Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jan 23.
Published in final edited form as: Annu Rev Pharmacol Toxicol. 2022 Sep 28;63:541–563. doi: 10.1146/annurev-pharmtox-051921-123023

Personalized Therapeutics for KATP-Dependent Pathologies

Colin G Nichols 1
PMCID: PMC9868118  NIHMSID: NIHMS1854017  PMID: 36170658

Abstract

Ubiquitously expressed throughout the body, ATP-sensitive potassium (KATP) channels couple cellular metabolism to electrical activity in multiple tissues; their unique assembly as four Kir6 pore-forming subunits and four sulfonylurea receptor (SUR) subunits has resulted in a large armory of selective channel opener and inhibitor drugs. The spectrum of monogenic pathologies that result from gain- or loss-of-function mutations in these channels, and the potential for therapeutic correction of these pathologies, is now clear. However, while available drugs can be effective treatments for specific pathologies, cross-reactivity with the other Kir6 or SUR subfamily members can result in drug-induced versions of each pathology and may limit therapeutic usefulness. This review discusses the background to KATP channel physiology, pathology, and pharmacology and consider the potential for more specific or effective therapeutic agents.

Keywords: Kir6.2, Kir6.1, SUR1, SUR2, congenital hyperinsulinism, neonatal diabetes, Cantú syndrome

KATP CHANNEL STRUCTURE AND PHYSIOLOGICAL REGULATION BY ADENINE NUCLEOTIDES

Present in multiple tissues throughout the body, ATP-sensitive potassium (KATP) channels are hetero-octameric complexes of four pore-forming inward rectifier potassium channel subunits (Kir6.1 or Kir6.2, encoded by KCNJ8 and KCNJ11, respectively) and four regulatory sulfonylurea receptor (SUR1 or SUR2, encoded by ABCC8 and ABCC9, respectively) subunits (1) (Figure 1). Alternative splicing of the 3′ exons of ABCC9 gives rise to distinct SUR2A and SUR2B subunits that differ in the carboxyl terminal 42 amino acids. Intriguingly, KCNJ8 is immediately downstream of ABCC9 on human chromosome 12, and KCNJ11 is immediately downstream of ABCC8 on chromosome 11. This indicates that one pair of genes is a duplication of the other and hints at each pair of genes being co-regulated.

Figure 1.

Figure 1

Molecular regulation of ATP-sensitive potassium (KATP) channels (a) KATP channels are octameric complexes in which four pore-forming inward rectifier potassium channel (Kir6) subunits generate the channel pore and four sulfonylurea receptor (SUR) subunits serve the regulatory role. Each Kir6 subunit consists of two transmembrane helices (M1, M2) and a reentrant pore loop that forms the K selectivity filter. Each SUR subunit consists of three major domains, the unique 5 helix TM0 and the ATP-binding cassette (ABC) core 6 helix TM1 and TM2 domains, each of which are followed by linker between TM0 and TM1 (L0), nucleotide-binding fold 1 (NBF1), and NBF2, respectively. (b) The major physiological regulation is via a gate at the cytoplasmic end of the inner cavity. ATP binding to the Kir6 cytoplasmic region provides the energetic push to close channels. Magnesium-bound ATP (MgATP) binding at the ATP-binding site (ABS1) formed at the NBF1-NBF2 interface, together with ATP hydrolysis or magnesium-bound ADP (MgADP) binding at ABS2, results in a conformational activated state that is transduced to override ATP inhibition. Phosphatidyl inositol 4,5 bisphosphate (PIP2) interaction at a site near the ATP inhibitory site also provides an energetic pull to open channels. Pharmacologically, sulfonylureas (SUs) or K channel openers (KCOs), interacting with sites at the TM1/2 interface, respectively, promote channel closure or opening. (c) Model of the Kir6.2/SUR1 channel constructed from a cryo-electron microscopy density map, viewed from the side (left) and from the extracellular side (right). Key features are indicated, including transmembrane domains (TMDs), intracellular NBFs [referred to as nucleotide-binding domains (NBDs)], and the C-terminal domain (CTD). Panel c adapted with permission from Reference 2.

As in all Kir channel subunits, Kir6 subunits comprise two membrane-spanning domains flanking a re-entrant pore loop that generates the K+-selective pore and a large cytoplasmic domain formed from the intracellular amino and carboxyl termini. SUR1 and SUR2 are members of the ATP-binding cassette (ABC) superfamily (subfamily C) of membrane proteins, with each SUR subunit containing 17 transmembrane domains and two intracellular nucleotide-binding folds (NBF1, NBF2) or domains (NBD1, NBD2) (Figure 1) that come together to form two adenine nucleotide-binding sites (ABS1, ABS2) at the interface between them. Recent cryo-electron microscopy (cryo-EM) structures of the pancreatic (Kir6.2/SUR1) KATP complex (25) confirm overall octameric assembly of the channel (6) and demonstrate the regulatory nucleotide-binding sites on Kir6.2 and SUR1 (Figure 1c). Physiologically, KATP channels are inhibited by nonhydrolytic binding of ATP at sites on the Kir6 subunits. By incorporating mutations in Kir6.2 that disrupt the ATP-binding site, enhance PIP2 activation, or stabilize the channel open state, potentially activated structures of the Kir6.2/SUR1 complex have recently been obtained (7, 8). In one such structure (7), the ATP-binding site is distorted, which may help to explain why ATP preferentially binds to the closed state, underlying the ATP inhibitory effect (9). KATP channels are also inactivated by magnesium-bound ADP and ATP (MgADP and MgATP) interactions with the NBDs of SUR1 (1) (Figure 1b). In each subunit, the interface between transmembrane domain 1 (TMD1) and TMD2 is open to the cytoplasm, and the two NBDs are separated in the absence of Mg nucleotides (2). The NBDs come together to form two adenine nucleotide-binding sites (ABSs) at the dimer interface in the presence of Mg nucleotides, with MgATP being bound at a closed, catalytically inactive ABS1 site and MgADP being bound in the potentially catalytically competent ABS2 site (4) (Figure 1b). Rigid body rotations of SUR1 TMD1 and TMD2 underlie distinct propeller and quatrefoil conformations of the channel (4). The latter disrupts the interaction between Kir6.2 and the linker between TM0 and TM1 (L0) that underlies increased ATP sensitivity of Kir6 subunits when coexpressed with SURs (10). Very recent cryo-EM structures of Kir6.1/SUR2B channels, determined in the presence of ATP and glibenclamide, also include distinct propeller and quatrefoil conformations (11), showing that both can be obtained in both NDB-dimerized (i.e., activatory) and glibenclamide-bound (i.e., inhibitory) conditions, leaving the functional relevance of the two conformations unclear.

Finally, phosphatidyl inositol 4,5 bisphosphate (PIP2) is a ubiquitous activator of Kir channels, binding to sites that overlap and functionally compete with inhibitory ATP binding on Kir6.2 (9). Intriguingly, the Kir6.1 cytoplasmic domain is translocated downward compared to Kir6.2 structures, similar to that in apo-Kir2.1 channel structures (3JYC), in which the PIP2-binding site is disrupted. The functional relevance is not yet clear but may be consistent with the apparently minimal effects of PIP2 depletion on SUR2B/Kir6.1 activity (12), which suggests that Kir6.1 channels may not require PIP2 for activity.

MONOGENIC KATP DISEASES

Functional KATP channels can be reconstituted by expressing any combination of Kir6.1 or Kir6.2 with SUR1, SUR2A, or SUR2B (13, 14), and more than one SUR or Kir6 isoform can coexist in a single channel (1517). Native KATP channel composition could thus vary continuously across tissues (18), and each subunit gene could, in principle, have its own associated pathologic syndrome. However, as the monogenic pathologies associated with each subunit have become clear, it has also become clear that these are tightly matched. Despite the fact that certain tissues appear to express noncanonical pairings, notably striated cardiac and skeletal muscle (in which Kir6.2 and SUR2 predominate), the monogenic Kir6.2 and SUR1 pathologies are largely indistinguishable from one another, and thus far the same appears to be the case for Kir6.1 and SUR2 pathologies.

Kir6.2 and SUR1 Loss-of-Function Mutations Cause Congenital Hyperinsulinism

Loss-of-function (LOF) mutations in KCNJ11 and ABCC8 were first causally associated with congenital hyperinsulinism (CHI; MIM 256450) over 25 years ago (1921). CHI is typically identified in the neonatal period by insulin hypersecretion despite low blood glucose levels that, in the absence of treatment, can lead to severe mental retardation and epilepsy. CHI may be morphologically diffuse or focal: In the diffuse form, mutations can be recessively or dominantly inherited (22, 23), and all pancreatic β cells are affected, whereas in the focal form, the affected cell population is in a localized region of the pancreas (24). The focal form is typically a result of inheriting one copy of the mutated, inactive gene from the unaffected father. During embryonic development, a mutation occurs in the other, active copy of the gene and is then found within only a subset of the pancreatic islets. Many recessive ABCC8 mutations are intronic. These lead to splicing defects or exon skipping (25). Small insertions and deletions and a large number of missense mutations typically either (a) reduce channel biosynthesis, trafficking, or assembly, or (b) decrease intrinsic channel activity (26). Many of the latter localize to the NBFs and result in a reduced stimulatory effect of MgADP (26). In a subset of NBF mutations, residual channel activity is still present due to a partial MgADP response (23), and, as a consequence, the associated hyperinsulinism phenotype is generally milder than that associated with complete loss of function (22) and can be treated by KATP channel-opening (KCO) drugs that increase the residual KATP activity (see below).

Kir6.2 and SUR1 Loss-of-Function Mutations Cause Neonatal Diabetes and DEND Syndrome

Conversely, more than 70 gain-of-function (GOF) mutations in KCNJ11 and ABCC8 have now been associated with the corollary disorder, neonatal diabetes mellitus (NDM; MIM 606176) (2730). Typically diagnosed soon after birth, with low birth weight, marked hyperglycemia, and ketoacidosis, NDM is a rare disorder with incidence of up to approximately 1 in 90,000 live births (31). It can be permanent (PNDM), requiring antidiabetic treatment for life, or a milder transient form (TNDM) in which hyperglycemia usually resolves within the first two years but can later relapse. The exact mechanisms underlying TNDM remission are unclear but may reflect a secondary increase in insulin sensitivity, compensatory increase in β cell function, or both (see below). In the most extreme KATP-dependent PNDM cases, patients also exhibit developmental delay in motor and intellectual skills, epilepsy, and neonatal diabetes (DEND) syndrome or intermediate DEND without epilepsy (32, 33), reflecting KATP overactivity outside the pancreas (34).

In the pancreas, elevated glucose raises the [ATP]:[ADP] ratio, normally closing Kir6.2/SUR1-dependent KATP channels and triggering action potential firing and insulin secretion. Overactive KATP channels suppress electrical activity, and as a result, insulin secretion is inhibited and glucose rises (3537). Kir6.2 and SUR1 are also widely expressed in the hippocampus and cerebellum as well as the midbrain and brainstem (38). Transgenic mice overexpressing the Kir6.2[V59M] DEND mutation in either muscle or nerve indicate that motor impairments originate in the central nervous system (39). Hippocampus-specific KATP-GOF transgenic mice exhibit mostly learning and memory deficiencies (40), pointing to a specific hippocampal involvement, but new models and approaches will be needed to fully define the origins of nonpancreatic DEND features.

All functionally assessed mutations result in enhanced channel activity at any given [ATP]:[ADP] ratio by either reducing ATP inhibition (4143) or increasing sensitivity to stimulatory Mg2+ adenosine (28, 42, 44). Residues located in or near the inhibitory sites on Kir6.2 reduce ATP binding directly (2). Mutations outside the ATP-binding site, or in the SUR1 TMD0 domain, increase channel open state stability, allosterically reducing ATP sensitivity (41, 45). Since ATP interacts predominantly with the closed state of the channel (9), access to the binding site is reduced when the open state is favored, and as a result, ATP sensitivity is decreased. Regardless of the molecular mechanism, disease severity generally correlates with the magnitude of channel overactivity (42, 45, 46). The association of DEND with more severe activating mutations suggests that extrapancreatic tissues are less sensitive to changes in KATP activity than the pancreatic β cell and that a threshold of channel overactivity must be reached before neuronal firing rates are suppressed and neurological features become evident.

Interestingly, phenotypic heterogeneity has been reported within families carrying a single NDM-causing KATP mutation. In a pedigree with the activating Kir6.2[C42R] mutation, cross-generational phenotypes of affected family members ranged from PNDM and gestational diabetes to late-onset type 2 diabetes mellitus (T2DM) (47). Similarly, in several pedigrees with SUR1 mutations, carriers were variously unaffected, type 2 diabetic, or neonatally diabetic (28, 48, 49). Such findings suggest that the severity of the β cell defect in KATP-induced NDM can be modulated by additional, unknown genetic and/or environmental factors that integrate to determine the clinical presentation.

Kir6.1 and SUR2 Gain-of-Function Mutations Cause Cantú Syndrome

First described as a distinct syndrome in 1982 (50), Cantú syndrome (CS; MIM 239850) is a multiorgan disorder characterized by congenital hypertrichosis, distinctive facial features, osteochondrodysplasia, multiple cardiovascular features (51), and potentially lethal pulmonary hypertension and bronchopulmonary dysplasia (52). Genetically associated with ABCC9 in 2012 (53, 54) and with KCNJ8 in 2014 (55, 56), over 30 mutations have since been identified in more than 100 patients (51, 52, 5762). All assessed mutations lead to recombinant KATP channel GOF (56, 63, 64). Similar to NDM mutations in SUR1 and Kir6.2, the underlying mechanisms include decreased ATP inhibition and enhanced MgADP activation (6365).

The recognition that essentially identical CS features are seen with KCNJ8 or ABCC9 mutations (56, 64) definitively ties the syndrome to Kir6.1/SUR2 channel overactivity but does not immediately explain the pathophysiologic consequences. Smooth muscle (SM) KATP is predominantly formed of SUR2B/Kir6.1 subunits (66), and recent animal experiments have led to a mechanistic framework for understanding many CS features as a consequence of SM dysfunction. Hypotension, vascular dilation, and twisted blood vessels, as well as cardiomegaly, are reiterated in Cantú mice, in which CS-associated SUR2[A478V] and Kir6.1[V65M] mutations were introduced to the equivalent mouse loci (67). Enhanced basal KATP conductance in vascular smooth muscle (VSM) provides a ready explanation for vasodilation and hence reduced systemic vascular resistance (SVR), leading to lower blood pressure (BP).

Striated muscle KATP channels may have various subunit combinations, with the noncanonical SUR2A/Kir6.2 being prominent in both heart and skeletal muscle. Consistent with cardiac channels being formed of SUR2 and Kir6.2 (but not Kir6.1), the ATP sensitivity of the ventricular myocyte KATP channel is reduced in SUR2[A478V] but normal in Kir6.1[V65M] mice. However, cardiac enlargement is even greater in the latter (67) and must therefore arise as a secondary consequence of KATP GOF in other cells. Decreased SVR in these animals is accompanied by elevated renin-angiotensin signaling (RAS), and cardiac enlargement can be reversed by blockade of RAS signaling (68) or by dominant-negative suppression of SM KATP (69), indicating that cardiac remodeling is a secondary consequence of the vascular GOF. It is accompanied by expression of genes associated with pathological hypertrophy and by decreased exercise tolerance, suggestive of reduced cardiac reserve (68). Longitudinal studies of CS patients (G.K. Singh, C. McClenaghan, M. Aggarwal, H. Gu, M.S. Remedi, et al., unpublished data) suggest that the long-term consequence of this reduced reserve is the gradual development of heart failure, a potentially important therapeutic issue.

Muscular fatigue is a common experience in CS patients (51). Consistent with skeletal muscle KATP being primarily composed of Kir6.2/SUR1, there is also a KATP channel GOF in flexor digitorum brevis myocytes from SUR2[A478V] mice (70) but not from Kir6.1[V65M] mice (71). However, reduced forelimb muscle strength, with evidence of atrophy and noninflammatory edema, is observed in both genotypes (70, 71), paralleling the finding of reduced channel ATP sensitivity in cardiac myocytes from A478V but not V65M, yet pathologic enlargement and hypercontractility in both (67). RAS is also a known mediator of atrophy and sarcopenia in skeletal muscle (72) and is potentially a major driver of the muscle pathology, but further study is needed.

Reduced SM contractility might underlie other CS features, including hypertrichosis. By opening surface blood vessels, more oxygen and nutrients are directed to hair follicles, potentially causing follicles in the telogen phase to shed and be replaced by thicker hairs in a new anagen phase (73). Kir6.1/SUR2 subunits underlie KATP channels in SM throughout the gastrointestinal (GI) tract (74), and GI dysmotility is also common in CS (51). Cantú mice exhibit reduced intrinsic contractility throughout the intestine, resulting in death in the most severely affected homozygous Kir6.1[V65M] animals when weaned onto solid food (74). Death is avoided by weaning onto a liquid gel diet, and GI transit is normalized by treatment with the KATP inhibitor glibenclamide (74). Kir6.1 and SUR2B are also expressed in mouse lymphatic SM (7577), but Kir6.1 alone is expressed in lymphatic endothelium (75). Spontaneous popliteal lymphatic vessel contraction is inhibited by SM-specific expression of a Kir6.1 GOF subunit, but it is essentially unaffected by expression in lymphatic endothelium (75), suggesting that KATP GOF in lymphatic SM also underlies lymphedema in CS.

Potential GOF mutations in KCNJ8 and ABCC9 have been reported in association with other pathologies. We recently examined a de novo case of idiopathic lymphedema associated with a SUR2 mutation. Patch-clamp analysis showed this mutation to generate a marked GOF in recombinant KATP channels, and retrospective analysis led to a clear rediagnosis of the patient as suffering from CS (J. Gao, C. McClenaghan, I. Christiaans, M. Alders, K. van Duinen, et al., unpublished data). SUR2[V734I], reported to reduce MgATP sensitivity in mutant Kir6.2/SUR2B channels but not in Kir6.2/SUR2A or Kir6.1/SUR2B channels, was detected in patients with myocardial infarction (78, 79) and in patients with cardiac early repolarization syndrome (ERS) (80). Whether these patients might also actually represent a mild CS genotype is unclear. Kir6.1[S422L] has also been reported to underlie ERS (81), and some studies suggest that a recombinant KATP channel GOF may also exist (8284). However, we could not demonstrate any such effect (56), and the recognition that this variant is common in Ashkenazim (85) suggests no causal association with early repolarization, highlighting the need for careful functional characterization and replication of potential pathological associations.

SUR2 Loss-of-Function Mutation Causes AIMS

LOF mutations in ABCC9 and KCNJ8 have been reported in association with various pathologies, including idiopathic dilated cardiomyopathy and rhythm disturbances (86), predisposition to adrenergic atrial fibrillation (87), and sudden unexplained nocturnal death syndrome (88, 89). However, there is as yet no fully established pathology arising from Kir6.1 or SUR2 LOF. We recently identified six patients from two families with a consistent phenotype of mild intellectual disability, similar facial appearance, myopathy, and cerebral white matter hyperintensities and with cardiac systolic dysfunction in the oldest patients (90). The patients are homozygous for an ABCC9 variant that causes in-frame deletion of SUR2 exon 8, which generates nonfunctional KATP channels in recombinant assays (90). There is similar fatigability and cardiac dysfunction in SUR2−/− mice and zebrafish. We termed the condition ABCC9-related intellectual disability myopathy syndrome [AIMS; also referred to as intellectual disability and myopathy syndrome (IDMYS); MIM 619719]. Further studies are needed, but we suggest that AIMS represents the primary ABCC9 LOF syndrome.

Association of KATP Genes with More Common Diseases: Type 2 Diabetes and Pulmonary Hypertension

With a high allelic frequency (>30%), the common Kir6.2[E23K] variant is a prominent risk allele for T2DM (91, 92). Recombinant functional analyses consistently report slightly enhanced channel activity due to Kir6.2[E23K] (91, 93) or to SUR1[S1369A], which exists in linkage dis-equilibrium (94). As in NDM, variants that increase KATP activity are predicted to impair β cell response and decrease serum insulin levels. The variant is associated with reduced insulin secretion in humans (93, 95), and in the presence of diet-induced insulin resistance, this may be expected to exacerbate hyperglycemia. Homozygous Kir6.2[E23K] knock-in mice (96) show no effect on body weight or blood glucose, and insignificant effects on insulin secretion, on a normal diet. On a high-fat diet, there is a trend toward greater weight gain and worsened glucose tolerance in E23K females yet enhancement of insulin secretion (96). Thus, it is still not clear exactly how the variant predisposes individuals to T2DM nor, interestingly, why E23K carriers with T2DM appear to be at increased risk of secondary failure to sulfonylurea (SU) treatment (97).

SUR1 is also a regulatory subunit of KATP channels in mouse atria (98) and of ventricular KATP in humans (99). It may also be prominent in pulmonary arteries (100), and we recently reported multiple SUR1 LOF variants, some previously associated with CHI, in patients with pulmonary arterial hypertension (101). Whether these variants are causal, and how pulmonary hypertension might be linked to SUR1 LOF, but also to SUR2 GOF in CS, is not clear. Potentially, the precise KATP subunit composition in any given cell type may vary subtly or be more labile than is currently perceived, making it critical to focus on precise subunit distributions, and further detailed comparative analysis of cardiovascular phenotypes and outcomes in SUR1−/− and SUR2−/− mice may be warranted.

KATP CHANNEL PHARMACOLOGY

KATP Channel Inhibitors

The unique involvement of SUR subunits in KATP channel complexes underlies a rich pharmacology specific to this class of channels. This includes the SU class of inhibitors, the first-generation tolbutamide being introduced clinically in 1952 before the existence of KATP channels was even recognized (102). Increasingly potent and longer-acting SUs [gliclazide, glipizide, glibenclamide (glyburide), glimepiride], as well as a new structural class [glinides (metiglinide, nateglinide, and repaglinide)], have since been introduced (103).

SU inhibition of KATP channel activity depends in a complex way on cellular nucleotide levels (65, 104). Inhibition is also typically biphasic, reflecting two inhibitory binding sites (104). High-affinity binding at the SUR, which underlies clinical action, is transduced allosterically to closure at the Kir6 subunit, and the fraction of high- versus low-affinity inhibition depends critically on the open state stability of the channel, that is, the stability of the open versus closed state, which is in turn affected by the membrane PIP2 level (9). Open state stability can also be markedly affected by mutations throughout Kir6 subunits and the SUR TMD0 domains. As a consequence, SU inhibition of Kir6.2/SUR1-dependent NDM mutations (41) and Kir6.1/SUR2-dependent CS mutations (64, 65) can be markedly reduced (Figure 2a), an important consideration for their use in these conditions and in the search for novel agents.

Figure 2.

Figure 2

Sulfonylurea inhibition of KATP channels. (a) Kir6.2/sulfonylurea receptor subunit 1 (SUR1)-dependent KATP channel activity as a function of [tolbutamide], recorded in inside-out patches, shows biphasic inhibition. Dashed lines and blue circles show wild-type (WT) channel activity. Tolbutamide sensitivity is unaffected by transient neonatal diabetes mellitus (NDM) mutant Kir6.2[I182V] but is reduced by permanent NDM mutants V59M and Q52R and abolished by the developmental delay, epilepsy, and neonatal diabetes (DEND) syndrome mutation Kir6.2[I296L] (significance indicated: *P < 0.05 and **P < 0.01). Even in the heterozygous condition (mixed wild type and I296L), high-affinity tolbutamide sensitivity is markedly reduced. Panel a adapted from Reference 41. (b) A structural model of the repaglinide (RPG)-bound SUR1 ATP-binding cassette transporter core module viewed from the side shows the slice viewed from the top and side (indicated by the two gray lines) at higher magnification to the right. The pharmacochaperone pocket is shown from the top and the side of the channel in the states indicated. Ligand density corresponding to RPG is shown in magenta, carbamazepine (CBZ) in red, and glibenclamide (GBC) in blue. (c) The Kir6.2 N-terminal cryo-electron microscopy density (pink mesh) is superposed with the polyalanine model shown in the RPG-bound (green) SUR1 structural model. The piperidino moiety of RPG is highlighted to show close proximity to the N-terminal methionine of the modeled Kir6.2 N-terminal peptide. The close proximity of SUR1 residue C1142 and Kir6.2 residue L2 is also highlighted. Panels b and c adapted with permission from Reference 105.

Cryo-EM studies reveal a common binding site for SUs and other KATP channel inhibitors (3, 105107) between TMD1 and TMD2 of SUR1 (Figure 2b). Glibenclamide binding, involving two overlapping, so-called A and B sites that bridge TMD1 and TMD2, likely prevents NBF dimerization, explaining its inhibitory action (105, 108). The chlorobenzamidoethyl group interacts with the B site formed by residues on transmembrane helices 6–8 and 11, while the cyclo-hexyl moiety binds to the hydrophobic A site pocket formed by residues on transmembrane helices 16 and 17 (Figure 2b), in close proximity to S1238 in helix 16. SUR1-dependent channels are generally more sensitive to SUs than are SUR2-dependent channels, and interestingly, the equivalent residue in SUR2 is a more bulky Tyr. Substituting these residues converts high-affinity inhibition of SUR1-dependent channels to a lower SUR2-like sensitivity (109) and increases the affinity of binding and potency of inhibition of SUR2B-dependent channels (110). Deletion of up to 30 amino acids from the Kir6.2 N terminus intrinsically stabilizes the open channel, thereby reducing SU action (104, 111). Potentially explaining this finding, additional cryo-EM structures have revealed a previously unimagined insertion of the full Kir6.2 N terminus into the central cavity of SUR1, between TM1 and TMD2, adjacent to the SU-binding pocket (Figure 2c). In so doing, this potentially stabilizes bound SUs and increases inhibitory potency (105107).

KATP Channel Openers

Conversely, structurally diverse benzopyrans, benzothiadiazines, cyanoguanidines, pyridyl nitrates, and thioformamides and second-generation cyclobutenediones, dihydropyridines, and tertiary carbinols (112), collectively referred to as KCOs, activate KATP channels. Mutagenesis studies suggest a common KCO binding site (113115), with two regions (between Thr1059-Leu1087 and Arg1218-Asn1320 in TMD2 of SUR2) being critical for binding of the KCO P1075 (114). The first cryo-EM structure with a KCO (NN414) bound to Kir6.2/SUR1 channels (8) (Figure 3a) reveals the ligand in the cleft between TMD1 and TMD2, in contact with residues I1030, D1031, C1072, and T1286 of TMD2 as well as residues P551-V555, H584, and L580 in TMD1, confirming key findings of earlier studies. A recent paper places both the P1075 and levcromakalim activators in essentially the same site (116). As with inhibitory SUs, KCO potency and selectivity are modulated by channel open state stability and by the interactions of Mg nucleotides at NBFs (104, 117), and as discussed below, it is likely that KCO sensitivities are also strongly affected by disease mutations.

Figure 3.

Figure 3

KCO action and crossover drug-induced pathologies in KATP channels. (a, left) Cryo- electron microscopy density map shows SUR1 in complex with Mg nucleotides and NN414 (viewed from the side). TMD1-NBD1, TMD2-NBD2, and NN414 are colored in pink, blue, and red, respectively. (Right) Close-up views of the NN414-binding site. TMD1 and TMD2 are colored in pink and blue, respectively. NN414 (orange) and residues that interact with NN414 are shown as sticks. Panel a adapted with permission from Reference 8. (b) Canonical KATP channels are formed by the products of the ABCC8/KCNJ11 and ABCC9/KCNJ8 gene pairs. LOF mutations in either subunit of each pair (causing CHI and AIMS, respectively) and GOF mutations (causing NDM and Cantú syndrome, respectively) may be treatable with channel-activating KCOs or inhibitory SUs. However, crossover effects on the unaffected channels encoded by the other gene pair cause inevitable side effects. (c) Hypertrichosis and fluid retention characterize both Cantú syndrome, caused by a Kir6.1/SUR2 GOF mutation, and CHI when treated with diazoxide. Images in panel c reproduced with permission from References 53 and 179. Abbreviations: AIMS, ABCC9-related intellectual disability myopathy syndrome; CHI, congenital hyperinsulinism; GI, gastrointestinal; GOF, gain-of-function; KATP, ATP-sensitive potassium; KCO, KATP channel opener; Kir, pore-forming inward rectifier potassium channel subunit; LOF, loss-of-function; NBD, nucleotide-binding domain; NDM, neonatal diabetes mellitus; SU, sulfonylurea; SUR, sulfonylurea receptor subunit; SVR, systemic vascular resistance; TM, transmembrane; TMD, transmembrane domain.

MONOGENIC KATP DISEASE THERAPIES

Current Pharmacological Therapies for Congenital Hyperinsulinism

First sold as Hyperstat, for emergent hypertension, diazoxide was recognized 60 years ago as inducing VSM relaxation and hence blood vessel vasodilation (118). Almost as long ago, it was realized that it also inhibited insulin secretion from pancreatic β cells (119). Some CHI patients with KATP LOF mutations must maintain some KATP channel activity since they are responsive to diazoxide (120), which (sold as Proglycem) is now the preferred treatment for responsive CHI patients (121, 122). Other KATP-dependent CHI cases completely lack KATP channels. In these and other cases that are unresponsive to diazoxide (or other drugs, including chlorothiazide, nifedipine, glucagon, and somatostatin, all of which aim to suppress insulin secretion), partial or total pancreatectomy is generally required (121). Surgical resection is also effective in patients with focal hyperinsulinism once the lesion has been preoperatively located (123). Importantly, in nonsurgically treated KATP-dependent and non-KATP-dependent CHI, hyperinsulinism symptoms typically lessen with age and diazoxide may no longer be needed (124127). One study found that in adult carriers of dominant KATP mutations, all of whom would be expected to have suffered hyperinsulinism as children, essentially half were symptomless (potentially indicating a preceding remission), and several were diabetic (22). Multiple other studies report CHI patients with dominant ABCC8 LOF mutations who remitted and then progressed to diabetes later in life (23, 125, 128130).

Partial loss of KATP in mice causes hyperinsulinism (131, 132), but mice with complete loss of KATP cross over to undersecretion and glucose intolerance (133135), and overt diabetes after being fed a high-fat diet (136). Potentially this crossover reflects the same progression as that seen in human CHI. Strikingly, SUR1[E1506K] knock-in mice, carrying the same mutation as a diabetes-prone Finnish CHI pedigree (23), show hyperinsulinism in the neonatal period but again rapidly progress to undersecretion (137). Also seen in patients with CHI caused by GOF mutations in glucokinase (GCK), which lead to an elevated ATP:ADP ratio and consequent suppression of KATP activity (138, 139), this progression from hyperinsulinism to diabetes is unexplained but needs to be understood for appropriate long-term treatment and management of CHI.

Current Pharmacological Therapies for Neonatal Diabetes Mellitus

The realization that KATP channel GOF mutations cause NDM rapidly shifted therapy from insulin injections to oral SUs. By directly inhibiting overactive KATP channels, these drugs can provide successful control of blood glucose levels, removing (or markedly reducing) insulin requirements and significantly improving glycemic control and overall lifestyle for most KATP-induced NDM patients (140, 141). Unlike injected insulin, which indiscriminately lowers blood glucose by promoting peripheral uptake, SUs directly target KATP overactivity, and insulin secretion becomes quasi-physiological, with much better glucose control than when insulin treated (142).

However, the many mutations that reduce both ATP and SU sensitivity (41) contribute to SU dose requirements being generally higher than those for treatment of T2DM (140, 143), and the success rate for SU treatment is lower in patients with the more severe DEND symptoms (140, 144147). Moreover, while SUs can resolve diabetes and improve neurological function in some DEND cases (32, 142, 148), neurologic features are unresponsive in most. SUs are transported out of the brain by the ABC protein P-glycoprotein, potentially lowering central nervous system (CNS) concentrations (149, 150). Together with the marked loss of SU sensitivity in many DEND mutations (41, 151) (Figure 2a), this concentration lowering in the CNS provides an explanation for variable SU response in DEND. As discussed below, new agents with better brain-penetrating abilities, avoiding P-glycoprotein export, as well as new mechanisms of action, are needed to resolve this issue.

There is a correlation between the age at which SU transfer is attempted in human NDM and overall β cell responsiveness: The younger the patient, the greater the chance for successful SU therapy (140, 143). One case report showed that the number of β cells in an insulin-treated 2-year-old female with a KCNJ11 mutation was significantly lower than in normal age-matched children (152), and loss of β cell mass is also consistently observed in diabetic pancreatic KATP GOF mice (36, 37). This loss of β-cell mass is associated with dedifferentiation to progenitor cells (153), but importantly, these dedifferentiated cells can redifferentiate to mature insulin-secreting β cells after normalization of blood glucose by intensive insulin therapy (153). If initial hyperglycemia is avoided, either by early SU therapy or by syngeneic islet transplantation, loss of β cell mass is prevented (36), and acute SU treatment at the onset of transgene induction in a subset of pancreatic KATP GOF mice actually led to persistently near-normal glucose levels once the SU therapy was terminated (154). Conceivably, this normalization could reflect the mechanistic progression of human TNDM, wherein early avoidance of hyperglycemia might allow a long-term compensatory mechanism to provide remission. While unexplained, the finding predicts that early intervention with SUs in KATP-dependent NDM may help preserve β cell function. Retrospective study shows that initiation of SU therapy before genetic testing results are available is generally safe and often successful in NDM patients (155), supporting aggressive early SU intervention.

One potential caveat in the undoubted success of SU therapy for NDM is that, by analogy to the progressive loss of SU responsiveness seen in T2DM (156), SU treatment might eventually fail, in which case insulin therapy may be needed. Experience to date has not borne this out (140, 157, 158), suggesting that SU treatment for NDM—which is intended to normalize insulin secretion, not hyperstimulate it—may be safe and effective in the long-term.

Therapies for Cantú Syndrome and AIMS

Reduced vascular contractility probably underlies the common finding in CS of persistent patent ductus arteriosus (51), often requiring surgical closure. Pericardial effusion sometimes requires pericardiocentesis and, ultimately, pericardial stripping to prevent reaccumulation. High incidence of cerebrovascular abnormalities in CS (159) may lead to magnetic resonance angiograms to evaluate persistent headaches or other neurological symptoms becoming standard of care. Treatments for hypertrichosis include shaving, use of depilatories, or laser hair removal. CS patients also require monitoring for edema, which often develops in adolescence or early adulthood and may benefit from compression stockings. Other than these generally palliative treatments, there is currently no specific therapy for CS. Some treatments are essential and effective, others are incompletely effective, and some currently prescribed cardiac agents (51) such as angiotensin-converting enzyme inhibitors or β-blockers may even be contraindicated: Captopril causes extreme BP lowering in CS mice (68), and propranolol causes significant cardiac contractile inhibition (160).

Clearly, the use of appropriate KATP channel inhibitors that could reverse the underlying molecular defect is a potential magic-bullet therapy, as SUs have proven to be for NDM. In SUR2[A478V] mice, chronic glibenclamide treatment (1-month slow-release pellets), initiated at 2 months of age, does substantially reverse cardiac enlargement and normalize BP, although it fails to reverse aortic enlargement, and normalization of cardiac size and BP is incomplete in the more severely affected V65M mice (69). In one case study, glibenclamide did appear to be beneficial in a CS child with SUR2[R1116H] (161). At the time of writing, the child is 5 years old and still on the same glibenclamide dose, without overt cardiac or other complications (A. Ma, personal communication) but with no resolution of hypertrichosis. Additional unpublished efforts in infants thus far have not shown obvious symptom improvement. Nevertheless, the US Food and Drug Administration (FDA)-approved status of SUs, the availability of validated animals in which to assess their action (67), and the availability of a cohort of motivated CS patients in whom to assess clinical outcomes (51) justify an urgent need to examine the clinical potential for SU use in CS, and to this end, we are currently carrying out initial glibenclamide tolerance tests in older CS patients.

Unfortunately, all commercially approved and available KATP channel inhibitors are also active on Kir6.2/SUR1 channels, and while it seems likely that cardiovascular, cerebrovascular, and lymphatic dysfunction in human CS will all respond to SUs, there is a risk of hypoglycemia, as discussed below.

Crossover drug-induced pathologies.

As discussed above, current therapies for KATP pathologies rely on reversing channel GOF or LOF mutations by using SU inhibitors or KCOs, respectively. In each case, the target is the mutated Kir6.2/SUR1 or Kir6.1/SUR2 channel pair, but the poor selectivity of currently available agents will inevitably have an undesired action on the unaffected channel pair (Figure 3b).

KATP channel openers.

This crossover action is clearly a problem for diazoxide treatment of CHI, with extensive documentation of complications, most notably hypertrichosis but also the development of abnormal facial features, low BP, and edema, as well as potentially lethal respiratory failure and pulmonary hypertension (162), as seen in CS (52), prompting a black box warning by the FDA in 2015. Suspected necrotizing enterocolitis with abdominal distension (which resolved after discontinuation of diazoxide) has also been reported in both diazoxide treatment (162) and CS (52).

Marketed as Rogaine in the United States, and Regaine in Europe, the KCO minoxidil is a popular over-the-counter treatment for hair loss. The mechanism of action remains unclear (73), but since it is an effective opener of Kir6.1/SUR2B channels, the association of Kir6.1/SUR2 GOF mutations with hypertrichosis in CS definitively ties hair growth to KATP channel activation. Since minoxidil preferentially acts on SUR2-dependent channels, blood-glucose lowering is not generally reported as a side effect, but other drug-induced Cantú features similar to those seen with diazoxide therapy are common and have led to very severe problems in people using the drug for hair growth (73) and even in their pets after accidental ingestion (163).

KATP channel inhibitors.

Since Kir6.2/SUR1 channels are generally more sensitive to glibenclamide than are Kir6.1/SUR2 channels, side effects of glibenclamide therapy for NDM are generally minimal, although hypersensitivity of the skin, stomach upset, and diarrhea can be common (164). It is tempting to suggest that the latter results from inhibition of KATP channels in the GI SM, given that glibenclamide markedly increases motility in Cantú animals (74). More significantly, the higher sensitivity of Kir6.2/SUR1 channels to SUs presents the potential for severe blood-glucose lowering if SUs are used to treat CS. As discussed above, the glucose-lowering effect of high-dose SU is only transient in mice, and then the animals rapidly lose glucose-sensitive insulin secretion, resulting in slight glucose intolerance, as in KATP knockout animals (165). Whether the same occurs in humans is unclear. The evidence from treatment of NDM, in which the SU dose is adjusted to provide pseudonormal excitability and insulin secretion, indicates that there is a sustained effect. However, in T2DM, where SU treatment promotes supraphysiological insulin secretion by inducing supranormal excitability, the drugs gradually become ineffective. SU sensitivity can be restored, however, by so-called β cell rest with intensive insulin therapy, conceivably reversing the same desensitization that occurs in mice with high-dose SU treatment. If so, then the same desensitization of the otherwise normal pancreas might occur if SU is used at high doses to reverse CS features. Intriguingly, in the single reported human CS case treated with glibenclamide, transient hypoglycemia was observed with each dose escalation (161), suggesting the concern may not be preclusive.

POTENTIAL FOR NEW THERAPEUTICS

Crossover drug-induced pathologies, compounded by reduced sensitivity to channel inhibitors with GOF mutations, highlight a need for new inhibitors and new activators for KATP and especially a need for sharper therapeutics (103) with greater isoform specificity.

New Small Molecules

High-throughput screening may identify novel or more potent inhibitors or KCOs acting through the identified binding sites in the SUR cores (8), but the structural similarity of these sites in SUR1 and SUR2 may ultimately preclude true isoform specificity, making crossover drug-induced complications unavoidable. One potential lead is the non-SU inhibitor U37883A, which has prohibitive off-target effects (166) but does have a novel site of action on Kir6.1 (167). To overcome the potential that new inhibitors may still be ineffective because of open state stabilization with GOF channel mutations, Kharade et al. (103) proposed to identify KATP channel correctors by screening chemical libraries using functional assays with disease-causing mutant channels. With the use of rapid cloning techniques, including landing-pad cells, in which a stably integrated cloning site has been genomically incorporated (168), it is now feasible to rapidly develop the multiple necessary cell lines (169). The use of tetracycline induction of the active channel minimizes cell line degeneration and loss of assay performance associated with constitutive KATP channel overexpression, making it possible to repeatedly assay the same lines (170).

As initially implemented, fluorescent assays are performed in multiwell plates using a standard kinetic imaging plate reader to measure the flux of thallium (Tl+) as a K+ surrogate with a Tl+-sensitive dye (171). Both Kir6.2/SUR1 and Kir6.1/SUR2B channels are mostly closed under basal physiologic conditions due to the relatively high cytoplasmic [ATP]:[ADP] ratio in HEK-293 cells, and channels must first be opened with metabolic inhibitors or a KCO. However, as we have recently shown (169), voltage-sensitive DiBAC4(3) [bis-(1,3-dibutylbarbituric acid)trimethine oxonol] fluorescence provides a very sensitive assay for the hyperpolarizing effect of even a very low basal KATP conductance. Such an approach makes high-throughput screening for viable inhibitors of Kir6.2/SUR1 or Kir6.1/SUR2B channels with GOF mutations possible under basal physiological conditions. Screening for activators can be achieved by simply leaving the channels closed and looking for compounds that induce Tl+ flux. A novel-scaffold xanthine derivative (VU063), discovered using this approach, is an effective KCO that acts on Kir6.2/SUR1 channels (171), is approximately 10 times more potent than diazoxide, and is an effective inhibitor of glucose-stimulated calcium entry in primary mouse pancreatic β cells.

New Biological Therapeutics

Advances in gene editing may achieve the ultimate treatment for monogenic pathologies—correction of the underlying mutations (172). In the meantime, new biological agents, particularly targeted antibodies and nanobodies, are revolutionizing disease therapeutics. Facilitated by unique cryo-EM-identified structural motifs, such agents may be ideal for achieving subunit specificity. For instance, the unique structure of the Kir6.1 extracellular turret region has now been resolved (11) and might represent a targetable Kir6.1-specific domain. Conceptually, an inhibitor acting on the channel from the extracellular side might also avoid the issue of drug interactions with nucleotide modulation.

A natural venom screen recently identified a novel 54-residue protein toxin (SpTx-1) that inhibits both wild-type and NDM mutant Kir6.2/SUR1 channels with low nanomolar potency from the extracellular side (173). Intriguingly, toxin sensitivity was dependent on human-specific residue E108 (174), and in KATP GOF mice expressing humanized Kir6.2[ΔN30, K185Q, V108E], the toxin was effective at controlling blood sugar levels (174), a promising indicator that such an approach may provide another avenue to identifying novel channel inhibitors. High-resolution structures also open up the possibility of developing new modulators using in silico approaches. Molecular dynamics simulations of DEND syndrome Kir6.2[Q52R] and [L164P] mutations recently identified betaxolol and levobetaxolol as effective pore blockers, as well as tavoprost, a SUR-binding prostaglandin with potencies in the micromolar range (175).

Finally, both SUs and KCOs can rescue surface expression of mistrafficked mutant channels in vitro (176178), raising the possibility of molecular chaperone drugs as a novel therapy to treat trafficking defects in HI. Since SUs present a Catch-22 situation by inhibiting channels even as they increase trafficking, translation to a viable therapy may be difficult, but it does illustrate the concept that there may be additional routes to modifying channel activity.

ACKNOWLEDGMENTS

I am very grateful to the numerous colleagues and collaborators that have contributed to work in this area. My own research is supported by National Institutes of Health grant R35 HL140024.

Footnotes

DISCLOSURE STATEMENT

The author is not aware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the objectivity of this review.

LITERATURE CITED

  • 1.Nichols CG. 2006. KATP channels as molecular sensors of cellular metabolism. Nature 440:471–76 [DOI] [PubMed] [Google Scholar]
  • 2.Martin GM, Yoshioka C, Rex EA, Fay JF, Xie Q, et al. 2017. Cryo-EM structure of the ATP-sensitive potassium channel illuminates mechanisms of assembly and gating. eLife 6:e24149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Martin GM, Kandasamy B, DiMaio F, Yoshioka C, Shyng SL. 2017. Anti-diabetic drug binding site in a mammalian KATP channel revealed by Cryo-EM. eLife 6:e31054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Lee KPK, Chen J, MacKinnon R. 2017. Molecular structure of human KATP in complex with ATP and ADP. eLife 6:e32481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Li N, Wu JX, Ding D, Cheng J, Gao N, et al. 2017. Structure of a pancreatic ATP-sensitive potassium channel. Cell 168:101–110.e10 [DOI] [PubMed] [Google Scholar]
  • 6.Shyng S, Nichols CG. 1997. Octameric stoichiometry of the KATP channel complex. J. Gen. Physiol 110:655–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zhao C, MacKinnon R. 2021. Molecular structure of an open human KATP channel. PNAS 118:e2112267118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wang M, Wu J-X, Ding D, Duan X, Ma S, et al. 2022. Structural insights into the mechanism of nucleotide regulation of pancreatic KATP channel. bioRxiv 2021.11.29.470334. 10.1101/2021.11.29.470334 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Enkvetchakul D, Nichols CG. 2003. Gating mechanism of KATP channels: Function fits form. J. Gen. Physiol 122:471–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Babenko AP, Bryan J. 2003. SUR domains that associate with and gate KATP pores define a novel gatekeeper. J. Biol. Chem 278:41577–80 [DOI] [PubMed] [Google Scholar]
  • 11.Sung MW, Yang Z, Driggers CM, Patton BL, Mostofian B, et al. 2021. Vascular KATP channel structural dynamics reveal regulatory mechanism by Mg-nucleotides. PNAS 118:e2109441118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Quinn KV, Cui Y, Giblin JP, Clapp LH, Tinker A. 2003. Do anionic phospholipids serve as cofactors or second messengers for the regulation of activity of cloned ATP-sensitive K+ channels? Circ. Res 93:646–55 [DOI] [PubMed] [Google Scholar]
  • 13.Hu H, Sato T, Seharaseyon J, Liu Y, Johns DC, et al. 1999. Pharmacological and histochemical distinctions between molecularly defined sarcolemmal KATP channels and native cardiac mitochondrial KATP channels. Mol. Pharmacol 55:1000–5 [PubMed] [Google Scholar]
  • 14.Inagaki N, Gonoi T, Clement JP, Wang CZ, Aguilar-Bryan L, et al. 1996. A family of sulfonylurea receptors determines the pharmacological properties of ATP-sensitive K+ channels. Neuron 16:1011–17 [DOI] [PubMed] [Google Scholar]
  • 15.Cheng WWL, Tong A, Flagg TP, Nichols CG. 2008. Random assembly of SUR subunits in KATP channel complexes. Channels 2:34–38 [DOI] [PubMed] [Google Scholar]
  • 16.Chan KW, Wheeler A, Csanady L. 2008. Sulfonylurea receptors type 1 and 2A randomly assemble to form heteromeric KATP channels of mixed subunit composition. J. Gen. Physiol 131:43–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Pountney DJ, Sun ZQ, Porter LM, Nitabach MN, Nakamura TY, et al. 2001. Is the molecular composition of KATP channels more complex than originally thought? J. Mol. Cell. Cardiol 33:1541–46 [DOI] [PubMed] [Google Scholar]
  • 18.Flagg TP, Nichols CG. 2011. “Cardiac KATP”: a family of ion channels. Circ. Arrhythm. Electrophysiol 4:796–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Nichols CG, Shyng SL, Nestorowicz A, Glaser B, Clement JP, et al. 1996. Adenosine diphosphate as an intracellular regulator of insulin secretion. Science 272:1785–87 [DOI] [PubMed] [Google Scholar]
  • 20.Thomas PM, Cote GJ, Hallman DM, Mathew PM. 1995. Homozygosity mapping, to chromosome 11p, of the gene for familial persistent hyperinsulinemic hypoglycemia of infancy. Am. J. Hum. Genet 56:416–21 [PMC free article] [PubMed] [Google Scholar]
  • 21.Nestorowicz A, Wilson BA, Schoor KP, Inoue H, Glaser B, et al. 1996. Mutations in the sulonylurea receptor gene are associated with familial hyperinsulinism in Ashkenazi Jews. Hum. Mol. Genet 5:1813–22 [DOI] [PubMed] [Google Scholar]
  • 22.Pinney SE, MacMullen C, Becker S, Lin YW, Hanna C, et al. 2008. Clinical characteristics and biochemical mechanisms of congenital hyperinsulinism associated with dominant KATP channel mutations. J. Clin. Investig 118:2877–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Huopio H, Reimann F, Ashfield R, Komulainen J, Lenko HL, et al. 2000. Dominantly inherited hyperinsulinism caused by a mutation in the sulfonylurea receptor type 1. J. Clin. Investig 106:897–906 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Dunne MJ, West-Jordan JA, Abraham RJ, Edwards RHT, Peterson OH. 1988. The gating of nucleotide-sensitive K+ channels in insulin- secreting cells can be modulated by changes in the ratio ATP4−/ATP3− and by nonhydrolyzable derivatives of both ATP and ADP. J. Membr. Biol 104:165–77 [DOI] [PubMed] [Google Scholar]
  • 25.Saint-Martin C, Cauchois-Le Miere M, Rex E, Soukarieh O, Arnoux JB, et al. 2021. Functional characterization of ABCC8 variants of unknown significance based on bioinformatics predictions, splicing assays, and protein analyses: benefits for the accurate diagnosis of congenital hyperinsulinism. Hum. Mutat 42:408–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Shyng SL, Ferrigni T, Shepard JB, Nestorowicz A, Glaser B, et al. 1998. Functional analyses of novel mutations in the sulfonylurea receptor 1 associated with persistent hyperinsulinemic hypoglycemia of infancy. Diabetes 47:1145–51 [DOI] [PubMed] [Google Scholar]
  • 27.Gloyn AL, Pearson ER, Antcliff JF, Proks P, Bruining GJ, et al. 2004. Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. New Engl. J. Med 350:1838–49 [DOI] [PubMed] [Google Scholar]
  • 28.Babenko AP, Polak M, Cave H, Busiah K, Czernichow P, et al. 2006. Activating mutations in the ABCC8 gene in neonatal diabetes mellitus. N. Engl. J. Med 355:456–66 [DOI] [PubMed] [Google Scholar]
  • 29.Tornovsky S, Crane A, Cosgrove KE, Hussain K, Lavie J, et al. 2004. Hyperinsulinism of infancy: novel ABCC8 and KCNJ11 mutations and evidence for additional locus heterogeneity. J. Clin. Endocrinol. Metab 89:6224–34 [DOI] [PubMed] [Google Scholar]
  • 30.Pipatpolkai T, Usher S, Stansfeld PJ, Ashcroft FM. 2020. New insights into KATP channel gene mutations and neonatal diabetes mellitus. Nat. Rev. Endocrinol 16:378–93 [DOI] [PubMed] [Google Scholar]
  • 31.Iafusco D, Massa O, Pasquino B, Colombo C, Iughetti L, et al. 2012. Minimal incidence of neonatal/infancy onset diabetes in Italy is 1:90,000 live births. Acta Diabetol. 49:405–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Mlynarski W, Tarasov AI, Gach A, Girard CA, Pietrzak I, et al. 2007. Sulfonylurea improves CNS function in a case of intermediate DEND syndrome caused by a mutation in KCNJ11. Nat. Clin. Pract. Neurol 3:640–45 [DOI] [PubMed] [Google Scholar]
  • 33.Hattersley AT, Pearson ER. 2006. Minireview: pharmacogenetics and beyond: the interaction of therapeutic response, β-cell physiology, and genetics in diabetes. Endocrinology 147:2657–63 [DOI] [PubMed] [Google Scholar]
  • 34.Gloyn AL, Siddiqui J, Ellard S. 2006. Mutations in the genes encoding the pancreatic beta-cell KATP channel subunits Kir6.2 (KCNJ11) and SUR1 (ABCC8) in diabetes mellitus and hyperinsulinism. Hum. Mutat 27:220–31 [DOI] [PubMed] [Google Scholar]
  • 35.Koster JC, Marshall BA, Ensor N, Corbett JA, Nichols CG. 2000. Targeted overactivity of β cell KATP channels induces profound neonatal diabetes. Cell 100:645–54 [DOI] [PubMed] [Google Scholar]
  • 36.Remedi MS, Kurata HT, Scott A, Wunderlich FT, Rother E, et al. 2009. Secondary consequences of β cell inexcitability: identification and prevention in a murine model of KATP-induced neonatal diabetes mellitus. Cell Metab. 9:140–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Girard CA, Wunderlich FT, Shimomura K, Collins S, Kaizik S, et al. 2009. Expression of an activating mutation in the gene encoding the KATP channel subunit Kir6.2 in mouse pancreatic β cells recapitulates neonatal diabetes. J. Clin. Investig 119:80–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Karschin C, Ecke C, Ashcroft FM, Karschin A. 1997. Overlapping distribution of KATP channel-forming Kir6.2 subunit and the sulfonylurea receptor SUR1 in rodent brain. FEBS Lett. 401:59–64 [DOI] [PubMed] [Google Scholar]
  • 39.Clark RH, McTaggart JS, Webster R, Mannikko R, Iberl M, et al. 2010. Muscle dysfunction caused by a KATP channel mutation in neonatal diabetes is neuronal in origin. Science 329:458–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Yahil S, Wozniak DF, Yan Z, Mennerick S, Remedi MS. 2021. Cognitive deficits and impaired hippocampal long-term potentiation in KATP-induced DEND syndrome. PNAS 118:e2109721118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Koster JC, Remedi MS, Dao C, Nichols CG. 2005. ATP and sulfonylurea sensitivity of mutant ATP-sensitive K+ channels in neonatal diabetes: implications for pharmacogenomic therapy. Diabetes 54:2645–54 [DOI] [PubMed] [Google Scholar]
  • 42.Masia R, De Leon DD, MacMullen C, McKnight H, Stanley CA, et al. 2007. A mutation in the TMD0-L0 region of sulfonylurea receptor-1 (L225P) causes permanent neonatal diabetes mellitus (PNDM). Diabetes 56:1357–62 [DOI] [PubMed] [Google Scholar]
  • 43.Proks P, Antcliff JF, Lippiat J, Gloyn AL, Hattersley AT, et al. 2004. Molecular basis of Kir6.2 mutations associated with neonatal diabetes plus neurological features. PNAS 101:17539–544 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.de Wet H, Rees MG, Shimomura K, Aittoniemi J, Patch AM, et al. 2007. Increased ATPase activity produced by mutations at arginine-1380 in nucleotide-binding domain 2 of ABCC8 causes neonatal diabetes. PNAS 104:18988–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Proks P, Girard C, Ashcroft FM. 2005. Functional effects of KCNJ11 mutations causing neonatal diabetes: enhanced activation by MgATP. Hum. Mol. Genet 14:2717–26 [DOI] [PubMed] [Google Scholar]
  • 46.Tammaro P, Proks P, Ashcroft FM. 2006. Functional effects of naturally occurring KCNJ11 mutations causing neonatal diabetes on cloned cardiac KATP channels. J. Physiol 571:3–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Yorifuji T, Nagashima K, Kurokawa K, Kawai M, Oishi M, et al. 2005. The C42R mutation in the Kir6.2 (KCNJ11) gene as a cause of transient neonatal diabetes, childhood diabetes, or later-onset, apparently type 2 diabetes mellitus. J. Clin. Endocrinol. Metab 90:3174–78 [DOI] [PubMed] [Google Scholar]
  • 48.Flanagan SE, Patch AM, Mackay DJ, Edghill EL, Gloyn AL, et al. 2007. Mutations in ATP-sensitive K+ channel genes cause transient neonatal diabetes and permanent diabetes in childhood or adulthood. Diabetes 56:1930–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Patch AM, Flanagan SE, Boustred C, Hattersley AT, Ellard S. 2007. Mutations in the ABCC8 gene encoding the SUR1 subunit of the KATP channel cause transient neonatal diabetes, permanent neonatal diabetes or permanent diabetes diagnosed outside the neonatal period. Diabetes Obes. Metab 9(Suppl. 2):28–39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Cantú JM, Garcia-Cruz D, Sanchez-Corona J, Hernandez A, Nazar Z. 1982. A distinct osteochon-drodysplasia with hypertrichosis? Individualization of a probable autosomal recessive entity. Hum. Genet 60:36–41 [DOI] [PubMed] [Google Scholar]
  • 51.Grange DK, Roessler HI, McClenaghan C, Duran K, Shields K, et al. 2019. Cantú syndrome: findings from 74 patients in the International Cantú Syndrome Registry. Am. J. Med. Genet. C Semin. Med. Genet 181:658–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Park JY, Koo SH, Jung YJ, Lim YJ, Chung ML. 2014. A patient with Cantú syndrome associated with fatal bronchopulmonary dysplasia and pulmonary hypertension. Am. J. Med. Genet. A 164A:2118–20 [DOI] [PubMed] [Google Scholar]
  • 53.Harakalova M, van Harssel JJ, Terhal PA, van Lieshout S, Duran K, et al. 2012. Dominant missense mutations in ABCC9 cause Cantú syndrome. Nat. Genet 44:793–96 [DOI] [PubMed] [Google Scholar]
  • 54.van Bon BW, Gilissen C, Grange DK, Hennekam RC, Kayserili H, et al. 2012. Cantú syndrome is caused by mutations in ABCC9. Am. J. Hum. Genet 90:1094–101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Brownstein CA, Towne MC, Luquette LJ, Harris DJ, Marinakis NS, et al. 2013. Mutation of KCNJ8 in a patient with Cantú syndrome with unique vascular abnormalities—support for the role of KATP channels in this condition. Eur. J. Med. Genet 56:678–82 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Cooper PE, Reutter H, Woelfle J, Engels H, Grange DK, et al. 2014. Cantú syndrome resulting from activating mutation in the KCNJ8 gene. Hum. Mutat 35:809–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kirk EP, Scurr I, van Haaften G, van Haelst MM, Nichols CG, et al. 2017. Clinical utility gene card for: Cantú syndrome. Eur. J. Hum. Genet 25:512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Afifi HH, Abdel-Hamid MS, Eid MM, Mostafa IS, Abdel-Salam GM. 2016. De novo mutation in ABCC9 causes hypertrichosis acromegaloid facial features disorder. Pediatr. Dermatol 33:e109–13 [DOI] [PubMed] [Google Scholar]
  • 59.Hiraki Y, Miyatake S, Hayashidani M, Nishimura Y, Matsuura H, et al. 2014. Aortic aneurysm and craniosynostosis in a family with Cantú syndrome. Am. J. Med. Genet. A 164A:231–36 [DOI] [PubMed] [Google Scholar]
  • 60.Czeschik JC, Voigt C, Goecke TO, Ludecke HJ, Wagner N, et al. 2013. Wide clinical variability in conditions with coarse facial features and hypertrichosis caused by mutations in ABCC9. Am. J. Med. Genet. A 161A:295–300 [DOI] [PubMed] [Google Scholar]
  • 61.Kim H, Kim S, Jeon H, Kim J, Yoo J, et al. 2017. Clinical and molecular delineation of a novel Cys1050Phe missense mutation in the ABCC9 gene in a Korean patient with Cantú syndrome. Clin. Lab 63:991–95 [DOI] [PubMed] [Google Scholar]
  • 62.Marques P, Spencer R, Morrison PJ, Carr IM, Dang MN, et al. 2018. Cantú syndrome with coexisting familial pituitary adenoma. Endocrine 59:677–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Cooper PE, Sala-Rabanal M, Lee SJ, Nichols CG. 2015. Differential mechanisms of Cantú syndrome-associated gain of function mutations in the ABCC9 (SUR2) subunit of the KATP channel. J. Gen. Physiol 146:527–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.McClenaghan C, Hanson A, Sala-Rabanal M, Roessler HI, Josifova D, et al. 2018. Cantú syndrome-associated SUR2 (ABCC9) mutations in distinct structural domains result in KATP channel gain-of-function by differential mechanisms. J. Biol. Chem 293:2041–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Houtman MJC, Chen X, Qile M, Duran K, van Haaften G, et al. 2019. Glibenclamide and HMR1098 normalize Cantú syndrome-associated gain-of-function currents. J. Cell. Mol. Med 23:4962–69 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Flagg TP, Enkvetchakul D, Koster JC, Nichols CG. 2010. Muscle KATP channels: recent insights to energy sensing and myoprotection. Physiol. Rev 90:799–829 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Huang Y, McClenaghan C, Harter TM, Hinman K, Halabi CM, et al. 2018. Cardiovascular consequences of KATP overactivity in Cantú syndrome. JCI Insight 3:e121153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.McClenaghan C, Huang Y, Matkovich SJ, Kovacs A, Weinheimer CJ, et al. 2020. The mechanism of high-output cardiac hypertrophy arising from potassium channel gain-of-function in Cantú syndrome. Function 1:zqaa004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.McClenaghan C, Huang Y, Yan Z, Harter T, Halabi CM, et al. 2020. Glibenclamide reverses cardiovascular abnormalities of Cantu syndrome driven by KATP channel overactivity. J. Clin. Investig 130(3):1116–21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Scala R, Maqoud F, Zizzo N, Passantino G, Mele A, et al. 2021. Consequences of SUR2[A478V] mutation in skeletal muscle of murine model of Cantu syndrome. Cells 10:1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Scala R, Maqoud F, Zizzo N, Mele A, Camerino GM, et al. 2020. Pathophysiological consequences of KATP channel overactivity and pharmacological response to glibenclamide in skeletal muscle of a murine model of Cantú syndrome. Front. Pharmacol 11:604885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Powers SK, Morton AB, Hyatt H, Hinkley MJ. 2018. The renin-angiotensin system and skeletal muscle. Exerc. Sport Sci. Rev 46:205–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Rossi A, Cantisani C, Melis L, Iorio A, Scali E, et al. 2012. Minoxidil use in dermatology, side effects and recent patents. Recent Pat. Inflamm. Allergy Drug Discov 6:130–36 [DOI] [PubMed] [Google Scholar]
  • 74.York NW, Parker H, Xie Z, Tyus D, Waheed MA, et al. 2020. Kir6.1- and SUR2-dependent KATP over-activity disrupts intestinal motility in murine models of Cantú syndrome. JCI Insight 5:e141443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Davis MJ, Kim HJ, Zawieja SD, Castorena-Gonzalez JA, Gui P, et al. 2020. Kir6.1-dependent KATP channels in lymphatic smooth muscle and vessel dysfunction in mice with Kir6.1 gain-of-function. J. Physiol 598:3107–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.von der Weid PY. 1998. ATP-sensitive K+ channels in smooth muscle cells of guinea-pig mesenteric lymphatics: role in nitric oxide and β-adrenoceptor agonist-induced hyperpolarizations. Br. J. Pharmacol 125:17–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Mizuno R, Ono N, Ohhashi T. 1999. Involvement of ATP-sensitive K+ channels in spontaneous activity of isolated lymph microvessels in rats. Am. J. Physiol 277:H1453–56 [DOI] [PubMed] [Google Scholar]
  • 78.Minoretti P, Falcone C, Aldeghi A, Olivieri V, Mori F, et al. 2006. A novel Val734Ile variant in the ABCC9 gene associated with myocardial infarction. Clin. Chim. Acta 370:124–28 [DOI] [PubMed] [Google Scholar]
  • 79.Smith KJ, Chadburn AJ, Adomaviciene A, Minoretti P, Vignali L, et al. 2013. Coronary spasm and acute myocardial infarction due to a mutation (V734I) in the nucleotide binding domain 1 of ABCC9. Int. J. Cardiol 168:3506–13 [DOI] [PubMed] [Google Scholar]
  • 80.Hu D, Barajas-Martinez H, Terzic A, Park S, Pfeiffer R, et al. 2014. ABCC9 is a novel Brugada and early repolarization syndrome susceptibility gene. Int. J. Cardiol 171:431–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Haïssaguerre M, Chatel S, Sacher F, Weerasooriya R, Probst V, et al. 2009. Ventricular fibrillation with prominent early repolarization associated with a rare variant of KCNJ8/KATP channel. J. Cardiovasc. Electrophysiol 20:93–98 [DOI] [PubMed] [Google Scholar]
  • 82.Medeiros-Domingo A, Tan B-H, Crotti L, Tester DJ, Eckhardt L, et al. 2010. Gain-of-function mutation S422L in the KCNJ8-encoded cardiac KATP channel Kir6.1 as a pathogenic substrate for J-wave syndromes. Heart Rhythm 7:1466–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Barajas-Martinez H, Hu D, Ferrer T, Onetti CG, Wu Y, et al. 2012. Molecular genetic and functional association of Brugada and early repolarization syndromes with S422L missense mutation in KCNJ8. Heart Rhythm 9:548–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Delaney JT, Muhammad R, Blair MA, Kor K, Fish FA, et al. 2012. A KCNJ8 mutation associated with early repolarization and atrial fibrillation. EP Europace 14:1428–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Veeramah KR, Karafet TM, Wolf D, Samson RA, Hammer MF. 2014. The KCNJ8-S422L variant previously associated with J-wave syndromes is found at an increased frequency in Ashkenazi Jews. Eur. J. Hum. Genet 22:94–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Bienengraeber M, Olson TM, Selivanov VA, Kathmann EC, O’Cochlain F, et al. 2004. ABCC9 mutations identified in human dilated cardiomyopathy disrupt catalytic KATP channel gating. Nat. Genet 36:382–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Olson TM, Alekseev AE, Moreau C, Liu XK, Zingman LV, et al. 2007. KATP channel mutation confers risk for vein of Marshall adrenergic atrial fibrillation. Nat. Clin. Pract. Cardiovasc. Med 4:110–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zhang L, Tester DJ, Lang D, Chen Y, Zheng J, et al. 2016. Does sudden unexplained nocturnal death syndrome remain the autopsy-negative disorder: a gross, microscopic, and molecular autopsy investigation in southern China. Mayo Clin. Proc 91:1503–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Tester DJ, Tan BH, Medeiros-Domingo A, Song C, Makielski JC, et al. 2011. Loss-of-function mutations in the KCNJ8-encoded Kir6.1 KATP channel and sudden infant death syndrome. Circ. Cardiovasc. Genet 4:510–15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Smeland MF, McClenaghan C, Roessler HI, Savelberg S, Hansen GAM, et al. 2019. ABCC9-related Intellectual disability Myopathy Syndrome is a KATP channelopathy with loss-of-function mutations in ABCC9. Nat. Commun 10:4457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Schwanstecher C, Meyer U, Schwanstecher M. 2002. KIR6.2 polymorphism predisposes to type 2 diabetes by inducing overactivity of pancreatic β-cell ATP-sensitive K+ channels. Diabetes 51:875–79 [DOI] [PubMed] [Google Scholar]
  • 92.Hani EH, Boutin P, Durand E, Inoue H, Permutt MA, et al. 1998. Missense mutations in the pancreatic islet beta cell inwardly rectifying K+ channel gene (KIR6.2/BIR): A meta-analysis suggests a role in the polygenic basis of Type II diabetes mellitus in Caucasians. Diabetologia 41:1511–15 [DOI] [PubMed] [Google Scholar]
  • 93.Villareal DT, Koster JC, Robertson H, Akrouh A, Miyake K, et al. 2009. Kir6.2 variant E23K increases ATP-sensitive K+ channel activity and is associated with impaired insulin release and enhanced insulin sensitivity in adults with normal glucose tolerance. Diabetes 58:1869–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hamming KS, Soliman D, Matemisz LC, Niazi O, Lang Y, et al. 2009. Coexpression of the type 2 diabetes susceptibility gene variants KCNJ11 E23K and ABCC8 S1369A alter the ATP and sulfonylurea sensitivities of the ATP-sensitive K+ channel. Diabetes 58:2419–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Florez JC, Burtt N, de Bakker PI, Almgren P, Tuomi T, et al. 2004. Haplotype structure and genotype-phenotype correlations of the sulfonylurea receptor and the islet ATP-sensitive potassium channel gene region. Diabetes 53:1360–68 [DOI] [PubMed] [Google Scholar]
  • 96.Sachse G, Haythorne E, Hill T, Proks P, Joynson R, et al. 2021. The KCNJ11-E23K gene variant hastens diabetes progression by impairing glucose-induced insulin secretion. Diabetes 70:1145–56 [DOI] [PubMed] [Google Scholar]
  • 97.Sesti G, Laratta E, Cardellini M, Andreozzi F, Del Guerra S, et al. 2006. The E23K variant of KCNJ11 encoding the pancreatic β-cell adenosine 5’-triphosphate-sensitive potassium channel subunit Kir6.2 is associated with an increased risk of secondary failure to sulfonylurea in patients with type 2 diabetes. J. Clin. Endocrinol. Metab 91:2334–39 [DOI] [PubMed] [Google Scholar]
  • 98.Flagg TP, Kurata HT, Masia R, Caputa G, Magnuson MA, et al. 2008. Differential structure of atrial and ventricular KATP: Atrial KATP channels require SUR1. Circ. Res 103:1458–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Fedorov VV, Glukhov AV, Ambrosi CM, Kostecki G, Chang R, et al. 2011. Effects of KATP channel openers diazoxide and pinacidil in coronary-perfused atria and ventricles from failing and non-failing human hearts. J. Mol. Cell Cardiol 51:215–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Le Ribeuz H, Masson B, Capuano V, Dutheil M, Gooroochurn H, et al. 2022. SUR1 as a new therapeutic target for pulmonary arterial hypertension. Am. J. Respir. Cell Mol. Biol 66:539–54 [DOI] [PubMed] [Google Scholar]
  • 101.Bohnen MS, Ma L, Zhu N, Qi H, McClenaghan C, et al. 2018. Loss-of-function ABCC8 mutations in pulmonary arterial hypertension. Circ. Genom. Precis Med 11:e002087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Loubatières A 1969. [The discovery of hypoglycemic sulfonamides and particularly of their action mechanism]. Acta Diabetol. Lat 6(Suppl. 1):20–56 (In French) [PubMed] [Google Scholar]
  • 103.Kharade SV, Nichols C, Denton JS. 2016. The shifting landscape of KATP channelopathies and the need for ‘sharper’ therapeutics. Future Med. Chem 8:789–802 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Koster JC, Sha Q, Nichols CG. 1999. Sulfonylurea and K+-channel opener sensitivity of KATP channels. Functional coupling of Kir6.2 and SUR1 subunits. J. Gen. Physiol 114:203–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Martin GM, Sung MW, Yang Z, Innes LM, Kandasamy B, et al. 2019. Mechanism of pharmacochaperoning in a mammalian KATP channel revealed by cryo-EM. eLife 8:e46417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ding D, Wang M, Wu J-X, Kang Y, Chen L. 2019. The structural basis for the binding of repaglinide to the pancreatic KATP channel. Cell Rep 27:1848–57.e4 [DOI] [PubMed] [Google Scholar]
  • 107.Wu JX, Ding D, Wang M, Kang Y, Zeng X, et al. 2018. Ligand binding and conformational changes of SUR1 subunit in pancreatic ATP-sensitive potassium channels. Protein Cell 9:553–67 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Wu JX, Ding D, Wang M, Chen L. 2020. Structural insights into the inhibitory mechanism of insulin secretagogues on the pancreatic ATP-sensitive potassium channel. Biochemistry 59:18–25 [DOI] [PubMed] [Google Scholar]
  • 109.Ashfield R, Gribble FM, Ashcroft SJ, Ashcroft FM. 1999. Identification of the high-affinity tolbutamide site on the SUR1 subunit of the KATP channel. Diabetes 48:1341–47 [DOI] [PubMed] [Google Scholar]
  • 110.Hambrock A, Loffler-Walz C, Russ U, Lange U, Quast U. 2001. Characterization of a mutant sulfonylurea receptor SUR2B with high affinity for sulfonylureas and openers: differences in the coupling to Kir6.x subtypes. Mol. Pharmacol 60:190–99 [DOI] [PubMed] [Google Scholar]
  • 111.Kuhner P, Prager R, Stephan D, Russ U, Winkler M, et al. 2012. Importance of the Kir6.2 N-terminus for the interaction of glibenclamide and repaglinide with the pancreatic KATP channel. Naunyn Schmiedebergs Arch Pharmacol 385:299–311 [DOI] [PubMed] [Google Scholar]
  • 112.Mannhold R 2004. KATP channel openers: structure-activity relationships and therapeutic potential. Med. Res. Rev 24:213–66 [DOI] [PubMed] [Google Scholar]
  • 113.Schwanstecher M, Sieverding C, Dorschner H, Gross I, Aguilar-Bryan L, et al. 1998. Potassium channel openers require ATP to bind to and act through sulfonylurea receptors. EMBO J 17:5529–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Uhde I, Toman A, Gross I, Schwanstecher C, Schwanstecher M. 1999. Identification of the potassium channel opener site on sulfonylurea receptors. J. Biol. Chem 274:28079–82 [DOI] [PubMed] [Google Scholar]
  • 115.Matsuoka T, Matsushita K, Katayama Y, Fujita A, Inageda K, et al. 2000. C-terminal tails of sulfonylurea receptors control ADP-induced activation and diazoxide modulation of ATP-sensitive K+ channels. Circ. Res 87:873–80 [DOI] [PubMed] [Google Scholar]
  • 116.Ding D, Wu J-X, Duan X, Ma S, Lai L, Chen L. 2022. Structural identification of vasodilator binding sites on the SUR2 subunit. Nat. Commun 13:2675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Reimann F, Gribble FM, Ashcroft FM. 2000. Differential response of KATP channels containing SUR2A or SUR2B subunits to nucleotides and pinacidil. Mol. Pharmacol 58:1318–25 [DOI] [PubMed] [Google Scholar]
  • 118.Rubin AA, Roth FE, Taylor RM, Rosenkilde H. 1962. Pharmacology of diazoxide, an antihypertensive, nondiuretic benzothiadiazine. J. Pharmacol. Exp. Ther 136:344–52 [PubMed] [Google Scholar]
  • 119.Drash A, Wolff F. 1964. Drug therapy in leucine-sensitive hypoglycemia. Metabolism 13:487–92 [DOI] [PubMed] [Google Scholar]
  • 120.Henwood MJ, Kelly A, Macmullen C, Bhatia P, Ganguly A, et al. 2005. Genotype-phenotype correlations in children with congenital hyperinsulinism due to recessive mutations of the adenosine triphosphate-sensitive potassium channel genes. J. Clin. Endocrinol. Metab 90:789–94 [DOI] [PubMed] [Google Scholar]
  • 121.De Leon DD, Stanley CA. 2007. Mechanisms of disease: advances in diagnosis and treatment of hyperinsulinism in neonates. Nat. Clin. Pract. Endocrinol. Metab 3:57–68 [DOI] [PubMed] [Google Scholar]
  • 122.Hussain K, Aynsley-Green A. 2000. Management of hyperinsulinism in infancy and childhood. Ann. Med 32:544–51 [DOI] [PubMed] [Google Scholar]
  • 123.Hardy OT, Hernandez-Pampaloni M, Saffer JR, Suchi M, Ruchelli E, et al. 2007. Diagnosis and localization of focal congenital hyperinsulinism by 18F-fluorodopa PET scan. J. Pediatr 150:140–45 [DOI] [PubMed] [Google Scholar]
  • 124.Banerjee I, Skae M, Flanagan SE, Rigby L, Patel L, et al. 2011. The contribution of rapid KATP channel gene mutation analysis to the clinical management of children with congenital hyperinsulinism. Eur. J. Endocrinol 164:733–40 [DOI] [PubMed] [Google Scholar]
  • 125.Salomon-Estebanez M, Flanagan SE, Ellard S, Rigby L, Bowden L, et al. 2016. Conservatively treated congenital hyperinsulinism (CHI) due to K-ATP channel gene mutations: reducing severity over time. Orphanet J. Rare Dis 11:163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Mazor-Aronovitch K, Gillis D, Lobel D, Hirsch HJ, Pinhas-Hamiel O, et al. 2007. Long-term neurodevelopmental outcome in conservatively treated congenital hyperinsulinism. Eur. J. Endocrinol 157:491–97 [DOI] [PubMed] [Google Scholar]
  • 127.Martinez R, Fernandez-Ramos C, Vela A, Velayos T, Aguayo A, et al. 2016. Clinical and genetic characterization of congenital hyperinsulinism in Spain. Eur. J. Endocrinol 174:717–26 [DOI] [PubMed] [Google Scholar]
  • 128.Kapoor RR, Flanagan SE, Arya VB, Shield JP, Ellard S, et al. 2013. Clinical and molecular characterisation of 300 patients with congenital hyperinsulinism. Eur. J. Endocrinol 168:557–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Abdulhadi-Atwan M, Bushman J, Tornovsky-Babaey S, Perry A, Abu-Libdeh A, et al. 2008. Novel de novo mutation in sulfonylurea receptor 1 presenting as hyperinsulinism in infancy followed by overt diabetes in early adolescence. Diabetes 57:1935–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Gussinyer M, Clemente M, Cebrian R, Yeste D, Albisu M, et al. 2008. Glucose intolerance and diabetes are observed in the long-term follow-up of nonpancreatectomized patients with persistent hyperinsulinemic hypoglycemia of infancy due to mutations in the ABCC8 gene. Diabetes Care 31:1257–59 [DOI] [PubMed] [Google Scholar]
  • 131.Remedi MS, Rocheleau JV, Tong A, Patton BL, McDaniel ML, et al. 2006. Hyperinsulinism in mice with heterozygous loss of KATP channels. Diabetologia 49:2368–78 [DOI] [PubMed] [Google Scholar]
  • 132.Koster JC, Remedi MS, Flagg TP, Johnson JD, Markova KP, et al. 2002. Hyperinsulinism induced by targeted suppression of beta cell KATP channels. PNAS 99:16992–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Miki T, Nagashima K, Tashiro F, Kotake K, Yoshitomi H, et al. 1998. Defective insulin secretion and enhanced insulin action in KATP channel-deficient mice. PNAS 95:10402–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Miki T, Tashiro F, Iwanaga T, Nagashima K, Yoshitomi H, et al. 1997. Abnormalities of pancreatic islets by targeted expression of a dominant-negative KATP channel. PNAS 94:11969–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Seghers V, Nakazaki M, DeMayo F, Aguilar-Bryan L, Bryan J. 2000. Sur1 knockout mice: a model for KATP channel-independent regulation of insulin secretion. J. Biol. Chem 275:9270–77 [DOI] [PubMed] [Google Scholar]
  • 136.Remedi MS, Koster JC, Markova K, Seino S, Miki T, et al. 2004. Diet-induced glucose intolerance in mice with decreased β-cell ATP-sensitive K+ channels. Diabetes 53:3159–67 [DOI] [PubMed] [Google Scholar]
  • 137.Shimomura K, Tusa M, Iberl M, Brereton MF, Kaizik S, et al. 2013. A mouse model of human hyperinsulinism produced by the E1506K mutation in the sulphonylurea receptor SUR1. Diabetes 62:3797–806 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Sakura H, Ashcroft SJ, Terauchi Y, Kadowaki T, Ashcroft FM. 1998. Glucose modulation of ATP-sensitive K-currents in wild-type, homozygous and heterozygous glucokinase knock-out mice. Diabetologia 41:654–59 [DOI] [PubMed] [Google Scholar]
  • 139.Glaser B, Kesavan P, Heyman M, Davis E, Cuesta A, et al. 1998. Familial hyperinsulinism caused by an activating glucokinase mutation. N. Engl. J. Med 338:226–30 [DOI] [PubMed] [Google Scholar]
  • 140.Pearson ER, Flechtner I, Njolstad PR, Malecki MT, Flanagan SE, et al. 2006. Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N. Engl. J. Med 355:467–77 [DOI] [PubMed] [Google Scholar]
  • 141.Remedi MS, Koster JC. 2010. KATP channelopathies in the pancreas. Pflugers Arch. 460:307–20 [DOI] [PubMed] [Google Scholar]
  • 142.Koster JC, Cadario F, Peruzzi C, Colombo C, Nichols CG, et al. 2008. The G53D mutation in Kir6.2 (KCNJ11) is associated with neonatal diabetes and motor dysfunction in adulthood that is improved with sulfonylurea therapy. J. Clin. Endocrinol. Metab 93:1054–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Wambach JA, Marshall BA, Koster JC, White NH, Nichols CG. 2009. Successful sulfonylurea treatment of an insulin-naive neonate with diabetes mellitus due to a KCNJ11 mutation. Pediatr. Diabetes 11:286–88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Sagen JV, Raeder H, Hathout E, Shehadeh N, Gudmundsson K, et al. 2004. Permanent neonatal diabetes due to mutations in KCNJ11 encoding Kir6.2: patient characteristics and initial response to sulfonylurea therapy. Diabetes 53:2713–18 [DOI] [PubMed] [Google Scholar]
  • 145.Tonini G, Bizzarri C, Bonfanti R, Vanelli M, Cerutti F, et al. 2006. Sulfonylurea treatment outweighs insulin therapy in short-term metabolic control of patients with permanent neonatal diabetes mellitus due to activating mutations of the KCNJ11 (KIR6.2) gene. Diabetologia 49:2210–13 [DOI] [PubMed] [Google Scholar]
  • 146.Sumnik Z, Kolouskova S, Wales JK, Komarek V, Cinek O. 2007. Sulphonylurea treatment does not improve psychomotor development in children with KCNJ11 mutations causing permanent neonatal diabetes mellitus accompanied by developmental delay and epilepsy (DEND syndrome). Diabet. Med 24:1176–78 [DOI] [PubMed] [Google Scholar]
  • 147.Masia R, Koster JC, Tumini S, Chiarelli F, Colombo C, et al. 2007. An ATP-binding mutation (G334D) in KCNJ11 is associated with a sulfonylurea-insensitive form of developmental delay, epilepsy, and neonatal diabetes. Diabetes 56:328–36 [DOI] [PubMed] [Google Scholar]
  • 148.Slingerland AS, Nuboer R, Hadders-Algra M, Hattersley AT, Bruining GJ. 2006. Improved motor development and good long-term glycaemic control with sulfonylurea treatment in a patient with the syndrome of intermediate developmental delay, early-onset generalised epilepsy and neonatal diabetes associated with the V59M mutation in the KCNJ11 gene. Diabetologia 49:2559–63 [DOI] [PubMed] [Google Scholar]
  • 149.Takanaga H, Murakami H, Koyabu N, Matsuo H, Naito M, et al. 1998. Efflux transport of tolbutamide across the blood-brain barrier. J. Pharm. Pharmacol 50:1027–33 [DOI] [PubMed] [Google Scholar]
  • 150.Bessadok A, Garcia E, Jacquet H, Martin S, Garrigues A, et al. 2011. Recognition of sulfonylurea receptor (ABCC8/9) ligands by the multidrug resistance transporter P-glycoprotein (ABCB1): functional similarities based on common structural features between two multispecific ABC proteins. J. Biol. Chem 286:3552–69 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Proks P, de Wet H, Ashcroft FM. 2013. Molecular mechanism of sulphonylurea block of KATP channels carrying mutations that impair ATP inhibition and cause neonatal diabetes. Diabetes 62:3909–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Greeley SA, Zielinski MC, Poudel A, Ye H, Berry S, et al. 2017. Preservation of reduced numbers of insulin-positive cells in sulfonylurea-unresponsive KCNJ11-related diabetes. J. Clin. Endocrinol. Metab 102:1–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Wang Z, York NW, Nichols CG, Remedi MS. 2014. Pancreatic β cell dedifferentiation in diabetes and redifferentiation following insulin therapy. Cell Metab. 19:872–82 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Remedi MS, Agapova SE, Vyas AK, Hruz PW, Nichols CG. 2011. Acute sulfonylurea therapy at disease onset can cause permanent remission of KATP-induced diabetes. Diabetes 60:2515–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Carmody D, Bell CD, Hwang JL, Dickens JT, Sima DI, et al. 2014. Sulfonylurea treatment before genetic testing in neonatal diabetes: pros and cons. J. Clin. Endocrinol. Metab 99:E2709–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Matthews DR, Cull CA, Stratton IM, Holman RR, Turner RC. 1998. UKPDS 26: sulphonylurea failure in non-insulin-dependent diabetic patients over six years. UK Prospective Diabetes Study (UKPDS) Group. Diabet. Med 15:297–303 [DOI] [PubMed] [Google Scholar]
  • 157.Bowman P, Sulen A, Barbetti F, Beltrand J, Svalastoga P, et al. 2018. Effectiveness and safety of long-term treatment with sulfonylureas in patients with neonatal diabetes due to KCNJ11 mutations: an international cohort study. Lancet Diabet. Endocrinol 6:637–46 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Iafusco D, Bizzarri C, Cadario F, Pesavento R, Tonini G, et al. 2011. No beta cell desensitisation after a median of 68 months on glibenclamide therapy in patients with KCNJ11-associated permanent neonatal diabetes. Diabetologia 54:2736–38 [DOI] [PubMed] [Google Scholar]
  • 159.Leon Guerrero CR, Pathak S, Grange DK, Singh GK, Nichols CG, et al. 2016. Neurologic and neuroimaging manifestations of Cantú syndrome: a case series. Neurology 87:270–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Levin MD, Singh GK, Zhang HX, Uchida K, Kozel BA, et al. 2016. KATP channel gain-of-function leads to increased myocardial L-type Ca2+ current and contractility in Cantú syndrome. PNAS 113:6773–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Ma A, Gurnasinghani S, Kirk EP, McClenaghan C, Singh GK, et al. 2019. Glibenclamide treatment in a Cantú syndrome patient with a pathogenic ABCC9 gain-of-function variant: initial experience. Am. J. Med. Genet. A 179:1585–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Desai J, Key L, Swindall A, Gaston K, Talati AJ. 2021. The danger of diazoxide in the neonatal intensive care unit. Ther. Adv. Drug Saf 12:20420986211011338 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Tater KC, Gwaltney-Brant S, Wismer T. 2021. Topical minoxidil exposures and toxicoses in dogs and cats: 211 cases (2001–2019). J. Am. Anim. Hosp. Assoc 57:225–31 [DOI] [PubMed] [Google Scholar]
  • 164.Beltrand J, Baptiste A, Busiah K, Bouazza N, Godot C, et al. 2019. Glibenclamide oral suspension: suitable and effective in patients with neonatal diabetes. Pediatr. Diabetes 20:246–54 [DOI] [PubMed] [Google Scholar]
  • 165.Remedi MS, Nichols CG. 2008. Chronic antidiabetic sulfonylureas in vivo: reversible effects on mouse pancreatic β-cells. PLOS Med. 5:e206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Teramoto N 2006. Pharmacological profile of U-37883A, a channel blocker of smooth muscle-type ATP-sensitive K channels. Cardiovasc. Drug Rev 24:25–32 [DOI] [PubMed] [Google Scholar]
  • 167.Smith MP, Humphrey SJ, Jackson WF. 1994. Selective in vivo antagonism of pinacidil-induced hypotension by the guanidine U37883A in anesthetized rats. Pharmacology 49:363–75 [DOI] [PubMed] [Google Scholar]
  • 168.Gaidukov L, Wroblewska L, Teague B, Nelson T, Zhang X, et al. 2018. A multi-landing pad DNA integration platform for mammalian cell engineering. Nucleic Acids Res. 46:4072–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Gao J, Nichols C. 2022. Rapid characterization of the functional and pharmacological consequences of Cantú syndrome KATP channel mutations in intact cells. Biophys. J 121:388A–89A [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Raphemot R, Weaver CD, Denton JS. 2013. High-throughput screening for small-molecule modulators of inward rectifier potassium channels. J. Vis. Exp. Genet 71:e4209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Raphemot R, Swale DR, Dadi PK, Jacobson DA, Cooper P, et al. 2014. Direct activation of β-cell KATP channels with a novel xanthine derivative. Mol. Pharmacol 85:858–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Bekaert B, Boel A, Cosemans G, De Witte L, Menten B, et al. 2022. CRISPR/Cas gene editing in the human germline. Semin. Cell Dev. Biol In press [DOI] [PubMed] [Google Scholar]
  • 173.Ramu Y, Xu Y, Lu Z. 2018. A novel high-affinity inhibitor against the human ATP-sensitive Kir6.2 channel. J. Gen. Physiol 150:969–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Ramu Y, Yamakaze J, Zhou Y, Hoshi T, Lu Z. 2022. Blocking Kir6.2 channels with SpTx1 potentiates glucose-stimulated insulin secretion from murine pancreatic β cells and lowers blood glucose in diabetic mice. eLife 11:e77026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Houtman MJC, Friesacher T, Chen X, Zangerl-Plessl EM, van der Heyden MAG, et al. 2021. Development of IKATP ion channel blockers targeting sulfonylurea resistant mutant KIR6.2 based channels for treating DEND syndrome. Front. Pharmacol 12:814066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Partridge CJ, Beech DJ, Sivaprasadarao A. 2001. Identification and pharmacological correction of a membrane trafficking defect associated with a mutation in the sulfonylurea receptor causing familial hyperinsulinism. J. Biol. Chem 276:35947–52 [DOI] [PubMed] [Google Scholar]
  • 177.Yan FF, Casey J, Shyng SL. 2006. Sulfonylureas correct trafficking defects of disease-causing ATP-sensitive potassium channels by binding to the channel complex. J. Biol. Chem 281:33403–13 [DOI] [PubMed] [Google Scholar]
  • 178.Chen PC, Olson EM, Zhou Q, Kryukova Y, Sampson HM, et al. 2013. Carbamazepine as a novel small molecule corrector of trafficking-impaired ATP-sensitive potassium channels identified in congenital hyperinsulinism. J. Biol. Chem 288:20942–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Salido-Vallejo R, Gómez-García FJ, Garnacho-Saucedo G, Galán-Gutiérrez M. 2013. Acquired generalized hypertrichosis due to diazoxide. Actas Dermosifiliogr. 104:166–67 [DOI] [PubMed] [Google Scholar]

RESOURCES