Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jul 5.
Published in final edited form as: Chembiochem. 2022 Mar 21;23(13):e202100625. doi: 10.1002/cbic.202100625

Chemical and Biological Tools for the Study of Voltage-Gated Sodium Channels in Electrogenesis and Nociception

Anna V Elleman a, J Du Bois a
PMCID: PMC9359671  NIHMSID: NIHMS1822658  PMID: 35315190

Abstract

The malfunction and misregulation of voltage-gated sodium channels (NaVs) underlie in large part the electrical hyper-excitability characteristic of chronic inflammatory and neuropathic pain. NaVs are responsible for the initiation and propagation of electrical impulses (action potentials) in cells. Tissue and nerve injury alter the expression and localization of multiple NaV isoforms, including NaV1.1, 1.3, and 1.6–1.9, resulting in aberrant action potential firing patterns. To better understand the role of NaV regulation, localization, and trafficking in electrogenesis and pain pathogenesis, a number of chemical and biological reagents for interrogating NaV function have been advanced. The development and application of such tools for understanding NaV physiology are the focus of this review.

Keywords: chemical biology, ion channels, molecular physiology, neurochemistry

1. Voltage-Gated Sodium Channels

1.1. Channel structure

Voltage-gated sodium channels comprise a 240–260 kDa sodium-conducting α-subunit (Figure 1) and one or two auxiliary β-subunits. The α-subunit includes four homologous repeats (DI-DIV), each containing six membrane-spanning α-helices (S1-S6). Helices S5 and S6 compose the so-called pore domain (PD), whereas S1–S4 form the voltage-sensing domains (VSD).[1] At the base of the extracellular mouth, four residues—aspartate, glutamate, lysine, alanine (DEKA)—form a filter that selects for hydrated sodium ions.[2] High resolution structures of α-subunits for different mammalian NaV subtypes, including hNaV1.2,[3] 1.4,[4] 1.5,[5] and 1.7,[6] as well as αβ-heteromeric complexes[3,4,6,7] are now available owing to recent advances in cryogenic electron microscopy (cryo-EM).

Figure 1.

Figure 1.

Human voltage-gated sodium channel, hNaV1.7 (PDB 6J8J).[6] A) Side view. IFM fast inactivation particle shown in yellow (space filling model).[4] B) Top view. Domain I (orange), Domain II (red), Domain III (gray), Domain IV (cyan).

The S4 helix of each voltage sensor includes positively charged arginine residues that are sensitive to the membrane potential. As the cell depolarizes, the S4 helices move outward, triggering conformational changes that prompt the opening of the channel pore.[8] Movement of the DIV S4 helix is the last to occur,[9] resulting in a synchronous conformational change in the intracellular loop between DIII and DIV. Within this loop, the position of the isoleucine-phenylalanine-methionine (IFM) particle is altered to inhibit Na+ conduction—a process termed fast inactivation, which occurs within milliseconds of channel opening.[10] Slow inactivation, which ensues on the timescale of tens of milliseconds to seconds, arises from additional movement of the VS domains as a result of prolonged and/or repetitive stimulation.[11] Subsequent membrane repolarization resets the conformation of the voltage-sensing domains, returning the channel to the closed, resting state. Auxiliary proteins, so-called β subunits (β1A, β1B, β2, β3, and β4), are positioned on the extracellular face of the channel and modulate the movement of the voltage sensors, thereby altering the voltage dependence and gating properties of the α-subunit.[12] The totality of the conformational changes of this heteromeric protein complex in response to membrane voltage regulates the Na+ component (i.e., rising phase) of the action potential (AP) (Figure 2).

Figure 2.

Figure 2.

A) NaV component of the action potential. (1) Closed (2) Open (3) Inactivated. B) Limiting conformational states of NaV.

Voltage dependence and gating properties differ among NaV isoforms (see Section 1.2) (Table 1).[8] Subtle variations in channel expression levels and cellular distributions and in the intrinsic gating properties of each subtype give rise to the diversity of AP signal outputs. Specifically, NaV isoforms display disparate voltage dependences of activation and inactivation, time constants of inactivation, and rate constants for transitioning between conformational states. The initial transition from closed to open channels results in Na+ influx, termed transient current, which ceases within milliseconds as channels inactivate. In some cases, incomplete inactivation can produce a longlasting, so-called persistent Na+ current.[13] Membrane repolarization generally returns channels to the reprimed, closed state. Some channels, however, enter a unique nonconducting state after channel opening that, unlike the canonical inactivated state, transitions to the open rather than closed state upon repolarization, producing an aptly named resurgent current.[14] The composite of these responses contributes to action potential waveform, amplitude, and frequency.[15]

Table 1.

Characteristic properties of NaV α-subunit isoforms.

Isoform V1/2 activation/inactivation [mv][8] τinactivation[24] τrepriming Persistent current Resurgent current Ramp current Window current Location[8]

1.1 − 33 − 72 Fast Fast[25] + [26] C/P/H
1.2 − 24 − 63[a] Fast Fast[27] + C/P[d]/H
1.3 − 24 − 70 Fast Fast[28] + [1] + C/P[d]/H
1.4 − 26 − 56[a] Fast Fast[28] [29] M
1.5 − 44 − 87 Slow Fast[30] [29] H/C/P[d]
1.6 − 29 − 72 Fast Fast[31] + + [29] C/P/H
1.7 − 25 − 74 Fast Slow[31] [29] + P/C
1.8 − 02 − 36[b] Slow Fast[32] + [33] + [22] + [33] P/H[34]
1.9 − 54 − 54[c] Slow Fast[21] + + [21] + P

All V1/2 data were collected in HEK293 unless otherwise noted.+, yes; −, no. Isoform locations are listed in order of decreasing prevalence.

[a]

CHO.

[b]

Co-expressed with β1.

[c]

DRG.

[d]

Embryonic.

Abbreviations: τ, time constant; C, central nervous system; P, peripheral nervous system; H, heart; M, skeletal muscle. Data on current properties (persistent, resurgent, ramp, window) taken from Rush et al.[20] unless otherwise noted.

1.2. Channel isoforms

Ten gene loci have been identified that encode NaV α-subunits, nine of which—NaV1.1 to 1.9—are voltage-dependent, and a tenth, NaVx, that is involved in salt-sensing.[16] Each isoform exhibits distinct voltage dependence and gating properties (Table 1), which are further modulated by alterations in channel splicing,[17] post-translational modification,[18] and the binding of auxiliary proteins.[19] In the peripheral nervous system, for example, isoforms 1.3, 1.6, 1.8, and 1.9 exhibit incomplete inactivation,[20] producing persistent currents that facilitate action potential initiation. NaV1.3, 1.7, 1.8, and 1.9[21] also have the capacity to amplify slow, subthreshold stimuli through so-called ramp currents. Similarly, resurgent currents produced by rapidly recovering NaV1.6 and 1.8[22] increase AP firing frequency. The overlapping voltage dependence of activation and inactivation of NaV1.9 furnishes an open, non-inactivating channel population primed to contribute to AP firing.[23] Accordingly, the identity and distribution of NaV isoforms determines signal threshold and dictates electrical responses to generator stimuli.

In both human and animal models of pain there is significant evidence that injury-induced NaV redistribution produces ectopic AP firing patterns resulting in mechanical allodynia, thermal hyperalgesia, and spontaneous nociception.[35] In healthy dorsal root ganglion (DRG) cells, NaVs are concentrated at the proximal segment and the nodes of Ranvier. NaV1.2 and 1.3 appear only in embryonic DRGs, whereas adult cells express predominantly NaV1.7, 1.8, and 1.9, and to a lesser extent NaV1.1 and 1.6. In animal models of neuropathic pain, however, NaV1.3 expression is consistently upregulated.[3638] Further, NaV1.8 has been shown to redistribute to the site of injury.[39] These changes, as well as the concomitant downregulation of NaV1.9,[40] alter the excitability of injured neurons. Specifically, the upregulation of NaV1.3 and NaV1.8 facilitates channel repriming and generator potential amplification, respectively. Simultaneously, the downregulation of NaV1.9 produces a hyperpolarized resting potential, enabling the recycling of otherwise inactivated channels (see hyperpolarized V1/2 of inactivation: Table 1, column 2).[41] Consistent with animal models, NaV1.3, 1.7, and 1.8 have been found to accumulate in the axonal tips of human neuromas.[4245] Recently, NaV1.1[46,47] and 1.6[48] upregulation has also been implicated in animal models of neuropathic pain, though the underlying pathophysiology of either isoform has not been investigated extensively. By contrast, in models of inflammatory pain, increased expression of NaV1.3, 1.7, and 1.8 has been noted.[49,50,51] However, as compared to neuropathic pain, fewer studies of inflammation-induced changes in NaV expression have been conducted. Similarly, the effect of injury or inflammation on channel splicing, post-translational modification, and trafficking is not well understood at this time.

1.3. Channel modulation in nociception

The dysregulation and redistribution of neuronally-expressed NaV channels following injury are prompted by signals arising from immune- and/or glial cell-derived mediators.[52] Briefly, in inflammatory pain or tissue injury, immune cells at the site of injury secrete interleukins and growth factors that bind tyrosine kinase receptors, G-protein-coupled receptors, and cytokine receptors on nociceptors. Subsequent activation of several protein kinases, including protein kinase A, protein kinase C, and extracellular signal-regulated kinases, results in phosphorylation of internal stores of NaVs and promotes translocation to the cell membrane (Figure 3).[53,54] Following nerve injury, similar pathways are activated by Schwann cell-released glial mediators.[5557] Increased expression of NaVs at the cell membrane, among other changes, facilitates action potential initiation and propagation, which are perceived as painful stimuli. Repetitive AP firing induces changes in the central nervous system (CNS)[58] that manifest as chronic inflammatory or neuropathic pain.

Figure 3.

Figure 3.

Upstream regulators of channel function in inflammatory and neuropathic pain. Created with BioRender.com. Receptors (blue): tyrosine kinase, G-protein coupled receptors, cytokine receptors. Circles (blue): protein kinase A, protein kinase C, extracellular signal-regulated kinases. Channels (red): NaVs. Sheath (blue): Schwann cells. Abbreviations: TRP, transient receptor potential channel; ASIC, acid-sensing ion channel; KV, voltage-gated potassium channel; CaV, voltage-gated calcium channel; GABAR, gamma-aminobutyric acid receptor; GluR, glutamate receptors; TACR1, tachykinin receptor 1.

Although the involvement of NaVs in inflammatory and neuropathic pain has been studied extensively for the past several decades, there are significant gaps in our understanding of pain pathogenesis. Injury-induced changes in the subcellular distribution of specific NaV isoforms are poorly characterized, as are the trafficking mechanisms responsible for redistribution. Similarly, the regulatory pathways that control channel localization have not been fully elucidated. The contributions of specific NaV isoforms to inflammatory and neuropathic pain vary, and how such changes alter APs and the corresponding sensory experience remains unclear. In an effort to answer these outstanding questions, multiple, disparate technologies have been developed to study NaVs. Herein, we discuss two categories of tools: chemical tools (derivatized small molecules and proteins) and biological tools (channel tagging, knockdown, and knockout). The full complement of these methods and their advantages and disadvantages for investigations of NaV function and physiology are detailed.

2. Chemical Tools to Study NaVs

A large and diverse number of eukaryotic organisms utilize NaV-specific small molecules and peptides for the purposes of selfdefense and/or predation.[59,60] These toxins can induce paralysis, pain, heart arrhythmia, and death depending on their relative potency and mode of action against NaV subtypes. Research to examine the physiological effects of natural poisons has shaped our current understanding of NaVs and their role in electrical signaling. Animal toxins, in particular tetrodotoxin and saxitoxin, were the first tool compounds used to identify and to classify NaV subtypes and to interrogate channel function.[61,62] Following these investigations, additional insights into NaV physiology have been aided with the identification of subtype-selective NaV modulators, semi-synthetic modification of natural toxins, and small molecule design. Other molecular probes that target NaVs include radiolabeled, protein affinity, and fluorescent imaging agents. Although a small number of these tool compounds have been used in specific studies aimed at understanding the action of NaVs in electrogenesis and nociception, most remain underutilized and many require additional optimization. The remainder of this section will discuss the historic contributions of chemical reagents to NaV research, as well as potential applications of these tool compounds to measure NaV expression, localization, trafficking, and turnover.

2.1. Animal toxins

Tetrodotoxin (TTX) and saxitoxin (STX) were the first small molecule animal toxins discovered to act on NaVs.[61,62] Originally identified as the paralysis-inducing components of poisonous pufferfish and bivalves, respectively, TTX and STX were determined to specifically block sodium currents in electrically excitable cells. These toxins have become mainstays in the field of neuroscience due to their low nanomolar potency and selectivity for a subset of NaV channel subtypes, so-called TTX-sensitive NaVs (NaV1.1, 1.2, 1.3, 1.4, 1.6, 1.7). Markedly higher concentrations of toxins are needed to block TTX-resistant channels (NaV1.5, 1.8, 1.9). The classification of NaVs as TTX-sensitive (TTX-S) or resistant (TTX-R) is still employed today, and TTX in particular finds considerable use for blocking TTX-S sodium currents in studies to understand neuronal function, including those related to pain perception.[63,64]

Since the discovery of TTX and STX, numerous toxins have been isolated from venomous animals and toxic algae that are potent towards NaVs. No less than eight different receptor sites have been identified on NaVs;[65] small molecules and peptides binding to any one of these locations modulate channel activity (Figure 4). Most interact directly or allosterically with the voltage-sensing domains (Sites 2–7), acting as channel agonists by modifying NaV gating and voltage dependence. In contrast, simple conductance blockers target Sites 1 and 8 (note: Site 8 and ‘local anesthetic binding site’ have been used interchangeably). Cationic TTX, STX, and μ-conotoxins occlude Na+ passage by binding to the extracellular mouth of the pore domain (Site 1), whereas hydrophobic small molecule drugs and anesthetics such as lidocaine, cocaine, and carbamazepine achieve a similar effect by targeting the inner pore (Site 8).[65] Almost all toxins exhibit selective binding to a particular channel conformer (so-called state dependence) and/or higher affinity upon repeated channel opening (use dependence). The majority of toxins that interact with the voltage sensors are state-dependent, preferentially associating with the closed, open, or inactivated form of the channel. Pore blockers such as TTX and STX tend to be modestly use-dependent, displaying enhanced potency upon more frequent channel opening (Table 2). The multifarious activities of NaV-targeting toxins have been reviewed extensively.[6568]

Figure 4.

Figure 4.

Voltage-gated sodium channel animal toxin binding sites. A) Side view of S5–S6 helices and binding sites therein. B) Top view of NaV and binding sites therein. C) Secondary structure motifs of animal toxin binding sites. Domain I (orange), Domain II (red), Domain III (gray), Domain IV (cyan).

Table 2.

Animal toxins that target NaVs.[65,6972]

Effect Binding site INa+ V1/2 activation τinactivationrepriming State/use dependence Toxin

Antagonism 1 −/− Use saxitoxin tetrodotoxin μ-conotoxin
8 −/↓ State (I) Use anesthetic anticonvulsant antiarrhythmic antidepressant
UD[7375] UD UD UD kalkitoxin jamaicamides polygodial
Mixed 4 −/− State (varied) δ-palutoxin μ O-conotoxin β-scorpion, spider toxins
Agonism 2 ↓/− State (O) aconitine batrachotoxin grayanotoxin veratridine hoiamide A antillatoxin
3 ↓/− State (C) δ-atracotoxin α-scorpion, sea anemone, wasp toxins
5 −/↑ State (O) brevetoxin ciguatoxin
6 ↓/− δ-conotoxin
7 ↓/− State (O) Use pyrethroid

↑/↓, increase/decrease; –, no effect. These properties are general to the binding site but may vary somewhat for individual toxins. Abbreviations: INa +, sodium channel current; τ, time constant; UD, undetermined; O, open; C, closed; I, inactivated.

Despite the structural and functional diversity of existing small molecule and peptide toxins, relatively few specifically target single NaV subtypes. In general, toxins that bind in the large, hydrophobic inner pore (Sites 2, 5, and 8) tend to be the least selective for different NaV isoforms. By contrast, those that bind the polar extracellular surface (Sites 3 and 4) or the anionically charged extracellular pore (Site 1) better differentiate among subtypes. TTX and STX show little to no subtype-selectivity among the six TTX-S channels. Conversely, members of the μ-conotoxin family, as well as scorpion and spider toxins, target single NaV isoforms (vide infra, section 2.5).[76] These selective channel antagonists have been utilized for single isoform ‘knock-out’ to elucidate the roles of NaV subtypes in nociception.[7780] In the same vein, studies with NaV agonists give insight into how alterations in channel gating and voltage dependence underlie inflammatory pain.[81] Recent cryo-EM structures of NaVs in complex with animal toxins—including STX,[6] TTX,[6] the μ-conotoxin KIIIA,[3] and the spider toxins HwTx-IV[6] and ProTx-II[82]—should facilitate the rational design of more selective agents with potential therapeutic benefits.[8385] Future investigations with natural toxins and derivatives thereof present opportunities to quantify, isolate, and/or image select channel subtypes in models of injury or inflammation.

2.2. Radiolabeled probes

Radiolabeled probes offer several advantages for in vitro and in vivo studies of sodium channels. First, due to the minimal structural perturbation of radioisotope incorporation—as compared to the addition of a sterically large, hydrophobic fluorophore, for example—the potency, binding kinetics, and isoform selectivity of a radiolabeled probe typically mirror that of the parent toxin or small molecule investigational drug. Further, because radioactivity can be accurately measured, radioligands make possible precise quantification of protein target concentration. Although the spatial resolution of autoradiography is low, recent advances in positron emission tomography (PET)[86] and single-photon emission computed tomography (SPECT)[87] now enable 3D in vivo imaging on the order of a few hundred micrometers resolution. As such, beyond their uses in the quantification of protein receptors in cells, NaV-specific radiolabeled probes have potential as diagnostics for monitoring channel accumulation at sites of injury or inflammation.

Two classes of radiolabeled probes have been developed for NaV study. The first class comprises channel pore blockers, most notably saxitoxin (STX) and tetrodotoxin (TTX), both of which were radiolabeled through tritium exchange[88,89] and used extensively in early experiments to understand the role of NaVs in electrogenesis (Figure 5). [14C]Guanidine[90] and [35S]gonyautoxin[91] also fall into this class, although these probes have been employed only sparingly in assays designed for neurotoxin detection and never in the study of electrical signaling. The second class of probes is composed of state-dependent molecules, such as the Site 2 toxin, [3H]batrachotoxinin-A 20-α-benzoate ([3H]BTX-B),[92,93] and the anti-ischemia drug, [3H]lifarizine.[94] These molecules have been used almost exclusively for competition experiments to locate ligand binding sites on the channel. The remainder of this section will focus on studies performed and insights gained with the Site 1 radiolabeled TTX and STX probes, as these agents are, by far, the most widely described in the literature.

Figure 5.

Figure 5.

Radiolabeled probes targeting voltage-gated sodium channels at different receptor sites. A) Site 1. B) Site 2. C) Site 8. References: [3H]STX,[88] 11-[3H]TTX,[114] [18F]STX,[125] [35S]GTX II/III,[91] [3H]BTX-B,[92] [3H]lifarizine.[94]

[3H]Saxitoxin ([3H]STX) has been most generally used as a radioprobe for NaV studies due to its ready access from naturally isolated toxin and tritiated water.[88] This procedure rapidly and efficiently exchanges the C11 protons of STX, a significant advantage over the previously established, low-yielding method for labeling TTX with tritium gas.[89] With its high affinity for TTX-S channels (Ki =3.4 nM)[99] and favorable binding kinetics, [3H]STX is arguably the preeminent reagent for measuring NaV expression levels in cell membranes. Channels labeled by [3H]STX can be quantified by scintillation counting and/or isolated by protein liquid chromatography and gel electrophoresis.[9597] Purification of NaVs facilitated by [3H]STX advanced the subsequent design of NaV-specific antibodies (see Section 3.1),[98] which have become the de facto proxies for NaV expression.

Early estimates of single channel conductance and NaV density in neuronal cells were made by bathing giant squid axons in [3H]STX.[99] Other experiments measured channel density at the nodes of Ranvier (NaV density ~12,000/μm2) compared to the internodal space (NaV density ~25/μm2),[100] which provided a physical rationale for saltatory conduction and paved the way for subsequent investigations of electrogenesis. Studies with [3H]STX also quantified the effects of upstream regulators of NaV surface expression, including nerve growth factor (NGF),[101] PKC,[102,103] and ERK,[104] and aided in the characterization of auxiliary proteins such as the beta subunits[105,106,107] and ankyrin G.[108] In addition, some of the first work describing changes in NaV expression in response to physical injury[109,110] and diabetes[111] was enabled with [3H]STX.

The experimental utility of [3H]STX is limited by washout of the C11 tritium labels in aqueous solution, which necessitates working with this compound at temperatures ≤4°C to prevent errors in channel quantification.[112] While [3H]tetrodotoxin ([3H]TTX) can be employed for purposes identical to [3H]STX, difficulties in obtaining this material have severely limited its use.[89] Hence, modified radiolabeled TTX derivatives have been designed with non-exchangeable tritium labels such as [3H]ethylenediamine ditetrodotoxin[113] and 11-[3H]tetrodotoxin[114] in order to facilitate experiments in live cells at 25–37°C. However, preparing these probes is also quite challenging and has greatly restricted their utility. Of note, a 2017 report employed 11-[3H]TTX for in vivo determination of ADME (absorption, distribution, metabolism, and excretion) in rats, preclinical work to validate the application of TTX as a low-dose pain relief drug.[115]

In the past two decades, radiolabeled STX and TTX have witnessed declining application for NaV studies, passed over in favor of NaV-specific antibodies (see Section 3.1). For STX, this may be due in part to lack of accessibility, as this compound remains on the Schedule 1 Chemical Weapons list. With the availability of de novo chemical syntheses of STX,[116122] it should be possible to generate new tritiated derivatives that are stable to solution exchange. The physicochemical properties of this molecule are optimally suited for its development as a chemical probe.[123]

De novo synthesis of STX has provided access to a novel [18F]-labeled STX derivative. Despite structural modification of the parent toxin, [18F]STX retains high affinity for STX-sensitive NaVs (IC50 =46±7 nM vs. NaV1.4;[124] 10.6 nM vs. PC12 cells (rNaV1.2 and 1.7)) and has been used successfully in positron emission tomography (PET) imaging of ectopically expressed NaVs in a rat model of neuropathic pain (spared nerve injury). Findings from this work confirm previous reports of elevated NaV concentrations at a nerve injury site.[125] Currently, this study with [18F]STX remains one of a small number of reports highlighting the use of a chemical tool for labeling endogenous TTX-S NaVs in vivo. Others describe 125I-derived NaV radioligands for SPECT imaging; however, rapid metabolism of these agents severely limits their use.[126,127] New radiolabeled small molecules specific for NaV subtypes have great potential to enable studies of NaV physiology.

2.3. Crosslinking probes

Photoaffinity probes derived from toxins and other small molecules have been applied to great effect to map NaV toxin receptor sites[128130] and, in select cases, have helped reveal elements of the mechanisms for voltage dependence.[131] More than a dozen unique photo-crosslinking probes targeting Sites 1–5 of NaVs have been described. In general, these tools bear either a phenylazide or phenyldiazirine moiety appended from an alcohol or amine group on the ligand (Figure 6). Incorporation of these photoreactive groups can be synthetically challenging and the sizeable steric modification can markedly alter ligand potency and binding kinetics. Irreversible channel modification with photo-crosslinking agents is typically inefficient and therefore complete inhibition (or activation) of a population of channels is not possible. Nevertheless, photoreactive reagents that bind individual channel subtypes would have value for targeting endogenous channels and for introducing fluorescent labels or cofactors that could enable real-time monitoring of channel turnover in healthy and injured neurons. Some of the more promising probes for these purposes are discussed herein.

Figure 6.

Figure 6.

Photo-crosslinking probes for the study of voltage-gated sodium channels. Abbreviations: NAP, 4-azido-2-nitrophenyl; OAB, ortho-azidobenzoate; ANB, azidonitrobenzoyl; AB, azidobenzoate. References: NAP[3H]lysine TTX,[136] [3C]BTX-OAB,[131] GTX diazirine,[129] ANB-125I1ScTx,[133] HwTx L-photomethionine,[130] p-AB PbTx,[132] riluzole azide.[138]

Seminal investigations describing NaV identification and purification utilized photoaffinity labeling with scorpion toxinderived tool compounds.[133,134,135] In these studies, an 125I-radiolabelled, photoactivatable azidonitrobenzoyl scorpion toxin (ANB-ScTx) was prepared from Leiurus quinquestriatus scorpion venom. Such tools are advantaged over previously developed radioligands (i.e., [3H]STX) given the covalent nature of the photo-crosslinking reaction–NaVs can be located and quantified without any concern for probe washout. ANB-ScTx proved instrumental for the initial characterization of NaVs. Unfortunately, the low yield of the photolysis reaction limits the application of this probe for protein quantification, and its use has largely fallen out of favor along with the aforementioned radioligands.

Several small molecule-based photoaffinity probes have been prepared for the purpose of inhibiting NaV activity. 4-Azido-2-nitrophenyl-[3H]lysine tetrodotoxin (NAP[3H]lysine TTX) has been demonstrated to irreversibly inhibit the rising phase of action potentials in the crab giant axon following irradiation.[136] Other TTX derivatives, including NAP[3H]ethylenediamine TTX and arylazido-β-alanine TTX, have been used for analogous experiments in toad muscle.[137] No other reports highlighting the application of these reagents have appeared, however, perhaps due to the extreme difficulties in their preparation. Given the reported efficiencies of the photo-crosslinking reactions, the development of subtype-selective variants of these tools warrants further investigation.

Recent research to develop photoaffinity probes for NaVs has focused on the design of small molecules capable of modulating channel voltage dependence. For example, riluzole, a state-dependent ligand that blocks sodium conductance and hyperpolarizes the voltage dependence of activation, has been functionalized with an azido group to yield a photoactivatable derivative. Although photo-crosslinking increases the inhibitory effects of riluzole on sodium conduction (~70% block after crosslinking under saturating conditions), the ability of this probe to hyperpolarize voltage dependence is preserved.[138] This reagent is a first-in-class tool compound given its ability to alter the energetics of NaV gating rather than simply block channel conduction. These types of probes may hold promise for determining the contributions of sodium channel activation (in the absence of indeterminate upstream signaling pathways) to ectopic electrical signaling.

To date, only one NaV-specific covalent probe has been reported that does not require photoactivation. Saxitoxin ethyl maleimide (Figure 7A) rapidly and efficiently binds to TTX-S channels (as validated against rNaV1.2 and 1.4) without the requirement of additional reagents or protein mutation (>80% irreversible channel block in six minutes under saturating conditions).[139] As with photoaffinity probes, the specific amino acid residue(s) with which STX ethyl maleimide reacts is currently unknown. If properly functionalized, this type of probe presents a potentially unique tool compound for tagging and tracking endogenous TTX-S NaVs.

Figure 7.

Figure 7.

A) Cysteine-reactive probe for irreversible inhibition of NaVs.[139] B) Photocaged STX derivative for spatiotemporal control of NaVs.[140]

The development of chemical probes that target and covalently react with NaVs is an underexplored research area given the potential of such compounds to selectively inhibit channel activity and to facilitate channel isolation and purification. Continued efforts to advance these types of reagents will empower research to understand NaV physiology.

2.4. Photocaged probes

Modified saxitoxins consisting of a photocleavable ‘caging’ group have been made available through de novo synthesis (Figure 7B).[140] These compounds enable rapid, precise, spatiotemporal control of NaVs through light-induced uncaging. The released inhibitor is a nanomolar potent, reversible binder of TTX-S NaVs. Electrophysiological experiments demonstrate the value of these tool compounds for selectively inhibiting action potentials in dissociated neurons and brain slice. Further, recordings in a corpus collosum slice preparation reveal differences in the susceptibility of myelinated vs. unmyelinated axons to NaV block. These reagents are first-in-class photocaged, NaV-selective inhibitors, and will enable studies of the dependence of neuronal function on NaV spatial organization. This class of tools has been recently expanded with the development of the photocaged spider toxin HwTxIV-Nvoc.[141 The caged peptide toxin has demonstrable in vivo efficacy in both mice and zebrafish after several minutes of ultraviolet light application.

2.5. Fluorescent probes

The development of NaV-targeting fluorescent probes to monitor changes in channel trafficking and localization in healthy cells and following response to injury has proven particularly challenging. Attachment of large fluorogenic dye molecules to ligands that associate to NaVs often adversely affects solubility, potency, and binding kinetics. Partly for this reason, the earliest fluorescent reporters were designed to measure Na+ influx[142] rather than to mark the location and/or expression level of the channel itself. The most common Na+ ionophore, benzofuran isophthalate (SBFI), has been used to examine the mechanisms of action of antidepressants, among other studies. Unfortunately, Na+-binding dyes[143] remain plagued by low sensitivity and poor signal-to-noise, and are considerably less effective than Ca2+ indicators like Fura-2.[144] To overcome these limitations, mutation of NaV to increase Ca2+ permeability has been demonstrated, which enables application of a fluorescent Ca2+ sensor (e.g., GECO) to measure channel activity.[145] While potentially valuable for drug discovery, this solution for measuring NaV response is not suitable for studies in neuronal cells.

In an effort to develop NaV fluorescent imaging agents, a number of small molecule channel binders have been modified with fluorophores (Table 3). N-Methylanthranilate (NMA), a weakly emissive fluorophore, was affixed to batrachotoxinin A (BTX A) to generate an analogue of bactrachotoxin (BTX), a potent channel agonist. This probe has been utilized for visualizing channel populations at the nodes of Ranvier in a mouse nerve fiber, but is too dim for tracking channels in cells expressing lower NaV concentrations.[146] NMA-BTX also has the disadvantage of being time-limited, as prolonged channel activation results in NaV internalization.[147,148] Given the development of improved imaging technologies and the relative ease of esterification of the C20-alcohol in batrachotoxinin-A, this tool and others like it may very well warrant a second look. The hydrophobicity of molecules like NMA-BTX can, however, present problems due to nonspecific protein and membrane binding. Other disadvantages of NMA-BTX stem from the suboptimal photophysical properties of NMA; nevertheless, replacing this fluorophore with larger, brighter dyes may come with a serious detriment to ligand-channel affinity and specificity.

Table 3.

Fluorescent probes for the study of voltage-gated sodium channels. IC50 values were collected against listed model system unless otherwise noted.

graphic file with name nihms-1822658-t0009.jpg
[a]

Data provided by the manufacturer.

[b]

Modified at W50X.

[c]

Collected in HEK. Similar potencies exhibited vs. hNaV1.1–1.3, hNaV1.6.

Toxin-fluorophore conjugates derived from STX, including STX-Cy5 and STX-DCDHF, offer a number of salient advantages over NMA-BTX and related probes, foremost of which is readily reversible channel binding. These probes have been successfully employed for single-molecule and super-resolution imaging experiments to visualize and track channels in PC12 cells.[149] Unfortunately, extending the application of these molecules to neuronal cells has been plagued by nonspecific ligand binding and poor signal-to-noise. Further optimization of probes such as STX-Cy5 may ultimately give way to a high-precision reagent for real-time, live cell NaV imaging.

Alternative approaches for generating NaV-selective imaging agents have exploited lysine modification of peptide toxins (e.g., Tityus serrulatus scorpion β-toxin, Conus geographus and Conus kinoshitai μ-conotoxins) with AlexaFluor and BODIPY dyes or unnatural amino acid incorporation.[154,155] The availability of these toxins from nature or through de novo peptide synthesis greatly facilitates this research. Thus far, fluorescent peptide toxin conjugates have been primarily employed for proof-of-concept imaging and luminescence resonance energy transfer (LRET) experiments.[156] However, given the potencies of these toxin derivatives, future experiments to visualize NaV expression in live cells may be possible. In one such example, Gonzales, et al.[157] used a modified Haplopelma schmidti spider toxin, Hs1a-Cy7.5, to image NaV1.7 populations in mouse sciatic nerve ex vivo. With nanomolar potency against neuronal TTX-S NaV isoforms and an infrared absorption/emission spectrum, Hs1a-Cy7.5 has potential application for in vivo NaV imaging. To our knowledge, such studies have not yet been reported.

In addition to potent small molecule- and peptide-based fluorophores, chemically modified nanoparticles have been described for NaV imaging. Europium-doped yttrium vanadate particles coated with guanidine groups represent one such design. Although n-alkylguanidine derivatives are millimolar inhibitors of NaVs,[158] the coated nanoparticles display myriad copies of the guanidine ligand and thus the particle has high avidity for the channel. Using this probe, NaVs in cardiac myocytes have been imaged.[159] Unfortunately, as with other fluorescent agents, follow-up studies in neuronal cells or live tissue have not appeared. Perhaps not surprisingly, the guanidine-studded nanoparticle does not display isoform selectivity. Recent disclosures of a number of isoform-specific NaV ligands, as discussed below, may portend the coming of age of imaging probes that target NaV subtypes.

2.6. Isoform-selective, investigational drugs

With the explosion of interest in NaVs as therapeutic targets for analgesic development, ignited by the disclosure of genetic data in human subjects incapable of sensing pain, a rush to develop potent, isoform-selective molecules has ensued.[160] Much of this effort has targeted hNaV1.7, as defects in this subtype have been implicated in paroxysmal pain disorders (gain-of-function)[161] and congenital insensitivity to pain (lossof-function).[162] This portion of the review will briefly highlight a select number of examples of publicly accessible, nanomolar potent tool compounds used for the study of NaVs in neuronal signaling and nociception (Table 4). The topic of NaV drug development has been reviewed extensively, and interested readers are directed to the following works.[163165] Disappointingly, the in vitro and preclinical efficacy of investigational analgesic medicines has witnessed little in the way of clinical success, for reasons fully elucidated elsewhere.[166,167]

Table 4.

IC50 values of NaV1.7 selective probes, given in μM. All IC50 values were collected in HEK stably expressing NaVs unless otherwise noted.

graphic file with name nihms-1822658-t0010.jpg
[a]

Data collected in CHO.

Despite the lack of clinical success,[168,169] studies with Pfizer’s PF-05089771 have greatly informed our understanding of the contributions of NaV1.7 to electrogenesis and the threshold for signal conduction.[170] Unfortunately, PF-05089771 differentiates poorly between NaV1.2 and NaV1.7 (<10-fold selectivity),[171] likely resulting in unwanted off-target effects at concentrations necessary to achieve clinical efficacy. The challenge of distinguishing among the nine NaV isoforms and avoiding other off-target channels and receptors is a recurring problem for NaV1.7-selective small molecule and protein design.[172174] Crystal structure determinations of a NaV1.7 chimera with similarly potent, yet nonselective, sulfonamides analogous to PF-05089771 have revealed the structural basis for their mechanism of action and enabled the development of subtype-specific inhibitors.[175,176] Advancement of PF-06456384, which displays more than 300-fold selectivity for NaV1.7 over other NaV subtypes, affords the most potent, state-dependent NaV1.7-specific small molecule known to date (IC50 =0.01 nM).[177] Alternative sulfonamide-based ligands are also reported to be highly selective, state-dependent NaV inhibitors, with AM0466 and AM-8379 exhibiting 100- to 1000-fold affinity for NaV1.7 over any other isoform.[178181] Recently, SiteOne Therapeutics has disclosed a first-in-class, state-independent NaV1.7-selective inhibitor, ST-2262, modeled after the natural product saxitoxin.[182] This work demonstrates the potential for targeting the outer vestibule of the channel pore to achieve isoform specificity.

In parallel with NaV1.7 research, NaV1.8 and NaV1.9 have also found interest as analgesic targets, studies prompted by human genetic data.[184,185] Several exquisitely selective, state-dependent investigational drugs have been developed that bind to NaV1.8, including Pfizer’s PF-04885614[186] and Abbott’s (now Abbvie) A-803467 (Table 5), both of which are commercially available. A-803467 has been instrumental for elucidating the contributions of NaV1.8 to nociception in models of spinal injury and osteoarthritis.[187189] These experiments were especially important given ambiguous results from NaV1.8 knockdown and knockout studies (vide infra, section 3). In contrast, while NaV1.9 has been implicated in several models of neuropathic pain,[190192] no NaV1.9-selective inhibitors have been disclosed to date.

Table 5.

IC50 values of isoform selective probes, given in μM. All IC50 values were collected in HEK stably expressing NaVs unless otherwise noted.

graphic file with name nihms-1822658-t0011.jpg
[a]

Data collected in Xenopus oocytes; subsequent studies in HEK cells indicate that this compound is also potent against NaV1.1.[195]

[b]

EC50.

Unlike the number of selective inhibitors available for investigations of NaV1.7 and 1.8, few exist that target other potentially interesting channel isoforms (i.e., NaV1.1, 1.2, 1.3, 1.6). Without compelling genetic evidence that links these subtypes to nociception, there has been less interest in developing selective inhibitors as analgesics. Currently, two molecules have been reported to bind selectively to NaV1.6 (Table 5): (1) 4,9-anhydrotetrodotoxin, a natural derivative of TTX with >40-fold selectivity for NaV1.6;[193] and (2) the Centruroides noxius beta-scorpion peptide (Cn2),[194] an animal toxin NaV agonist that evokes resurgent current in this isoform. Neither of these compounds has enjoyed widespread use for channel studies. In fact, a recent report has shown that 4,9anhydroTTX also inhibits NaV1.1 and 1.3 (particularly the former).[195] Two additional probes are modestly selective for NaV1.1 (Table 5): Icagen’s ICA 121431,[196] a channel blocker that is ~6-fold less potent for NaV1.3; and Heteroscodra maculata δ-theraphotoxin-Hm1a,[197] a tarantula peptide toxin that inhibits fast inactivation in NaV1.1 with ~6-fold selectivity over NaV1.2 and 1.3. While studies with Hm1a suggest a role for NaV1.1 in mechanical pain sensation, neither this probe nor the Icagen molecule are selective enough for investigations of single NaV isoforms in neuronal cells. To the best of our knowledge, no tools capable of selectively targeting NaV1.2 have been described. Continued efforts to advance potent, subtype-specific NaV modulators is warranted in order to deconvolute the physiological function(s) of each isoform in electrogenesis and nociception.

3. Biological Tools to Study NaVs

The successful development and application of small molecule, NaV-selective reagents notwithstanding, much of our understanding of NaV physiology and nociception is owed to the availability of biological techniques and tools. While these methods do not afford the temporal response displayed by most chemical probes, antibody labeling, oligonucleotide-based knockdown, genetic knockout, and protein epitope tagging offer greater precision for targeting NaVs. The advent and use of biological methods for NaV investigations are highlighted below.

3.1. Antibodies

NaV antibodies (Abs) that bind to large intracellular loop regions of NaVs have been employed extensively in studies of NaV physiology, including nociception.[3648] NaV Abs were originally developed in individual labs and screened for specificity by monitoring heart/CNS/PNS cross-reactivity[198,199,200] or accumulation in NaV knockout mice.[201] Several companies (e.g., Alomone and Abcam) have since developed a host of orthologue- and isoform-specific NaV Abs. While generally useful for immunohistochemistry and Western blot experiments, intracellular antibodies are unfit for live cell imaging due to the necessity of cell permeabilization. In part for this reason, studies of NaV trafficking in response to cell or tissue injury have been lacking. Recent developments in channel epitope tagging, however, are changing this landscape (Section 3.5).

The preparation of Abs that target extracellular regions of NaVs has witnessed some key advances in the past five years. Amgen has synthesized several exquisitely potent Ab-toxin conjugates using peptide toxins GpTx-1[202] and JzTx-V[203] from Grammostola porteri and Chilobrachys jingzhao spider venom, respectively. Following optimization studies to improve the potencies and selectivities of these otherwise nonspecific toxins for NaV1.7, these molecules were ligated to human immunoglobulin G-derived Abs, which exploit binding to the neonatal Fc receptor (FcRn) to reduce lysosomal degradation and extend serum half-life.[204] The resulting conjugates display nanomolar potency against the target and long circulating half-lives (t1/2 = 80 hours, as compared to 0.6 h for the unconjugated toxin in the case of GpTx-1), and can be used in vivo for extended block of NaV function. Derivatives of these novel Ab conjugates could prove quite useful as research tools for the study of channel localization.

Three additional extracellular Abs are available for channel study, targeting NaV1.3,[205] 1.5,[206] and 1.8.[207] While the NaV1.5 Ab has been successfully employed for the study of cancer progression,[208] NaV1.3 and 1.8 Abs, both from Alomone, have found limited use. The former is nonselective, associating with NaV1.1 and 1.2 as well as 1.3, and the latter does not bind the human NaV1.8 orthologue. No reports describing the application of these reagents for cellular imaging have appeared.

3.2. Oligonucleotide-based knockdown

One of the most prominent techniques for studying NaV physiology is oligonucleotide-based knockdown.[209] Antisense and RNA interference methods make possible selective depletion of specific NaV isoforms with a level of precision that is unrivaled by other technologies (genome engineering, notwithstanding). The major drawbacks of such methods are that protein knockdown is never quantitative and the process is slow (i.e., tens of hours are generally required to lower protein levels).[210] Since the late 1990s, antisense knockdown featured as the preeminent method for deconvoluting the roles of individual NaV isoforms in various animal models of pain. Early experiments were largely conducted by microinjection of NaV-specific antisense oligodeoxynucleotides.[211] Later, as the field of RNA silencing developed, lentiviral delivery of siRNA was popularized.[212] Depending on the choice of method, model organism, and oligonucleotide sequence, mRNA and protein concentrations can be reduced by 20–80%; however, the exact percent reduction of mRNA and protein is unpredictable and mRNA and protein concentrations are not strictly correlated.

Knockdown of different NaV isoforms results in pain reduction in a variety of animal models (Table 6). Decreasing expression of NaV1.3,[213,214] NaV1.6,[48,215,216] and NaV1.8[211,217] was found to reduce mechanical allodynia in murine models of neuropathic pain. Knockdown of either NaV1.3[218,219] or NaV1.8[212,220222] was also shown to reduce thermal hyperalgesia, although the contributions of the latter subtype are better documented than the former. In contrast, NaV1.6 was only recently implicated in models of neuropathic pain and has been comparatively underexplored. NaV1.7, while aggressively pursued as a pain target, has not been studied in animal models of neuropathic pain by antisense knockdown or RNA interference. However, knockdown experiments of NaV1.7 have been performed in models of inflammatory pain and skin burn.[223225] Analogous studies of inflammatory pain have examined the role of NaV1.8[211,226228] and to a lesser extent NaV1.6,[229] NaV1.9,[190] and NaX.[230] NaV1.7 also plays a role in acute pain signaling.[231,232] Although present in sensory neurons, albeit at low levels based on transcriptional profiling, the possible involvement of NaV1.1 and 1.2 in nociception has not been explored using oligonucleotide-based knockdown methods.

Table 6.

Isoform-selective knockdown of NaVs.

Pain Type Model 1.3 1.6 1.7 1.8 1.9 1.x

Neuropathic Chronic constriction sciatic nerve [218] [216][b] [220,211,217][b]
Sciatic nerve entrapment [212][c]
Chronic compression DRG [216][b]
Spinal cord contusion [213,219]
Spared nerve injury NE[239], ↓[238][a]
L5/L6 spinal nerve ligation [215][b] [211,221,222] NE[222]
Chemotherapy [48] NE[211] [191]
Diabetes [214][a]
Inflammatory Complete Freund’s Adjuvant [224] [211,228] NE[228]
Zymosan [229][b]
Carrageenan [190]
Acetic/citric acid [223][a] [226]
PGE2 [227]
Imiquimod [230][d]
Other Incision [231][c] NE[211]
Burn [232][c]
Cancer [233][c]
Venom [234][c] [235]

↓ = reduced pain-associated behavior. NE=no effect on pain-associated behavior. Knockdowns were performed with the injection of antisense oligodeoxynucleotides unless otherwise noted.

[a]

Adeno-associated virus (AAV)-shRNA knockdown.

[b]

siRNA transfection.

[c]

lentivirus.

[d]

Dicer-substrate RNAi (DsiRNAs).

Red indicates disagreement among published data.

Due to the number of and variability among animal pain models,[236,237] knockdown data collected across disparate experimental protocols are often difficult to compare. Even among studies of the identical NaV isoform in the same model system, conclusions can vary, as is the case for investigations of NaV1.3 in spared nerve injury. Separate reports offer conflicting evidence as to the involvement,[238] or lack thereof,[239] of this subtype in nociception. Whether these inconsistencies are due to variations in experimental setup or off-target effects of the oligonucleotide[33] remains unknown. With some notable exceptions,[231,223,212] the effects of single isoform knockdown on the expression of off-target NaVs is not routinely measured. Compensatory upregulation of alternative NaV subtypes, which is known to occur in genetic knockouts, may be responsible for some of the ambiguities associated with knockdown experiments.[240242] Further, whether knockdown alters the subcellular distribution of NaVs—critical for proper electrical signaling[243]—has not been examined.

3.3. Knockouts

Mouse genetic knockouts (KOs) of every neuronal voltage-gated sodium channel have been described. Most global knockouts, including those for NaV1.1,[244] 1.2,[245] 1.6,[246] and 1.7,[247] are lethal within one month of birth. While improvements in cross breeding have facilitated the generation of a NaV1.7 KO that survives to adulthood,[248] the study of other isoforms in animal models of pain has proven challenging. One solution involves the development of a NaV1.8-Cre mouse,[249] which allows for selective, conditional knockout of individual NaV subtypes (or other protein targets of interest). Although NaV1.8-expressing neurons were thought to be almost exclusively nociceptive in nature, it has since been determined that certain mechanoreceptors express NaV1.8 as well,[250] thus complicating analysis of the results from experiments with NaV1.8-Cre lines. Such conditional KOs, as well as null models, have been used extensively to examine the contributions of NaV isoforms to signaling. Nevertheless, using KO techniques to understand NaV function and pain pathology is rife with opportunity. Questions regarding the involvement of certain NaV isoforms in nociception remain outstanding—only a single report of a 1.3 null mouse has appeared[251] and, as yet, KOs of NaV1.1 and 1.2 have only been used for investigating specific CNS disorders.

Neuropathic pain can be induced in select NaV KO models; some of these findings undercut previously established conclusions (from knockdown studies) regarding the involvement of NaV1.3[251,252] and 1.8[247,253,192] in nociception. Conversely, results from KO studies of NaV1.6[246,254] and 1.9[191,192] suggest that these subtypes contribute to neuropathic pain signaling. In inflammatory pain models, genetic KO experiments have implicated NaV1.7,[248,255] 1.8,[241,253,256] and 1.9.[242,257,258] Unfortunately, there is a considerable amount of disparity in the KO literature (Table 7, red). In one example, conflicting results appear between publications describing the same inflammatory pain model and, more broadly, knockout animal. Comparative analysis of knockout and knockdown data also reveals inconsistencies (Table 7, italicized font). While these differences might be explained by the choice of animal subject (most often rat, in case of knockdown, vs. mouse, in KO models), the discrepancies among KO data are more difficult to rationalize. One probable explanation arises from the fact that NaV KO, for example of NaV1.1,[244] 1.8,[241] or 1.9,[242] promotes the concomitant upregulation of alternative isoforms. Moreover, single isoform KO can result in subcellular redistribution of the expressed subtypes.[240,244] Further, the choice of method for generating KO animals can produce mice with different channel expression profiles.[242,257] To address these problems, NaV1.6-floxed mice[254] have been developed; NaV deletion by AAV-Cre viral induction in adult mice limits the timeframe during which compensatory upregulation can occur. Similarly, NaV1.8-Cre-diptheria toxin mice[256] have been genetically engineered to promote selective apoptosis of NaV1.8-expressing neurons. Studies with these animals are complicated by the significant biochemical and physiological changes accompanying programmed cell death.

Table 7.

Genetic knockouts of NaV isoforms.

Pain Type Model 1.3 1.6 1.7 1.8 1.9

Neuropathic Chronic constriction sciatic nerve NE [192] [192]
L5/L6 spinal nerve ligation NE[251][a] NE[259,247][d]
[259][e]
NE[247][f]
Sciatic nerve ligation NE[254][b]
[254][c]
NE[253] NE[242,257]
Spared nerve injury NE[192] NE[192,257]
Chemotherapy NE[252] [246] NE[252] NE [252]
[191]
Diabetes [260]
Inflammatory Complete Freund’s Adjuvant NE[251] NE[254][b] [248,255][b]
NE [259][d]
NE[192]
[256][f]
[242,257,258]
NE [192]
Carrageenan [255][b]
NE[259][d]
[241]
NE[192]
[242,258]
NE [192]
Formalin NE[251] [255][b],[248]
NE[259][d]
NE[192]
[256][f]
[242,258]
NE[192]
NGF [255][b] [253] NE[257]
PGE2 NE [253] [242,257]
Other Itch [248] [261]

↓ = reduced pain-associated behavior. NE=no effect on pain-associated behavior. Whole mouse genetic knockouts were used unless otherwise noted. Red indicates disagreement among published data. Italic font indicates disagreement between knockout (Table 7) and knockdown (Table 6) data.

[a]

Ligation of spinal nerve L5 only.

[b]

NaV1.8-Cre.

[c]

Floxed mice; AAV-Cre induced KO.

[d]

Advillin-Cre (sensory neuron specific).

[e]

Wnt1-Cre (sensory and sympathetic neuron specific).

[f]

NaV1.8-Cre-diptheria toxin.

3.4. Optogenetics

One of the most interesting developments to arise from the design of conditional NaV knockouts is the crossbreeding of loxP-STOP-loxP-rhodopsin mice with the NaV1.8-Cre mouse line. The selective expression of channelrhodopsin ChR2 or archaerhodopsin Arch3 in NaV1.8-positive neurons enables light-based control of electrical signaling. Using this optogenetic method, activation of NaV1.8+ neurons has been demonstrated to induce nocifensive behavior,[262] whereas silencing reduces mechanical allodynia and thermal hyperalgesia in models of both inflammatory and neuropathic pain (Figure 8).[263] These findings have led to the development of an implantable optoelectronic system for the continuous modulation of bladder pain in mice.[264] The specific targeting of an electrically excitable cell type, rather than a single NaV isoform, is a paradigm shift in the study and potential treatment of pain pathogenesis.

Figure 8.

Figure 8.

Rhodopsin-modulated electrical signaling.[265] Created with BioRender.com. A) Blue light activation of non-specific cation channel ChR2 depolarizes the membrane potential, causing NaV opening and action potential firing. B) Yellow (or green) light activation of proton pump Arch3 prevents NaV opening thereby reducing excitability.[266]

3.5. Fluorescent and epitope tagging

Recombinant genetic technologies that enable cellular expression of fluorescent protein- and epitope-tagged NaVs are essential for the study of NaV trafficking. Specific challenges, however, have prevented these methods from realizing their full promise. NaV1.1, 1.2, and 1.6 plasmids, in particular, mutate and recombine extensively when expressed in E. coli,[267] rendering the synthesis of functionally tagged channels quite challenging. As such, examples of epitope-tagged channels derived from NaV1.4 and 1.5—subtypes with comparatively stable plasmid DNA—are most prevalent in the literature (Table 8). Unfortunately, the most broadly applicable of these tools, sodium channel protein-based activity reporting construct (SPARC),[268] which produces an optical readout of channel opening, was found to aggregate intracellularly, with less than 5% of the total protein reaching the membrane.[269] Early exploration with NaV1.5-based constructs demonstrated the importance of protein kinase activation[270] and β-subunit co-expression[271] for ensuring proper trafficking and membrane localization. Thus, the use of model cell lines for the purpose of screening NaV-GFP fusion or epitope-tagged constructs may be intrinsically limited. In light of these problems, much of the work reported with epitope-tagged NaVs involves protein isolation rather than live cell imaging studies.

Table 8.

Genetically-modified Nav α-subunits.

Isoform Modification Location (E/I)[a] Model systems Use

1.2 FLAG N-terminus (I) Transgenic mouse, MDCK Pull-down[278,279]
HA N-terminus (I) tsA201 Purification[280]
EGFP N-terminus (I) MDCK, mouse neuroglial cocultures Immunohistochemistry[279]
1.4 GFP L850/S851 (I) X. oocytes, HEK, rat E18 hippocampal neurons Sodium channel protein-based activity reporting
construct[268,269]
EVIFQTPL[b] H599L/F600 (I) X. oocytes, tsA201, primary muscle Structural analysis, live-cell imaging[115]
1.5 GFP N-terminus, C-terminus (I) X. oocytes, HEK, dog cardiomyocytes Live-cell imaging[271, 281, 282,286]
YFP N-terminus (I) X. oocytes, HEK Live-cell imaging,[278,280] FRET[282]
CFP N-terminus (I) HEK, dog cardiomyocytes Live-cell imaging,[280] FRET[281]
FLAG L299/V300 (I) HEK Validation[281]
FLAG DI S5-S6 (E) tsA201, rat cardiomyocytes Immunohistochemistry[283,284]
HA or 3xHA DI S5-S6 (E) X. oocytes, HEK, rat cardiomyocytes Immunohistochemistry,[285] Chemiluminescence[270]
1.6 (TTX- myc N-terminus (I) MDCK Pull-down, Immunohistochemistry[279]
R) ECFP N-terminus (I) MDCK, mouse neuroglial cocultures Immunohistochemistry[279]
GFP; BAD C-terminus (I); S1545/K1546 (E) ND7/23, rat E18 hippocampal neurons Live-cell imaging, single molecule tracking[272274]
1.7 TAP C-terminus (I) HEK, transgenic mouse Protein interaction mapping[276]
1.7 (TTX- GFP C-terminus (I) HEK, ND7/23 Live-cell imaging[286]
R) Venus; BAD N-terminus (I);M1529/V1530 (E) Rat P2–4 DRGs Live-cell imaging[275]
3xmyc-Halo-3xHA N-terminus (I) Rat P2–4 DRGs Live-cell imaging, single molecule tracking[275]
1.8 Venus C-terminus (I) HeLa, rat superior cervical ganglion neurons Live cell imaging[287]
EGFP N-terminus (I) HeLa, rat superior cervical ganglion neurons Live cell imaging[287]
1.9 sfGFP N-terminus (I) ND7/23, Nav1.8-Cre transgenic mouse Immunohistochemistry[277]
[a]

(E/I) = (Extracellular/Intracellular).

[b]

Antigen for mAb 142.[22]

Epitope tags: FLAG, DYKDDDDK; HA, YPYDVPDYA; myc, EQKLISEEDL. Acronyms: G/Y/CFP, green/yellow/cyan fluorescent protein; TAP, tandem affinity purification; sfGFP, superfolder GFP. Cell lines: HEK, human embryonic kidney; CHO, Chinese hamster ovary; ND7/23, mouse neuroblastoma; DRG, dorsal root ganglion.

Recent advances have highlighted the design of dually-tagged sodium channels—affixed with both a biotin-affinity domain (BAD) and a fluorescent protein–for the study of NaV1.6 and NaV1.7. Incorporation of the BAD domain in the S1—S2 extracellular loop of domain IV enables fluorescent imaging of channels expressed in the membrane following extracellular application of a streptavidin-dye conjugate (e.g., SACF640, SA-568, SA-594); inclusion of the fluorescent protein facilitates monitoring of total protein concentration. This technology is extremely powerful for live cell monitoring of membrane NaVs, and has made possible seminal imaging and single-molecule tracking studies of NaVs in neurons. Such experiments have begun to unravel the mechanisms of NaV insertion into and removal from the cellular membrane, as well as the role of channel nanoclusters in electrical signaling.[272274] Studies with NaV1.7 have, for the first time, measured changes in channel trafficking in response to mediators of inflammatory pain.[275] Undoubtedly, further application of these unique tools will be instrumental for elucidating the regulatory pathways that underlie NaV physiology in healthy and aberrant cells.

The rapid evolution of CRISPR technology for gene editing has enabled the design of knock-in (KI) mice that express engineered NaV constructs. In the case of a NaV1.7-tandem affinity purification tag (TAP) knock-in, transgenic mice were used to identify several hundred proteins with direct channel interactions.[276] In a separate study involving a superfolder GFPNaV1.9 KI model, the GFP tag made possible analysis of channel expression patterns in DRGs by immunohistochemistry.[277] Compared to other genetic methods (e.g., knockdown, knockout), CRISPR-enabled knock-in has several advantages, foremost of which is the ability to study NaV function in fully intact systems where compensatory changes in NaV expression and subcellular distribution are no longer complicating factors. Future studies involving CRISPR-engineered NaV KI animal subjects are expected to give unprecedented insight into NaV physiology.

4. Opportunities and Challenges

The study of NaV function and physiology has been empowered with the invention of chemical and biological tools that make possible the selective manipulation of specific channel subtypes. These advances notwithstanding, all technologies have limitations of some form in terms of efficacy, selectivity, and/or utility for in vitro or in vivo studies. The development of new, high-precision reagents for labeling and modulating NaV subtypes in primary cells and tissue presents tremendous research opportunities. Such pursuits are challenged by the immutable fact that channels rapidly cycle through different conformational states and that most ligands, be they small molecules or peptides, display state and use dependence. For these reasons, discoveries made in dissociated cells using electrophysiology measurements are often confounded once translated to in vivo applications. A better understanding of channel dynamics, state-dependent ligand binding, and how conformational distributions are influenced under different physiological conditions would accelerate efforts to advance new reagents and therapeutic leads that modulate NaV activity.

Potential applications for radioligand and fluorescent probes that target single NaV isoforms in live cells are manifold, particularly given the current limitations of NaV antibodies and the challenges with engineering epitope- and GFP-tagged channels. Other tool compounds that enable isolation of membrane-expressed NaVs would facilitate proteomic studies to examine how post-translational modifications influence channel function in healthy and injured neurons. Improvements in CRISPR genome editing and protein engineering are, undoubtedly, going to usher in new research opportunities to study NaV expression and trafficking in vivo. Similarly, advances in antibody production will deliver potent, subtype-specific reagents that bind to extracellular sites on the channel. All told, the continued expansion of the NaV ‘toolbox’ will drive research aimed at illuminating the complex physiological mechanisms that underpin electrogenesis and nociception.

Acknowledgements

We are grateful to the NIH (R01GM117263-05) for support of our program. A.V.E. was supported by the Stanford Center for Molecular Analysis and Design (CMAD) as well as a Stanford Interdisciplinary Graduate Fellowship (SIGF) through the Stanford Bio-X Interdisciplinary Biosciences Institute.

Biography

graphic file with name nihms-1822658-b0012.gif

Anna Elleman received her B.S. in chemical biology from the University of California, Berkeley in 2015 while researching in the laboratory of Professor John Kuriyan. She completed her Ph.D. in Chemistry in 2021 under the direction of Professor Justin Du Bois at Stanford University, where she was a Bio-X Stanford Interdisciplinary Graduate and Center for Molecular Analysis and Design Fellow. She has since returned to the University of California, Berkeley, where she works as a postdoctoral researcher with Professors Richard H. Kramer and Stephen Brohawn.

graphic file with name nihms-1822658-b0013.gif

Justin Du Bois received his B.S. degree from the University of California, Berkeley in 1992, where he conducted undergraduate research with Professor Ken Raymond. In 1997 he earned his Ph.D. from the California Institute of Technology under the direction of Professor Erick Carreira. Following a two-year NIH postdoctoral position with Professor Stephen Lippard at MIT, he joined the faculty at Stanford University. He currently holds the title of Henry Dreyfus Professor of Chemistry, and is a professor, by courtesy, in Chemical and Systems Biology and a Bass University Fellow in Undergraduate Education.

Footnotes

Conflict of Interest

J.D. is a cofounder and holds equity shares in SiteOne Therapeutics, Inc., a start-up company interested in developing subtype-selective modulators of NaV.

References

  • [1].Wang J, Ou SW, Wang YJ, Channels 2017, 11, 534–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Catterall WA, J. Physiol 2012, 590, 2577–2589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Pan X, Li Z, Huang Z, Huang G, Shen H, Liu L, Lei J, Yan N, Science 2019, 363, 1309–1313. [DOI] [PubMed] [Google Scholar]
  • [4].Pan X, Li Z, Zhou Q, Shen H, Wu K, Huang X, Chen J, Zhang J, Zhu X, Lei J, Xong W, Gong H, Xiao B, Yan N, Science 2018, 362, 305–314. [DOI] [PubMed] [Google Scholar]
  • [5].Li Z, Jin Z, Wu T, Huang G, Wu K, Lei J, Pan X, Yan N, Angew. Chem.Int. Ed 2021, 60, 11474–11480. [DOI] [PubMed] [Google Scholar]
  • [6].Shen H, Liu D, Wu K, Lei J, Yan N, Science 2019, 363, 1303–1308. [DOI] [PubMed] [Google Scholar]
  • [7].Yan Z, Zhou Q, Wang L, Wu J, Zhao Y, Haung G, Peng W, Shen H, Lei J, Yan N, Cell 2017, 170, 470–482. [DOI] [PubMed] [Google Scholar]
  • [8].Bertran D, Biton B, Licher T, Chambard JM, Lanneau C, Partiseti M, Lefevre IA, Curr. Protoc. Pharmacol 2016, 75, 9.21.1–9.21.35. [DOI] [PubMed] [Google Scholar]
  • [9].Bosmans F, Martin-Eauclaire MF, Swartz KJ, Nature 2008, 456, 202–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Vassilev PM, Scheuer T, Catterall WA, Science 1988, 241, 1658–1661. [DOI] [PubMed] [Google Scholar]
  • [11].Silva JR, Goldstein SAN, J. Gen. Physiol 2013, 141, 309–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Brackenbury WJ, Isom LL, Front. Pharmacol 2011, 53, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Aman TK, Grieco-Calub TM, Chen C, Rusconi R, Slat EA, Isom LL, Raman IM, J. Neurosci 2009, 29, 2027–2042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Lewis AH, Raman IM, J. Physiol 2014, 592, 4825–4838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Nusser Z, Trends Neurosci. 2009, 32, 267–274. [DOI] [PubMed] [Google Scholar]
  • [16].Watanabe E, Fujikawa A, Matsunaga H, Yasoshima Y, Sako N, Yamamoto T, Saegusa C, Noda M, J. Neurosci 2000, 20, 7743–7751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Lin WH, Wright DE, Muraro NI, Baines RA, J. Neurophysiol 2009, 102, 1994–2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Scheuer T, Semin. Cell Dev. Biol 2011, 22, 160–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Pitt GS, Lee SY, Protein Sci. 2016, 25, 1573–1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Rush AM, Cummins TR, Waxman SG, J. Physiol 2007, 579, 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Zhou X, Xiao Z, Xu Y, Zhang Y, Tang D, Wu X, Tang C, Chen M, Shi X, Chen P, Liang S, Liu Z, Front. Pharmacol 2017, 8, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Xiao Y, Barbosa C, Pei Z, Xie W, Strong JA, Zhang JM, Cummins TR, J. Neurosci 2019, 39, 1539–1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-Hajj SD, Am. Physiol. Rev 2019, 99, 1079–1151. [DOI] [PubMed] [Google Scholar]
  • [24].Jukic M, Kikelj D, Anderluh M, Curr. Med. Chem 2013, 21, 164–186. [DOI] [PubMed] [Google Scholar]
  • [25].Lossin C, Rhodes TH, Desai RR, Vanoye CG, Wang D, Carniciu S, Devinsky O, George AL, J. Neurosci 2003, 23, 11289–11295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Mantegazza M, Yu FH, Powell AJ, Clare JJ, Catterall WA, Scheuer T, J. Neurosci 2005, 25, 3341–3349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Ye M, Yang J, Tian C, Zhu Q, Yin L, Jiang S, Yang M, Shu Y, Sci. Rep 2018, 8, 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Cummins TR, Aglieco F, Renganathan M, Herzog RI, Dib-Hajj SD, Waxman SG, J. Neurosci 2001, 21, 5952–5961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Jarecki BW, Piekarz AD, Jackson JO, Cummins TR, J. Clin. Invest 2010, 120, 369–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Liang W, Cho HC, Marbán E, J. Physiol 2015, 593, 1147–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Herzog RI, Cummins TR, Ghassemi F, Dib-Hajj SD, Waxman SG, J. Physiol 2003, 551, 741–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Zhou X, Zhang Y, Tang D, Liang S, Chen P, Tang C, Liu ZA, Front. Pharmacol 2018, 9, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Han C, Estacion M, Huang J, Vasylyev D, Zhao P, Dib-Hajj SD, Waxman SG, J. Neurophysiol 2015, 113, 3172–3185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Facer P, Punjabi PP, Abrari A, Kaba RA, Severs NJ, Chambers J, Kooner JS, Anand P, Int. Heart J 2011, 52, 146–152. [DOI] [PubMed] [Google Scholar]
  • [35].Dib-Hajj SD, Black JA, Waxman SG, Pain Med. 2009, 10, 1260–1269. [DOI] [PubMed] [Google Scholar]
  • [36].Hains BC, Saab CY, Klein JP, Craner MJ, Waxman SG, J. Neurosci 2004, 24, 4832–4839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Huang XJ, Mao Q, Lin Y, Feng JF, Jiang JY, J. Neurotrauma 2013, 30, 39–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Lindia JA, Köhler MG, Martin WJ, Abbadie C, Pain 2005, 117, 145–153. [DOI] [PubMed] [Google Scholar]
  • [39].Novakovic SD, Tzoumaka E, McGivern JG, Haraguchi M, Sangameswaran L, Gogas KR, Eglen RM, Hunter JC, J. Neurosci 1998, 18, 2174–2187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Sleeper AA, Cummins TR, Dib-Hajj SD, Hormuzdiar W, Tyrrell L, Waxman SG, Black JA, J. Neurosci 2000, 20, 7279–7289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Dib-Hajj SD, Black JA, Waxman SG, Nat. Rev. Neurosci 2015, 16, 511–519. [DOI] [PubMed] [Google Scholar]
  • [42].Black JA, Nikolajsen L, Kroner K, Jensen TS, Waxman SG, Ann. Neurol 2008, 64, 644–653. [DOI] [PubMed] [Google Scholar]
  • [43].Bird EV, Robinson PP, Boissonade FM, Arch. Oral Biol 2007, 52, 494–502. [DOI] [PubMed] [Google Scholar]
  • [44].Yiangou Y, Birch R, Sangameswaran L, Eglen R, Anand P, FEBS Lett. 2000, 467, 249–252. [DOI] [PubMed] [Google Scholar]
  • [45].England JD, Happel LT, Kline DG, Gamboni F, Thouron CL, Liu ZP, Levinson SR, Neurology 1996, 47, 272–276. [DOI] [PubMed] [Google Scholar]
  • [46].Wang W, Atianjoh F, Gauda EB, Yaster M, Li Y, Tao YX, Anat. Rec 2011, 194, 1406–1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Goodwin G, McMahon SB, Nat. Rev. Neurosci 2021, 22, 263–274. [DOI] [PubMed] [Google Scholar]
  • [48].Zhang XL, Ding HH, Xu T, Liu M, Ma C, Wu SL, Wei JY, Liu CC, Zhang SB, Xin WJ, Sci. Signaling 2018, 11, 1–13. [DOI] [PubMed] [Google Scholar]
  • [49].Black JA, Liu S, Tanaka M, Cummins TR, Waxman SG, Pain 2004, 108, 237–247. [DOI] [PubMed] [Google Scholar]
  • [50].Gould HJ, England JD, Soignier RD, Nolan P, Minor LD, Liu ZP, Levinson SR, Paul D, J. Pain 2004, 5, 270–280. [DOI] [PubMed] [Google Scholar]
  • [51].Tanaka M, Cummins TR, Ishikawa K, Dib-Hajj SD, Black JA, Waxman SG, NeuroReport 1998, 9, 967–972. [DOI] [PubMed] [Google Scholar]
  • [52].McMahon SB, Cafferty WBJ, Marchand F, Exp. Neurol 2005, 192, 444–462. [DOI] [PubMed] [Google Scholar]
  • [53].Laedermann CJ, Abriel H, Decosterd I, Front. Pharmacol 2015, 6, 1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Su YS, World J. Anesthesiol. 2014, 3, 71–81. [Google Scholar]
  • [55].Devor M, J. Pain 2006, 7, S3–S12. [DOI] [PubMed] [Google Scholar]
  • [56].Wei Z, Fei Y, Su W, Chen G, Front. Cell. Neurosci 2019, 13, 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Ji RR, Berta T, Nedergaard M, Pain 2013. 154, S10–S28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Henry DE, Chiodo AE, Yang W, Phys. Med. Rehab 2011, 3, 1116–1125. [DOI] [PubMed] [Google Scholar]
  • [59].Billen B, Bosmans F, Tytgat J, Curr. Pharm. Des 2008, 14, 2492–2502. [DOI] [PubMed] [Google Scholar]
  • [60].Catterall W, Cestèle S, Yarov-Yarovoy V, Yu FH, Konoki K, Scheuer T, Toxicon 2007, 49, 124–141. [DOI] [PubMed] [Google Scholar]
  • [61].Evans MH, Br. J. Pharmacol. Chemother 1964, 22, 478–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Takata M, Moore JW, Kao CY, Fuhrman FA, J. Gen. Physiol 1966, 49, 977–988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Pinto V, Derkach VA, Safronov BV, J. Neurophysiol 2008, 99, 617–628. [DOI] [PubMed] [Google Scholar]
  • [64].Blair NT, Bean BP, J. Neurosci 2002, 22, 10277–10290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Stevens M, Peigneur S, Tytgat J, Front. Pharmacol 2011, 2, 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Xu L, Ding X, Wang T, Mou S, Sun H, Hou T, Drug Discovery Today 2019, 24, 1389–1397. [DOI] [PubMed] [Google Scholar]
  • [67].Ahern CA, Payandeh J, Bosmans F, B. Chanda, J. Gen. Physiol 2016, 147, 1–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Kalia J, Milescu M, Salvatierra J, Wagner J, Klint J, King G, Olivera B, Bosmans F, J. Mol. Biol 2015, 427, 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Lipscombe D, Toro CP, in From Molecules to Networks: An Introduction to Cellular and Molecular Neuroscience, 3rd ed. (Eds.: Byrne JH, Heidelberger R, Waxham MN), Elsevier, Cambridge, 2014, 377–407. [Google Scholar]
  • [70].Gilchrist J, Olivera BM, Bosmans F, in Voltage Gated Sodium Channels (Ed.: Ruben PC), Springer, New York, 2013, 203–230. [Google Scholar]
  • [71].Canning BJ, Spina D, in Sensory Nerves (Eds.: Canning BJ, Spina D), Springer, New York, 2009, 519–562. [Google Scholar]
  • [72].Liu Z, Cai T, Zhu Q, Deng M, Li J, Zhou X, Zhang D, Li D, Li J, Liu Y, Hu W, Liang S, J. Biol. Chem 2013, 288, 20392–20403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].LePage KT, Goeger D, Yokokawa F, Asano T, Shioiri T, Gerwick WH, Murray TF, Toxicol. Lett 2005, 158, 133–139. [DOI] [PubMed] [Google Scholar]
  • [74].Edwards DJ, Marquez BL, Mogle LM, McPhail K, Goeger DE, Roberts MA, Cell Chem. Biol 2004, 11, 817–883. [DOI] [PubMed] [Google Scholar]
  • [75].Paz C, Ortiz L, Deuis JR, Vetter I, Nat. Prod. Res 2022, 10.1080/14786419.2022.2025592. [DOI] [PubMed] [Google Scholar]
  • [76].Gilchrist J, Olivera BM, Bosmans F, Handb. Exp. Pharmacol 2014, 221, 203–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Wilson MJ, Yoshikami D, Azam L, Gajewiak J, Olivera BM, Bulaj G, Zhang MM, Proc. Natl. Acad. Sci. USA 2011, 108, 10302–10307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Ekberg J, Jayamanne A, Vaughan CW, Aslan S, Thomas L, Mould J, Drinkwater R, Baker MD, Abrahamsen B, Wood JN, Adams DJ, Christie MJ, Lewis RJ, Proc. Natl. Acad. Sci. USA 2006, 103, 17030–17035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Nieto FR, Cobos EJ, Tejada MÁ, Sánchez-Fernández C, González-Cano R, Cendán CM, Mar. Drugs 2012, 10, 281–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Mueller A, Starobova H, Morgan M, Dekan Z, Cheneval O, Schroeder CI, Alewood PF, Deuis JR, Vetter I, Pain 2019, 160, 1766–1780. [DOI] [PubMed] [Google Scholar]
  • [81].Bai Z-T, Liu T, Jiang F, Cheng M, Pang X-Y, Hua L-M, Shi J, Zhou J-J, Shu X-Q, Zhang J-W, Ji Y-H, Exp. Neurol 2010, 226, 159–172. [DOI] [PubMed] [Google Scholar]
  • [82].Xu H, Li T, Rohou A, Arthur CP, Tzakoniati F, Wong E, Estevez A, Kugel C, Franke Y, Chen J, Ciferri C, Hackos DH, Koth CM, Payandeh J, Cell 2019, 176, 702–715. [DOI] [PubMed] [Google Scholar]
  • [83].Beckley JT, Pajouhesh H, Luu G, Klas S, Delwig A, Monteleone D, Zhou X, Giuvelis D, Meng ID, Yeomans DC, Hunter JC, Mulcahy JV, Pain 2021, 162, 1250–1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Hu H, Mawlawi SE, Zhao T, Deuis JR, Jami S, Vetter I, Lewis RJ, Cardoso FC, Front. Mol. Biosci 2021, 8, 742457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Moyer BD, Murray JK, Ligutti J, Andrews K, Favreau P, Jordan JB, Lee JH, Liu D, Long J, Sham K, Shi L, Stöcklin R, Wu B, Yin R, Yu V, Zou A, Biswas K, Miranda LP, PLoS One 2018, 13, e1096791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Kim K, Dutta J, Groll A, El Fakhri G, Meng LJ, Li Q, Phys. Med. Biol 2018, 63, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Der Van Have F, Vastenhouw B, Ramakers RM, Branderhorst W, Krah JO, Ji C, Staelens SG, Beekman FJ, J. Nucl. Med 2009, 50, 599–605. [DOI] [PubMed] [Google Scholar]
  • [88].Ritchie JM, Rogart RB, Strichartz GR, J. Physiol 1976, 261, 477–494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Wilzbach KE, J. Am. Chem. Soc 1957, 79, 1013. [Google Scholar]
  • [90].Reith MEA, Eur. J. Pharmacol. Mol. Pharmacol. Sect 1990, 188, 33–41. [DOI] [PubMed] [Google Scholar]
  • [91].Laycock MV, Kralovec J, Richards R, Nat. Toxins 1997, 5, 36–42. [DOI] [PubMed] [Google Scholar]
  • [92].Brown GB, Tieszen SC, Daly JW, Warnick JE, Albuquerque EX, Cell. Mol. Neurobiol 1981, 1, 19–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Strichartz GR, Rogart RB, Ritchie JM, J. Membr. Biol 1979, 48, 357–364. [DOI] [PubMed] [Google Scholar]
  • [94].MacKinnon AC, Wyatt KM, McGivern JG, Sheridan RD, Brown CM, Br. J. Pharmacol 1995, 115, 1103–1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Hartshorne RP, Catterall WA, Proc. Natl. Acad. Sci. USA 1981, 78, 4620–4624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Hartshorne RP, Messner DJ, Coppersmith JC, Catterall WA, J. Biol. Chem 1982, 257, 13888–13895. [PubMed] [Google Scholar]
  • [97].Corbett AM, Krueger BK, J. Membr. Biol 1990, 117, 163–176. [DOI] [PubMed] [Google Scholar]
  • [98].Moore HH, Fritz LC, Raftery MA, Brockes JP, Proc. Natl. Acad. Sci. USA 1982, 79, 1673–1677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Strichartz GR, Rogart RB, Ritchie JM, J. Membr. Biol 1979, 48, 357–364. [DOI] [PubMed] [Google Scholar]
  • [100].Ritchie JM, Rogart RB, Proc. Natl. Acad. Sci. USA 1977, 74, 211–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Rudy B, Kirschenbaum B, Greene LA, J. Neurosci 1982, 2, 1405–1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Yanagita T, Wada A, Yamamoto R, Kobayashi H, Yuhi T, Urabe M, Niina H, J. Neurochem 1996, 66, 1249–1253. [DOI] [PubMed] [Google Scholar]
  • [103].Yanagita T, Kobayashi H, Yamamoto R, Kataoka H, Yokoo H, Shiraishi S, Minami SI, Koono M, Wada A, J. Neurochem 2000, 74, 1674–1684. [DOI] [PubMed] [Google Scholar]
  • [104].Yanagita T, Kobayashi H, Uezono Y, Yokoo H, Sugano T, Saitoh T, Minami SI, Shiraishi S, Wada A, Mol. Pharmacol 2003, 63, 1125–1136. [DOI] [PubMed] [Google Scholar]
  • [105].Kazen-Gillespie KA, Ragsdale DS, D’Andreall MR, Mattei LN, Rogers KE, Isom LL, J. Biol. Chem 2000, 275, 1079–1088. [DOI] [PubMed] [Google Scholar]
  • [106].Chen C, Bharucha V, Chen Y, Westenbroek RE, Brown A, Malhotra JD, Jones D, Avery C, Gillespie PJ, Kazen-Gillespie KA, Kazarinova-Noyes K, Shrager P, Saunders TL, Macdonald RL, Ransom BR, Scheuer T, Catterall WA, Isom LL, Proc. Natl. Acad. Sci. USA 2002, 99, 17072–17077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Patino GA, Brackenbury WJ, Bao B, Lopez-Santiago LF, O’Malley HA, Chen C, Calhoun JD, Lafrenière RG, Cossette P, Rouleau GA, Isom LL, J. Neurosci 2011, 31, 14577–14591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Srinivasan Y, Elmer L, Davis J, Bennett V, Angelides K, Nature 1988, 333, 177–180. [DOI] [PubMed] [Google Scholar]
  • [109].Pellegrino RG, Politis MJ, Ritchie JM, Spencer PS, J. Neurocytol 1986, 15, 17–28. [DOI] [PubMed] [Google Scholar]
  • [110].Politis MJ, Pellegrino RG, Oaklander AL, Ritchie JM, Brain Res. 1983, 273, 392–395. [DOI] [PubMed] [Google Scholar]
  • [111].Yorek MA, Stefani MR, Wachtel RE, J. Neurochem 1994, 62, 63–69. [DOI] [PubMed] [Google Scholar]
  • [112].Ritchie JM, Rogart RB, in Reviews of Physiology Biochemistry and Pharmacology (Eds.: Adrian RH, Holzer H, Kramer K, Linden RJ, Miescher PA, Rasmussen H, Trendelenburg W, Vogt W), Springer, New York, 1977, 1–50. [DOI] [PubMed] [Google Scholar]
  • [113].Bontemps J, Cantineau R, Grandfils C, Leprince P, Dandrifosse G, Schoffeniels E, Anal. Biochem 1984, 139, 149–157. [DOI] [PubMed] [Google Scholar]
  • [114].Wu BQ, Yang L, Kao CY, Levinson SR, Yotsu-Yamashita M, Yasumoto T, Toxicon 1996, 34, 407–416. [DOI] [PubMed] [Google Scholar]
  • [115].Hong B, Chen H, Han J, Xie Q, He J, Bai K, Dong Y, Yi R, Mar. Drugs 2017, 15, 159–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Tanino H, Nakata T, Kaneko T, Kishi Y, J. Am. Chem. Soc 1977, 99, 2818–2819. [DOI] [PubMed] [Google Scholar]
  • [117].Jacobi PA, Martinelli MJ, Polanc S, J. Am. Chem. Soc 1984, 106, 5594–5598. [Google Scholar]
  • [118].Fleming JJ, Du Bois J, J. Am. Chem. Soc 2006, 128, 3926–3927. [DOI] [PubMed] [Google Scholar]
  • [119].Iwamoto O, Shinohara R, Nagasawa K, Asian J. Chem. 2009, 4, 277–285. [DOI] [PubMed] [Google Scholar]
  • [120].Bhonde VR, Looper RE, J. Am. Chem. Soc 2011, 133, 20172–20174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Sawayama Y, Nishikawa T, J. Synth. Org. Chem. Jpn 2012, 70, 1178–1186. [Google Scholar]
  • [122].Martinelli MJ, Brownstein AD, Jacobi PA, Polanc S, Croat. Chem. Acta 1986, 59, 267–295. [Google Scholar]
  • [123].Thottumkara AP, Parsons WH, Du Bois J, Angew. Chem. Int. Ed 2014, 53, 5760–5784; Angew. Chem. 2014, 126, 5868–5894. [DOI] [PubMed] [Google Scholar]
  • [124].Andresen BM, Du Bois J, J. Am. Chem. Soc 2009, 131, 12524–12525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Hoehne A, Behera D, Parsons WH, James ML, Shen B, Borgohain P, Bodapati D, Prabhakar A, Gambhir SS, Yeomans DC, Biswal S, Chin FT, Du Bois J, J. Am. Chem. Soc 2013, 135, 18012–18015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Pérez-Medina C, Patel N, Robson M, Lythgoe MF, Årstad E, Bioorg. Med. Chem. Lett 2013, 23, 5170–5173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Pérez-Medina C, Patel N, Robson M, Lythgoe MF, Årstad E, Org. Biomol. Chem 2012, 10, 9474–9480. [DOI] [PubMed] [Google Scholar]
  • [128].Uehara S, Uyemura K, Neurochem. Res 1985, 10, 1119–1128. [DOI] [PubMed] [Google Scholar]
  • [129].Iwasa J, Kamano H, Baba N, Nakajima S, Tsuji K, Seyama I, Biosci. Biotechnol. Biochem 1992, 56, 1169–1171. [DOI] [PubMed] [Google Scholar]
  • [130].Tzakoniati F, Xu H, Li T, Garcia N, Kugel C, Payandeh J, Koth CM, Tate EW, Cell Chem. Biol 2020, 27, 306–313.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Casebolt TL, Brown GB, Toxicon 1993, 31, 1113–1122. [DOI] [PubMed] [Google Scholar]
  • [132].Trainer VL, Thomsen WJ, Catterall WA, Baden DG, Mol. Pharmacol 1991, 40, 988–994. [PubMed] [Google Scholar]
  • [133].Beneski DA, Catterall WA, Proc. Natl. Acad. Sci. USA 1980, 77, 639–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Catterall WA, Hartshorne RP, Beneski DA, Toxicon 1982, 20, 27–40. [DOI] [PubMed] [Google Scholar]
  • [135].de Lima ME, Couraud F, Lapied B, Pelhate M, Ribeiro Diniz C, Rochat H, Biochem. Biophys. Res. Commun 1988, 151, 187–192. [DOI] [PubMed] [Google Scholar]
  • [136].Balerna M, Lombert A, Chicheportiche R, Romey G, Lazdunski M, Biochim. Biophys. Acta 1981, 664, 219–225. [DOI] [PubMed] [Google Scholar]
  • [137].Guillory RJ, Rayner MD, D’arrigo JS, Science 1977, 196, 883–886. [DOI] [PubMed] [Google Scholar]
  • [138].Lukacs P, Földi MC, Valánszki L, Casanova E, Biri-Kovács B, Nyitray L, Málnási-Csizmadia A, Mike A, Sci. Rep 2018, 8, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Parsons WH, Du Bois J, J. Am. Chem. Soc 2013, 135, 10582–10585. [DOI] [PubMed] [Google Scholar]
  • [140].Elleman AV, Devienne G, Makinson CD, Haynes AL, Huguenard JR, Du Bois J, Nat. Commun 2021, 12, 4171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Montnach J, Blömer LA, Lopez L, Filipis L, Meudal H, Lafoux A, Nicolas S, Chu D, Caumes C, Béroud R, Jopling C, Bosmans F, Huchet C, Landon C, Canepari M, De Waard M, Nat. Commun 2022, 13, 417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Minta A, Tsien RY, J. Biol. Chem 1989, 264, 19449–19457. [PubMed] [Google Scholar]
  • [143].Roder P, Hille C, Photochem. Photobiol. Sci 2014, 13, 1699–1710. [DOI] [PubMed] [Google Scholar]
  • [144].Tsien RY, Rink RJ, Poenie M, Cell Calcium 1985, 6, 145–157. [DOI] [PubMed] [Google Scholar]
  • [145].Zhang H, Zou B, Du F, Xu K, Li M, Mol. Pharmacol 2015, 87, 207–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Brown GB, Bradley RJ, Neurosci J. Methods 1985, 13, 119–129. [DOI] [PubMed] [Google Scholar]
  • [147].Alcaraz G, Sampo B, Tricaud N, Giraud P, Martin-Eauclaire MF, Couraud F, Dargent B, Mol. Brain Res 1997, 51, 143–153. [DOI] [PubMed] [Google Scholar]
  • [148].Dargent B, Couraud F, Proc. Natl. Acad. Sci. USA 1990, 87, 5907–5911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Ondrus AE, Lee HLD, Iwanaga S, Parsons WH, Andresen BM, Moerner WE, Du Bois J, Chem. Biol 2012, 19, 902–912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Lord SJ, Conley NR, Lee HD, Nichimura SY, Pomerantz AK, Willets KA, Lu Z, Wang H, Liu N, Samuel R, Weber R, Semyonov A, He M, Twieg RJ, Moerner WE, ChemPhysChem 2009, 10, 55–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Gonzales J, Pirovano G, Chow CY, Demetrio P, Franca DS, Carter LM, Klint JK, Guru N, Lewis JS, King GF, Reiner T, EJNMMI Res. 2020, 10, 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Sakai A, Yoshikawa K, Kurata H, Tanimura A, Food Hyg J. Soc. Japan 1978, 19, 85–90. [Google Scholar]
  • [153].Melhuish WH, J. Phys. Chem 1961, 65, 229–235. [Google Scholar]
  • [154].Massensini AR, Suckling J, Brammer MJ, Moraes-Santos T, Gomez MV, Romano-Silva MA, Neurosci J. Methods 2002, 116, 189–196. [DOI] [PubMed] [Google Scholar]
  • [155].Dang B, Kubota T, Correa AM, Bezanilla F, Kent SBH, Angew. Chem. Int. Ed 2014, 53, 8970–8974; Angew. Chem. 2014, 126, 9116–9120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Kubota T, Durek T, Dang B, Finol-Urdaneta RK, Craik DJ, Kent SBH, French RJ, Bezanilla F, Correa AM, Proc. Natl. Acad. Sci. USA 2017, 114, E1857–E1865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Gonzales J, Pirovano G, Chow CY, Demetrio P, Franca DS, Carter LM, Klint JK, Guru N, Lewis JS, King GF, Reiner T, EJNMMI Res. 2020, 10, 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Kirsch GE, Yeh JZ, Farley JM, Narahishi T, J. Gen. Physiol 1980, 76, 315–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Beaurepaire E, Buissette V, Sauviat MP, Giaume D, Lahlil K, Mercuri A, Casanova D, Huignard A, Martin JL, Gacoin T, Boilot JP, Alexandrou A, Nano Lett. 2004, 4, 2079–2083. [Google Scholar]
  • [160].Mulcahy JV, Pajouhesh H, Beckley JT, Delwig A, Du Bois J, Hunter JC, J. Med. Chem 2019, 62, 8695–8710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Fertleman CR, Ferrie CD, Aicardi J, Bednarek NAF, Eeg-Olofsson O, Elmslie FV, Griesemer DA, Goutières F, Kirkpatrick M, Malmros INO, Pollitzer M, Rossiter M, Roulet-Perez E, Schubert R, Smith VV, Testard H, Wong V, Stephenson JBP, Neurology 2007, 69, 586–595. [DOI] [PubMed] [Google Scholar]
  • [162].Cox JJ, Reimann F, Nicholas AK, Thornton G, Roberts E, Springell K, Karbani G, Jafri H, Mannan J, Raashid Y, Al-Gazali L, Hamamy H, Valente EM, Gorman S, Williams R, McHale DP, Wood JN, Gribble FM, Woods CG, Nature 2006, 444, 894–898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Nardi A, Damann N, Hertrampf T, Kless A, ChemMedChem. 2012, 10, 1712–1740. [DOI] [PubMed] [Google Scholar]
  • [164].Bagal SK, Chapman ML, Marron BE, Prime R, Storer I, Swain NA, Bioorg. Med. Chem. Lett 2014, 24, 3690–3699. [DOI] [PubMed] [Google Scholar]
  • [165].Payandeh J, Hackos DH, Handb. Exp. Pharmacol 2018, 246, 271–306. [DOI] [PubMed] [Google Scholar]
  • [166].Eagles DA, Chow CY, King GF, Br. J. Pharmacol 2020, 10.1111/bph.15327. [DOI] [PubMed] [Google Scholar]
  • [167].Mogil JS, Nat. Rev. Neurosci 2009, 10, 283–294. [DOI] [PubMed] [Google Scholar]
  • [168].Siebenga P, van Amerongen G, Hay JL, McDonnell A, Gorman D, Butt R, Groeneveld GJ, Clin. Transl. Sci 2020, 13, 318–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Mcdonnell A, Collins S, Ali Z, Iavarone L, Surujbally R, Kirby S, Butt RP, Pain 2018, 159, 1465–1476. [DOI] [PubMed] [Google Scholar]
  • [170].Alexandrou AJ, Brown AR, Chapman ML, Estacion M, Turner J, Mis MA, Wilbrey A, Payne EC, Gutteridge A, Cox PJ, Doyle R, Printzenhoff D, Lin Z, Marron BE, West C, Swain NA, Storer RI, Stupple PA, Castle NA, Hounshell JA, Rivara M, Randall A, Dib-Hajj SD, Krafte D, Waxman SG, Patel MK, Butt RP, Stevens EB, PLoS One 2016, 11, 1–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Swain NA, Batchelor D, Beaudoin S, Bechle BM, Bradley PA, Brown AD, Brown B, Butcher KJ, Butt RP, Chapman ML, Denton S, Ellis D, Galan SRG, Gaulier SM, Greener BS, de Groot MJ, Glossop MS, Gurrell IK, Hannam J, Johnson MS, Lin Z, Markworth CJ, Marron BE, Millan DS, Nakagawa S, Pike A, Printzenhoff D, Rawson DJ, Ransley SJ, Reister SM, Sasaki K, Storer RI, Stupple PA, West CW, J. Med. Chem 2017, 60, 7029–7042. [DOI] [PubMed] [Google Scholar]
  • [172].Sun S, Jia Q, Zenova AY, Wilson MS, Chowdhury S, Focken T, Li J, Decker S, Grimwood ME, Andrez J-C, Hemeon I, Sheng T, Chen C-A, White A, Hackos DH, Deng L, Bankar G, Khakh K, Chang E, Kwan R, Lin S, Nelkenbrecher K, Sellers BD, DiPasquale AG, Chang J, Pang J, Sojo L, Lindgren A, Waldbrook M, Xie Z, Young C, Johnson JP, Robinette CL, Cohen CJ, Safina BS, Sutherlin DP, Ortwine DF, Dehnhardt CM, J. Med. Chem 2019, 62, 908–927. [DOI] [PubMed] [Google Scholar]
  • [173].Neff RA, Flinspach M, Gibbs A, Shih AY, Minassian NA, Liu Y, Fellows R, Libiger O, Young S, Pennington MW, Hunter MJ, Wickenden AD, J. Biol. Chem 2020, 295, 1315–1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Mueller A, Dekan Z, Kaas Q, Agwa AJ, Starobova H, Alewood PF, Schroeder CI, Mobli M, Deuis JR, Vetter I, ACS Pharmacol. Transl. Sci 2020, 3, 535–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Ahuja S, Mukund S, Deng L, Khakh K, Chang E, Ho H, Shriver S, Young C, Lin S, Johnson JP, Wu P, Li J, Coons M, Tam C, Brillantes B, Sampang H, Mortara K, Bowman KK, Clark KR, Estevez A, Xie Z, Verschoof H, Grimwood M, Dehnhardt C, Andrez J, Focken T, Sutherlin DP, Safina BS, Starovasnik MA, Ortwine DF, Franke Y, Cohen CJ, Hackos DH, Koth CM, Payandeh J, Science 2015, 350, aac5464. [DOI] [PubMed] [Google Scholar]
  • [176].Noreng S, Li T, Payandeh J, J. Mol. Biol 2021, 433, 166967. [DOI] [PubMed] [Google Scholar]
  • [177].Storer RI, Pike A, Swain NA, Alexandrou AJ, Bechle BM, Blakemore DC, Brown AD, Castle NA, Corbett MS, Flanagan NJ, Fengas D, Johnson MS, Jones LH, Marron BE, Payne CE, Printzenhofff D, Rawson DJ, Rose CR, Ryckmans T, Sun J, Theile JW, Torella R, Tseng E, Warmus JS, Bioorg. Med. Chem. Lett 2017, 27, 4805–4811. [DOI] [PubMed] [Google Scholar]
  • [178].Graceffa RF, Boezio AA, Able J, Altmann S, Berry LM, Boezio C, Butler JR, Chu-Moyer M, Cooke M, DiMauro EF, Dineen TA, Bojic EF, Foti RS, Fremeau RT Jr., Guzman-Perez A, Gao H, Gunaydin H, Huang H, Huang L, Ilch C, Jarosh M, Kornecook T, Kreiman CR, La DS, Ligutti J, Milgram BC, Lin M-HJ, Marx IE, Nguyen HN, Peterson EA, Rescourio G, Roberts J, Schenkel L, Shimanovich R, Sparling BA, Stellwagen J, Taborn K, Vaida KR, Wang J, Yeoman J, Yu V, Zhu D, Moyer BD, Weiss MM, J. Med. Chem 2017, 60, 5990–6017. [DOI] [PubMed] [Google Scholar]
  • [179].Luo G, Chen L, Easton A, Newton A, Bourin C, Shields E, Mosure K, Soars MG, Knox RJ, Matchett M, Pieschl LRL, Post-Munson DJ, Wang S, Herrington J, Graef J, Newberry K, Sivarao DV, Senapati A, Bristow LJ, Meanwell NA, Thompson LA, Dzierba C, J. Med. Chem 2019, 62, 831–856. [DOI] [PubMed] [Google Scholar]
  • [180].Kornecook TJ, Yin R, Altmann S, Be X, Berry V, Ilch CP, Jarosh M, Johnson D, Lee JH, Lehto SG, Ligutti J, Liu D, Luther J, Matson D, Ortuno D, Roberts J, Taborn K, Wang J, Weiss MM, Yu V, Zhu DXD, Fremeau RT Jr., Moyer BD, J. Pharmacol. Exp. Ther 2017, 362, 146–160. [DOI] [PubMed] [Google Scholar]
  • [181].Weiss MM, Dineen TA, Marx IE, Altmann S, Boezio A, Bregman H, Chu-Moyer M, Dimauro EF, Feric Bojic E, Foti RS, Gao H, Graceffa R, Gunaydin H, Guzman-Perez A, Huang H, Huang L, Jarosh M, Kornecook T, Kreiman CR, Ligutti J, La DS, Jasmine Lin M-H, Liu D, Moyer BD, Nguyen HN, Peterson EA, Rose PE, Taborn K, Youngblood BD, Yu V, Fremeau RT Jr., J. Med. Chem 2017, 60, 5969–5989. [DOI] [PubMed] [Google Scholar]
  • [182].Pajouhesh H, Beckley JT, Delwig A, Hajare HS, Luu G, Monteleone D, Zhou X, Ligutti J, Amagasu S, Moyer BD, Yeomans DC, Du Bois J, Mulcahy JV, Sci. Rep 2020, 10, 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Schmalhofer WA, Calhoun J, Burrows R, Bailey T, Kohler MG, Weinglass AB, Kaczorowski GJ, Garcia ML, Koltzenburg M, Priest BT, Mol. Pharmacol 2008, 74, 1476–1484. [DOI] [PubMed] [Google Scholar]
  • [184].Faber CG, Lauria G, Merkies ISJ, Cheng X, Han C, Ahn H, Persson A, Hoeijmakers JGJ, Gerrits MM, Pierro T, Lombardi R, Kapetis D, Dib-Hajj SD, Waxman SG, Proc. Natl. Acad. Sci. USA 2012, 20, 19444–19449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Zhang XY, Wen J, Yang W, Wang C, Gao L, Zheng LH, Wang T, Ran K, Li Y, Li X, Xu M, Luo J, Feng S, Ma X, Chai Z, Zhou Z, Yao J, Zhang X, Liu JY, Am. J. Hum. Genet 2013, 93, 957–966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Maher B, 2017, WO2017075222A1.
  • [187].Jarvis MF, Honore P, Shieh CC, Chapman M, Joshi S, Zhang XF, Kort M, Carroll W, Marron B, Atkinson R, Thomas J, Liu D, Krambis M, Liu Y, McGaraughty S, Chu K, Roeloffs R, Zhong C, Mikusa JP, Hernandez G, Gauvin D, Wade C, Zhu C, Pai M, Scanio M, Shi L, Drizin I, Gregg R, Matulenko M, Hakeem A, Gross M, Johnson M, Marsh K, Wagoner PK, Sullivan JP, Faltynek CR, Krafte DS, Proc. Natl. Acad. Sci. USA 2007, 104, 8520–8525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [188].Mcgaraughty S, Chu KL, Scanio MJC, Kort ME, Faltynek CR, Jarvis MF, Pharmacology 2008, 324, 1204–1211. [DOI] [PubMed] [Google Scholar]
  • [189].Schuelert N, McDougall JJ, Arthritis Res. Ther 2012, 14, R5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Lolignier S, Amsalem M, Maingret F, Padilla F, Gabriac M, Chapuy E, Eschalier A, Delmas P, Busserolles J, PLoS One 2011, 6, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Lolignier S, Bonnet C, Gaudioso C, Noël J, Ruel J, Amsalem M, Ferrier J, Rodat-Despoix L, Bouvier V, Aissouni Y, Prival L, Chapuy E, Padilla F, Eschalier A, Delmas P, Busserolles J, Cell Rep. 2015, 11, 1067–1078. [DOI] [PubMed] [Google Scholar]
  • [192].Leo S, D’Hooge R, Meert T, Behav. Brain Res 2010, 208, 149–157. [DOI] [PubMed] [Google Scholar]
  • [193].Rosker C, Lohberger B, Hofer D, Steinecker B, Quasthoff S, Schreibmayer W, Am. J. Physiol. Cell Physiol 2007, 293, 783–789. [DOI] [PubMed] [Google Scholar]
  • [194].Schiavon E, Sacco T, Cassulini RR, Gurrola G, Tempia F, Possani LD, Wanke E, J. Biol. Chem 2006, 281, 20326–20337. [DOI] [PubMed] [Google Scholar]
  • [195].Denomme N, Lukowski AL, Hull JM, Jameson MB, Bouza AA, Narayan ARH, Isom LL, Neurosci. Lett 2020, 724, 134853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].McCormack K, Santos S, Chapman ML, Krafte DS, Marron BE, West CW, Krambis MJ, Antonio BM, Zellmer SG, Printzenhoff D, Padilla KM, Lin Z, Wagoner PK, Swain NA, Stupple PA, de Groot M, Butt RP, Castle NA, Proc. Natl. Acad. Sci. USA 2013, 110, 2724–2732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Osteen JD, Herzig V, Gilchrist J, Emrick JJ, Zhang C, Wang X, Castro J, Garcia-Caraballo S, Grundy L, Rychkov GY, Nature 2016, 534, 494–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Hains BC, Black JA, Waxman SG, J. Neurosci. Res 2002, 70, 546–552. [DOI] [PubMed] [Google Scholar]
  • [199].Toledo-Aral JJ, Moss BL, He BZ-J, Koszowski AG, Whisenand T, Levinson SR, Wolf JJ, Silos-Santiago I, Halegoua S, Mandel G, Proc. Natl. Acad. Sci. USA 1997, 94, 1527–1532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [200].Okuse K, Chaplan SR, Mcmahon SB, Luo ZD, Calcutt NA, Scott BP, Akopian AN, Wood JN, Mol. Cell. Neurosci 1997, 10, 196–207. [DOI] [PubMed] [Google Scholar]
  • [201].Black JA, Renganathan M, Waxman SG, Mol. Brain Res 2002, 105, 19–28. [DOI] [PubMed] [Google Scholar]
  • [202].Biswas K, Nixey TE, Murray JK, Falsey JR, Yin L, Liu H, Gingras J, Hall BE, Herberich B, Holder JR, Li H, Ligutti J, Lin M-HJ, Liu D, Soriano BD, Soto M, Tran L, Tegley CM, Zou A, Gunasekaran K, Moyer BD, Doherty L, Miranda LP, ACS Chem. Biol 2017, 12, 2427–2435. [DOI] [PubMed] [Google Scholar]
  • [203].Murray JK, Wu B, Tegley CM, Nixey TE, Falsey JR, Herberich B, Yin L, Sham K, Long J, Aral J, Cheng Y, Netirojjanakul C, Doherty L, Glaus C, Ikotun T, Li H, Tran L, Soto M, Salimi-Moosavi H, Ligutti J, Amagasu S, Andrews KL, Be X, Lin M-HJ, Foti RS, Ilch CP, Youngblood B, Kornecook TJ, Karow M, Walker KW, Moyer BD, Biswas K, Miranda LP, ACS Chem. Biol 2019, 14, 806–818. [DOI] [PubMed] [Google Scholar]
  • [204].Kuo TT, Aveson VG, mAbs 2011, 3, 422–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].“Anti-SCN3 A NaV1.3 extracellular Antibody: #ASC-023,” can be found under: https://www.alomone.com/p/anti-nav1-3-extracellular/ASC-023, 2022.
  • [206].Chioni AM, Fraser SP, Pani F, Foran P, Wilkin GP, Diss JKJ, Djamgoz J. Neurosci. Methods 2005, 147, 88–98. [DOI] [PubMed] [Google Scholar]
  • [207].“Anti-NaV1.8 SCN10 A extracellular Antibody: #ASC-028,” can be found under https://www.alomone.com/p/anti-nav1-8-extracellular/ASC-028, 2022.
  • [208].Gao R, Cao T, Chen H, Cai J, Lei M, Wang Z, Transl. Cancer Res 2019, 8, 44–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Achenbach TV, Brunner B, Heermeier K, ChemBioChem 2003, 4, 928–935. [DOI] [PubMed] [Google Scholar]
  • [210].Curtis CD, Nardulli AM, Methods Mol. Biol 2009, 505, 187–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [211].Joshi SK, Mikusa JP, Hernandez G, Baker S, Shieh CC, Neelands T, Zhang XF, Niforatos W, Kage K, Han P, Krafte D, Faltynek C, Sullivan JP, Jarvis MF, Honore P, Pain 2006, 123, 75–82. [DOI] [PubMed] [Google Scholar]
  • [212].Ruangsri S, Lin A, Mulpuri Y, Lee K, Spigelman I, Nishimura I, J. Biol. Chem 2011, 286, 39836–39847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [213].Hains BC, Saab CY, Waxman SG, Brain 2005, 128, 2359–2371. [DOI] [PubMed] [Google Scholar]
  • [214].Tan AM, Samad OA, Dib-Hajj SD, Waxman SG, Mol. Med 2015, 21, 544–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [215].Xie W, Strong JA, Zhang J, J. Physiol. Behav 2015, 291, 317–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Xie W, Zhang J, Strong JA, Zhang J, Neurosci. 2019, 402, 51–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Dong XW, Goregoaker S, Engler H, Zhou X, Mark L, Crona J, Terry R, Hunter J, Priestley T, Neuroscience 2007, 146, 812–821. [DOI] [PubMed] [Google Scholar]
  • [218].Hains BC, Saab CY, Klein JP, Craner MJ, Waxman SG, J. Neurosci 2004, 24, 4832–4839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [219].Hains BC, Klein JP, Saab CY, Craner MJ, Black JA, Waxman SG, J. Neurosci 2003, 23, 8881–8892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [220].Liu Y, Yao S, Song W, Wang Y, Liu D, Zen L, Huazhong Univ J. Sci. Technol. Med. Sci 2005, 25, 696–699. [DOI] [PubMed] [Google Scholar]
  • [221].Lai J, Gold MS, Kim CS, Biana D, Ossipov MH, Hunter JC, Porreca F, Pain 2002, 95, 143–152. [DOI] [PubMed] [Google Scholar]
  • [222].Porreca F, Lai J, Bian DI, Wegert S, Ossipov MH, Eglen RM, Kassotakis L, Novakovic S, Rabert DK, Sangameswaran L, Hunter JC, Proc. Natl. Acad. Sci. USA 1999, 96, 7640–7644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].Muroi Y, Ru F, Kollarik M, Canning BJ, Hughes SA, Walsh S, Sigg M, Carr M, Undem BJ, J. Physiol 2011, 589, 5663–5676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [224].Yeomans DC, Levinson SR, Peters MC, Koszowski AG, Tzabazis AZ, Gilly WF, Wilson SP, Hum. Gene Ther 2005, 16, 271–277. [DOI] [PubMed] [Google Scholar]
  • [225].Cai W, Cao J, Ren X, Qiao L, Chen X, Li M, Zang W, BMC Anesthesiol. 2016, 16, 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Yoshimura N, Seki S, Novakovic SD, Tzoumaka E, Erickson VL, Erickson KA, Chancellor MB, De Groat WC, J. Neurosci 2001, 21, 8690–8696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Khasar SG, Gold MS, Levine JD, Neurosci. Lett 1998, 256, 17–20. [DOI] [PubMed] [Google Scholar]
  • [228].Yu YQ, Zhao F, Guan SM, Chen J, PLoS One 2011, 6, e19865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [229].Xie W, Strong JA, Ye L, Mao J, Zhang J, Pain 2013, 154, 1170–1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [230].Zhao J, Xie P, Galiano RD, Qi S, Mao R, Mustoe TA, Hong SJ, Exp. Dermatol 2019, 28, 576–584. [DOI] [PubMed] [Google Scholar]
  • [231].Sun J, Li N, Duan G, Liu Y, Guo S, Wang C, Zhu C, Zhang X, Mol. Pain 2018, 14, 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [232].Cai W, Cao J, Ren X, Qiao L, Chen X, Li M, Zang W, BMC Anesthesiol. 2016, 16, 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [233].Ke CB, He WS, Li CJ, Shi D, Gao F, Tian YK, Neuroscience 2012, 227, 80–89. [DOI] [PubMed] [Google Scholar]
  • [234].Qin S, Jiang F, Zhou Y, Zhou G, Ye P, Ji Y, Acta Biochim. Biophys. Sin 2017, 49, 713–721. [DOI] [PubMed] [Google Scholar]
  • [235].Ye P, Hua L, Jiao Y, Li Z, Qin S, Fu J, Jiang F, Liu T, Ji Y, Acta Biochim. Biophys. Sin 2015, 48, 132–144. [DOI] [PubMed] [Google Scholar]
  • [236].Kaliyaperumal S, Wilson K, Aeffner F, Dean C, Toxicol. Pathol 2020, 48, 202–219. [DOI] [PubMed] [Google Scholar]
  • [237].Lemmon VP, Ferguson AR, Popovich PG, Xu XM, Snow DM, Igarashi M, Beattie CE, Bixby JL, J. Neurotrauma 2014, 31, 1354–1361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [238].Samad OA, Tan AM, Cheng X, Foster E, Dib-Hajj SD, Waxman SG, Mol. Ther 2013, 21, 49–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [239].Lindia JA, Köhler MG, Martin WJ, Abbadie C, Pain 2005, 117, 145–153. [DOI] [PubMed] [Google Scholar]
  • [240].Kearney JA, Hum. Mol. Genet 2002, 11, 2765–2775. [DOI] [PubMed] [Google Scholar]
  • [241].Wood JN, Akopian AN, Souslova V, England S, Okuse K, Ogata N, Ure J, Smith A, Kerr BJ, McMahon SB, Boyce S, Hill R, Stanfa LC, Dickenson AH, Wood JN, Nat. Neurosci 1999, 2, 541–548. [DOI] [PubMed] [Google Scholar]
  • [242].Priest BT, Murphy BA, Lindia JA, Diaz C, Abbadie C, Ritter AM, Liberator P, Iyer LM, Kash SF, Kohler MG, Kaczorowski GH, MacIntyre DE, Martin WJ, Proc. Natl. Acad. Sci. USA 2005, 102, 9382–9387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [243].Coward K, Aitken A, Powell A, Plumpton C, Birch R, Tate S, Bountra C, Anand P, NeuroReport 2001, 12, 495–500. [DOI] [PubMed] [Google Scholar]
  • [244].Yu FH, Mantegazza M, Westenbroek RE, Robbins CA, Kalume F, Burton KA, Spain WJ, McKnight GS, Scheuer T, Catterall WA, Nat. Neurosci 2006, 9, 1142–1149. [DOI] [PubMed] [Google Scholar]
  • [245].Planells-Cases R, Caprini M, Zhang J, Rockenstein EM, Rivera RR, Murre C, Masliah E, Montal M, Biophys. J 2000, 78, 2878–2891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [246].Sittl R, Lampert A, Huth T, Schuy ET, Link AS, Fleckenstein J, Alzheimer C, Grafe P, Carr RW, Proc. Natl. Acad. Sci. USA 2012, 109, 6704–6709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [247].Nassar MA, Levato A, Stirling LC, Wood JN, Mol. Pain 2005, 1, 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [248].Gingras J, Smith S, Matson DJ, Johnson D, Nye K, Couture L, Feric E, Yin R, Moyer BD, Peterson ML, Rottman JB, Beiler RJ, Malmberg AB, McDonough SI, PLoS One 2014, 9, e105895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [249].Stirling LC, Forlani G, Baker MD, Wood JN, Matthews EA, Dickenson AH, Nassar MA, Pain 2005, 113, 27–36. [DOI] [PubMed] [Google Scholar]
  • [250].Shields SD, Ahn HS, Yang Y, Han C, Seal RP, Wood JN, Waxman SG, Dib-Hajj SD, Pain 2012, 153, 2017–2030. [DOI] [PubMed] [Google Scholar]
  • [251].Nassar MA, Baker MD, Levato A, Ingram R, Mallucci G, McMahon SB, Wood JN, Mol. Pain 2006, 2, 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Deuis JR, Zimmermann K, Romanovsky AA, Possani LD, Cabot PJ, Lewis RJ, Vetter I, Pain 2013, 154, 1749–1757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [253].Kerr BJ, Souslova V, McMahon SB, Wood JN, NeuroReport 2001, 12, 3077–3080. [DOI] [PubMed] [Google Scholar]
  • [254].Chen L, Huang J, Zhao P, Persson AK, Dib-Hajj FB, Cheng X, Tan A, Waxman SG, Dib-Hajj SD, Sci. Rep 2018, 8, 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [255].Stirling LC, Forlani G, Baker MD, Wood JN, Matthews EA, Dickenson AH, Nassar MA, Pain 2005, 113, 27–36. [DOI] [PubMed] [Google Scholar]
  • [256].Abrahamsen B, Zhao J, Asante CO, Cendan CM, Dickenson AH, Wood JN, Science 2008, 321, 702–706. [DOI] [PubMed] [Google Scholar]
  • [257].Amaya F, Wang H, Costigan M, Allchorne AJ, Hatcher JP, Egerton J, Stean T, Morisset V, Grose D, Gunthorpe MJ, Chessel IP, Tate S, Green PJ, Woolf CJ, J. Neurosci 2006, 26, 12852–12860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [258].Lolignier S, Amsalem M, Maingret F, Padilla F, Gabriac M, Chapuy E, Eschalier A, Delmas P, Busserolles J, PLoS One 2011, 6, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [259].Minett MS, Nassar MA, Clark AK, Passmore G, Dickenson AH, Wang F, Malcangio M, Wood JN, Nat. Commun 2012, 3, 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [260].Bierhaus A, Fleming T, Stoyanov S, Leffler A, Babes A, Neacsu C, Sauer SK, Eberhardt M, Schnölzer M, Lasischka F, Neuhuber WL, Kichko TI, Konrade I, Elvert R, Mier W, Pirags V, Lukic IK, Morcos M, Dehmer T, Rabbani N, Thornalley PJ, Edelstein D, Nau C, Forbes J, Humpert PM, Schwaninger M, Ziegler D, Stern DM, Cooper ME, Haberkorn U, Brownlee M, Reech PW, Nawroth PP, Nat. Med 2012, 18, 926–933. [DOI] [PubMed] [Google Scholar]
  • [261].Salvatierra J, Dong X, Bosmans F, J. Clin. Invest 2018, 128, 5434–5447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [262].Daou I, Tuttle AH, Longo G, Wieskopf JS, Bonin RP, Ase AR, Wood JN, De Koninck Y, Ribeiro-da-Silva A, Mogil JS, Séguéla P, J. Neurosci 2013, 33, 18631–18640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [263].Daou I, Beaudry H, Ase AR, Wieskopf JS, Ribeiro-da-Silva A, Mogil JS, Séguéla P, eNeuro 2016, 3, 702–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [264].Samineni VK, Mickle AD, Yoon J, Grajales-Reyes JG, Pullen MY, Crawford KE, Noh KN, Gereau GB, Vogt SK, Lai HH, Rogers JA, Gereau IV RW, Sci. Rep 2017, 7, 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [265].Madisen L, Mao T, Koch H, Zhuo JM, Berenyi A, Fujisawa S, Hsu YWA, Garcia AJ, Gu X, Zanella S, Kidney J, Gu H, Mao Y, Hooks BM, Boyden ES, Buzsáki G, Ramirez JM, Jones AR, Svoboda K, Hand X, Turner EE, Zeng H, Nat. Neurosci 2012, 15, 793–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [266].El-Gaby M, Zhang Y, Wolf K, Schwiening CJ, Paulsen O, Shipton OA, Cell Rep. 2016, 16, 2259–2268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [267].Feldman DH, Lossin C, Methods 2014, 1, 6–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [268].Ataka K, Pieribone VA, Biophys. J 2002, 82, 509–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [269].Baker BJ, Lee H, Pieribone VA, Cohen LB, Isacoff EY, Knopfel T, Kosmidis EK, Neurosci J. Methods 2007, 161, 32–38. [DOI] [PubMed] [Google Scholar]
  • [270].Hallaq H, Yang Z, Viswanathan PC, Fukuda K, Shen W, Wang DW, Wells KS, Zhou J, Yi J, Murray KT, Cardiovasc. Res 2006, 72, 250–261. [DOI] [PubMed] [Google Scholar]
  • [271].Zimmer T, Biskup C, Bollensdorff C, Benndorf K, J. Membr. Biol 2002, 186, 13–21. [DOI] [PubMed] [Google Scholar]
  • [272].Akin EJ, Solé L, Dib-Hajj SD, Waxman SG, Tamkun MM, PLoS One 2015, 10, 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [273].Akin EJ, Solé L, Johnson B, el Beheiry M, Masson JB, Krapf D, Tamkun MM, Biophys. J 2016, 111, 1235–1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [274].Solé L, Wagnon JL, Akin EJ, Meisler MH, Tamkun MM, Neurosci J. 2019, 39, 4238–4251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [275].Akin EJ, Higerd GP, Mis MA, Tanaka BS, Adi T, Liu S, Dib-Hajj FB, Waxman SG, Dib-Hajj SD, Sci. Adv 2019, 5, 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [276].Kanellopoulos AH, Koenig J, Huang H, Pyrski M, Millet Q, Lolignier S, Morohashi T, Gossage SJ, Jay M, Linley JE, Baskozos G, Kessler BM, Cox JJ, Dolphin AC, Zufall F, Wood JN, Zhao J, EMBO J. 2018, 37, 427–445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [277].Salvatierra J, Diaz-Bustamante M, Meixiong J, Tierney E, Dong X, Bosmans F, J. Clin. Invest 2018, 128, 5434–5447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [278].Kearney JA, Plummer NW, Smith MR, Kapur J, Cummins TR, Waxman SG, Goldin AL, Meisler MH, Neuroscience 2001, 102, 307–317. [DOI] [PubMed] [Google Scholar]
  • [279].Lee A, Goldin AL, Channels 2009, 3, 171–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [280].Yu FH, Westenbroek RE, Silos-Santiago I, McCormick KA, Lawson D, Ge P, Ferriera H, Lilly J, DiStefano PS, Catterall WA, Scheuer T, Curtis R, Neurosci J. 2003, 23, 7577–7585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [281].Reinhard K, Rougier J-S, Ogrodnik J, Abriel H, F1000Research 2013, 2, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [282].Biskup C, Zimmer T, Benndorf K, Nat. Biotechnol 2004, 22, 220–224. [DOI] [PubMed] [Google Scholar]
  • [283].Baroudi G, Pauliot V, Denjoy I, Gulcheney P, Shrier A, Chahine M, Circ. Res 2001, 88, e78–e83. [DOI] [PubMed] [Google Scholar]
  • [284].Baroudi G, Acharfi S, Larouche C, Chahine M, Circ. Res 2002, 90, e11–e16. [PubMed] [Google Scholar]
  • [285].Mohler PJ, Rivolta I, Napolitano C, LeMaillet G, Lambert S, Priori SG, Bennett V, Proc. Natl. Acad. Sci. USA 2004, 101, 17533–17358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [286].Li Y, Zhu T, Yang H, Dib-Hajj SD, Waxman SG, Yu Y, Xu T-L, Cheng X, Mol. Pain 2018, 14, 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [287].Scofield GG, Puhl HL 3rd, Ikeda SR, Neurophysiol J. 2008, 99, 1917–1927. [DOI] [PubMed] [Google Scholar]

RESOURCES