from Ensembl. Exons are indicated as grey bars and include both coding and noncoding exons. Exons undergoing alternative splicing are shown in color and are named in accordance with Quednau and colleagues 220. 5′ and 3′ regions of the gene are not included so that the gene size given extends from the beginning of the first exon and ends at the 3′ end of the last exon. Of special note, NCX2 does not undergo alternative splicing and all exons encode for protein. NCX1 and NCX3 have one noncoding exon, both undergo alternative splicing. An example of mRNA profile of the indicated exchangers is shown under each corresponding gene. Graphs are to scale. The alternative spliced exons (in colors), which at the protein level result in changes in the calcium‐binding domain 2 (CBD2) 220, are depicted in blue.


Figure 3. Tissue distribution of the exchanger isoforms. The three exchanger isoforms and their corresponding splice variants are differentially distributed within tissues 133,136,143,220,221. Examples are given for heart, brain, kidney, and skeletal muscle. The tissue distribution has been assessed using reverse transcriptase‐polymerase chain reaction as described in Ref. 220. Numerous evidence indicates that the isoform NCX1.1 136,143,220 is the only exchanger expressed in cardiomyocytes, although a study has identified isoforms NCX1.3 and NCX1.4 133. The splice variants NCX1.6 (ACD) and NCX3.3 (BC) are absent from the table. These exchangers have been localized in the brain 136,220, but their cell specific expression is undetermined.


Figure 4. Progression of Na+‐Ca2+ exchanger topology. The cartoons show how the topology of the mammalian exchanger has evolved through the years since its cloning in 1990 185. (A) Initially, NCX1.1 was predicted to be organized into 12 transmembrane segments (TMS). The two highly conserved regions among the exchangers which are known as α‐repeats 245 are highlighted. These regions are essential for ion transport 183,184,195. (B) Subsequent studies 50,60,106,239 revealed that the first helix was a signal peptide not necessary for activity, restricting the mature protein to 11 TMSs with an extracellular N terminus. (C) Cysteine scanning mutagenesis and epitope tagging were then applied to investigate NCX1.1 architecture. Based on these studies the topology of NCX was reorganized into nine transmembrane segments with an intracellular C‐terminus and two reentrant loops between TMSs 2‐3 and 7‐8 35,112,114,186,187,212. (D) The crystal structure of the archaebacterial exchanger (NCX_Mj) 150 has revealed a protein of 10 TMSs, contrasting with the predicted 9 TMSs for the mammalian exchanger as shown in (C). (E) Topology of the mammalian NCX based on NCX_Mj organization. The new architecture does not include the previously modeled reentrant loops, which are now predicted to be a short extracellular loop between TMSs 2‐3, within the α1‐repeat, and a new transmembrane segment (TMS 8) in the α2‐repeat. Future studies should be conducted to confirm the absence of either or both of these two reentrant structures and to validate that the mammalian exchanger and the archaebacterial are had a similar organization. The position of the cysteine residues involved in the intramolecular disulfide bond 241 is also shown. Amino acids are numbered based on the protein lacking the signal sequence.


Figure 5. Inconsistencies between the topology of the Na+‐Ca2+ exchanger and the archaebacterial exchanger. (A) Cysteine scanning mutagenesis and epitope mapping studies have been instrumental in deciphering the topology of the cardiac exchanger 35,50,112,114,186,187,212. The figure shows the amino acids that were replaced with cysteine and found accessible to membrane‐impermeable sulfhydryl agents either from the cytoplasm (blue circles) or the external side of the membrane (orange circles). Based on these investigations, NCX was modeled to have an extracellular N‐terminus, and a cytoplasmic C‐terminus. Moreover, residues 120 and 125 within the α1‐repeat were found to be exposed to the intracellular environment as well as several residues within the α2‐repeat, constraining these regions into reentrant loops. This organization corresponds to the one shown in Figure 4C. (B) With the atomic structure of the archaebacterial exchanger at hand 150, NCX1.1 is now assumed to be organized in 10 transmembrane segments 228,265, as shown in Figure 4E. However, this new arrangement places several residues previously found accessible to the cytoplasmic application of sulfhydryl reagents (blue circles), now facing the extracellular environment, including the C‐terminus. Note that the new architecture does not include reentrant loops. More investigations are needed to fully elucidate the topology of the mammalian exchanger.


Figure 6. Sequence alignment of the α‐repeats. (A) The sequence alignment illustrates the conservation of the α repeat regions within the Na+‐Ca2+ exchanger family. This is consistent with a broad range of evidence underlying their essential role in ion transport 125,183,184,195. The α1‐repeat encompasses residues 96 to 150 of NCX1.1, while the α2‐repeat stretches from 797 to 849 (amino acid numbering of NCX1.1 without the signal sequence). These boundaries were selected as previously described in Ref. 245. The residues that coordinate Na+ and Ca2+ ions, as revealed by the atomic structure of the archaebacterial exchanger 150, are indicated with a red arrow. (B) In addition to the conservation between species, the α‐repeats show intramolecular homology. This forms the foundation for the idea that exchangers are a result of gene duplication.


Figure 7. Domains and residues relevant for NCX1.1 regulation. (A) Highlighted are the residues of the canine cardiac Na+‐Ca2+ exchanger found important for its regulation. For a detailed role of these regions in controlling NCX transport activity please see the main text. Briefly, the residues shown in pink constitute the XIP region, which is intimately involved with the Na+‐dependent inactivation 166. The two helix bundle domain (THB) follows in yellow and its structure is shown in (B) (PDB# 6BV7) 295. The role of this domain remains to be determined. The residues encompassing the two Ca2+ regulatory domains are shown in light blue (Ca2+‐binding domain 1, CBD1) and dark blue (Ca2+‐binding domain 2, CBD2), their atomic structure is shown in (C) and (D), respectively [CBD1 PDB# 2PDK 189 and CBD2 PDB# 2QVM 16]. At the distal end of the large cytoplasmic loop, there is a stretch of residues (in dark grey) forming an amphipathic helix, required for palmitoylation of cysteine at position 739 207. Single point mutations that affect Na+, Ca2+, and pH regulation are indicated.


Figure 8. The XIP region is essential for Na+‐dependent inactivation. (A) Sequence alignment of the XIP region (amino acids 219 to 238, note NCX_Mj has no XIP region). (B) Mutations within this region either accelerate or abrogate the time course of current decay due to raised intracellular Na+, so‐called “Na+‐dependent inactivation”. Modified, with permission, from Matsuoka S, et al., 1997 166.


Figure 9. Properties of the exchanger Ca2+‐binding domain 1 (CBD1). (A) Protein sequence alignment of the CBD1 region among human exchanger isoforms. Numbering is reported according to NCX1.1 sequence, excluding the signal peptide. Residues involved in ion coordination are highlighted with red arrows. (B) This domain coordinates 4 Ca2+ ions (PDB code 2DPK, Ca2+ ions are shown in yellow) 189 but only the ion coordinating sites Ca3 and Ca4 have been shown to be relevant for regulation of the cardiac exchanger 196. (C) The ionic current generated by the cardiac exchanger can be evaluated using the giant patch technique 100. Shown is the Ca2+ dependency of WT NCX1.1 ionic currents and the indicated mutants 196. Mutations E385A and D446A‐D447A disrupt the binding of Ca2+ from sites 3 and 4 respectively and decreases the sensitivity of NCX1.1 to cytoplasmic Ca2+ of approximately 10‐fold.


Figure 10. Properties of the exchanger Ca2+‐binding domain 2 (CBD2). (A) Protein sequence alignment of CBD2 regions among human exchanger isoforms. The protein regions generated by alternative splicing (see Figure 2) are shown. Key residues for Ca2+ coordination are indicated. The lysine at position 585 does not directly coordinate Ca2+ ions but maintains CBD2 structural integrity by coordinating Asp 552 and Glu 648 in the absence of Ca2+ 16,93. Residue 683 (Glu) corresponds to Glu 648 in isoforms expressing exons AD and to Glu 647 in exon BD expressing exchangers. Numbering is reported according to NCX1.1 sequence, excluding the signal peptide. (B) The alternative splicing within CBD2 affects Ca2+ coordination. NMR and X‐ray structures show that isoforms expressing exon A allows coordination of two Ca2+ ions (PDB code 2QVM) 16,93,95, while those expressing exon B (PDB code 2KLT) 93 are unable to coordinate Ca2+. The presence of a positive charge at position 578 and a cysteine at position 585 in exon B expressing CBD2s prevents Ca2+ coordination 16,93,95. (C) Example of ionic currents recorded from exchangers expressing either exon A or B is shown. While the steady state current of the brain isoform NCX1.4 (AD exon) augmented in the presence of 10 μM cytoplasmic Ca2+, the kidney NCX1.3 (BD exon) steady state ionic current was insensitive to changes in intracellular Ca2+ concentration. The binding of Ca2+ to CBD2 enables exon A expressing isoforms to overcome the Na+‐dependent inactivation with elevation in intracellular Ca2+ 49. In contrast, exon B expressing exchangers lack this important regulatory feature. Modified, with permission, from Dunn J, et al., 2002 49.


Figure 11. The ion coordinating residues are conserved between the archaebacterial and mammalian exchangers. (A) The atomic structure of the archaebacterial exchanger revealed 12 amino acids within the α‐repeats (TMS 2 yellow, TMS 3 green, TMS 7 pink, and TMS 8 cyan) that coordinate Na+ and Ca2+ ions (PDB code 3V5U) 150. (B) The residues involved in ion coordination in NCX_Mj are shown. In parenthesis are reported the homologous residues in the mammalian NCX1.1 (numbered without taking into account the signal peptide). The three sodium coordinating sites are identified as SExt, SMid, and SInt. The Ca2+ ion binding site is referred to as SCa 150. New evidence indicates that SMid coordinates water instead of Na+ 151,162.


Figure 12. Residues relevant for NCX transport activity. The location of residues whose replacement perturbed NCX1.1 transport activity is shown. Amino acid numbering is based on the sequence of the canine NCX1.1 exchanger, excluding the signal peptide. The indicated residues were mutated one at a time and the activity of the resulting exchanger was analyzed either electrophysiologically or as Na‐dependent 45Ca2+ uptake 47,112,114,125,183,184,195. Mutations within the α‐repeats drastically affect transport activity. This is consistent with these regions harboring the residues involved in ion coordination, as revealed by the crystal structure of NCX_Mj 150 (see Figure 11). The amino acids coordinating the transported ions (marked with a red circle) are well conserved between NCX_Mj and the mammalian exchanger (see Figure 11) and their replacement impairs activity (grey circle).


Figure 13. Molecular mechanisms of ion transport. (A) Superimposed structures of NCX_Mj obtained in the presence of high Na+ (100 mM, gold, PDB code 5HXE) or low Na+ (10 mM, blue, PDB code 5HWY), Ca2+ ions were absent 151. Binding of Na+ (red spheres) elicits conformational changes throughout the protein. The movements of the external half of TMS 7 govern the transition from outward facing state to the occluded configuration 151. In the absence of Na+, TMS 7 is a contiguous “straight” helix. Upon binding of Na+ to SExt, TMS 7 bends (forming two helices) resulting in its movement away from TMS 6. This allows TMSs 1 and 6 to slide perpendicular to the membrane and transition the protein through occluded state (not shown) to the inward state (not shown). (B) Cysteines scanning mutagenesis allowed the mapping of the residues of the canine cardiac exchanger (NCX1.1) accessible from the cytoplasmic side. Shown are the homologous NCX_Mj residues mapped onto the inward model 125,150. Cysteine introduced at positions highlighted in cyan were accessible to cytoplasmically applied membrane‐impermeable sulfhydryl agent independently of NCX1.1 conformational state of. Residues labeled in orange were accessible to intracellular sulfhydryl agents only when the exchanger was constrained to the inward state. Residues labeled in grey were unaffected by sulfhydryl agents as assessed by ionic current inhibition. (C) Surface representation of NCX_Mj viewed from the cytoplasm. The inward‐facing model predicts two separate cytoplasmic pathways for ions to reach their binding sites 150 and cysteine scanning mutagenesis conducted in the mammalian exchanger support this hypothesis 125. Residues are color coded as described in (B). (D) NCX_Mj residues corresponding to those of NCX1.1 tested for reactivity to sulfhydryl reagents are highlighted and indicated in the table. (B–D) Modified, with permission, from John SA, et al., 2013 125, National Academy of Sciences.


Figure 14. pH regulation of NCX1.1. (A) Cytoplasmic acidification inhibits NCX activity; conversely, alkaline pH enhances NCX ionic currents and renders NCX insensitive to both Ca2+ and Na+ (not shown) 45,46,124. Outward exchanger currents were recorded from oocytes expressing the canine cardiac exchanger NCX1.1 using the giant patch technique. (B) Detailed electrophysiological studies show that mutations that disrupt binding of Ca2+ either to CBD1 (Δ446–499) or CBD2 (E516L) do not decrease NCX pH sensitivity 124. This result suggests that displacement of Ca2+ from the regulatory sites is not essential to confer pH sensitivity to the full‐length exchanger. This contrasts with experiments conducted with the isolated Ca2+‐binding domains (CBD1 and CBD2) showing that protons can displace Ca2+ from their binding sites 22. (C) Replacement of histidine 125 and 165 with alanine drastically decreases the sensitivity of the cardiac exchanger to cytosolic protons and in the case of position 165 also affected the cooperativity of the binding. Measurements were conducted on the canine cardiac exchanger expressed in Xenopus laevis oocytes as detailed in 124. (A,C) Modified, with permission, from John S, et al., 2018 124, © 2018, Rockfeller University Press.


Figure 15. NCX1.1 distribution in cardiac cells. The cartoon summarizes the distribution of NCX within adult cardiomyocytes and its coupling to other proteins essential for excitation‐contraction coupling. (A) The exchanger has been localized in proximity to voltage‐dependent Na+ (Nav,). The influx of Na+ via voltage‐dependent Na+ channels drives NCX to function in reverse mode and therefore prime the dyadic cleft with Ca2+ 140,271. (B) Changes in NCX1.1 expression (overexpression or knockout) regulate the extent of the voltage‐dependent Ca2+ channels (LTCCs) Ca2+‐dependent inactivation process, suggesting that these two proteins are in close proximity within the dyadic cleft. (C) NCX forms a macromolecular complex with the Na+/K+ ATPase (NKA), inositol triphosphate receptors (IP3R) and ankyrin (ANKB) 172. Genetic disorders due to defective expression of ankyrin result in disruption of this macromolecular complex leading to cardiac arrhythmia 173. (D) NCX appears to be in tight connection with the transient receptor potential channel (TRPC3) channels. The current model predicts that the influx of Na+ via TRPC3 tunes NCX transport activity by changing the Na+ levels in its proximity. If elevated enough, Na+ may reverse NCX augmenting intracellular Ca2+ levels 53,54.


Figure 16. NCX1 genetically modified mouse lines. (A) The cartoon shows a timeline of the mouse lines generated with altered expression levels of NCX. (B,C) Overexpression or ablation of NCX affects the cardiac action potential duration. The effects on the action potential waveforms have been connected to changes in K+ currents density 219, although both Ca2+ channels and NCX may contribute to this phenotype. Additional studies are needed to fully elucidate the impact of NCX on both the contractile and electrical properties of the heart. (B,C) Modified, with permission, from Bögeholz N, et al., 2016 20, © 2016, Elsevier.
References
 1.Adachi‐Akahane S, Lu L, Li Z, Frank JS, Philipson KD, Morad M. Calcium signaling in transgenic mice overexpressing cardiac Na+‐Ca2+2+ exchanger. J Gen Physiol 109: 717‐729, 1997.
 2.Ander BP, Hurtado C, Raposo CS, Maddaford TG, Deniset JF, Hryshko LV, Pierce GN, Lukas A. Differential sensitivities of the NCX1.1 and NCX1.3 isoforms of the Na+‐Ca2+ exchanger to alpha‐linolenic acid. Cardiovasc Res 73: 395‐403, 2007.
 3.Armoundas AA, Hobai IA, Tomaselli GF, Winslow RL, O'Rourke B. Role of sodium‐calcium exchanger in modulating the action potential of ventricular myocytes from normal and failing hearts. Circ Res 93: 46‐53, 2003.
 4.Artman M. Sarcolemmal Na+‐Ca2+ exchange activity and exchanger immunoreactivity in developing rabbit hearts. Am J Physiol 263: H1506‐H1513, 1992.
 5.Artman M, Ichikawa H, Avkiran M, Coetzee WA. Na+/Ca2+ exchange current density in cardiac myocytes from rabbits and guinea pigs during postnatal development. Am J Physiol 268: H1714‐H1722, 1995.
 6.Asteggiano C, Berberian G, Beauge L. Phosphatidyl inositol‐4,5‐bisphosphate bound to bovine cardiac Na+/Ca2+ exchanger displays a MgATP regulation similar to that of the exchange fluxes. Eur J Biochem 268: 437‐442, 2001.
 7.Baker PF, Blaustein MP. Sodium‐dependent uptake of calcium by crab nerve. Biochim Biophys Acta 150: 167‐170, 1968.
 8.Baker PF, Blaustein MP, Hodgkin AL, Steinhardt RA. The influence of calcium on sodium efflux in squid axons. J Physiol 200: 431‐458, 1969.
 9.Bano D, Young KW, Guerin CJ, Lefeuvre R, Rothwell NJ, Naldini L, Rizzuto R, Carafoli E, Nicotera P. Cleavage of the plasma membrane Na+/Ca2+ exchanger in excitotoxicity. Cell 120: 275‐285, 2005.
 10.Barnes KV, Cheng G, Dawson MM, Menick DR. Cloning of cardiac, kidney, and brain promoters of the feline ncx1 gene. J Biol Chem 272: 11510‐11517, 1997.
 11.Bartholomaus I, Milan‐Lobo L, Nicke A, Dutertre S, Hastrup H, Jha A, Gether U, Sitte HH, Betz H, Eulenburg V. Glycine transporter dimers: Evidence for occurrence in the plasma membrane. J Biol Chem 283: 10978‐10991, 2008.
 12.Baruscotti M, Bucchi A, Viscomi C, Mandelli G, Consalez G, Gnecchi‐Rusconi T, Montano N, Casali KR, Micheloni S, Barbuti A, DiFrancesco D. Deep bradycardia and heart block caused by inducible cardiac‐specific knockout of the pacemaker channel gene Hcn4. Proc Natl Acad Sci U S A 108: 1705‐1710, 2011.
 13.Bassani RA, Bassani JW, Bers DM. Mitochondrial and sarcolemmal Ca2+ transport reduce [Ca2+]i during caffeine contractures in rabbit cardiac myocytes. J Physiol 453: 591‐608, 1992.
 14.Beauge L, Asteggiano C, Berberian G. Regulation of phosphatidylinositol‐4,5‐biphosphate bound to the bovine cardiac Na+/Ca2+ exchanger. Ann N Y Acad Sci 976: 288‐299, 2002.
 15.Bers DM. Cardiac excitation‐contraction coupling. Nature 415: 198‐205, 2002.
 16.Besserer GM, Ottolia M, Nicoll DA, Chaptal V, Cascio D, Philipson KD, Abramson J. The second Ca2+‐binding domain of the Na+ Ca2+ exchanger is essential for regulation: Crystal structures and mutational analysis. Proc Natl Acad Sci U S A 104: 18467‐18472, 2007.
 17.Blaustein MP, Lederer WJ. Sodium/calcium exchange: Its physiological implications. Physiol Rev 79: 763‐854, 1999.
 18.Boerth SR, Zimmer DB, Artman M. Steady‐state mRNA levels of the sarcolemmal Na+‐Ca2+ exchanger peak near birth in developing rabbit and rat hearts. Circ Res 74: 354‐359, 1994.
 19.Bogeholz N, Pauls P, Bauer BK, Schulte JS, Dechering DG, Frommeyer G, Kirchhefer U, Goldhaber JI, Muller FU, Eckardt L, Pott C. Suppression of early and late afterdepolarizations by heterozygous knockout of the Na+/Ca2+ exchanger in a murine model. Circ Arrhythm Electrophysiol 8: 1210‐1218, 2015.
 20.Bogeholz N, Pauls P, Kaese S, Schulte JS, Lemoine MD, Dechering DG, Frommeyer G, Goldhaber JI, Seidl MD, Kirchhefer U, Eckardt L, Muller FU, Pott C. Triggered activity in atrial myocytes is influenced by Na+/Ca2+ exchanger activity in genetically altered mice. J Mol Cell Cardiol 101: 106‐115, 2016.
 21.Bountra C, Vaughan‐Jones RD. Effect of intracellular and extracellular pH on contraction in isolated, mammalian cardiac muscle. J Physiol 418: 163‐187, 1989.
 22.Boyman L, Hagen BM, Giladi M, Hiller R, Lederer WJ, Khananshvili D. Proton‐sensing Ca2+ binding domains regulate the cardiac Na+/Ca2+ exchanger. J Biol Chem 286: 28811‐28820, 2011.
 23.Boyman L, Mikhasenko H, Hiller R, Khananshvili D. Kinetic and equilibrium properties of regulatory calcium sensors of NCX1 protein. J Biol Chem 284: 6185‐6193, 2009.
 24.Brette F, Orchard CH. Density and sub‐cellular distribution of cardiac and neuronal sodium channel isoforms in rat ventricular myocytes. Biochem Biophys Res Commun 348: 1163‐1166, 2006.
 25.Breukels V, Vuister GW. Binding of calcium is sensed structurally and dynamically throughout the second calcium‐binding domain of the sodium/calcium exchanger. Proteins 78: 1813‐1824, 2010.
 26.Bridge JH, Smolley JR, Spitzer KW. The relationship between charge movements associated with ICa and INa‐Ca in cardiac myocytes. Science 248: 376‐378, 1990.
 27.Bucchi A, Barbuti A, Difrancesco D, Baruscotti M. Funny current and cardiac rhythm: Insights from HCN knockout and transgenic mouse models. Front Physiol 3: 240, 2012.
 28.Bukowska A, Lendeckel U, Bode‐Boger SM, Goette A. Physiologic and pathophysiologic role of calpain: Implications for the occurrence of atrial fibrillation. Cardiovasc Ther 30: e115‐127, 2012.
 29.Chaptal V, Besserer GM, Ottolia M, Nicoll DA, Cascio D, Philipson KD, Abramson J. How does regulatory Ca2+ regulate the Na+‐Ca2+ exchanger? Channels (Austin) 1: 397‐399, 2007.
 30.Cheng G, Hagen TP, Dawson ML, Barnes KV, Menick DR. The role of GATA, CArG, E‐box, and a novel element in the regulation of cardiac expression of the Na+‐Ca2+ exchanger gene. J Biol Chem 274: 12819‐12826, 1999.
 31.Cheung JY, Rothblum LI, Moorman JR, Tucker AL, Song J, Ahlers BA, Carl LL, Wang J, Zhang XQ. Regulation of cardiac Na+/Ca2+ exchanger by phospholemman. Ann N Y Acad Sci 1099: 119‐134, 2007.
 32.Cho CH, Kim SS, Jeong MJ, Lee CO, Shin HS. The Na+ ‐Ca2+ exchanger is essential for embryonic heart development in mice. Mol Cells 10: 712‐722, 2000.
 33.Chou AC, Ju YT, Pan CY. Calmodulin interacts with the sodium/calcium exchanger NCX1 to regulate activity. PLoS One 10: e0138856, 2015.
 34.Cingolani E, Goldhaber JI, Marban E. Next‐generation pacemakers: From small devices to biological pacemakers. Nat Rev Cardiol 15: 139‐150, 2018.
 35.Cook O, Low W, Rahamimoff H. Membrane topology of the rat brain Na+‐Ca2+ exchanger. Biochim Biophys Acta 1371: 40‐52, 1998.
 36.Cross HR, Lu L, Steenbergen C, Philipson KD, Murphy E. Overexpression of the cardiac Na+/Ca2+ exchanger increases susceptibility to ischemia/reperfusion injury in male, but not female, transgenic mice. Circ Res 83: 1215‐1223, 1998.
 37.Cunha SR, Bhasin N, Mohler PJ. Targeting and stability of Na+/Ca2+ exchanger 1 in cardiomyocytes requires direct interaction with the membrane adaptor ankyrin‐B. J Biol Chem 282: 4875‐4883, 2007.
 38.Dan P, Lin E, Huang J, Biln P, Tibbits GF. Three‐dimensional distribution of cardiac Na+‐Ca2+ exchanger and ryanodine receptor during development. Biophys J 93: 2504‐2518, 2007.
 39.Despa S, Brette F, Orchard CH, Bers DM. Na/Ca exchange and Na/K‐ATPase function are equally concentrated in transverse tubules of rat ventricular myocytes. Biophys J 85: 3388‐3396, 2003.
 40.Despa S, Islam MA, Weber CR, Pogwizd SM, Bers DM. Intracellular Na+ concentration is elevated in heart failure but Na/K pump function is unchanged. Circulation 105: 2543‐2548, 2002.
 41.Despa S, Lingrel JB, Bers DM. Na+/K+‐ATPase alpha2‐isoform preferentially modulates Ca2+ transients and sarcoplasmic reticulum Ca2+ release in cardiac myocytes. Cardiovasc Res 95: 480‐486, 2012.
 42.DiPolo R. Calcium influx in internally dialyzed squid giant axons. J Gen Physiol 73: 91‐113, 1979.
 43.DiPolo R, Beauge L. In the squid axon Na+/Ca2+ exchanger the state of the Ca i‐regulatory site influences the affinities of the intra‐ and extracellular transport sites for Na+ and Ca2+. Pflugers Arch 456: 623‐633, 2008.
 44.Dixon RE, Yuan C, Cheng EP, Navedo MF, Santana LF. Ca2+ signaling amplification by oligomerization of L‐type Cav1.2 channels. Proc Natl Acad Sci U S A 109: 1749‐1754, 2012.
 45.Doering AE, Lederer WJ. The mechanism by which cytoplasmic protons inhibit the sodium‐calcium exchanger in guinea‐pig heart cells. J Physiol 466: 481‐499, 1993.
 46.Doering AE, Lederer WJ. The action of Na+ as a cofactor in the inhibition by cytoplasmic protons of the cardiac Na+‐Ca2+ exchanger in the guinea‐pig. J Physiol 480 (Pt 1): 9‐20, 1994.
 47.Doering AE, Nicoll DA, Lu Y, Lu L, Weiss JN, Philipson KD. Topology of a functionally important region of the cardiac Na+/Ca2+ exchanger. J Biol Chem 273: 778‐783, 1998.
 48.Doleschal B, Primessnig U, Wolkart G, Wolf S, Schernthaner M, Lichtenegger M, Glasnov TN, Kappe CO, Mayer B, Antoons G, Heinzel F, Poteser M, Groschner K. TRPC3 contributes to regulation of cardiac contractility and arrhythmogenesis by dynamic interaction with NCX1. Cardiovasc Res 106: 163‐173, 2015.
 49.Dunn J, Elias CL, Le HD, Omelchenko A, Hryshko LV, Lytton J. The molecular determinants of ionic regulatory differences between brain and kidney Na+/Ca2+ exchanger NCX1 isoforms. J Biol Chem 277: 33957‐33962, 2002.
 50.Durkin JT, Ahrens DC, Pan YC, Reeves JP. Purification and amino‐terminal sequence of the bovine cardiac sodium‐calcium exchanger: Evidence for the presence of a signal sequence. Arch Biochem Biophys 290: 369‐375, 1991.
 51.Dyck C, Maxwell K, Buchko J, Trac M, Omelchenko A, Hnatowich M, Hryshko LV. Structure‐function analysis of CALX1.1, a Na+‐Ca2+ exchanger from Drosophila. Mutagenesis of ionic regulatory sites. J Biol Chem 273: 12981‐12987, 1998.
 52.Dyck C, Omelchenko A, Elias CL, Quednau BD, Philipson KD, Hnatowich M, Hryshko LV. Ionic regulatory properties of brain and kidney splice variants of the NCX1 Na+‐Ca2+ exchanger. J Gen Physiol 114: 701‐711, 1999.
 53.Eder P, Poteser M, Romanin C, Groschner K. Na+ entry and modulation of Na+/Ca2+ exchange as a key mechanism of TRPC signaling. Pflugers Arch 451: 99‐104, 2005.
 54.Eder P, Probst D, Rosker C, Poteser M, Wolinski H, Kohlwein SD, Romanin C, Groschner K. Phospholipase C‐dependent control of cardiac calcium homeostasis involves a TRPC3‐NCX1 signaling complex. Cardiovasc Res 73: 111‐119, 2007.
 55.Eisner DA, Caldwell JL, Kistamas K, Trafford AW. Calcium and excitation‐contraction coupling in the heart. Circ Res 121: 181‐195, 2017.
 56.Faham S, Watanabe A, Besserer GM, Cascio D, Specht A, Hirayama BA, Wright EM, Abramson J. The crystal structure of a sodium galactose transporter reveals mechanistic insights into Na+/sugar symport. Science 321: 810‐814, 2008.
 57.Farhan H, Freissmuth M, Sitte HH. Oligomerization of neurotransmitter transporters: A ticket from the endoplasmic reticulum to the plasma membrane. Handb Exp Pharmacol: 233‐249, 2006.
 58.Frank JS, Mottino G, Reid D, Molday RS, Philipson KD. Distribution of the Na+‐Ca2+ exchange protein in mammalian cardiac myocytes: An immunofluorescence and immunocolloidal gold‐labeling study. J Cell Biol 117: 337‐345, 1992.
 59.Fuller W, Reilly L, Hilgemann DW. S‐palmitoylation and the regulation of NCX1. Channels Austin 10: 75‐77, 2016.
 60.Furman I, Cook O, Kasir J, Low W, Rahamimoff H. The putative amino‐terminal signal peptide of the cloned rat brain Na+‐Ca2+ exchanger gene RBE‐1 is not mandatory for functional expression. J Biol Chem 270: 19120‐19127, 1995.
 61.Fuster D, Moe OW, Hilgemann DW. Steady‐state function of the ubiquitous mammalian Na/H exchanger NHE1 in relation to dimer coupling models with 2Na/2H stoichiometry. J Gen Physiol 132: 465‐480, 2008.
 62.Gabellini N, Bortoluzzi S, Danieli GA, Carafoli E. The gene promoter of human Na+/Ca2+ exchanger isoform 3 SLC8A3 is controlled by cAMP and calcium. Ann N Y Acad Sci 976: 282‐284, 2002.
 63.Gabellini N, Bortoluzzi S, Danieli GA, Carafoli E. The human SLC8A3 gene and the tissue‐specific Na+/Ca2+ exchanger 3 isoforms. Gene 298: 1‐7, 2002.
 64.Gabellini N, Bortoluzzi S, Danieli GA, Carafoli E. Control of the Na+/Ca2+ exchanger 3 promoter by cyclic adenosine monophosphate and Ca2+ in differentiating neurons. J Neurochem 84: 282‐293, 2003.
 65.Gao Z, Rasmussen TP, Li Y, Kutschke W, Koval OM, Wu Y, Wu Y, Hall DD, Joiner ML, Wu XQ, Swaminathan PD, Purohit A, Zimmerman K, Weiss RM, Philipson KD, Song LS, Hund TJ, Anderson ME. Genetic inhibition of Na+‐Ca2+ exchanger current disables fight or flight sinoatrial node activity without affecting resting heart rate. Circ Res 112: 309‐317, 2013.
 66.Garciarena CD, Youm JB, Swietach P, Vaughan‐Jones RD. H+‐activated Na+ influx in the ventricular myocyte couples Ca2+‐signalling to intracellular pH. J Mol Cell Cardiol 61: 51‐59, 2013.
 67.Gershome C, Lin E, Kashihara H, Hove‐Madsen L, Tibbits GF. Colocalization of voltage‐gated Na+ channels with the Na+/Ca2+ exchanger in rabbit cardiomyocytes during development. Am J Physiol Heart Circ Physiol 300: H300‐H311, 2011.
 68.Giladi M, Bohbot H, Buki T, Schulze DH, Hiller R, Khananshvili D. Dynamic features of allosteric Ca2+ sensor in tissue‐specific NCX variants. Cell Calcium 51: 478‐485, 2012.
 69.Giladi M, Boyman L, Mikhasenko H, Hiller R, Khananshvili D. Essential role of the CBD1‐CBD2 linker in slow dissociation of Ca2+ from the regulatory two‐domain tandem of NCX1. J Biol Chem 285: 28117‐28125, 2010.
 70.Giladi M, Hiller R, Hirsch JA, Khananshvili D. Population shift underlies Ca2+‐induced regulatory transitions in the sodium‐calcium exchanger NCX. J Biol Chem 288: 23141‐23149, 2013.
 71.Giladi M, Khananshvili D. Molecular determinants of allosteric regulation in NCX proteins. Adv Exp Med Biol 961: 35‐48, 2013.
 72.Giladi M, Sasson Y, Fang X, Hiller R, Buki T, Wang YX, Hirsch JA, Khananshvili D. A common Ca2+‐driven interdomain module governs eukaryotic NCX regulation. PLoS One 7: e39985, 2012.
 73.Giladi M, Shor R, Lisnyansky M, Khananshvili D. Structure‐functional basis of ion transport in sodium‐calcium exchanger NCX proteins. Int J Mol Sci 17: 1949, 2016.
 74.Giladi M, Tal I, Khananshvili D. Structural features of ion transport and allosteric regulation in sodium‐calcium exchanger NCX proteins. Front Physiol 7: 30, 2016.
 75.Giladi M, van Dijk L, Refaeli B, Almagor L, Hiller R, Man P, Forest E, Khananshvili D. Dynamic distinctions in the Na+/Ca2+ exchanger adopting the inward‐ and outward‐facing conformational states. J Biol Chem 292: 12311‐12323, 2017.
 76.Ginsburg KS, Bers DM. Isoproterenol does not enhance Ca‐dependent Na/Ca exchange current in intact rabbit ventricular myocytes. J Mol Cell Cardiol 39: 972‐981, 2005.
 77.Ginsburg KS, Weber CR, Bers DM. Cardiac Na+‐Ca2+ exchanger: Dynamics of Ca2+‐dependent activation and deactivation in intact myocytes. J Physiol 591: 2067‐2086, 2013.
 78.Goel M, Zuo CD, Sinkins WG, Schilling WP. TRPC3 channels colocalize with Na+/Ca2+ exchanger and Na+ pump in axial component of transverse‐axial tubular system of rat ventricle. Am J Physiol Heart Circ Physiol 292: H874‐H883, 2007.
 79.Gok C, Plain F, Robertson AD, Howie J, Baillie GS, Fraser NJ, Fuller W. Dynamic palmitoylation of the sodium‐calcium exchanger modulates its structure, affinity for lipid‐ordered domains, and inhibition by XIP. Cell Rep 31: 107697, 2020.
 80.Goldhaber JI, Philipson KD. Cardiac sodium‐calcium exchange and efficient excitation‐contraction coupling: Implications for heart disease. Adv Exp Med Biol 961: 355‐364, 2013.
 81.Groenke S, Larson ED, Alber S, Zhang R, Lamp ST, Ren X, Nakano H, Jordan MC, Karagueuzian HS, Roos KP, Nakano A, Proenza C, Philipson KD, Goldhaber JI. Complete atrial‐specific knockout of sodium‐calcium exchange eliminates sinoatrial node pacemaker activity. PLoS One 8: e81633, 2013.
 82.Haddock PS, Coetzee WA, Cho E, Porter L, Katoh H, Bers DM, Jafri MS, Artman M. Subcellular [Ca2+]i gradients during excitation‐contraction coupling in newborn rabbit ventricular myocytes. Circ Res 85: 415‐427, 1999.
 83.Hafver TL, Hodne K, Wanichawan P, Aronsen JM, Dalhus B, Lunde PK, Lunde M, Martinsen M, Enger UH, Fuller W, Sjaastad I, Louch WE, Sejersted OM, Carlson CR. Protein phosphatase 1c associated with the cardiac sodium calcium exchanger 1 regulates its activity by dephosphorylating Serine 68‐phosphorylated phospholemman. J Biol Chem 291: 4561‐4579, 2016.
 84.Hale CC, Bliler S, Quinn TP, Peletskaya EN. Localization of an exchange inhibitory peptide XIP binding site on the cardiac sodium‐calcium exchanger. Biochem Biophys Res Commun 236: 113‐117, 1997.
 85.Hampton TG, Wang JF, DeAngelis J, Amende I, Philipson KD, Morgan JP. Enhanced gene expression of Na+/Ca2+ exchanger attenuates ischemic and hypoxic contractile dysfunction. Am J Physiol Heart Circ Physiol 279: H2846‐H2854, 2000.
 86.He Z, Feng S, Tong Q, Hilgemann DW, Philipson KD. Interaction of PIP2 with the XIP region of the cardiac Na/Ca exchanger. Am J Physiol Cell Physiol 278: C661‐C666, 2000.
 87.He Z, Petesch N, Voges K, Roben W, Philipson KD. Identification of important amino acid residues of the Na+‐Ca2+ exchanger inhibitory peptide, XIP. J Membr Biol 156: 149‐156, 1997.
 88.He Z, Tong Q, Quednau BD, Philipson KD, Hilgemann DW. Cloning, expression, and characterization of the squid Na+‐Ca2+ exchanger NCX‐SQ1. J Gen Physiol 111: 857‐873, 1998.
 89.Henderson SA, Goldhaber JI, So JM, Han T, Motter C, Ngo A, Chantawansri C, Ritter MR, Friedlander M, Nicoll DA, Frank JS, Jordan MC, Roos KP, Ross RS, Philipson KD. Functional adult myocardium in the absence of Na+‐Ca2+ exchange: Cardiac‐specific knockout of NCX1. Circ Res 95: 604‐611, 2004.
 90.Herrmann S, Lipp P, Wiesen K, Stieber J, Nguyen H, Kaiser E, Ludwig A. The cardiac sodium‐calcium exchanger NCX1 is a key player in the initiation and maintenance of a stable heart rhythm. Cardiovasc Res 99: 780‐788, 2013.
 91.Herz K, Rimon A, Jeschke G, Padan E. Beta‐sheet‐dependent dimerization is essential for the stability of NhaA Na+/H+ antiporter. J Biol Chem 284: 6337‐6347, 2009.
 92.Hilge M. Ca2+ regulation of ion transport in the Na+/Ca2+ exchanger. J Biol Chem 287: 31641‐31649, 2012.
 93.Hilge M, Aelen J, Foarce A, Perrakis A, Vuister GW. Ca2+ regulation in the Na+/Ca2+ exchanger features a dual electrostatic switch mechanism. Proc Natl Acad Sci U S A 106: 14333‐14338, 2009.
 94.Hilge M, Aelen J, Perrakis A, Vuister GW. Structural basis for Ca2+ regulation in the Na+/Ca2+ exchanger. Ann N Y Acad Sci 1099: 7‐15, 2007.
 95.Hilge M, Aelen J, Vuister GW. Ca2+ regulation in the Na+/Ca2+ exchanger involves two markedly different Ca2+ sensors. Mol Cell 22: 15‐25, 2006.
 96.Hilgemann DW. Regulation and deregulation of cardiac Na+‐Ca2+ exchange in giant excised sarcolemmal membrane patches. Nature 344: 242‐245, 1990.
 97.Hilgemann DW. Unitary cardiac Na+, Ca2+ exchange current magnitudes determined from channel‐like noise and charge movements of ion transport. Biophys J 71: 759‐768, 1996.
 98.Hilgemann DW, Ball R. Regulation of cardiac Na+,Ca2+ exchange and KATP potassium channels by PIP2. Science 273: 956‐959, 1996.
 99.Hilgemann DW, Collins A. Mechanism of cardiac Na+‐Ca2+ exchange current stimulation by MgATP: Possible involvement of aminophospholipid translocase. J Physiol 454: 59‐82, 1992.
 100.Hilgemann DW, Collins A, Cash DP, Nagel GA. Cardiac Na+‐Ca2+ exchange system in giant membrane patches. Ann N Y Acad Sci 639: 126‐139, 1991.
 101.Hilgemann DW, Collins A, Matsuoka S. Steady‐state and dynamic properties of cardiac sodium‐calcium exchange. Secondary modulation by cytoplasmic calcium and ATP. J Gen Physiol 100: 933‐961, 1992.
 102.Hilgemann DW, Lu CC. Giant membrane patches: Improvements and applications. Methods Enzymol 293: 267‐280, 1998.
 103.Hilgemann DW, Matsuoka S, Nagel GA, Collins A. Steady‐state and dynamic properties of cardiac sodium‐calcium exchange. Sodium‐dependent inactivation. J Gen Physiol 100: 905‐932, 1992.
 104.Hnatowich M, Le HD, DeMoissac D, Ranson K, Yurkov V, Gilchrist JS, Omelchenko A. mu‐Calpain‐mediated deregulation of cardiac, brain, and kidney NCX1 splice variants. Cell Calcium 51: 164‐170, 2012.
 105.Hoshi M, Du XX, Shinlapawittayatorn K, Liu H, Chai S, Wan X, Ficker E, Deschenes I. Brugada syndrome disease phenotype explained in apparently benign sodium channel mutations. Cir Cardiovasc Genet 7: 123‐131, 2014.
 106.Hryshko LV, Nicoll DA, Weiss JN, Philipson KD. Biosynthesis and initial processing of the cardiac sarcolemmal Na+‐Ca2+ exchanger. Biochim Biophys Acta 1151: 35‐42, 1993.
 107.Huang J, Hove‐Madsen L, Tibbits GF. Na+/Ca2+ exchange activity in neonatal rabbit ventricular myocytes. Am J Physiol Cell Physiol 288: C195‐C203, 2005.
 108.Hunte C, Screpanti E, Venturi M, Rimon A, Padan E, Michel H. Structure of a Na+/H+ antiporter and insights into mechanism of action and regulation by pH. Nature 435: 1197‐1202, 2005.
 109.Huser J, Lipsius SL, Blatter LA. Calcium gradients during excitation‐contraction coupling in cat atrial myocytes. J Physiol 494 (Pt 3): 641‐651, 1996.
 110.Imahashi K, Pott C, Goldhaber JI, Steenbergen C, Philipson KD, Murphy E. Cardiac‐specific ablation of the Na+‐Ca2+ exchanger confers protection against ischemia/reperfusion injury. Circ Res 97: 916‐921, 2005.
 111.Inserte J, Hernando V, Garcia‐Dorado D. Contribution of calpains to myocardial ischaemia/reperfusion injury. Cardiovasc Res 96: 23‐31, 2012.
 112.Iwamoto T, Nakamura TY, Pan Y, Uehara A, Imanaga I, Shigekawa M. Unique topology of the internal repeats in the cardiac Na+/Ca2+ exchanger. Febs Letters 446: 264‐268, 1999.
 113.Iwamoto T, Pan Y, Wakabayashi S, Imagawa T, Yamanaka HI, Shigekawa M. Phosphorylation‐dependent regulation of cardiac Na+/Ca2+ exchanger via protein kinase C. J Biol Chem 271: 13609‐13615, 1996.
 114.Iwamoto T, Uehara A, Imanaga I, Shigekawa M. The Na+/Ca2+ exchanger NCX1 has oppositely oriented reentrant loop domains that contain conserved aspartic acids whose mutation alters its apparent Ca2+ affinity. J Biol Chem 275: 38571‐38580, 2000.
 115.Iwamoto T, Uehara A, Nakamura TY, Imanaga I, Shigekawa M. Chimeric analysis of Na+/Ca2+ exchangers NCX1 and NCX3 reveals structural domains important for differential sensitivity to external Ni2+ or Li+. J Biol Chem 274: 23094‐23102, 1999.
 116.Iwamoto T, Wakabayashi S, Shigekawa M. Growth factor‐induced phosphorylation and activation of aortic smooth muscle Na+/Ca2+ exchanger. J Biol Chem 270: 8996‐9001, 1995.
 117.Iwamoto T, Watanabe Y, Kita S, Blaustein MP. Na+/Ca2+ exchange inhibitors: A new class of calcium regulators. Cardiovasc Hematol Disord Drug Targets 7: 188‐198, 2007.
 118.James PF, Grupp IL, Grupp G, Woo AL, Askew GR, Croyle ML, Walsh RA, Lingrel JB. Identification of a specific role for the Na,K‐ATPase alpha 2 isoform as a regulator of calcium in the heart. Mol Cell 3: 555‐563, 1999.
 119.Jardetzky O. Simple allosteric model for membrane pumps. Nature 211: 969‐970, 1966.
 120.Jayasinghe ID, Cannell MB, Soeller C. Organization of ryanodine receptors, transverse tubules, and sodium‐calcium exchanger in rat myocytes. Biophys J 97: 2664‐2673, 2009.
 121.Jeffs GJ, Meloni BP, Bakker AJ, Knuckey NW. The role of the Na+/Ca2+ exchanger NCX in neurons following ischaemia. J Clin Neurosci 14: 507‐514, 2007.
 122.Jeffs GJ, Meloni BP, Sokolow S, Herchuelz A, Schurmans S, Knuckey NW. NCX3 knockout mice exhibit increased hippocampal CA1 and CA2 neuronal damage compared to wild‐type mice following global cerebral ischemia. Exp Neurol 210: 268‐273, 2008.
 123.Jeon D, Chu K, Jung KH, Kim M, Yoon BW, Lee CJ, Oh U, Shin HS. Na+/Ca2+ exchanger 2 is neuroprotective by exporting Ca2+ during a transient focal cerebral ischemia in the mouse. Cell Calcium 43: 482‐491, 2008.
 124.John S, Kim B, Olcese R, Goldhaber JI, Ottolia M. Molecular determinants of pH regulation in the cardiac Na+‐Ca2+ exchanger. J Gen Physiol 150: 245‐257, 2018.
 125.John SA, Liao J, Jiang Y, Ottolia M. The cardiac Na+‐Ca2+ exchanger has two cytoplasmic ion permeation pathways. Proc Natl Acad Sci U S A 110: 7500‐7505, 2013.
 126.John SA, Ribalet B, Weiss JN, Philipson KD, Ottolia M. Ca2+‐dependent structural rearrangements within Na+‐Ca2+ exchanger dimers. Proc Natl Acad Sci U S A 108: 1699‐1704, 2011.
 127.Jordan MC, Henderson SA, Han T, Fishbein MC, Philipson KD, Roos KP. Myocardial function with reduced expression of the sodium‐calcium exchanger. J Card Fail 16: 786‐796, 2010.
 128.Jovanovic S, Jovanovic A, Shen WK, Terzic A. Low concentrations of 17beta‐estradiol protect single cardiac cells against metabolic stress‐induced Ca2+ loading. J Am Coll Cardiol 36: 948‐952, 2000.
 129.Kang K, Bauer PJ, Kinjo TG, Szerencsei RT, Bonigk W, Winkfein RJ, Schnetkamp PP. Assembly of retinal rod or cone Na+/Ca2+‐K+ exchanger oligomers with cGMP‐gated channel subunits as probed with heterologously expressed cDNAs. Biochemistry 42: 4593‐4600, 2003.
 130.Kang TM, Hilgemann DW. Multiple transport modes of the cardiac Na+/Ca2+ exchanger. Nature 427: 544‐548, 2004.
 131.Keller M, Pignier C, Niggli E, Egger M. Mechanisms of Na+‐Ca2+ exchange inhibition by amphiphiles in cardiac myocytes: Importance of transbilayer movement. J Membr Biol 198: 159‐175, 2004.
 132.Khananshvili D. Structure‐dynamic coupling through Ca2+‐binding regulatory domains of mammalian NCX isoform/splice variants. Adv Exp Med Biol 981: 41‐58, 2017.
 133.Kim I, Koh GY, Lee CO. Identification of alternatively spliced Na+‐Ca2+ exchanger isoforms expressed in the heart. Comp Biochem Physiol B Biochem Mol Biol 119: 157‐161, 1998.
 134.Kim I, Lee CO. Cloning of the mouse cardiac Na+‐Ca2+ exchanger and functional expression in Xenopus oocytes. Ann N Y Acad Sci 779: 126‐128, 1996.
 135.Kockskamper J, Sheehan KA, Bare DJ, Lipsius SL, Mignery GA, Blatter LA. Activation and propagation of Ca2+ release during excitation‐contraction coupling in atrial myocytes. Biophys J 81: 2590‐2605, 2001.
 136.Kofuji P, Lederer WJ, Schulze DH. Mutually exclusive and cassette exons underlie alternatively spliced isoforms of the Na/Ca exchanger. J Biol Chem 269: 5145‐5149, 1994.
 137.Kohajda Z, Loewe A, Toth N, Varro A, Nagy N. The cardiac pacemaker story‐fundamental role of the Na+/Ca2+ exchanger in spontaneous automaticity. Front Pharmacol 11: 516, 2020.
 138.Koushik SV, Wang J, Rogers R, Moskophidis D, Lambert NA, Creazzo TL, Conway SJ. Targeted inactivation of the sodium‐calcium exchanger Ncx1 results in the lack of a heartbeat and abnormal myofibrillar organization. FASEB J 15: 1209‐1211, 2001.
 139.Lakatta EG, Maltsev VA, Vinogradova TM. A coupled SYSTEM of intracellular Ca2+ clocks and surface membrane voltage clocks controls the timekeeping mechanism of the heart's pacemaker. Circ Res 106: 659‐673, 2010.
 140.Larbig R, Torres N, Bridge JH, Goldhaber JI, Philipson KD. Activation of reverse Na+‐Ca2+ exchange by the Na+ current augments the cardiac Ca2+ transient: Evidence from NCX knockout mice. J Physiol 588: 3267‐3276, 2010.
 141.Leblanc N, Hume JR. Sodium current‐induced release of calcium from cardiac sarcoplasmic reticulum. Science 248: 372‐376, 1990.
 142.Lee C, Dhalla NS, Hryshko LV. Therapeutic potential of novel Na+‐Ca2+ exchange inhibitors in attenuating ischemia‐reperfusion injury. Can J Cardiol 21: 509‐516, 2005.
 143.Lee SL, Yu AS, Lytton J. Tissue‐specific expression of Na+‐Ca2+ exchanger isoforms. J Biol Chem 269: 14849‐14852, 1994.
 144.Lee SY, Giladi M, Bohbot H, Hiller R, Chung KY, Khananshvili D. Structure‐dynamic basis of splicing‐dependent regulation in tissue‐specific variants of the sodium‐calcium exchanger. FASEB J 30: 1356‐1366, 2016.
 145.Levitsky DO, Nicoll DA, Philipson KD. Identification of the high affinity Ca2+‐binding domain of the cardiac Na+‐Ca2+ exchanger. J Biol Chem 269: 22847‐22852, 1994.
 146.Li XF, Lytton J. A circularized sodium‐calcium exchanger exon 2 transcript. J Biol Chem 274: 8153‐8160, 1999.
 147.Li Z, Matsuoka S, Hryshko LV, Nicoll DA, Bersohn MM, Burke EP, Lifton RP, Philipson KD. Cloning of the NCX2 isoform of the plasma membrane Na+‐Ca2+ exchanger. J Biol Chem 269: 17434‐17439, 1994.
 148.Li ZP, Burke EP, Frank JS, Bennett V, Philipson KD. The cardiac Na+‐Ca2+ exchanger binds to the cytoskeletal protein ankyrin. J Biol Chem 268: 11489‐11491, 1993.
 149.Li ZP, Nicoll DA, Collins A, Hilgemann DW, Filoteo AG, Penniston JT, Weiss JN, Tomich JM, Philipson KD. Identification of a peptide inhibitor of the cardiac sarcolemmal Na+‐Ca2+ exchanger. J Biol Chem 266: 1014‐1020, 1991.
 150.Liao J, Li H, Zeng W, Sauer DB, Belmares R, Jiang Y. Structural insight into the ion‐exchange mechanism of the sodium/calcium exchanger. Science 335: 686‐690, 2012.
 151.Liao J, Marinelli F, Lee C, Huang Y, Faraldo‐Gomez JD, Jiang Y. Mechanism of extracellular ion exchange and binding‐site occlusion in a sodium/calcium exchanger. Nat Struct Mol Biol 23: 590‐599, 2016.
 152.Lin X, Jo H, Sakakibara Y, Tambara K, Kim B, Komeda M, Matsuoka S. Beta‐adrenergic stimulation does not activate Na+/Ca2+ exchange current in guinea pig, mouse, and rat ventricular myocytes. Am J Physiol Cell Physiol 290: C601‐C608, 2006.
 153.Linck B, Qiu Z, He Z, Tong Q, Hilgemann DW, Philipson KD. Functional comparison of the three isoforms of the Na+/Ca2+ exchanger NCX1, NCX2, NCX3. Am J Physiol 274: C415‐C423, 1998.
 154.Lindgren RM, Zhao J, Heller S, Berglind H, Nister M. Molecular cloning and characterization of two novel truncated isoforms of human Na+/Ca2+ exchanger 3, expressed in fetal brain. Gene 348: 143‐155, 2005.
 155.Lipp P, Niggli E. Sodium current‐induced calcium signals in isolated guinea‐pig ventricular myocytes. J Physiol 474: 439‐446, 1994.
 156.Liu T, Zhao J, Ibarra C, Garcia MU, Uhlen P, Nister M. Glycosylation controls sodium‐calcium exchanger 3 sub‐cellular localization during cell cycle. Eur J Cell Biol 97: 190‐203, 2018.
 157.Lytton J. Na+/Ca2+ Exchangers and Ca2+ Transport in Neurons. Boston, MA: Springer, 2007.
 158.Lytton J. Na+/Ca2+ exchangers: Three mammalian gene families control Ca2+ transport. Biochem J 406: 365‐382, 2007.
 159.Maack C, Ganesan A, Sidor A, O'Rourke B. Cardiac sodium‐calcium exchanger is regulated by allosteric calcium and exchanger inhibitory peptide at distinct sites. Circ Res 96: 91‐99, 2005.
 160.Maltsev VA, Lakatta EG. Normal heart rhythm is initiated and regulated by an intracellular calcium clock within pacemaker cells. Heart Lung Circ 16: 335‐348, 2007.
 161.Manfra O, Shen X, Hell JW, Edward Louch W. Super‐resolution dSTORM imaging of calcium handling proteins in cardiomyocytes. Biophysical Journal 114: 620a, 2018.
 162.Marinelli F, Almagor L, Hiller R, Giladi M, Khananshvili D, Faraldo‐Gomez JD. Sodium recognition by the Na+/Ca2+ exchanger in the outward‐facing conformation. Proc Natl Acad Sci U S A 111: E5354‐E5362, 2014.
 163.Marshall CR, Fox JA, Butland SL, Ouellette BF, Brinkman FS, Tibbits GF. Phylogeny of Na+/Ca2+ exchanger NCX genes from genomic data identifies new gene duplications and a new family member in fish species. Physiol Genomics 21: 161‐173, 2005.
 164.Matsuoka S, Hilgemann DW. Steady‐state and dynamic properties of cardiac sodium‐calcium exchange. Ion and voltage dependencies of the transport cycle. J Gen Physiol 100: 963‐1001, 1992.
 165.Matsuoka S, Hilgemann DW. Inactivation of outward Na+‐Ca2+ exchange current in guinea‐pig ventricular myocytes. J Physiol 476: 443‐458, 1994.
 166.Matsuoka S, Nicoll DA, He Z, Philipson KD. Regulation of cardiac Na+‐Ca2+ exchanger by the endogenous XIP region. J Gen Physiol 109: 273‐286, 1997.
 167.Matsuoka S, Nicoll DA, Hryshko LV, Levitsky DO, Weiss JN, Philipson KD. Regulation of the cardiac Na+‐Ca2+ exchanger by Ca2+. Mutational analysis of the Ca2+‐binding domain. J Gen Physiol 105: 403‐420, 1995.
 168.Matsuoka S, Nicoll DA, Reilly RF, Hilgemann DW, Philipson KD. Initial localization of regulatory regions of the cardiac sarcolemmal Na+‐Ca2+ exchanger. Proc Natl Acad Sci U S A 90: 3870‐3874, 1993.
 169.Matsuoka S, Philipson KD, Hilgemann DW. Multiple functional states of the cardiac Na+‐Ca2+ exchanger. Whole‐cell, native‐excised, and cloned‐excised properties. Ann N Y Acad Sci 779: 159‐170, 1996.
 170.Maxwell K, Scott J, Omelchenko A, Lukas A, Lu L, Lu Y, Hnatowich M, Philipson KD, Hryshko LV. Functional role of ionic regulation of Na+/Ca2+ exchange assessed in transgenic mouse hearts. Am J Physiol 277: H2212‐H2221, 1999.
 171.Miura Y, Kimura J. Sodium‐calcium exchange current. Dependence on internal Ca and Na and competitive binding of external Na and Ca. J Gen Physiol 93: 1129‐1145, 1989.
 172.Mohler PJ, Davis JQ, Bennett V. Ankyrin‐B coordinates the Na/K ATPase, Na/Ca exchanger, and InsP3 receptor in a cardiac T‐tubule/SR microdomain. PLoS Biol 3: e423, 2005.
 173.Mohler PJ, Schott JJ, Gramolini AO, Dilly KW, Guatimosim S, duBell WH, Song LS, Haurogne K, Kyndt F, Ali ME, Rogers TB, Lederer WJ, Escande D, Le Marec H, Bennett V. Ankyrin‐B mutation causes type 4 long‐QT cardiac arrhythmia and sudden cardiac death. Nature 421: 634‐639, 2003.
 174.Molinaro P, Cuomo O, Pignataro G, Boscia F, Sirabella R, Pannaccione A, Secondo A, Scorziello A, Adornetto A, Gala R, Viggiano D, Sokolow S, Herchuelz A, Schurmans S, Di Renzo G, Annunziato L. Targeted disruption of Na+/Ca2+ exchanger 3 NCX3 gene leads to a worsening of ischemic brain damage. J Neurosci 28: 1179‐1184, 2008.
 175.Molinaro P, Natale S, Serani A, Calabrese L, Secondo A, Tedeschi V, Valsecchi V, Pannaccione A, Scorziello A, Annunziato L. Genetically modified mice to unravel physiological and pathophysiological roles played by NCX isoforms. Cell Calcium 87: 102189, 2020.
 176.Molinaro P, Pannaccione A, Sisalli MJ, Secondo A, Cuomo O, Sirabella R, Cantile M, Ciccone R, Scorziello A, di Renzo G, Annunziato L. A new cell‐penetrating peptide that blocks the autoinhibitory XIP domain of NCX1 and enhances antiporter activity. Mol Ther 23: 465‐476, 2015.
 177.Morad M, Cleemann L, Menick DR. NCX1 phosphorylation dilemma: A little closer to resolution. Focus on “Full‐length cardiac Na+/Ca2+ exchanger 1 protein is not phosphorylated by protein kinase A”. Am J Physiol Cell Physiol 300: C970‐C973, 2011.
 178.Muller JG, Isomatsu Y, Koushik SV, O'Quinn M, Xu L, Kappler CS, Hapke E, Zile MR, Conway SJ, Menick DR. Cardiac‐specific expression and hypertrophic upregulation of the feline Na+‐Ca2+ exchanger gene H1‐promoter in a transgenic mouse model. Circ Res 90: 158‐164, 2002.
 179.Nakasaki Y, Iwamoto T, Hanada H, Imagawa T, Shigekawa M. Cloning of the rat aortic smooth muscle Na+/Ca2+ exchanger and tissue‐specific expression of isoforms. J Biochem 114: 528‐534, 1993.
 180.Neco P, Rose B, Huynh N, Zhang R, Bridge JH, Philipson KD, Goldhaber JI. Sodium‐calcium exchange is essential for effective triggering of calcium release in mouse heart. Biophys J 99: 755‐764, 2010.
 181.Nicholas SB, Philipson KD. Cardiac expression of the Na+/Ca2+ exchanger NCX1 is GATA factor dependent. Am J Physiol 277: H324‐H330, 1999.
 182.Nicholas SB, Yang W, Lee SL, Zhu H, Philipson KD, Lytton J. Alternative promoters and cardiac muscle cell‐specific expression of the Na+/Ca2+ exchanger gene. Am J Physiol 274: H217‐H232, 1998.
 183.Nicoll DA, Hryshko LV, Matsuoka S, Frank JS, Philipson KD. Mutagenesis studies of the cardiac Na+‐Ca2+ exchanger. Ann N Y Acad Sci 779: 86‐92, 1996.
 184.Nicoll DA, Hryshko LV, Matsuoka S, Frank JS, Philipson KD. Mutation of amino acid residues in the putative transmembrane segments of the cardiac sarcolemmal Na+‐Ca2+ exchanger. J Biol Chem 271: 13385‐13391, 1996.
 185.Nicoll DA, Longoni S, Philipson KD. Molecular cloning and functional expression of the cardiac sarcolemmal Na+‐Ca2+ exchanger. Science 250: 562‐565, 1990.
 186.Nicoll DA, Ottolia M, Lu L, Lu Y, Philipson KD. A new topological model of the cardiac sarcolemmal Na+‐Ca2+ exchanger. J Biol Chem 274: 910‐917, 1999.
 187.Nicoll DA, Ottolia M, Philipson KD. Toward a topological model of the NCX1 exchanger. Ann N Y Acad Sci 976: 11‐18, 2002.
 188.Nicoll DA, Quednau BD, Qui Z, Xia YR, Lusis AJ, Philipson KD. Cloning of a third mammalian Na+‐Ca2+ exchanger, NCX3. J Biol Chem 271: 24914‐24921, 1996.
 189.Nicoll DA, Sawaya MR, Kwon S, Cascio D, Philipson KD, Abramson J. The crystal structure of the primary Ca2+ sensor of the Na+/Ca2+ exchanger reveals a novel Ca2+ binding motif. J Biol Chem 281: 21577‐21581, 2006.
 190.Niggli E, Lederer WJ. Molecular operations of the sodium‐calcium exchanger revealed by conformation currents. Nature 349: 621‐624, 1991.
 191.On C, Marshall CR, Chen N, Moyes CD, Tibbits GF. Gene structure evolution of the Na+‐Ca2+ exchanger NCX family. BMC Evol Biol 8: 127, 2008.
 192.Ottolia M, John S, Qiu Z, Philipson KD. Split Na+‐Ca2+ exchangers. Implications for function and expression. J Biol Chem 276: 19603‐19609, 2001.
 193.Ottolia M, John S, Ren X, Philipson KD. Fluorescent Na+‐Ca+ exchangers: Electrophysiological and optical characterization. J Biol Chem 282: 3695‐3701, 2007.
 194.Ottolia M, Nicoll DA, John S, Philipson KD. Interactions between Ca2+ binding domains of the Na+‐Ca2+ exchanger and secondary regulation. Channels Austin 4: 159‐162, 2010.
 195.Ottolia M, Nicoll DA, Philipson KD. Mutational analysis of the alpha‐1 repeat of the cardiac Na+‐Ca2+ exchanger. J Biol Chem 280: 1061‐1069, 2005.
 196.Ottolia M, Nicoll DA, Philipson KD. Roles of two Ca2+‐binding domains in regulation of the cardiac Na+‐Ca2+ exchanger. J Biol Chem 284: 32735‐32741, 2009.
 197.Ottolia M, Philipson KD, John S. Conformational changes of the Ca2+ regulatory site of the Na+‐Ca2+ exchanger detected by FRET. Biophys J 87: 899‐906, 2004.
 198.Ottolia M, Torres N, Bridge JH, Philipson KD, Goldhaber JI. Na/Ca exchange and contraction of the heart. J Mol Cell Cardiol 61: 28‐33, 2013.
 199.Perez C, Khafizov K, Forrest LR, Kramer R, Ziegler C. The role of trimerization in the osmoregulated betaine transporter BetP. EMBO Rep 12: 804‐810, 2011.
 200.Philipson KD. Interaction of charged amphiphiles with Na+‐Ca2+ exchange in cardiac sarcolemmal vesicles. J Biol Chem 259: 13999‐14002, 1984.
 201.Philipson KD, Bersohn MM, Nishimoto AY. Effects of pH on Na+‐Ca2+ exchange in canine cardiac sarcolemmal vesicles. Circ Res 50: 287‐293, 1982.
 202.Philipson KD, Frank JS, Nishimoto AY. Effects of phospholipase C on the Na+‐Ca2+ exchange and Ca2+ permeability of cardiac sarcolemmal vesicles. J Biol Chem 258: 5905‐5910, 1983.
 203.Philipson KD, Longoni S, Ward R. Purification of the cardiac Na+‐Ca2+ exchange protein. Biochim Biophys Acta 945: 298‐306, 1988.
 204.Philipson KD, Nishimoto AY. Na+‐Ca2+ exchange in inside‐out cardiac sarcolemmal vesicles. J Biol Chem 257: 5111‐5117, 1982.
 205.Philipson KD, Nishimoto AY. Stimulation of Na+‐Ca2+ exchange in cardiac sarcolemmal vesicles by phospholipase D. J Biol Chem 259: 16‐19, 1984.
 206.Philipson KD, Ward R. Effects of fatty acids on Na+‐Ca2+ exchange and Ca2+ permeability of cardiac sarcolemmal vesicles. J Biol Chem 260: 9666‐9671, 1985.
 207.Plain F, Congreve SD, Yee RSZ, Kennedy J, Howie J, Kuo CW, Fraser NJ, Fuller W. An amphipathic alpha‐helix directs palmitoylation of the large intracellular loop of the sodium/calcium exchanger. J Biol Chem 292: 10745‐10752, 2017.
 208.Pogwizd SM. Clinical potential of sodium‐calcium exchanger inhibitors as antiarrhythmic agents. Drugs 63: 439‐452, 2003.
 209.Pogwizd SM, Bers DM. Na/Ca exchange in heart failure: Contractile dysfunction and arrhythmogenesis. Ann N Y Acad Sci 976: 454‐465, 2002.
 210.Pogwizd SM, Bers DM. Cellular basis of triggered arrhythmias in heart failure. Trends Cardiovasc Med 14: 61‐66, 2004.
 211.Pogwizd SM, Qi M, Yuan W, Samarel AM, Bers DM. Upregulation of Na+/Ca2+ exchanger expression and function in an arrhythmogenic rabbit model of heart failure. Circ Res 85: 1009‐1019, 1999.
 212.Porzig H, Li Z, Nicoll DA, Philipson KD. Mapping of the cardiac sodium‐calcium exchanger with monoclonal antibodies. Am J Physiol 265: C748‐C756, 1993.
 213.Pott C, Eckardt L, Goldhaber JI. Triple threat: The Na+/Ca2+ exchanger in the pathophysiology of cardiac arrhythmia, ischemia and heart failure. Curr Drug Targets 12: 737‐747, 2011.
 214.Pott C, Goldhaber JI, Philipson KD. Genetic manipulation of cardiac Na+/Ca2+ exchange expression. Biochem Biophys Res Commun 322: 1336‐1340, 2004.
 215.Pott C, Goldhaber JI, Philipson KD. Homozygous overexpression of the Na+‐Ca2+ exchanger in mice: Evidence for increased transsarcolemmal Ca2+ fluxes. Ann N Y Acad Sci 1099: 310‐314, 2007.
 216.Pott C, Henderson SA, Goldhaber JI, Philipson KD. Na+/Ca2+ exchanger knockout mice: Plasticity of cardiac excitation‐contraction coupling. Ann N Y Acad Sci 1099: 270‐275, 2007.
 217.Pott C, Muszynski A, Ruhe M, Bogeholz N, Schulte JS, Milberg P, Monnig G, Fabritz L, Goldhaber JI, Breithardt G, Schmitz W, Philipson KD, Eckardt L, Kirchhof P, Muller FU. Proarrhythmia in a non‐failing murine model of cardiac‐specific Na+/Ca 2+ exchanger overexpression: Whole heart and cellular mechanisms. Basic Res Cardiol 107: 247, 2012.
 218.Pott C, Philipson KD, Goldhaber JI. Excitation‐contraction coupling in Na+‐Ca2+ exchanger knockout mice: Reduced transsarcolemmal Ca2+ flux. Circ Res 97: 1288‐1295, 2005.
 219.Pott C, Ren X, Tran DX, Yang MJ, Henderson S, Jordan MC, Roos KP, Garfinkel A, Philipson KD, Goldhaber JI. Mechanism of shortened action potential duration in Na+‐Ca2+ exchanger knockout mice. Am J Physiol Cell Physiol 292: C968‐C973, 2007.
 220.Quednau BD, Nicoll DA, Philipson KD. Tissue specificity and alternative splicing of the Na+/Ca2+ exchanger isoforms NCX1, NCX2, and NCX3 in rat. Am J Physiol 272: C1250‐C1261, 1997.
 221.Quednau BD, Nicoll DA, Philipson KD. The sodium/calcium exchanger family‐SLC8. Pflugers Arch 447: 543‐548, 2004.
 222.Radwanski PB, Brunello L, Veeraraghavan R, Ho HT, Lou Q, Makara MA, Belevych AE, Anghelescu M, Priori SG, Volpe P, Hund TJ, Janssen PM, Mohler PJ, Bridge JH, Poelzing S, Gyorke S. Neuronal Na+ channel blockade suppresses arrhythmogenic diastolic Ca2+ release. Cardiovasc Res 106: 143‐152, 2015.
 223.Ramos‐Franco J, Aguilar‐Sanchez Y, Escobar AL. Intact heart loose patch photolysis reveals ionic current kinetics during ventricular action potentials. Circ Res 118: 203‐215, 2016.
 224.Ranu HK, Terracciano CM, Davia K, Bernobich E, Chaudhri B, Robinson SE, Bin Kang Z, Hajjar RJ, MacLeod KT, Harding SE. Effects of Na+/Ca2+‐exchanger overexpression on excitation‐contraction coupling in adult rabbit ventricular myocytes. J Mol Cell Cardiol 34: 389‐400, 2002.
 225.Reeves JP, Hale CC. The stoichiometry of the cardiac sodium‐calcium exchange system. J Biol Chem 259: 7733‐7739, 1984.
 226.Reilly L, Howie J, Wypijewski K, Ashford ML, Hilgemann DW, Fuller W. Palmitoylation of the Na/Ca exchanger cytoplasmic loop controls its inactivation and internalization during stress signaling. FASEB J 29: 4532‐4543, 2015.
 227.Ren X, Nicoll DA, Galang G, Philipson KD. Intermolecular cross‐linking of Na+‐Ca2+ exchanger proteins: Evidence for dimer formation. Biochemistry 47: 6081‐6087, 2008.
 228.Ren X, Philipson KD. The topology of the cardiac Na+/Ca2+ exchanger, NCX1. J Mol Cell Cardiol 57: 68‐71, 2013.
 229.Reuter H, Han T, Motter C, Philipson KD, Goldhaber JI. Mice overexpressing the cardiac sodium‐calcium exchanger: Defects in excitation‐contraction coupling. J Physiol 554: 779‐789, 2004.
 230.Reuter H, Henderson SA, Han T, Mottino GA, Frank JS, Ross RS, Goldhaber JI, Philipson KD. Cardiac excitation‐contraction coupling in the absence of Na+ ‐ Ca2+ exchange. Cell Calcium 34: 19‐26, 2003.
 231.Reuter H, Henderson SA, Han T, Ross RS, Goldhaber JI, Philipson KD. The Na+‐Ca2+ exchanger is essential for the action of cardiac glycosides. Circ Res 90: 305‐308, 2002.
 232.Reuter H, Seitz N. The dependence of calcium efflux from cardiac muscle on temperature and external ion composition. J Physiol 195: 451‐470, 1968.
 233.Riedel MJ, Baczko I, Searle GJ, Webster N, Fercho M, Jones L, Lang J, Lytton J, Dyck JR, Light PE. Metabolic regulation of sodium‐calcium exchange by intracellular acyl CoAs. Embo J 25: 4605‐4614, 2006.
 234.Rimon A, Tzubery T, Padan E. Monomers of the NhaA Na+/H+ antiporter of Escherichia coli are fully functional yet dimers are beneficial under extreme stress conditions at alkaline pH in the presence of Na+ or Li+. J Biol Chem 282: 26810‐26821, 2007.
 235.Robertson JL, Kolmakova‐Partensky L, Miller C. Design, function and structure of a monomeric ClC transporter. Nature 468: 844‐847, 2010.
 236.Roos KP, Jordan MC, Fishbein MC, Ritter MR, Friedlander M, Chang HC, Rahgozar P, Han T, Garcia AJ, Maclellan WR, Ross RS, Philipson KD. Hypertrophy and heart failure in mice overexpressing the cardiac sodium‐calcium exchanger. J Card Fail 13: 318‐329, 2007.
 237.Rosker C, Graziani A, Lukas M, Eder P, Zhu MX, Romanin C, Groschner K. Ca2+ signaling by TRPC3 involves Na+ entry and local coupling to the Na+/Ca2+ exchanger. J Biol Chem 279: 13696‐13704, 2004.
 238.Ruknudin AM, Wei SK, Haigney MC, Lederer WJ, Schulze DH. Phosphorylation and other conundrums of Na/Ca exchanger, NCX1. Ann N Y Acad Sci 1099: 103‐118, 2007.
 239.Sahin‐Toth M, Nicoll DA, Frank JS, Philipson KD, Friedlander M. The cleaved N‐terminal signal sequence of the cardiac Na+‐Ca2+ exchanger is not required for functional membrane integration. Biochem Biophys Res Commun 212: 968‐974, 1995.
 240.Salinas RK, Bruschweiler‐Li L, Johnson E, Bruschweiler R. Ca2+ binding alters the interdomain flexibility between the two cytoplasmic calcium‐binding domains in the Na+/Ca2+ exchanger. J Biol Chem 286: 32123‐32131, 2011.
 241.Santacruz‐Toloza L, Ottolia M, Nicoll DA, Philipson KD. Functional analysis of a disulfide bond in the cardiac Na+‐Ca2+ exchanger. J Biol Chem 275: 182‐188, 2000.
 242.Schafer C, Ladilov Y, Inserte J, Schafer M, Haffner S, Garcia‐Dorado D, Piper HM. Role of the reverse mode of the Na+/Ca2+ exchanger in reoxygenation‐induced cardiomyocyte injury. Cardiovasc Res 51: 241‐250, 2001.
 243.Schulson MN, Scriven DR, Fletcher P, Moore ED. Couplons in rat atria form distinct subgroups defined by their molecular partners. J Cell Sci 124: 1167‐1174, 2011.
 244.Schulze DH, Muqhal M, Lederer WJ, Ruknudin AM. Sodium/calcium exchanger NCX1 macromolecular complex. J Biol Chem 278: 28849‐28855, 2003.
 245.Schwarz EM, Benzer S. Calx, a Na‐Ca exchanger gene of Drosophila melanogaster. Proc Natl Acad Sci U S A 94: 10249‐10254, 1997.
 246.Scranton K, Umar S, Calmettes G, Eghbali M, Goldhaber JI, John SA, Olcese R, Escobar AL, Ottolia M. The Na+‐dependent inactivation of NCX1.1 is physiologically relevant to cardiac function. Biophys J 118: 100a‐101a, 2020.
 247.Scriven DR, Dan P, Moore ED. Distribution of proteins implicated in excitation‐contraction coupling in rat ventricular myocytes. Biophys J 79: 2682‐2691, 2000.
 248.Shannon TR, Hale CC, Milanick MA. Interaction of cardiac Na‐Ca exchanger and exchange inhibitory peptide with membrane phospholipids. Am J Physiol 266: C1350‐C1356, 1994.
 249.Shattock MJ, Ottolia M, Bers DM, Blaustein MP, Boguslavskyi A, Bossuyt J, Bridge JH, Chen‐Izu Y, Clancy CE, Edwards A, Goldhaber J, Kaplan J, Lingrel JB, Pavlovic D, Philipson K, Sipido KR, Xie ZJ. Na+/Ca2+ exchange and Na+/K+‐ATPase in the heart. J Physiol 593: 1361‐1382, 2015.
 250.Shen C, Lin MJ, Yaradanakul A, Lariccia V, Hill JA, Hilgemann DW. Dual control of cardiac Na+ Ca2+ exchange by PIP2: Analysis of the surface membrane fraction by extracellular cysteine PEGylation. J Physiol 582: 1011‐1026, 2007.
 251.Shieh BH, Xia Y, Sparkes RS, Klisak I, Lusis AJ, Nicoll DA, Philipson KD. Mapping of the gene for the cardiac sarcolemmal Na+‐Ca2+ exchanger to human chromosome 2p21‐p23. Genomics 12: 616‐617, 1992.
 252.Shigekawa M, Iwamoto T, Uehara A, Kita S. Probing ion binding sites in the Na+/Ca2+ exchanger. Ann N Y Acad Sci 976: 19‐30, 2002.
 253.Shipston MJ. Ion channel regulation by protein palmitoylation. J Biol Chem 286: 8709‐8716, 2011.
 254.Shlosman I, Marinelli F, Faraldo‐Gomez JD, Mindell JA. The prokaryotic Na+/Ca2+ exchanger NCX_Mj transports Na+ and Ca2+ in a 3:1 stoichiometry. J Gen Physiol 150: 51‐65, 2018.
 255.Shuart NG, Haitin Y, Camp SS, Black KD, Zagotta WN. Molecular mechanism for 3:1 subunit stoichiometry of rod cyclic nucleotide‐gated ion channels. Nat Commun 2: 457, 2011.
 256.Sitte HH, Farhan H, Javitch JA. Sodium‐dependent neurotransmitter transporters: Oligomerization as a determinant of transporter function and trafficking. Mol Interv 4: 38‐47, 2004.
 257.Skogestad J, Aronsen JM, Tovsrud N, Wanichawan P, Hougen K, Stokke MK, Carlson CR, Sjaastad I, Sejersted OM, Swift F. Coupling of the Na+/K+‐ATPase to Ankyrin B controls Na+/Ca2+ exchanger activity in cardiomyocytes. Cardiovasc Res 116: 78‐90, 2020.
 258.Sokolow S, Manto M, Gailly P, Molgo J, Vandebrouck C, Vanderwinden JM, Herchuelz A, Schurmans S. Impaired neuromuscular transmission and skeletal muscle fiber necrosis in mice lacking Na/Ca exchanger 3. J Clin Invest 113: 265‐273, 2004.
 259.Sorimachi H, Ono Y. Regulation and physiological roles of the calpain system in muscular disorders. Cardiovasc Res 96: 11‐22, 2012.
 260.Steccanella F, Scranton K, Panday N, Angelini M, Zhang R, Lotteau S, John SA, Olcese R, Goldhaber JI, Ottolia M. Genetic ablation of NCX1.1 Na+‐dependent inactivation impacts cardiac action potential and Ca2+ transient. Biophys J 118: 100a, 2020.
 261.Stieber J, Herrmann S, Feil S, Loster J, Feil R, Biel M, Hofmann F, Ludwig A. The hyperpolarization‐activated channel HCN4 is required for the generation of pacemaker action potentials in the embryonic heart. Proc Natl Acad Sci U S A 100: 15235‐15240, 2003.
 262.Sugishita K, Su Z, Li F, Philipson KD, Barry WH. Gender influences [Ca2+]i during metabolic inhibition in myocytes overexpressing the Na+‐Ca2+ exchanger. Circulation 104: 2101‐2106, 2001.
 263.Swift F, Birkeland JA, Tovsrud N, Enger UH, Aronsen JM, Louch WE, Sjaastad I, Sejersted OM. Altered Na+/Ca2+‐exchanger activity due to downregulation of Na+/K+‐ATPase alpha2‐isoform in heart failure. Cardiovasc Res 78: 71‐78, 2008.
 264.Swift F, Tovsrud N, Enger UH, Sjaastad I, Sejersted OM. The Na+/K+‐ATPase alpha2‐isoform regulates cardiac contractility in rat cardiomyocytes. Cardiovasc Res 75: 109‐117, 2007.
 265.Szerencsei RT, Kinjo TG, Schnetkamp PP. The topology of the C‐terminal sections of the NCX1 Na +/Ca 2+ exchanger and the NCKX2 Na +/Ca 2+ ‐K + exchanger. Channels Austin 7: 109‐114, 2013.
 266.Terracciano CM, Souza AI, Philipson KD, MacLeod KT. Na+‐Ca2+ exchange and sarcoplasmic reticular Ca2+ regulation in ventricular myocytes from transgenic mice overexpressing the Na+‐Ca2+ exchanger. J Physiol 512 (Pt 3): 651‐667, 1998.
 267.Thomas MJ, Sjaastad I, Andersen K, Helm PJ, Wasserstrom JA, Sejersted OM, Ottersen OP. Localization and function of the Na+/Ca2+‐exchanger in normal and detubulated rat cardiomyocytes. J Mol Cell Cardiol 35: 1325‐1337, 2003.
 268.Tombola F, Ulbrich MH, Kohout SC, Isacoff EY. The opening of the two pores of the Hv1 voltage‐gated proton channel is tuned by cooperativity. Nat Struct Mol Biol 17: 44‐50, 2010.
 269.Torrente AG, Zhang R, Wang H, Zaini A, Kim B, Yue X, Philipson KD, Goldhaber JI. Contribution of small conductance K+ channels to sinoatrial node pacemaker activity: Insights from atrial‐specific Na+/Ca2+ exchange knockout mice. J Physiol 595: 3847‐3865, 2017.
 270.Torrente AG, Zhang R, Zaini A, Giani JF, Kang J, Lamp ST, Philipson KD, Goldhaber JI. Burst pacemaker activity of the sinoatrial node in sodium‐calcium exchanger knockout mice. Proc Natl Acad Sci U S A 112: 9769‐9774, 2015.
 271.Torres NS, Larbig R, Rock A, Goldhaber JI, Bridge JH. Na+ currents are required for efficient excitation‐contraction coupling in rabbit ventricular myocytes: A possible contribution of neuronal Na+ channels. J Physiol 588: 4249‐4260, 2010.
 272.van der Vusse GJ, van Bilsen M, Reneman RS. Ischemia and reperfusion induced alterations in membrane phospholipids: An overview. Ann N Y Acad Sci 723: 1‐14, 1994.
 273.van Dijk L, Giladi M, Refaeli B, Hiller R, Cheng MH, Bahar I, Khananshvili D. Key residues controlling bidirectional ion movements in Na+/Ca2+ exchanger. Cell Calcium 76: 10‐22, 2018.
 274.Van Eylen F, Kamagate A, Herchuelz A. A new Na/Ca exchanger splicing pattern identified in situ leads to a functionally active 70kDa NH2‐terminal protein. Cell Calcium 30: 191‐198, 2001.
 275.Vaughan‐Jones RD, Spitzer KW, Swietach P. Intracellular pH regulation in heart. J Mol Cell Cardiol 46: 318‐331, 2009.
 276.Vemuri R, Philipson KD. Phospholipid composition modulates the Na+‐Ca2+ exchange activity of cardiac sarcolemma in reconstituted vesicles. Biochim Biophys Acta 937: 258‐268, 1988.
 277.Waight AB, Pedersen BP, Schlessinger A, Bonomi M, Chau BH, Roe‐Zurz Z, Risenmay AJ, Sali A, Stroud RM. Structural basis for alternating access of a eukaryotic calcium/proton exchanger. Nature 499: 107‐110, 2013.
 278.Wakimoto K, Fujimura H, Iwamoto T, Oka T, Kobayashi K, Kita S, Kudoh S, Kuro‐o M, Nabeshima Y, Shigekawa M, Imai Y, Komuro I. Na+/Ca2+ exchanger‐deficient mice have disorganized myofibrils and swollen mitochondria in cardiomyocytes. Comp Biochem Physiol B Biochem Mol Biol 135: 9‐15, 2003.
 279.Wakimoto K, Kobayashi K, Kuro OM, Yao A, Iwamoto T, Yanaka N, Kita S, Nishida A, Azuma S, Toyoda Y, Omori K, Imahie H, Oka T, Kudoh S, Kohmoto O, Yazaki Y, Shigekawa M, Imai Y, Nabeshima Y, Komuro I. Targeted disruption of Na+/Ca2+ exchanger gene leads to cardiomyocyte apoptosis and defects in heartbeat. J Biol Chem 275: 36991‐36998, 2000.
 280.Wang J, Chan TO, Zhang XQ, Gao E, Song J, Koch WJ, Feldman AM, Cheung JY. Induced overexpression of Na+/Ca2+ exchanger transgene: Altered myocyte contractility, [Ca2+]i transients, SR Ca2+ contents, and action potential duration. Am J Physiol Heart Circ Physiol 297: H590‐H601, 2009.
 281.Wanichawan P, Hafver TL, Hodne K, Aronsen JM, Lunde IG, Dalhus B, Lunde M, Kvaloy H, Louch WE, Tonnessen T, Sjaastad I, Sejersted OM, Carlson CR. Molecular basis of calpain cleavage and inactivation of the sodium‐calcium exchanger 1 in heart failure. J Biol Chem 289: 33984‐33998, 2014.
 282.Wanichawan P, Louch WE, Hortemo KH, Austbo B, Lunde PK, Scott JD, Sejersted OM, Carlson CR. Full‐length cardiac Na+/Ca2+ exchanger 1 protein is not phosphorylated by protein kinase A. Am J Physiol Cell Physiol 300: C989‐C997, 2011.
 283.Weber CR, Ginsburg KS, Bers DM. Cardiac submembrane [Na+] transients sensed by Na+‐Ca2+ exchange current. Circ Res 92: 950‐952, 2003.
 284.Weber CR, Ginsburg KS, Philipson KD, Shannon TR, Bers DM. Allosteric regulation of Na/Ca exchange current by cytosolic Ca in intact cardiac myocytes. J Gen Physiol 117: 119‐131, 2001.
 285.Weber CR, Piacentino V 3rd, Ginsburg KS, Houser SR, Bers DM. Na+‐Ca2+ exchange current and submembrane [Ca2+] during the cardiac action potential. Circ Res 90: 182‐189, 2002.
 286.Weber CR, Piacentino V 3rd, Houser SR, Bers DM. Dynamic regulation of sodium/calcium exchange function in human heart failure. Circulation 108: 2224‐2229, 2003.
 287.Wetzel GT, Chen F, Klitzner TS. Na+/Ca2+ exchange and cell contraction in isolated neonatal and adult rabbit cardiac myocytes. Am J Physiol 268: H1723‐H1733, 1995.
 288.Wu M, Le HD, Wang M, Yurkov V, Omelchenko A, Hnatowich M, Nix J, Hryshko LV, Zheng L. Crystal structures of progressive Ca2+ binding states of the Ca2+ sensor Ca2+ binding domain 1 CBD1 from the CALX Na+/Ca2+ exchanger reveal incremental conformational transitions. J Biol Chem 285: 2554‐2561, 2009.
 289.Wu M, Tong S, Gonzalez J, Jayaraman V, Spudich JL, Zheng L. Structural basis of the Ca2+ inhibitory mechanism of Drosophila Na+/Ca2+ exchanger CALX and its modification by alternative splicing. Structure 19: 1509‐1517, 2011.
 290.Wu M, Tong S, Waltersperger S, Diederichs K, Wang M, Zheng L. Crystal structure of Ca2+/H+ antiporter protein YfkE reveals the mechanisms of Ca2+ efflux and its pH regulation. Proc Natl Acad Sci U S A 110: 11367‐11372, 2013.
 291.Xiao YF, Ke Q, Chen Y, Morgan JP, Leaf A. Inhibitory effect of n‐3 fish oil fatty acids on cardiac Na+/Ca2+ exchange currents in HEK293t cells. Biochem Biophys Res Commun 321: 116‐123, 2004.
 292.Xie Y, Ottolia M, John SA, Chen JN, Philipson KD. Conformational changes of a Ca2+‐binding domain of the Na+/Ca2+ exchanger monitored by FRET in transgenic zebrafish heart. Am J Physiol Cell Physiol 295: C388‐C393, 2008.
 293.Yao A, Su Z, Nonaka A, Zubair I, Lu L, Philipson KD, Bridge JH, Barry WH. Effects of overexpression of the Na+‐Ca2+ exchanger on [Ca2+]i transients in murine ventricular myocytes. Circ Res 82: 657‐665, 1998.
 294.Yaradanakul A, Feng S, Shen C, Lariccia V, Lin MJ, Yang J, Kang TM, Dong P, Yin HL, Albanesi JP, Hilgemann DW. Dual control of cardiac Na+ Ca2+ exchange by PIP2: Electrophysiological analysis of direct and indirect mechanisms. J Physiol 582: 991‐1010, 2007.
 295.Yuan J, Yuan C, Xie M, Yu L, Bruschweiler‐Li L, Bruschweiler R. The intracellular loop of the Na+/Ca2+ exchanger contains an "Awareness Ribbon"‐shaped two‐helix bundle domain. Biochemistry 57: 5096‐5104, 2018.
 296.Yue X, Hazan A, Lotteau S, Zhang R, Torrente AG, Philipson KD, Ottolia M, Goldhaber JI. Na/Ca exchange in the atrium: Role in sinoatrial node pacemaking and excitation‐contraction coupling. Cell Calcium 87: 102167, 2020.
 297.Yue X, Zhang R, Kim B, Ma A, Philipson KD, Goldhaber JI. Heterogeneity of transverse‐axial tubule system in mouse atria: Remodeling in atrial‐specific Na+‐Ca2+ exchanger knockout mice. J Mol Cell Cardiol 108: 50‐60, 2017.
 298.Zhang J, Lee MY, Cavalli M, Chen L, Berra‐Romani R, Balke CW, Bianchi G, Ferrari P, Hamlyn JM, Iwamoto T, Lingrel JB, Matteson DR, Wier WG, Blaustein MP. Sodium pump alpha2 subunits control myogenic tone and blood pressure in mice. J Physiol 569: 243‐256, 2005.
 299.Zhang XQ, Ahlers BA, Tucker AL, Song J, Wang J, Moorman JR, Mounsey JP, Carl LL, Rothblum LI, Cheung JY. Phospholemman inhibition of the cardiac Na+/Ca2+ exchanger. Role of phosphorylation. J Biol Chem 281: 7784‐7792, 2006.
 300.Zhao J, Shigaki T, Mei H, Guo YQ, Cheng NH, Hirschi KD. Interaction between Arabidopsis Ca2+/H+ exchangers CAX1 and CAX3. J Biol Chem 284: 4605‐4615, 2009.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Michela Ottolia, Scott John, Adina Hazan, Joshua I. Goldhaber. The Cardiac Na+‐Ca2+ Exchanger: From Structure to Function. Compr Physiol 2021, 12: 2681-2717. doi: 10.1002/cphy.c200031