1
|
Elsheikh A, Driggers CM, Truong HH, Yang Z, Allen J, Henriksen NM, Walczewska-Szewc K, Shyng SL. AI-based discovery and cryoEM structural elucidation of a K ATP channel pharmacochaperone. eLife 2025; 13:RP103159. [PMID: 40135739 PMCID: PMC11942174 DOI: 10.7554/elife.103159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025] Open
Abstract
Pancreatic KATP channel trafficking defects underlie congenital hyperinsulinism (CHI) cases unresponsive to the KATP channel opener diazoxide, the mainstay medical therapy for CHI. Current clinically used KATP channel inhibitors have been shown to act as pharmacochaperones and restore surface expression of trafficking mutants; however, their therapeutic utility for KATP trafficking-impaired CHI is hindered by high affinity binding, which limits functional recovery of rescued channels. Recent structural studies of KATP channels employing cryo-electron microscopy (cryoEM) have revealed a promiscuous pocket where several known KATP pharmacochaperones bind. The structural knowledge provides a framework for discovering KATP channel pharmacochaperones with desired reversible inhibitory effects to permit functional recovery of rescued channels. Using an AI-based virtual screening technology AtomNet followed by functional validation, we identified a novel compound, termed Aekatperone, which exhibits chaperoning effects on KATP channel trafficking mutations. Aekatperone reversibly inhibits KATP channel activity with a half-maximal inhibitory concentration (IC50) ~9 μM. Mutant channels rescued to the cell surface by Aekatperone showed functional recovery upon washout of the compound. CryoEM structure of KATP bound to Aekatperone revealed distinct binding features compared to known high affinity inhibitor pharmacochaperones. Our findings unveil a KATP pharmacochaperone enabling functional recovery of rescued channels as a promising therapeutic for CHI caused by KATP trafficking defects.
Collapse
Affiliation(s)
- Assmaa Elsheikh
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
- Department of Medical Biochemistry, College of Medicine, Tanta UniversityTantaEgypt
| | - Camden M Driggers
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
| | | | - Zhongying Yang
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
| | - John Allen
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
| | | | - Katarzyna Walczewska-Szewc
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in ToruńToruńPoland
| | - Show-Ling Shyng
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
| |
Collapse
|
2
|
Li L, Wang S, Fu S, Chen Z, Wang P, Zhao Y. Human ATP-binding proteins: Structural features, functional diversity, and pharmacotherapeutic potential in disease: A review. Int J Biol Macromol 2025; 308:142303. [PMID: 40118416 DOI: 10.1016/j.ijbiomac.2025.142303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/03/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
ATP-binding proteins (ABPs) form diverse and essential protein families across living organisms. Early life forms likely relied on simple chemical reactions for energy, but with the emergence of ABPs and their evolving functions, organisms became capable of more efficient energy storage and utilization, which drove the complexity of metabolic and life processes. By binding and hydrolyzing ATP through conserved structural motifs such as the Walker motifs, ABPs play critical roles in material transport, signal transduction, cellular structure maintenance, motility, and cell cycle regulation. Dysfunctions arising from mutations, deletions, or misregulation of ABPs are linked to a variety of human diseases, including cancer, neurodegenerative disorders, and cardiovascular diseases. The growing recognition of ABPs' significance in disease progression highlights their relevance not only in basic biology but also in clinical applications, particularly as biomarkers and therapeutic targets. This review provides a comprehensive overview of human ABPs, detailing their structural and functional roles, their involvement in disease mechanisms, and the latest advances in understanding their clinical relevance. Additionally, it identifies current research gaps and offers new perspectives for future investigations and therapeutic strategies.
Collapse
Affiliation(s)
- Letong Li
- School of Pharmacy, Health Science Center, Ningbo University, Ningbo 315211, PR China; Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, PR China
| | - Shanshan Wang
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, PR China.
| | - Songsen Fu
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, PR China
| | - Zhen Chen
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, PR China
| | - Pengjun Wang
- College of Electrical and Electronic Engineering, Wenzhou University, Wenzhou 325035, PR China.
| | - Yufen Zhao
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, PR China; Department of Chemical Biology, College of Chemistry and Chemical Engineering, and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, Xiamen 361005, PR China; Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, PR China.
| |
Collapse
|
3
|
ElSheikh A, Driggers CM, Truong HH, Yang Z, Allen J, Henriksen N, Walczewska-Szewc K, Shyng SL. AI-Based Discovery and CryoEM Structural Elucidation of a K ATP Channel Pharmacochaperone. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.05.611490. [PMID: 39282384 PMCID: PMC11398524 DOI: 10.1101/2024.09.05.611490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Pancreatic KATP channel trafficking defects underlie congenital hyperinsulinism (CHI) cases unresponsive to the KATP channel opener diazoxide, the mainstay medical therapy for CHI. Current clinically used KATP channel inhibitors have been shown to act as pharmacochaperones and restore surface expression of trafficking mutants; however, their therapeutic utility for KATP trafficking impaired CHI is hindered by high-affinity binding, which limits functional recovery of rescued channels. Recent structural studies of KATP channels employing cryo-electron microscopy (cryoEM) have revealed a promiscuous pocket where several known KATP pharmacochaperones bind. The structural knowledge provides a framework for discovering KATP channel pharmacochaperones with desired reversible inhibitory effects to permit functional recovery of rescued channels. Using an AI-based virtual screening technology AtomNet® followed by functional validation, we identified a novel compound, termed Aekatperone, which exhibits chaperoning effects on KATP channel trafficking mutations. Aekatperone reversibly inhibits KATP channel activity with a half-maximal inhibitory concentration (IC50) ~ 9 μM. Mutant channels rescued to the cell surface by Aekatperone showed functional recovery upon washout of the compound. CryoEM structure of KATP bound to Aekatperone revealed distinct binding features compared to known high affinity inhibitor pharmacochaperones. Our findings unveil a KATP pharmacochaperone enabling functional recovery of rescued channels as a promising therapeutic for CHI caused by KATP trafficking defects.
Collapse
Affiliation(s)
- Assmaa ElSheikh
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Medical Biochemistry, College of Medicine, Tanta University, Tanta, Egypt
| | - Camden M. Driggers
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ha H. Truong
- Atomwise Inc., 250 Sutter St., Suite 650, San Francisco, CA, USA
| | - Zhongying Yang
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - John Allen
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Niel Henriksen
- Atomwise Inc., 250 Sutter St., Suite 650, San Francisco, CA, USA
| | - Katarzyna Walczewska-Szewc
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, ul. Grudziądzka 5, 87-100 Toruń, Poland
| | - Show-Ling Shyng
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
4
|
Antonacci M, Maqoud F, Di Turi A, Miciaccia M, Perrone MG, Scilimati A, Tricarico D. KATP Channel Inhibitors Reduce Cell Proliferation Through Upregulation of H3K27ac in Diffuse Intrinsic Pontine Glioma: A Functional Expression Investigation. Cancers (Basel) 2025; 17:358. [PMID: 39941728 PMCID: PMC11816144 DOI: 10.3390/cancers17030358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/04/2025] [Accepted: 01/14/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Diffuse intrinsic pontine glioma [DIPG] is a fatal pediatric disease characterized by a post-translational modification, a replacement of lysine by methionine in position 27 of the N-terminal [H3K27M] tail of histone 3 isoform-1 [H3.1] or histone 3 isoform-3 [H3.3], respectively, expressed in the DIPG-36 and DIPG-50 cells. We investigated the role of cation channels in DIPG cells for the first time and the effects of ATP-sensitive K+[KATP] and TRPV1 channel modulators. METHODS Experiments were performed using "in vitro" cytotoxic assays combined with the patch clamp technique, RT-PCR, Western blot, and flow cytometry assays. RESULTS The most effective anti-proliferative drugs were repaglinide and glibenclamide after short and long-term incubation [6-96 h]. These drugs reduced macroscopic currents of the DIPG cells recorded in whole-cell patch clamp. Repaglinide concentration dependently enhanced the target protein H3K27ac in Western blotting after 48 h of incubation. This drug reduced cell diameter and enhanced cleaved caspase-3 in DIPG cells; total AKT/mTOR levels and phospho-mTOR were downregulated in DIPG-36. CONCLUSIONS KATP and TRPV1 channels are functionally expressed, and sulphonylureas are effective antiproliferative upregulating H3K27ac with apoptosis in DIPG cells and the sub-micromolar concentrations in DIPG-50.
Collapse
Affiliation(s)
- Marina Antonacci
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
| | - Fatima Maqoud
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology Saverio de Bellis, I.R.C.C.S. Research Hospital, 70013 Castellana Grotte, Italy
| | - Annamaria Di Turi
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
| | - Morena Miciaccia
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
| | - Maria Grazia Perrone
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
| | - Antonio Scilimati
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
| | - Domenico Tricarico
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
| |
Collapse
|
5
|
Li K, Satpute Janve V, Denton J. Characterization of four structurally diverse inhibitors of SUR2-containing K ATP channels. Channels (Austin) 2024; 18:2398565. [PMID: 39303216 PMCID: PMC11418212 DOI: 10.1080/19336950.2024.2398565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Vascular smooth muscle ATP-sensitive potassium (KATP) channels play critical roles in modulating vascular tone and thus represent important drug targets for diverse cardiovascular pathologies. Despite extensive research efforts spanning several decades, the search for selective inhibitors that can discriminate between vascular KATP (i.e. Kir6.1/SUR2B) and pancreatic and brain KATP (i.e. Kir6.2/SUR1) channels has, until recently, been unsuccessful. Our group therefore carried out a high-throughput screen of chemically diverse compounds with the goal of discovering specific Kir6.1/SUR2B inhibitors. This screen identified several novel classes of Kir6.1/SUR2B inhibitors, including the first potent (IC50 ~100 nM) and selective inhibitor published to date, termed VU0542270. Here, we expand on this work by disclosing the identity and pharmacological properties of four additional Kir6.1/SUR2B inhibitors that are structurally unrelated to Kir to VU0542270. These inhibitors, named VU0212387, VU0543336, VU0605768, and VU0544086, inhibit Kir6.1/SUR2B with IC50 values ranging from approximately 100 nM to 1 µM and exhibit no apparent inhibitory activity toward Kir6.2/SUR1. Functional analysis of heterologously expressed subunit combinations of Kir6.1, Kir6.2, SUR1, SUR2A, and SUR2B and demonstrated that all four inhibitors act on SUR2 to induce channel inhibition. Interestingly, VU0543336 and VU0212387 exhibit paradoxical stimulatory effects on Kir6.2/SUR1 at higher doses. This study broadens our understanding of KATP channel pharmacology, generally, and reveals novel chemical matter for the development of Kir6.1/SUR2-selective drugs, specifically.
Collapse
Affiliation(s)
- Kangjun Li
- Department of Pharmacology, Vanderbilt University, Nashville, TN
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| | | | - Jerod Denton
- Department of Pharmacology, Vanderbilt University, Nashville, TN
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN
| |
Collapse
|
6
|
Su J, Xu J, Hu S, Ye H, Xie L, Ouyang S. Advances in small-molecule insulin secretagogues for diabetes treatment. Biomed Pharmacother 2024; 178:117179. [PMID: 39059347 DOI: 10.1016/j.biopha.2024.117179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024] Open
Abstract
Diabetes, a metabolic disease caused by abnormally high levels of blood glucose, has a high prevalence rate worldwide and causes a series of complications, including coronary heart disease, stroke, peripheral vascular disease, end-stage renal disease, and retinopathy. Small-molecule compounds have been developed as drugs for the treatment of diabetes because of their oral advantages. Insulin secretagogues are a class of small-molecule drugs used to treat diabetes, and include sulfonylureas, non-sulfonylureas, glucagon-like peptide-1 receptor agonists, dipeptidyl peptidase 4 inhibitors, and other novel small-molecule insulin secretagogues. However, many small-molecule compounds cause different side effects, posing huge challenges to drug monotherapy and drug selection. Therefore, the use of different small-molecule drugs must be improved. This article reviews the mechanism, advantages, limitations, and potential risks of small-molecule insulin secretagogues to provide future research directions on small-molecule drugs for the treatment of diabetes.
Collapse
Affiliation(s)
- Jingqian Su
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China.
| | - Jingran Xu
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Shan Hu
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Hui Ye
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Lian Xie
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Songying Ouyang
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China.
| |
Collapse
|
7
|
Díaz-García JD, Leyva-Leyva M, Sánchez-Aguillón F, de León-Bautista MP, Fuentes-Venegas A, Torres-Viloria A, Tenorio-Aguirre EK, Morales-Lázaro SL, Olivo-Díaz A, González-Ramírez R. Association Study of CACNA1D, KCNJ11, KCNQ1, and CACNA1E Single-Nucleotide Polymorphisms with Type 2 Diabetes Mellitus. Int J Mol Sci 2024; 25:9196. [PMID: 39273144 PMCID: PMC11395491 DOI: 10.3390/ijms25179196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex chronic disease characterized by decreased insulin secretion and the development of insulin resistance. Previous genome-wide association studies demonstrated that single-nucleotide polymorphisms (SNPs) present in genes coding for ion channels involved in insulin secretion increase the risk of developing this disease. We determined the association of 16 SNPs found in CACNA1D, KCNQ1, KCNJ11, and CACNA1E genes and the increased probability of developing T2DM. In this work, we performed a case-control study in 301 Mexican adults, including 201 cases with diabetes and 100 controls without diabetes. Our findings indicate a moderate association between T2DM and the C allele, and the C/C genotype of rs312480 within CACNA1D. The CAG haplotype surprisingly showed a protective effect, whereas the CAC and CGG haplotypes have a strong association with T2DM. The C allele and C/C genotype of rs5219 were significantly associated with diabetes. Also, an association was observed between diabetes and the A allele and the A/A genotype of rs3753737 and rs175338 in CACNA1E. The TGG and CGA haplotypes were also found to be significantly associated. The findings of this study indicate that the SNPs examined could serve as a potential diagnostic tool and contribute to the susceptibility of the Mexican population to this disease.
Collapse
Affiliation(s)
- Juan Daniel Díaz-García
- División de Medicina Interna, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (J.D.D.-G.); (A.F.-V.); (A.T.-V.); (E.K.T.-A.)
| | - Margarita Leyva-Leyva
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (M.L.-L.); (F.S.-A.); (A.O.-D.)
| | - Fabiola Sánchez-Aguillón
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (M.L.-L.); (F.S.-A.); (A.O.-D.)
| | - Mercedes Piedad de León-Bautista
- Escuela de Medicina, Universidad Vasco de Quiroga, Morelia 58090, Mexico;
- Laboratorio de Enfermedades Infecciosas y Genómica (INEX LAB), Morelia 58280, Mexico
| | - Abel Fuentes-Venegas
- División de Medicina Interna, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (J.D.D.-G.); (A.F.-V.); (A.T.-V.); (E.K.T.-A.)
| | - Alfredo Torres-Viloria
- División de Medicina Interna, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (J.D.D.-G.); (A.F.-V.); (A.T.-V.); (E.K.T.-A.)
| | - Erika Karina Tenorio-Aguirre
- División de Medicina Interna, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (J.D.D.-G.); (A.F.-V.); (A.T.-V.); (E.K.T.-A.)
| | - Sara Luz Morales-Lázaro
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
- Centro de Investigación Sobre el Envejecimiento, CINVESTAV, Mexico City 14330, Mexico
| | - Angélica Olivo-Díaz
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (M.L.-L.); (F.S.-A.); (A.O.-D.)
| | - Ricardo González-Ramírez
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (M.L.-L.); (F.S.-A.); (A.O.-D.)
- Centro de Investigación Sobre el Envejecimiento, CINVESTAV, Mexico City 14330, Mexico
| |
Collapse
|
8
|
Metwally E, Sanchez Solano A, Lavanderos B, Yamasaki E, Thakore P, McClenaghan C, Rios N, Radi R, Feng Earley Y, Nichols CG, Earley S. Mitochondrial Ca2+-coupled generation of reactive oxygen species, peroxynitrite formation, and endothelial dysfunction in Cantú syndrome. JCI Insight 2024; 9:e176212. [PMID: 39088268 PMCID: PMC11385080 DOI: 10.1172/jci.insight.176212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 07/25/2024] [Indexed: 08/03/2024] Open
Abstract
Cantú syndrome is a multisystem disorder caused by gain-of-function (GOF) mutations in KCNJ8 and ABCC9, the genes encoding the pore-forming inward rectifier Kir6.1 and regulatory sulfonylurea receptor SUR2B subunits, respectively, of vascular ATP-sensitive K+ (KATP) channels. In this study, we investigated changes in the vascular endothelium in mice in which Cantú syndrome-associated Kcnj8 or Abcc9 mutations were knocked in to the endogenous loci. We found that endothelium-dependent dilation was impaired in small mesenteric arteries from Cantú mice. Loss of endothelium-dependent vasodilation led to increased vasoconstriction in response to intraluminal pressure or treatment with the adrenergic receptor agonist phenylephrine. We also found that either KATP GOF or acute activation of KATP channels with pinacidil increased the amplitude and frequency of wave-like Ca2+ events generated in the endothelium in response to the vasodilator agonist carbachol. Increased cytosolic Ca2+ signaling activity in arterial endothelial cells from Cantú mice was associated with elevated mitochondrial [Ca2+] and enhanced reactive oxygen species (ROS) and peroxynitrite levels. Scavenging intracellular or mitochondrial ROS restored endothelium-dependent vasodilation in the arteries of mice with KATP GOF mutations. We conclude that mitochondrial Ca2+ overload and ROS generation, which subsequently leads to nitric oxide consumption and peroxynitrite formation, cause endothelial dysfunction in mice with Cantú syndrome.
Collapse
Affiliation(s)
- Elsayed Metwally
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Alfredo Sanchez Solano
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Boris Lavanderos
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Evan Yamasaki
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Pratish Thakore
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Conor McClenaghan
- Departments of Pharmacology and Medicine, Center for Advanced Biotechnology and Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Natalia Rios
- Departamento de Bioquímica, Facultad de Medicina, and
- Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, and
- Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Yumei Feng Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Colin G. Nichols
- Center for the Investigation of Membrane Excitability Diseases and Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Scott Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| |
Collapse
|
9
|
Li K, McClenahan SJ, Han C, Bungard JD, Rathnayake U, Boutaud O, Bauer JA, Days EL, Lindsley CW, Shelton EL, Denton JS. Discovery and Characterization of VU0542270, the First Selective Inhibitor of Vascular Kir6.1/SUR2B K ATP Channels. Mol Pharmacol 2024; 105:202-212. [PMID: 38302135 PMCID: PMC10877733 DOI: 10.1124/molpharm.123.000783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024] Open
Abstract
Vascular smooth muscle KATP channels critically regulate blood flow and blood pressure by modulating vascular tone and therefore represent attractive drug targets for treating several cardiovascular disorders. However, the lack of potent inhibitors that can selectively inhibit Kir6.1/SUR2B (vascular KATP) over Kir6.2/SUR1 (pancreatic KATP) has eluded discovery despite decades of intensive research. We therefore screened 47,872 chemically diverse compounds for novel inhibitors of heterologously expressed Kir6.1/SUR2B channels. The most potent inhibitor identified in the screen was an N-aryl-N'-benzyl urea compound termed VU0542270. VU0542270 inhibits Kir6.1/SUR2B with an IC50 of approximately 100 nM but has no apparent activity toward Kir6.2/SUR1 or several other members of the Kir channel family at doses up to 30 µM (>300-fold selectivity). By expressing different combinations of Kir6.1 or Kir6.2 with SUR1, SUR2A, or SUR2B, the VU0542270 binding site was localized to SUR2. Initial structure-activity relationship exploration around VU0542270 revealed basic texture related to structural elements that are required for Kir6.1/SUR2B inhibition. Analysis of the pharmacokinetic properties of VU0542270 showed that it has a short in vivo half-life due to extensive metabolism. In pressure myography experiments on isolated mouse ductus arteriosus vessels, VU0542270 induced ductus arteriosus constriction in a dose-dependent manner similar to that of the nonspecific KATP channel inhibitor glibenclamide. The discovery of VU0542270 provides conceptual proof that SUR2-specific KATP channel inhibitors can be developed using a molecular target-based approach and offers hope for developing cardiovascular therapeutics targeting Kir6.1/SUR2B. SIGNIFICANCE STATEMENT: Small-molecule inhibitors of vascular smooth muscle KATP channels might represent novel therapeutics for patent ductus arteriosus, migraine headache, and sepsis; however, the lack of selective channel inhibitors has slowed progress in these therapeutic areas. Here, this study describes the discovery and characterization of the first vascular-specific KATP channel inhibitor, VU0542270.
Collapse
Affiliation(s)
- Kangjun Li
- Departments of Anesthesiology (K.L., S.J.M., J.S.D.), Pharmacology (K.L., C.H., J.D.B., U.R., O.B., C.W.L., J.S.D.), Pediatrics (E.L.S.), and Biochemistry (J.A.B.), Vanderbilt University Medical Center, Nashville, Tennessee and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (J.A.B., E.L.D., J.S.D.)
| | - Samantha J McClenahan
- Departments of Anesthesiology (K.L., S.J.M., J.S.D.), Pharmacology (K.L., C.H., J.D.B., U.R., O.B., C.W.L., J.S.D.), Pediatrics (E.L.S.), and Biochemistry (J.A.B.), Vanderbilt University Medical Center, Nashville, Tennessee and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (J.A.B., E.L.D., J.S.D.)
| | - Changho Han
- Departments of Anesthesiology (K.L., S.J.M., J.S.D.), Pharmacology (K.L., C.H., J.D.B., U.R., O.B., C.W.L., J.S.D.), Pediatrics (E.L.S.), and Biochemistry (J.A.B.), Vanderbilt University Medical Center, Nashville, Tennessee and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (J.A.B., E.L.D., J.S.D.)
| | - Joseph D Bungard
- Departments of Anesthesiology (K.L., S.J.M., J.S.D.), Pharmacology (K.L., C.H., J.D.B., U.R., O.B., C.W.L., J.S.D.), Pediatrics (E.L.S.), and Biochemistry (J.A.B.), Vanderbilt University Medical Center, Nashville, Tennessee and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (J.A.B., E.L.D., J.S.D.)
| | - Upendra Rathnayake
- Departments of Anesthesiology (K.L., S.J.M., J.S.D.), Pharmacology (K.L., C.H., J.D.B., U.R., O.B., C.W.L., J.S.D.), Pediatrics (E.L.S.), and Biochemistry (J.A.B.), Vanderbilt University Medical Center, Nashville, Tennessee and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (J.A.B., E.L.D., J.S.D.)
| | - Olivier Boutaud
- Departments of Anesthesiology (K.L., S.J.M., J.S.D.), Pharmacology (K.L., C.H., J.D.B., U.R., O.B., C.W.L., J.S.D.), Pediatrics (E.L.S.), and Biochemistry (J.A.B.), Vanderbilt University Medical Center, Nashville, Tennessee and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (J.A.B., E.L.D., J.S.D.)
| | - Joshua A Bauer
- Departments of Anesthesiology (K.L., S.J.M., J.S.D.), Pharmacology (K.L., C.H., J.D.B., U.R., O.B., C.W.L., J.S.D.), Pediatrics (E.L.S.), and Biochemistry (J.A.B.), Vanderbilt University Medical Center, Nashville, Tennessee and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (J.A.B., E.L.D., J.S.D.)
| | - Emily L Days
- Departments of Anesthesiology (K.L., S.J.M., J.S.D.), Pharmacology (K.L., C.H., J.D.B., U.R., O.B., C.W.L., J.S.D.), Pediatrics (E.L.S.), and Biochemistry (J.A.B.), Vanderbilt University Medical Center, Nashville, Tennessee and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (J.A.B., E.L.D., J.S.D.)
| | - Craig W Lindsley
- Departments of Anesthesiology (K.L., S.J.M., J.S.D.), Pharmacology (K.L., C.H., J.D.B., U.R., O.B., C.W.L., J.S.D.), Pediatrics (E.L.S.), and Biochemistry (J.A.B.), Vanderbilt University Medical Center, Nashville, Tennessee and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (J.A.B., E.L.D., J.S.D.)
| | - Elaine L Shelton
- Departments of Anesthesiology (K.L., S.J.M., J.S.D.), Pharmacology (K.L., C.H., J.D.B., U.R., O.B., C.W.L., J.S.D.), Pediatrics (E.L.S.), and Biochemistry (J.A.B.), Vanderbilt University Medical Center, Nashville, Tennessee and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (J.A.B., E.L.D., J.S.D.)
| | - Jerod S Denton
- Departments of Anesthesiology (K.L., S.J.M., J.S.D.), Pharmacology (K.L., C.H., J.D.B., U.R., O.B., C.W.L., J.S.D.), Pediatrics (E.L.S.), and Biochemistry (J.A.B.), Vanderbilt University Medical Center, Nashville, Tennessee and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (J.A.B., E.L.D., J.S.D.)
| |
Collapse
|
10
|
Gao J, McClenaghan C, Matreyek KA, Grange DK, Nichols CG. Rapid Characterization of the Functional and Pharmacological Consequences of Cantú Syndrome K ATP Channel Mutations in Intact Cells. J Pharmacol Exp Ther 2023; 386:298-309. [PMID: 37527933 PMCID: PMC10449099 DOI: 10.1124/jpet.123.001659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/10/2023] [Accepted: 06/01/2023] [Indexed: 08/03/2023] Open
Abstract
Gain-of-function of KATP channels, resulting from mutations in either KCNJ8 (encoding inward rectifier sub-family 6 [Kir6.1]) or ABCC9 (encoding sulphonylurea receptor [SUR2]), cause Cantú syndrome (CS), a channelopathy characterized by excess hair growth, coarse facial appearance, cardiomegaly, and lymphedema. Here, we established a pipeline for rapid analysis of CS mutation consequences in Landing pad HEK 293 cell lines stably expressing wild type (WT) and mutant human Kir6.1 and SUR2B. Thallium-influx and cell membrane potential, reported by fluorescent Tl-sensitive Fluozin-2 and voltage-sensitive bis-(1,3-dibutylbarbituric acid)trimethine oxonol (DiBAC4(3)) dyes, respectively, were used to assess channel activity. In the Tl-influx assay, CS-associated Kir6.1 mutations increased sensitivity to the ATP-sensitive potassium (KATP) channel activator, pinacidil, but there was strikingly little effect of pinacidil for any SUR2B mutations, reflecting unexpected differences in the molecular mechanisms of Kir6.1 versus SUR2B mutations. Compared with the Tl-influx assay, the DiBAC4(3) assay presents more significant signal changes in response to subtle KATP channel activity changes, and all CS mutants (both Kir6.1 and SUR2B), but not WT channels, caused marked hyperpolarization, demonstrating that all mutants were activated under ambient conditions in intact cells. Most SUR2 CS mutations were markedly inhibited by <100 nM glibenclamide, but sensitivity to inhibition by glibenclamide, repaglinide, and PNU37883A was markedly reduced for Kir6.1 CS mutations. Understanding functional consequences of mutations can help with disease diagnosis and treatment. The analysis pipeline we have developed has the potential to rapidly identify mutational consequences, aiding future CS diagnosis, drug discovery, and individualization of treatment. SIGNIFICANCE STATEMENT: We have developed new fluorescence-based assays of channel activities and drug sensitivities of Cantú syndrome (CS) mutations in human Kir6.1/SUR2B-dependent KATP channels, showing that Kir6.1 mutations increase sensitivity to potassium channel openers, while SUR2B mutations markedly reduce K channel opener (KCO) sensitivity. However, both Kir6.1 and SUR2B CS mutations are both more hyperpolarized than WT cells under basal conditions, confirming pathophysiologically relevant gain-of-function, validating DiBAC4(3) fluorescence to characterize hyperpolarization induced by KATP channel activity under basal, non KCO-activated conditions.
Collapse
Affiliation(s)
- Jian Gao
- Department of Cell Biology and Physiology (J.G., C.M.C., C.G.N.), Center for the Investigation of Membrane Excitability Diseases (J.G., C.M.C., D.K.G., C.G.N.), and Division of Genetics and Genomic Medicine, Department of Pediatrics (D.K.G.), Washington University in St. Louis, St. Louis, Missouri; and Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio (K.A.M.)
| | - Conor McClenaghan
- Department of Cell Biology and Physiology (J.G., C.M.C., C.G.N.), Center for the Investigation of Membrane Excitability Diseases (J.G., C.M.C., D.K.G., C.G.N.), and Division of Genetics and Genomic Medicine, Department of Pediatrics (D.K.G.), Washington University in St. Louis, St. Louis, Missouri; and Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio (K.A.M.)
| | - Kenneth A Matreyek
- Department of Cell Biology and Physiology (J.G., C.M.C., C.G.N.), Center for the Investigation of Membrane Excitability Diseases (J.G., C.M.C., D.K.G., C.G.N.), and Division of Genetics and Genomic Medicine, Department of Pediatrics (D.K.G.), Washington University in St. Louis, St. Louis, Missouri; and Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio (K.A.M.)
| | - Dorothy K Grange
- Department of Cell Biology and Physiology (J.G., C.M.C., C.G.N.), Center for the Investigation of Membrane Excitability Diseases (J.G., C.M.C., D.K.G., C.G.N.), and Division of Genetics and Genomic Medicine, Department of Pediatrics (D.K.G.), Washington University in St. Louis, St. Louis, Missouri; and Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio (K.A.M.)
| | - Colin G Nichols
- Department of Cell Biology and Physiology (J.G., C.M.C., C.G.N.), Center for the Investigation of Membrane Excitability Diseases (J.G., C.M.C., D.K.G., C.G.N.), and Division of Genetics and Genomic Medicine, Department of Pediatrics (D.K.G.), Washington University in St. Louis, St. Louis, Missouri; and Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio (K.A.M.)
| |
Collapse
|
11
|
Wang J, Papanicolaou K, Tryon R, Sangalang J, Salazar B, Suarez-Pierre A, Dong J, Lee A, Larson E, Holmes S, O’Rourke B, Nichols C, Lawton J. Kir1.1 and SUR1 are not implicated as subunits of an adenosine triphosphate-sensitive potassium channel involved in diazoxide cardioprotection. JTCVS OPEN 2023; 15:231-241. [PMID: 37808059 PMCID: PMC10556815 DOI: 10.1016/j.xjon.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/29/2023] [Accepted: 06/06/2023] [Indexed: 10/10/2023]
Abstract
Objective The adenosine triphosphate-sensitive potassium channel opener diazoxide mimics ischemic preconditioning and is cardioprotective. Clarification of diazoxide's site and mechanism of action could lead to targeted pharmacologic therapies for patients undergoing cardiac surgery. Several mitochondrial candidate proteins have been investigated as potential adenosine triphosphate-sensitive potassium channel components. Renal outer medullary potassium (Kir1.1) and sulfonylurea sensitive regulatory subunit 1 have been suggested as subunits of a mitochondrial adenosine triphosphate-sensitive potassium channel. We hypothesized that pharmacologic blockade or genetic deletion (knockout) of renal outer medullary potassium and sensitive regulatory subunit 1 would result in loss of diazoxide cardioprotection in models of global ischemia with cardioplegia. Methods Myocyte volume and contractility were compared after Tyrode's physiologic solution (20 minutes), stress (hyperkalemic cardioplegia ± diazoxide, ± VU591 (Kir1.1 inhibitor), N = 9 to 23 each, 20 min), and Tyrode's (20 minutes). Isolated mouse (wild-type, sensitive regulatory subunit 1 [-/-], and cardiac knockout renal outer medullary potassium) hearts were given cardioplegia ± diazoxide (N = 9-16 each) before global ischemia (90 minutes) and 30 minutes reperfusion. Left ventricular pressures were compared before and after ischemia. Results Stress (cardioplegia) was associated with reduced myocyte contractility that was prevented by diazoxide. Isolated myocytes were not responsive to diazoxide in the presence of VU591. In isolated hearts, diazoxide improved left ventricular function after prolonged ischemia compared with cardioplegia alone in wild-type and knockout (sensitive regulatory subunit 1 [-/-] and cardiac knockout renal outer medullary potassium) mice. Conclusions Isolated myocyte and heart models may measure independent and separate actions of diazoxide. By definitive genetic deletion, these data indicate that sensitive regulatory subunit 1 and renal outer medullary potassium are not implicated in cardioprotection by diazoxide.
Collapse
Affiliation(s)
- Jie Wang
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| | - Kyriakos Papanicolaou
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Md
| | - Robert Tryon
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Mo
| | - Janelle Sangalang
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| | - Ben Salazar
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Md
| | | | - Jie Dong
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| | - Anson Lee
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| | - Emily Larson
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| | - Sari Holmes
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| | - Brian O’Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Md
| | - Colin Nichols
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Mo
| | - Jennifer Lawton
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University, Baltimore, Md
| |
Collapse
|
12
|
Witham MD, Granic A, Pearson E, Robinson SM, Sayer AA. Repurposing Drugs for Diabetes Mellitus as Potential Pharmacological Treatments for Sarcopenia - A Narrative Review. Drugs Aging 2023:10.1007/s40266-023-01042-4. [PMID: 37486575 PMCID: PMC10371965 DOI: 10.1007/s40266-023-01042-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2023] [Indexed: 07/25/2023]
Abstract
Sarcopenia, the age-related loss of muscle strength and mass or quality, is a common condition with major adverse consequences. Although the pathophysiology is incompletely understood, there are common mechanisms between sarcopenia and the phenomenon of accelerated ageing seen in diabetes mellitus. Drugs currently used to treat type 2 diabetes mellitus may have mechanisms of action that are relevant to the prevention and treatment of sarcopenia, for those with type 2 diabetes and those without diabetes. This review summarises shared pathophysiology between sarcopenia and diabetes mellitus, including the effects of advanced glycation end products, mitochondrial dysfunction, chronic inflammation and changes to the insulin signalling pathway. Cellular and animal models have generated intriguing, albeit mixed, evidence that supports possible beneficial effects on skeletal muscle function for some classes of drugs used to treat diabetes, including metformin and SGLT2 inhibitors. Most human observational and intervention evidence for the effects of these drugs has been derived from populations with type 2 diabetes mellitus, and there is a need for intervention studies for older people with, and at risk of, sarcopenia to further investigate the balance of benefit and risk in these target populations. Not all diabetes treatments will be safe to use in those without diabetes because of variable side effects across classes. However, some agents [including glucagon-like peptide (GLP)-1 receptor agonists and SGLT2 inhibitors] have already demonstrated benefits in populations without diabetes, and it is these agents, along with metformin, that hold out the most promise for further investigation in sarcopenia.
Collapse
Affiliation(s)
- Miles D Witham
- AGE Research Group, Newcastle University Institute for Translational and Clinical Research, Newcastle Upon Tyne, UK.
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne NHS Foundation Trust and Cumbria, Northumberland and Tyne and Wear NHS Foundation Trust, Newcastle Upon Tyne, UK.
| | - Antoneta Granic
- AGE Research Group, Newcastle University Institute for Translational and Clinical Research, Newcastle Upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne NHS Foundation Trust and Cumbria, Northumberland and Tyne and Wear NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Ewan Pearson
- Division of Population Health and Genomics, Dundee Medical School, University of Dundee, Dundee, UK
| | - Sian M Robinson
- AGE Research Group, Newcastle University Institute for Translational and Clinical Research, Newcastle Upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne NHS Foundation Trust and Cumbria, Northumberland and Tyne and Wear NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Avan A Sayer
- AGE Research Group, Newcastle University Institute for Translational and Clinical Research, Newcastle Upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne NHS Foundation Trust and Cumbria, Northumberland and Tyne and Wear NHS Foundation Trust, Newcastle Upon Tyne, UK
| |
Collapse
|
13
|
ElSheikh A, Shyng SL. K ATP channel mutations in congenital hyperinsulinism: Progress and challenges towards mechanism-based therapies. Front Endocrinol (Lausanne) 2023; 14:1161117. [PMID: 37056678 PMCID: PMC10086357 DOI: 10.3389/fendo.2023.1161117] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Congenital hyperinsulinism (CHI) is the most common cause of persistent hypoglycemia in infancy/childhood and is a serious condition associated with severe recurrent attacks of hypoglycemia due to dysregulated insulin secretion. Timely diagnosis and effective treatment are crucial to prevent severe hypoglycemia that may lead to life-long neurological complications. In pancreatic β-cells, adenosine triphosphate (ATP)-sensitive K+ (KATP) channels are a central regulator of insulin secretion vital for glucose homeostasis. Genetic defects that lead to loss of expression or function of KATP channels are the most common cause of HI (KATP-HI). Much progress has been made in our understanding of the molecular genetics and pathophysiology of KATP-HI in the past decades; however, treatment remains challenging, in particular for patients with diffuse disease who do not respond to the KATP channel activator diazoxide. In this review, we discuss current approaches and limitations on the diagnosis and treatment of KATP-HI, and offer perspectives on alternative therapeutic strategies.
Collapse
Affiliation(s)
- Assmaa ElSheikh
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, United States
- Department of Medical Biochemistry, Tanta University, Tanta, Egypt
| | - Show-Ling Shyng
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|