Cation Chloride Cotransporter NKCC1 Operates through a Rocking-Bundle MechanismClick to copy article linkArticle link copied!
- Manuel José Ruiz MunevarManuel José Ruiz MunevarLaboratory of Molecular Modelling & Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, ItalyMore by Manuel José Ruiz Munevar
- Valerio RizziValerio RizziBiomolecular & Pharmaceutical Modelling Group, Université de Genève, Rue Michel-Servet 1, Geneva CH-1211 4, SwitzerlandMore by Valerio Rizzi
- Corinne PortioliCorinne PortioliLaboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, ItalyLaboratory of Brain Development and Disease, Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, ItalyMore by Corinne Portioli
- Pietro VidossichPietro VidossichLaboratory of Molecular Modelling & Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, ItalyMore by Pietro Vidossich
- Erhu CaoErhu CaoDepartment of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112-5650, United StatesMore by Erhu Cao
- Michele Parrinello*Michele Parrinello*Email: [email protected]Laboratory of Atomistic Simulations, Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, ItalyMore by Michele Parrinello
- Laura Cancedda*Laura Cancedda*Email: [email protected]Laboratory of Brain Development and Disease, Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, ItalyMore by Laura Cancedda
- Marco De Vivo*Marco De Vivo*Email: [email protected]Laboratory of Molecular Modelling & Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, ItalyMore by Marco De Vivo
Abstract
The sodium, potassium, and chloride cotransporter 1 (NKCC1) plays a key role in tightly regulating ion shuttling across cell membranes. Lately, its aberrant expression and function have been linked to numerous neurological disorders and cancers, making it a novel and highly promising pharmacological target for therapeutic interventions. A better understanding of how NKCC1 dynamically operates would therefore have broad implications for ongoing efforts toward its exploitation as a therapeutic target through its modulation. Based on recent structural data on NKCC1, we reveal conformational motions that are key to its function. Using extensive deep-learning-guided atomistic simulations of NKCC1 models embedded into the membrane, we captured complex dynamical transitions between alternate open conformations of the inner and outer vestibules of the cotransporter and demonstrated that NKCC1 has water-permeable states. We found that these previously undefined conformational transitions occur via a rocking-bundle mechanism characterized by the cooperative angular motion of transmembrane helices (TM) 4 and 9, with the contribution of the extracellular tip of TM 10. We found these motions to be critical in modulating ion transportation and in regulating NKCC1’s water transporting capabilities. Specifically, we identified interhelical dynamical contacts between TM 10 and TM 6, which we functionally validated through mutagenesis experiments of 4 new targeted NKCC1 mutants. We conclude showing that those 4 residues are highly conserved in most Na+-dependent cation chloride cotransporters (CCCs), which highlights their critical mechanistic implications, opening the way to new strategies for NKCC1’s function modulation and thus to potential drug action on selected CCCs.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Figure 1
Figure 1. NKCC1’s structure embedded in the membrane. (A) 3D representation of full-length human NKCC1 (PDB 7MXO), with each monomer represented in dark and light gray. Each monomer is constructed by three main components: (i) conserved TM domain, composed of 12 helices, which contains all four ion binding sites, shown in green, blue and orange for Cl–, Na+, and K+ ions, respectively; (ii) disordered amino-terminal; and (iii) large carboxy-terminal domains. (13) All structures of NKCC1, and related CCC’s of the same family, (40) revealed that the TM helices are organized in two inverted repeats of five-helix bundles─also known as LeuT-fold. (2,16,17,21) (B) Schematic representation of NKCC1, showing its homodimeric structure with one monomer represented to highlight the transmembrane domains (TMs, dark gray); the other monomer represented to highlight the channel arrangement across the cell membrane (light brown); the two five-helix bundle inverted repeats (TM 1 to TM 5 and TM 6 to TM 10) and the dimeric interface (TM 11 and TM 12). The amino and carboxy-terminal domains are also highlighted.
Figure 2
Figure 2. Essential conformational transition for alternating accessibility of ion binding sites allows NKCC1 ion transport. (A) Schematic representation of outward open (left) and inward open (right) NKCC1 conformation. Ionic binding sites are exposed to the extra and intracellular side of the membrane, respectively. (B) In gray, atomic surface representation of outward open (left) and inward open (right) NKCC1 conformation. Ionic binding sites for Na+, K+, and Cl– are shown as colored surfaces (blue, orange, and green, respectively), and ions are represented as colored spheres. Outer (left) and inner (right) vestibules, open to the extra and intracellular sides of the membrane, are highlighted with discontinuous black lines.
Results
NKCC1 Operates via a Rocking-Bundle Mechanism for Conformational Transitions
Figure 3
Figure 3. Rocking-bundle angular motion of specific NKCC1 TMs facilitates alternate accessibility of ion binding sites. (A) Schematic representation of NKCC1’s angular motion, defined as the change of the angle (α) between TM 4 and TM 9 (in green) and TM 2 and TM 7 (in red), during the conformational transition between the outward open state (bright green) and inward open state (dim green), calculated from the centers of mass of the backbone atoms from the extracellular and intracellular tip of TM 4 and TM 9 and the intracellular tip of TM 2 and TM 7 (in red). (B) Quantification of TM 4 and TM 9 angular motion represented by the angle α through 16 conformational transitions from OPES Explore simulations. The light brown horizontal bars represent the outward open and inward open average angle α ± 1SD calculated from 1 μs of equilibrium molecular dynamics (MD). Circles represent the starting point of each transition, whereas the triangles represent the end point of the same transition and its direction. Circles and triangles are colored depending on the NKCC1 conformation they represent (bright green for outward open and dim green for inward open).
Figure 4
Figure 4. Stabilization of the hydrophobic interface between TM 4 and TM 9 allows for their cooperative action. Representation of human NKCC1 embedded in the cell membrane, with TM 4 and TM 9 highlighted in bright yellow. Inset on the right: Higher magnification of the hydrophobic interface between TM 4 and TM 9 (highlighted by the oval), which allows for their cooperative angular motion. Relevant residues are shown as sticks with their atomic surfaces pictured in red (oxygen), blue (nitrogen), gray (carbon), and white (hydrogen).
Figure 5
Figure 5. NKCC1 TM 10's corking motion modulates ion/water access to the outer vestibule. (A) Schematic representation of NKCC1 TM 10's corking motion, defined as the change of TM 10's (in green) intrahelical angle (θ), during the conformational transition between the outward open state (bright green) and inward open state (dim green), calculated from the centers of mass of the backbone atoms from TM 10’s intra and extracellular tips, and the backbone atoms where TM 10 bends. (B) Quantification of TM 10's corking motion represented by the angle θ through 16 conformational transitions from OPES Explore simulations. The light brown horizontal bars represent the outward open and inward open average angle θ ± 1SD calculated from 1 μs of equilibrium molecular dynamics (MD). Circles represent the starting point of each transition, whereas the triangles represent the end point of the same transition and its direction. Circles and triangles are colored depending on the NKCC1 conformation they represent (bright green for outward open and dim green for inward open).
Figure 6
Figure 6. Interface between TM 10 and TM 6 highlights crucial interactions that determine accessibility of extracellular binding sites. Representation of human NKCC1 embedded in the cell membrane, with TM 10 and TM 6 highlighted in bright yellow. Inset on the top right: In the IO state, as shown by the light green schematic representation, interacting residues at the TM 10 and TM 6 interface are shown as sticks with their atomic surfaces (pictured in red─oxygen, blue─nitrogen, gray─carbon, and white─hydrogen), blocking solvent access to the outer vestibule. Inset on the bottom right: In the OO state, as shown by the light orange schematic representation, previously interacting residues at the TM 10 and TM 6 interface are now shown to be too far apart to form bonds.
Figure 7
Figure 7. Mutagenesis targeting residues from TM 10 and TM 6 highlight their functional relevance. (A) Representation of human NKCC1 embedded in the cell membrane, with TM 10 and TM 6 highlighted in bright yellow. Inset on the right: The interface between TM 10 and TM 6 where homologous mutated residues are shown as sticks and are highlighted in green surface. Namely, these residues are mouse A490W (human Ala 497), mouse L664A (human Leu 671), mouse N665A (human Asn 672), and mouse A668W (human Ala 675). Gray surface represents the position of residues that mainly form/break interactions throughout TM 10's corking motion. (B) Example traces obtained in the Cl– influx assay on HEK293 cells transfected with the WT NKCC1 transporter or NKCC1 mutated at different residues. The arrow indicates the addition of NaCl (74 mM) to initiate the NKCC1-mediated Cl– influx. (C) Quantification of the mouse NKCC1 inhibitory activity using the Cl– influx fluorescence assay in HEK293 cells. A fluorescence signal decrease, corresponding to a decrease in NKCC1 transporter activity, was observed for all the cells transfected with NKCC1 mutants. Data are normalized and the average of the last 10 s of kinetics is plotted (ΔF/F0). Data are presented as a percentage of the WT. Data represent mean ± SEM from 3 to 4 independent experiments (Kruskal–Wallis one way ANOVA, H = 216, DF = 6, followed by Dunn’s post hoc test on multiple comparisons, *** P = 0.0002, **** P < 0.0001).
Free Energy Simulations Characterize the Previously Elusive NKCC1’s Occluded State
Figure 8
Figure 8. Free energy surface identifies relevant NKCC1 conformations for the inward open ↔ outward open transition. (A) Representation of the Free Energy Surface of the conformational transition between human NKCC1 IO and OO states computed by OPES Explore over the DeepLDA collective variable and the outer vestibule water coordination collective variable. Energetical basins are highlighted with a schematic representation of the conformation they identify. These are, namely: the IO state, the occluded state (labeled Occ), the OOd state (outward open “dry” – lower outer vestibule hydration) and OOw (outward open “wet” – higher outer vestibule hydration. (B) Higher magnification of the hexagon in A representing the main gating interactions that occlude the outer vestibule and block solvent access to the ionic binding sites. Relevant residues are shown as sticks with their atomic surfaces pictured in red (oxygen), blue (nitrogen), gray (carbon) and white (hydrogen). (C) Higher magnification of the hexagon in A representing of the main gating interactions that occlude the inner vestibule and block solvent access to the ionic binding sites. Relevant residues are shown as sticks with their atomic surfaces pictured in red (oxygen), blue (nitrogen), yellow (sulfur), gray (carbon) and white (hydrogen).
Figure 9
Figure 9. Pore profile confirms distinct binding-site accessibility of NKCC1 states along the inward open ↔ outward open transition. (A–C) Pore profile of the IO state (A), the occluded state (B), and OO state (C), schematically represented at the bottom. These profiles were obtained by calculating the radius of the largest sphere along the Z-axis of the ion translocation cavity, and then plotting the pore profile of several snapshots from their corresponding equilibrium MD simulations, computed by the software HOLE. Pore profiles were mirrored around radius 0 for visual clarity. The blue lines represent the outer vestibule, and the orange lines represent the inner vestibule of NKCC1.
NKCC1 Permeability Allows Water Transportation
Figure 10
Figure 10. NKCC1’s outward open conformations are permeable to water. (A) Representation of NKCC1 (gray cartoon) embedded in the membrane (black horizontal bars) in a water-permeable state. Water molecules are shown as red and white lines with a red atomic surface representation. A chain of water molecules whose oxygen atoms are within 4.0 Å of each other connecting the extracellular and intracellular solvent is present. (B, C) Schematic representation of both states that present permeability to water (outward open without ions bound, B, or fully loaded, C), and their respective plot, which tracks the appearance of permeable states during each state’s equilibrium MD simulation. Vertical red bars represent snapshots from the respective simulation where a chain of water molecules whose oxygen atoms are within 4.0 Å of each other connecting the extracellular and intracellular solvent through NKCC1 is observed.
Figure 11
Figure 11. NKCC1 passively transports water. Histogram of all transport events detected in the state equilibrium MD simulation of NKCC1 outward open conformation with no ions bound, organized by the length of each transport event. Bars show the frequency of efflux/influx (orange/green) events per transport event duration. Vertical continuous lines show the mean, and discontinuous lines show the standard deviation (efflux, orange; influx, green). Some longer transport events were excluded from the histogram for clarity.
Discussion
Extension of Our Mechanistic Implications to Na+-Dependent CCCs
Conclusions
Methods
Equilibrium MD
Conformational CV Design
Water CV Design
OPES Explore
Fully Loaded NKCC1 Conformations and Ion Binding Simulations
Water Permeability and Transport
Experimental Section
Generation of NKCC1 Mutants and Cl– Influx Assay
Generation of Mutants
Cl Influx Assay
Data Availability
Trajectories are available upon request, while representative structures from all states (inward open, occluded and outward open in both fully loaded and ion-free states) are available in MJRM’s github repository (https://github.com/themanuelr/NKCC1_rep_struc).
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/jacs.3c10258.
Alternating access mechanisms, collective variable design, and relevant TM helices sequence alignment, and further analysis of the equilibrium and enhanced sampling MD simulations (state stability, salt-bridge formation and water content, and others) (PDF)
Angular motion for the rocking-bundle mechanism (MP4)
Presence of water-permeable states (MP4)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We thank Narjes Ansari for useful scientific discussions. L.C. thanks Telethon for financial support (Grant TCP15021). L.C. thanks the European Research Council (ERC) for partial funding (Grant agreement no. 725563). C.P. thanks the Marie Skłodowska-Curie Action for financial support (Grant agreement no. 843239), under the European Union’s Horizon 2020 research and innovation programme. E.C. thanks the NIH for financial support (Grant DK128592).
References
This article references 57 other publications.
- 1Deidda, G.; Parrini, M.; Naskar, S.; Bozarth, I. F.; Contestabile, A.; Cancedda, L. Reversing Excitatory GABAAR Signaling Restores Synaptic Plasticity and Memory in a Mouse Model of Down Syndrome. Nat. Med. 2015, 21 (4), 318– 326, DOI: 10.1038/nm.3827Google Scholar1Reversing excitatory GABAAR signaling restores synaptic plasticity and memory in a mouse model of Down syndromeDeidda, Gabriele; Parrini, Martina; Naskar, Shovan; Bozarth, Ignacio F.; Contestabile, Andrea; Cancedda, LauraNature Medicine (New York, NY, United States) (2015), 21 (4), 318-326CODEN: NAMEFI; ISSN:1078-8956. (Nature Publishing Group)Down syndrome (DS) is the most frequent genetic cause of intellectual disability, and altered GABAergic transmission through Cl--permeable GABAA receptors (GABAARs) contributes considerably to learning and memory deficits in DS mouse models. However, the efficacy of GABAergic transmission has never been directly assessed in DS. Here GABAAR signaling was found to be excitatory rather than inhibitory, and the reversal potential for GABAAR-driven Cl- currents (ECl) was shifted toward more pos. potentials in the hippocampi of adult DS mice. Accordingly, hippocampal expression of the cation Cl- cotransporter NKCC1 was increased in both trisomic mice and individuals with DS. Notably, NKCC1 inhibition by the FDA-approved drug bumetanide restored ECl, synaptic plasticity and hippocampus-dependent memory in adult DS mice. Our findings demonstrate that GABA is excitatory in adult DS mice and identify a new therapeutic approach for the potential rescue of cognitive disabilities in individuals with DS.
- 2Zhao, Y.; Cao, E. Structural Pharmacology of Cation-Chloride Cotransporters. Membranes 2022, 12 (12), 1206, DOI: 10.3390/membranes12121206Google Scholar2Structural Pharmacology of Cation-Chloride CotransportersZhao, Yongxiang; Cao, ErhuMembranes (Basel, Switzerland) (2022), 12 (12), 1206CODEN: MBSEB6; ISSN:2077-0375. (MDPI AG)A review. Loop and thiazide diuretics have been cornerstones of clin. management of hypertension and fluid overload conditions for more than five decades. The hunt for their mol. targets led to the discovery of cation-chloride cotransporters (CCCs) that catalyze electroneutral movement of Cl- together with Na+ and/or K+. CCCs consist of two 1 Na+-1 K+-2 Cl- (NKCC1-2), one 1 Na+-1 Cl- (NCC), and four 1 K+-1 Cl- (KCC1-4) transporters in human. CCCs are fundamental in trans-epithelia ion secretion and absorption, homeostasis of intracellular Cl- concn. and cell vol., and regulation of neuronal excitability. Malfunction of NKCC2 and NCC leads to abnormal salt and water retention in the kidney and, consequently, imbalance in electrolytes and blood pressure. Mutations in KCC2 and KCC3 are assocd. with brain disorders due to impairments in regulation of excitability and possibly cell vol. of neurons. A recent surge of structures of CCCs have defined their dimeric architecture, their ion binding sites, their conformational changes assocd. with ion translocation, and the mechanisms of action of loop diuretics and small mol. inhibitors. These breakthroughs now set the stage to expand CCC pharmacol. beyond loop and thiazide diuretics, developing the next generation of diuretics with improved potency and specificity. Beyond drugging renal-specific CCCs, brain-penetrable therapeutics are sorely needed to target CCCs in the nervous system for the treatment of neurol. disorders and psychiatric conditions.
- 3Ben-Ari, Y. NKCC1 Chloride Importer Antagonists Attenuate Many Neurological and Psychiatric Disorders. Trends Neurosci. 2017, 40 (9), 536– 554, DOI: 10.1016/j.tins.2017.07.001Google Scholar3NKCC1 Chloride Importer Antagonists Attenuate Many Neurological and Psychiatric DisordersBen-Ari, YehezkelTrends in Neurosciences (2017), 40 (9), 536-554CODEN: TNSCDR; ISSN:0166-2236. (Elsevier Ltd.)In physiol. conditions, adult neurons have low intracellular Cl- [(Cl-)I] levels underlying the γ-aminobutyric acid (GABA)ergic inhibitory drive. In contrast, neurons have high (Cl-)I levels and excitatory GABA actions in a wide range of pathol. conditions including spinal cord lesions, chronic pain, brain trauma, cerebrovascular infarcts, autism, Rett and Down syndrome, various types of epilepsies, and other genetic or environmental insults. The diuretic highly specific NKCC1 chloride importer antagonist bumetanide (PubChem CID: 2461) efficiently restores low (Cl-)I levels and attenuates many disorders in exptl. conditions and in some clin. trials. Here, I review the mechanisms of action, therapeutic effects, promises, and pitfalls of bumetanide.
- 4Delpire, E.; Gagnon, K. B. Elusive Role of the Na-K-2Cl Cotransporter in the Choroid Plexus. Am. J. Physiol.-Cell Physiol. 2019, 316 (4), C522– C524, DOI: 10.1152/ajpcell.00490.2018Google ScholarThere is no corresponding record for this reference.
- 5Kaila, K.; Price, T. J.; Payne, J. A.; Puskarjov, M.; Voipio, J. Cation-Chloride Cotransporters in Neuronal Development, Plasticity and Disease. Nat. Rev. Neurosci. 2014, 15 (10), 637– 654, DOI: 10.1038/nrn3819Google Scholar5Cation-chloride cotransporters in neuronal development, plasticity and diseaseKaila, Kai; Price, Theodore J.; Payne, John A.; Puskarjov, Martin; Voipio, JuhaNature Reviews Neuroscience (2014), 15 (10), 637-654CODEN: NRNAAN; ISSN:1471-003X. (Nature Publishing Group)Elec. activity in neurons requires a seamless functional coupling between plasmalemmal ion channels and ion transporters. Although ion channels have been studied intensively for several decades, research on ion transporters is in its infancy. In recent years, it has become evident that one family of ion transporters, cation-chloride cotransporters (CCCs), and in particular K+-Cl- cotransporter 2 (KCC2), have seminal roles in shaping GABAergic signalling and neuronal connectivity. Studying the functions of these transporters may lead to major paradigm shifts in our understanding of the mechanisms underlying brain development and plasticity in health and disease.
- 6Pressey, J. C.; de Saint-Rome, M.; Raveendran, V. A.; Woodin, M. A. Chloride Transporters Controlling Neuronal Excitability. Physiol. Rev. 2023, 103 (2), 1095– 1135, DOI: 10.1152/physrev.00025.2021Google Scholar6Chloride transporters controlling neuronal excitabilityPressey, Jessica C.; de Saint-Rome, Miranda; Raveendran, Vineeth A.; Woodin, Melanie A.Physiological Reviews (2023), 103 (2), 1095-1135CODEN: PHREA7; ISSN:1522-1210. (American Physiological Society)A review. Synaptic inhibition plays a crucial role in regulating neuronal excitability, which is the foundation of nervous system function. This inhibition is largely mediated by the neurotransmitters GABA and glycine that activate Cl--permeable ion channels, which means that the strength of inhibition depends on the Cl- gradient across the membrane. In neurons, the Cl- gradient is primarily mediated by two secondarily active cation-chloride cotransporters (CCCs), NKCC1 and KCC2. CCC-mediated regulation of the neuronal Cl- gradient is crit. for healthy brain function, as dysregulation of CCCs has emerged as a key mechanism underlying neurol. disorders including epilepsy, neuropathic pain, and autism spectrum disorder. This review begins with an overview of neuronal chloride transporters before explaining the dependent relationship between these CCCs, Cl- regulation, and inhibitory synaptic transmission. We then discuss the evidence for how CCCs can be regulated, including by activity and their protein interactions, which underlie inhibitory synaptic plasticity. For readers who may be interested in conducting expts. on CCCs and neuronal excitability, we have included a section on techniques for estg. and recording intracellular Cl-, including their advantages and limitations. Although the focus of this review is on neurons, we also examine how Cl- is regulated in glial cells, which in turn regulate neuronal excitability through the tight relationship between this nonneuronal cell type and synapses. Finally, we discuss the relatively extensive and growing literature on how CCC-mediated neuronal excitability contributes to neurol. disorders.
- 7Savardi, A.; Borgogno, M.; De Vivo, M.; Cancedda, L. Pharmacological Tools to Target NKCC1 in Brain Disorders. Trends Pharmacol. Sci. 2021, 42 (12), 1009– 1034, DOI: 10.1016/j.tips.2021.09.005Google Scholar7Pharmacological tools to target NKCC1 in brain disordersSavardi, Annalisa; Borgogno, Marco; De Vivo, Marco; Cancedda, LauraTrends in Pharmacological Sciences (2021), 42 (12), 1009-1034CODEN: TPHSDY; ISSN:0165-6147. (Elsevier Ltd.)A review. The chloride importer NKCC1 and the chloride exporter KCC2 are key regulators of neuronal chloride concn. A defective NKCC1/KCC2 expression ratio is assocd. with several brain disorders. Preclin./clin. studies have shown that NKCC1 inhibition by the United States FDA-approved diuretic bumetanide is a potential therapeutic strategy in preclin./clin. studies of multiple neurol. conditions. However, bumetanide has poor brain penetration and causes unwanted diuresis by inhibiting NKCC2 in the kidney. To overcome these issues, a growing no. of studies have reported more brain-penetrating and/or selective bumetanide prodrugs, analogs, and new mol. entities. Here, we review the evidence for NKCC1 pharmacol. inhibition as an effective strategy to manage neurol. disorders. We also discuss the advantages and limitations of bumetanide repurposing and the benefits and risks of new NKCC1 inhibitors as therapeutic agents for brain disorders.
- 8Karimy, J. K.; Zhang, J.; Kurland, D. B.; Theriault, B. C.; Duran, D.; Stokum, J. A.; Furey, C. G.; Zhou, X.; Mansuri, M. S.; Montejo, J.; Vera, A.; DiLuna, M. L.; Delpire, E.; Alper, S. L.; Gunel, M.; Gerzanich, V.; Medzhitov, R.; Simard, J. M.; Kahle, K. T. Inflammation-Dependent Cerebrospinal Fluid Hypersecretion by the Choroid Plexus Epithelium in Posthemorrhagic Hydrocephalus. Nat. Med. 2017, 23 (8), 997– 1003, DOI: 10.1038/nm.4361Google Scholar8Inflammation-dependent cerebrospinal fluid hypersecretion by the choroid plexus epithelium in posthemorrhagic hydrocephalusKarimy, Jason K.; Zhang, Jinwei; Kurland, David B.; Theriault, Brianna Carusillo; Duran, Daniel; Stokum, Jesse A.; Furey, Charuta Gavankar; Zhou, Xu; Mansuri, M. Shahid; Montejo, Julio; Vera, Alberto; Di Luna, Michael L.; Delpire, Eric; Alper, Seth L.; Gunel, Murat; Gerzanich, Volodymyr; Medzhitov, Ruslan; Simard, J. Marc; Kahle, Kristopher T.Nature Medicine (New York, NY, United States) (2017), 23 (8), 997-1003CODEN: NAMEFI; ISSN:1078-8956. (Nature Publishing Group)The choroid plexus epithelium (CPE) secretes higher vols. of fluid (cerebrospinal fluid, CSF) than any other epithelium and simultaneously functions as the blood-CSF barrier to gate immune cell entry into the central nervous system. Posthemorrhagic hydrocephalus (PHH), an expansion of the cerebral ventricles due to CSF accumulation following intraventricular hemorrhage (IVH), is a common disease usually treated by suboptimal CSF shunting techniques. PHH is classically attributed to primary impairments in CSF reabsorption, but little exptl. evidence supports this concept. In contrast, the potential contribution of CSF secretion to PHH has received little attention. In a rat model of PHH, we demonstrate that IVH causes a Toll-like receptor 4 (TLR4)- and NF-κB-dependent inflammatory response in the CPE that is assocd. with a ∼3-fold increase in bumetanide-sensitive CSF secretion. IVH-induced hypersecretion of CSF is mediated by TLR4-dependent activation of the Ste20-type stress kinase SPAK, which binds, phosphorylates, and stimulates the NKCC1 co-transporter at the CPE apical membrane. Genetic depletion of TLR4 or SPAK normalizes hyperactive CSF secretion rates and reduces PHH symptoms, as does treatment with drugs that antagonize TLR4-NF-κB signaling or the SPAK-NKCC1 co-transporter complex. These data uncover a previously unrecognized contribution of CSF hypersecretion to the pathogenesis of PHH, demonstrate a new role for TLRs in regulation of the internal brain milieu, and identify a kinase-regulated mechanism of CSF secretion that could be targeted by repurposed US Food and Drug Administration (FDA)-approved drugs to treat hydrocephalus.
- 9Kharod, S. C.; Kang, S. K.; Kadam, S. D. Off-Label Use of Bumetanide for Brain Disorders: An Overview. Front. Neurosci. 2019, 13, 310, DOI: 10.3389/fnins.2019.00310Google Scholar9Off-Label Use of Bumetanide for Brain Disorders: An OverviewKharod Shivani C; Kang Seok Kyu; Kadam Shilpa D; Kadam Shilpa DFrontiers in neuroscience (2019), 13 (), 310 ISSN:1662-4548.Bumetanide (BTN or BUM) is a FDA-approved potent loop diuretic (LD) that acts by antagonizing sodium-potassium-chloride (Na-K-Cl) cotransporters, NKCC1 (SLc12a2) and NKCC2. While NKCC1 is expressed both in the CNS and in systemic organs, NKCC2 is kidney-specific. The off-label use of BTN to modulate neuronal transmembrane Cl(-) gradients by blocking NKCC1 in the CNS has now been tested as an anti-seizure agent and as an intervention for neurological disorders in pre-clinical studies with varying results. BTN safety and efficacy for its off-label use has also been tested in several clinical trials for neonates, children, adolescents, and adults. It failed to meet efficacy criteria for hypoxic-ischemic encephalopathy (HIE) neonatal seizures. In contrast, positive outcomes in temporal lobe epilepsy (TLE), autism, and schizophrenia trials have been attributed to BTN in studies evaluating its off-label use. NKCC1 is an electroneutral neuronal Cl(-) importer and the dominance of NKCC1 function has been proposed as the common pathology for HIE seizures, TLE, autism, and schizophrenia. Therefore, the use of BTN to antagonize neuronal NKCC1 with the goal to lower internal Cl(-) levels and promote GABAergic mediated hyperpolarization has been proposed. In this review, we summarize the data and results for pre-clinical and clinical studies that have tested off-label BTN interventions and report variable outcomes. We also compare the data underlying the developmental expression profile of NKCC1 and KCC2, highlight the limitations of BTN's brain-availability and consider its actions on non-neuronal cells.
- 10Wang, J.; Liu, R.; Hasan, M. N.; Fischer, S.; Chen, Y.; Como, M.; Fiesler, V. M.; Bhuiyan, M. I. H.; Dong, S.; Li, E.; Kahle, K. T.; Zhang, J.; Deng, X.; Subramanya, A. R.; Begum, G.; Yin, Y.; Sun, D. Role of SPAK–NKCC1 Signaling Cascade in the Choroid Plexus Blood–CSF Barrier Damage after Stroke. J. Neuroinflammation 2022, 19 (1), 91, DOI: 10.1186/s12974-022-02456-4Google Scholar10Role of SPAK-NKCC1 signaling cascade in the choroid plexus blood-CSF barrier damage after strokeWang, Jun; Liu, Ruijia; Hasan, Md Nabiul; Fischer, Sydney; Chen, Yang; Como, Matt; Fiesler, Victoria M.; Bhuiyan, Mohammad Iqbal H.; Dong, Shuying; Li, Eric; Kahle, Kristopher T.; Zhang, Jinwei; Deng, Xianming; Subramanya, Arohan R.; Begum, Gulnaz; Yin, Yan; Sun, DandanJournal of Neuroinflammation (2022), 19 (1), 91CODEN: JNOEB3; ISSN:1742-2094. (BioMed Central Ltd.)The mechanisms underlying dysfunction of choroid plexus (ChP) blood-cerebrospinal fluid (CSF) barrier and lymphocyte invasion in neuroinflammatory responses to stroke are not well understood. In this study, we investigated whether stroke damaged the blood-CSF barrier integrity due to dysregulation of major ChP ion transport system, Na+-K+-Cl- cotransporter 1 (NKCC1), and regulatory Ste20-related proline-alanine-rich kinase (SPAK). Sham or ischemic stroke was induced in C57Bl/6J mice. Changes on the SPAK-NKCC1 complex and tight junction proteins (TJs) in the ChP were quantified by immunofluorescence staining and immunoblotting. Immune cell infiltration in the ChP was assessed by flow cytometry and immunostaining. Cultured ChP epithelium cells (CPECs) and cortical neurons were used to evaluate H2O2-mediated oxidative stress in stimulating the SPAK-NKCC1 complex and cellular damage. In vivo or in vitro pharmacol. blockade of the ChP SPAK-NKCC1 cascade with SPAK inhibitor ZT-1a or NKCC1 inhibitor bumetanide were examd. Ischemic stroke stimulated activation of the CPECs apical membrane SPAK-NKCC1 complex, NF-κB, and MMP9, which was assocd. with loss of the blood-CSF barrier integrity and increased immune cell infiltration into the ChP. Oxidative stress directly activated the SPAK-NKCC1 pathway and resulted in apoptosis, neurodegeneration, and NKCC1-mediated ion influx. Pharmacol. blockade of the SPAK-NKCC1 pathway protected the ChP barrier integrity, attenuated ChP immune cell infiltration or neuronal death. Stroke-induced patholol. stimulation of the SPAK-NKCC1 cascade caused CPECs damage and disruption of TJs at the blood-CSF barrier. The ChP SPAK-NKCC1 complex emerged as a therapeutic target for attenuating ChP dysfunction and lymphocyte invasion after stroke.
- 11Chew, T. A.; Orlando, B. J.; Zhang, J.; Latorraca, N. R.; Wang, A.; Hollingsworth, S. A.; Chen, D.-H.; Dror, R. O.; Liao, M.; Feng, L. Structure and Mechanism of the Cation–Chloride Cotransporter NKCC1. Nature 2019, 572 (7770), 488– 492, DOI: 10.1038/s41586-019-1438-2Google Scholar11Structure and mechanism of the cation-chloride cotransporter NKCC1Chew, Thomas A.; Orlando, Benjamin J.; Zhang, Jinru; Latorraca, Naomi R.; Wang, Amy; Hollingsworth, Scott A.; Chen, Dong-Hua; Dror, Ron O.; Liao, Maofu; Feng, LiangNature (London, United Kingdom) (2019), 572 (7770), 488-492CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Cation-chloride cotransporters (CCCs) mediate the electroneutral transport of chloride, potassium and/or sodium across the membrane. They have crit. roles in regulating cell vol., controlling ion absorption and secretion across epithelia, and maintaining intracellular chloride homeostasis. These transporters are primary targets for some of the most commonly prescribed drugs. Here we detd. the cryo-electron microscopy structure of the Na-K-Cl cotransporter NKCC1, an extensively studied member of the CCC family, from Danio rerio. The structure defines the architecture of this protein family and reveals how cytosolic and transmembrane domains are strategically positioned for communication. Structural analyses, functional characterizations and computational studies reveal the ion-translocation pathway, ion-binding sites and key residues for transport activity. These results provide insights into ion selectivity, coupling and translocation, and establish a framework for understanding the physiol. functions of CCCs and interpreting disease-related mutations.
- 12Moseng, M. A.; Su, C.-C.; Rios, K.; Cui, M.; Lyu, M.; Glaza, P.; Klenotic, P. A.; Delpire, E.; Yu, E. W. Inhibition Mechanism of NKCC1 Involves the Carboxyl Terminus and Long-Range Conformational Coupling. Sci. Adv. 2022, 8 (43), eabq0952 DOI: 10.1126/sciadv.abq0952Google ScholarThere is no corresponding record for this reference.
- 13Yang, X.; Wang, Q.; Cao, E. Structure of the Human Cation–Chloride Cotransporter NKCC1 Determined by Single-Particle Electron Cryo-Microscopy. Nat. Commun. 2020, 11 (1), 1016, DOI: 10.1038/s41467-020-14790-3Google Scholar13Structure of the human cation-chloride cotransporter NKCC1 determined by single-particle electron cryo-microscopyYang, Xiaoyong; Wang, Qinzhe; Cao, ErhuNature Communications (2020), 11 (1), 1016CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)The secondary active cation-chloride cotransporters (CCCs) utilize the existing Na+ and/or K+ gradients to move Cl- into or out of cells. NKCC1 is an intensively studied member of the CCC family and plays fundamental roles in regulating trans-epithelial ion movement, cell vol., chloride homeostasis and neuronal excitability. Here, we report a cryo-EM structure of human NKCC1 captured in a partially loaded, inward-open state. NKCC1 assembles into a dimer, with the first ten transmembrane (TM) helixes harboring the transport core and TM11-TM12 helixes lining the dimer interface. TM1 and TM6 helixes break α-helical geometry halfway across the lipid bilayer where ion binding sites are organized around these discontinuous regions. NKCC1 may harbor multiple extracellular entryways and intracellular exits, raising the possibility that K+, Na+, and Cl- ions may traverse along their own routes for translocation. NKCC1 structure provides a blueprint for further probing structure-function relationships of NKCC1 and other CCCs.
- 14Zhang, S.; Zhou, J.; Zhang, Y.; Liu, T.; Friedel, P.; Zhuo, W.; Somasekharan, S.; Roy, K.; Zhang, L.; Liu, Y.; Meng, X.; Deng, H.; Zeng, W.; Li, G.; Forbush, B.; Yang, M. The Structural Basis of Function and Regulation of Neuronal Cotransporters NKCC1 and KCC2. Commun. Biol. 2021, 4 (1), 226, DOI: 10.1038/s42003-021-01750-wGoogle Scholar14The structural basis of function and regulation of neuronal cotransporters NKCC1 and KCC2Zhang, Sensen; Zhou, Jun; Zhang, Yuebin; Liu, Tianya; Friedel, Perrine; Zhuo, Wei; Somasekharan, Suma; Roy, Kasturi; Zhang, Laixing; Liu, Yang; Meng, Xianbin; Deng, Haiteng; Zeng, Wenwen; Li, Guohui; Forbush, Biff; Yang, MaojunCommunications Biology (2021), 4 (1), 226CODEN: CBOIDQ; ISSN:2399-3642. (Nature Research)NKCC and KCC transporters mediate coupled transport of Na++K++Cl- and K++Cl- across the plasma membrane, thus regulating cell Cl- concn. and cell vol. and playing crit. roles in transepithelial salt and water transport and in neuronal excitability. The function of these transporters has been intensively studied, but a mechanistic understanding has awaited structural studies of the transporters. Here, we present the cryo-electron microscopy (cryo-EM) structures of the two neuronal cation-chloride cotransporters human NKCC1 (SLC12A2) and mouse KCC2 (SLC12A5), along with computational anal. and functional characterization. These structures highlight essential residues in ion transport and allow us to propose mechanisms by which phosphorylation regulates transport activity.
- 15Zhao, Y.; Roy, K.; Vidossich, P.; Cancedda, L.; De Vivo, M.; Forbush, B.; Cao, E. Structural Basis for Inhibition of the Cation-Chloride Cotransporter NKCC1 by the Diuretic Drug Bumetanide. Nat. Commun. 2022, 13 (1), 2747, DOI: 10.1038/s41467-022-30407-3Google Scholar15Structural basis for inhibition of the Cation-chloride cotransporter NKCC1 by the diuretic drug bumetanideZhao, Yongxiang; Roy, Kasturi; Vidossich, Pietro; Cancedda, Laura; De Vivo, Marco; Forbush, Biff; Cao, ErhuNature Communications (2022), 13 (1), 2747CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)Cation-chloride cotransporters (CCCs) NKCC1 and NKCC2 catalyze electroneutral symport of 1 Na+, 1 K+, and 2 Cl- across cell membranes. NKCC1 mediates trans-epithelial Cl- secretion and regulates excitability of some neurons and NKCC2 is crit. to renal salt reabsorption. Both transporters are inhibited by the so-called loop diuretics including bumetanide, and these drugs are a mainstay for treating edema and hypertension. Here, our single-particle electron cryo-microscopy structures supported by functional studies reveal an outward-facing conformation of NKCC1, showing bumetanide wedged into a pocket in the extracellular ion translocation pathway. Based on these and the previously published inward-facing structures, we define the translocation pathway and the conformational changes necessary for ion translocation. We also identify an NKCC1 dimer with sepd. transmembrane domains and extensive transmembrane and C-terminal domain interactions. We further define an N-terminal phosphoregulatory domain that interacts with the C-terminal domain, suggesting a mechanism whereby (de)phosphorylation regulates NKCC1 by tuning the strength of this domain assocn.
- 16Krishnamurthy, H.; Gouaux, E. X-Ray Structures of LeuT in Substrate-Free Outward-Open and Apo Inward-Open States. Nature 2012, 481 (7382), 469– 474, DOI: 10.1038/nature10737Google Scholar16X-ray structures of LeuT in substrate-free outward-open and apo inward-open statesKrishnamurthy, Harini; Gouaux, EricNature (London, United Kingdom) (2012), 481 (7382), 469-474CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Neurotransmitter sodium symporters are integral membrane proteins that remove chem. transmitters from the synapse and terminate neurotransmission mediated by serotonin, dopamine, noradrenaline, glycine and GABA (γ-aminobutyric acid). Crystal structures of the bacterial homolog, LeuT, in substrate-bound outward-occluded and competitive inhibitor-bound outward-facing states have advanced our mechanistic understanding of neurotransmitter sodium symporters but have left fundamental questions unanswered. Here we report crystal structures of LeuT mutants in complexes with conformation-specific antibody fragments in the outward-open and inward-open states. In the absence of substrate but in the presence of sodium the transporter is outward-open, illustrating how the binding of substrate closes the extracellular gate through local conformational changes: hinge-bending movements of the extracellular halves of transmembrane domains 1, 2 and 6, together with translation of extracellular loop 4. The inward-open conformation, by contrast, involves large-scale conformational changes, including a reorientation of transmembrane domains 1, 2, 5, 6 and 7, a marked hinge bending of transmembrane domain 1a and occlusion of the extracellular vestibule by extracellular loop 4. These changes close the extracellular gate, open an intracellular vestibule, and largely disrupt the two sodium sites, thus providing a mechanism by which ions and substrate are released to the cytoplasm. The new structures establish a structural framework for the mechanism of neurotransmitter sodium symporters and their modulation by therapeutic and illicit substances.
- 17del Alamo, D.; Meiler, J.; Mchaourab, H. S. Principles of Alternating Access in LeuT-Fold Transporters: Commonalities and Divergences. J. Mol. Biol. 2022, 434 (19), 167746 DOI: 10.1016/j.jmb.2022.167746Google Scholar17Principles of Alternating Access in LeuT-fold Transporters: Commonalities and Divergencesdel Alamo, Diego; Meiler, Jens; Mchaourab, Hassane S.Journal of Molecular Biology (2022), 434 (19), 167746CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)A review. Found in all domains of life, transporters belonging to the LeuT-fold class mediate the import and exchange of hydrophilic and charged compds. such as amino acids, metals, and sugar mols. Nearly two decades of studies on the eponymous bacterial transporter LeuT have yielded a library of high-resoln. snapshots of its conformational cycle linked by soln.-state exptl. data obtained from multiple techniques. In parallel, its topol. was obsd. in symporters and antiporters characterized by a spectrum of substrate specificities and coupled to gradients of distinct ions. Here the authors review and compare mechanistic models of transport for LeuT, its well-studied homologs, as well as functionally distant members of the fold, emphasizing the commonalities and divergences in alternating access and the corresponding energy landscapes. The authors' integrated summary illustrates how fold conservation, a hallmark of the LeuT fold, coincides with divergent choreogs. of alternating access that nevertheless capitalize on recurrent structural motifs. In addn., it highlights the knowledge gap that hinders the leveraging of the current body of research into detailed mechanisms of transport for this important class of membrane proteins.
- 18Bonati, L.; Rizzi, V.; Parrinello, M. Data-Driven Collective Variables for Enhanced Sampling. J. Phys. Chem. Lett. 2020, 11 (8), 2998– 3004, DOI: 10.1021/acs.jpclett.0c00535Google Scholar18Data-Driven Collective Variables for Enhanced SamplingBonati, Luigi; Rizzi, Valerio; Parrinello, MicheleJournal of Physical Chemistry Letters (2020), 11 (8), 2998-3004CODEN: JPCLCD; ISSN:1948-7185. (American Chemical Society)Designing an appropriate set of collective variables is crucial to the success of several enhanced sampling methods. Here we focus on how to obtain such variables from information limited to the metastable states. We characterize these states by a large set of descriptors and employ neural networks to compress this information in a lower-dimensional space, using Fisher's linear discriminant as an objective function to maximize the discriminative power of the network. We test this method on alanine dipeptide, using the nonlinearly separable data set composed by at. distances. We then study an intermol. aldol reaction characterized by a concerted mechanism. The resulting variables are able to promote sampling by drawing nonlinear paths in the phys. space connecting the fluctuations between metastable basins. Lastly, we interpret the behavior of the neural network by studying its relation to the phys. variables. Through the identification of its most relevant features, we are able to gain chem. insight into the process.
- 19Invernizzi, M.; Parrinello, M. Exploration vs Convergence Speed in Adaptive-Bias Enhanced Sampling. J. Chem. Theory Comput. 2022, 18 (6), 3988– 3996, DOI: 10.1021/acs.jctc.2c00152Google Scholar19Exploration vs Convergence Speed in Adaptive-Bias Enhanced SamplingInvernizzi, Michele; Parrinello, MicheleJournal of Chemical Theory and Computation (2022), 18 (6), 3988-3996CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)In adaptive-bias enhanced sampling methods, a bias potential is added to the system to drive transitions between metastable states. The bias potential is a function of a few collective variables and is gradually modified according to the underlying free energy surface. We show that when the collective variables are suboptimal, there is an exploration-convergence tradeoff, and one must choose between a quickly converging bias that will lead to fewer transitions or a slower to converge bias that can explore the phase space more efficiently but might require a much longer time to produce an accurate free energy est. The recently proposed on-the-fly probability enhanced sampling (OPES) method focuses on fast convergence, but there are cases where fast exploration is preferred instead. For this reason, we introduce a new variant of the OPES method that focuses on quickly escaping metastable states at the expense of convergence speed. We illustrate the benefits of this approach in prototypical systems and show that it outperforms the popular metadynamics method.
- 20Invernizzi, M.; Parrinello, M. Rethinking Metadynamics: From Bias Potentials to Probability Distributions. J. Phys. Chem. Lett. 2020, 11 (7), 2731– 2736, DOI: 10.1021/acs.jpclett.0c00497Google Scholar20Rethinking Metadynamics: From Bias Potentials to Probability DistributionsInvernizzi, Michele; Parrinello, MicheleJournal of Physical Chemistry Letters (2020), 11 (7), 2731-2736CODEN: JPCLCD; ISSN:1948-7185. (American Chemical Society)Metadynamics is an enhanced sampling method of great popularity, based on the on-the-fly construction of a bias potential that is a function of a selected no. of collective variables. We propose here a change in perspective that shifts the focus from the bias to the probability distribution reconstruction while retaining some of the key characteristics of metadynamics, such as flexible on-the-fly adjustments to the free energy est. The result is an enhanced sampling method that presents a drastic improvement in convergence speed, esp. when dealing with suboptimal and/or multidimensional sets of collective variables. The method is esp. robust and easy to use and in fact requires only a few simple parameters to be set, and it has a straightforward reweighting scheme to recover the statistics of the unbiased ensemble. Furthermore, it gives more control of the desired exploration of the phase space since the deposited bias is not allowed to grow indefinitely and it does not push the simulation to uninteresting high free energy regions. We demonstrate the performance of the method in a no. of representative examples.
- 21Yamashita, A.; Singh, S. K.; Kawate, T.; Jin, Y.; Gouaux, E. Crystal Structure of a Bacterial Homologue of Na+/Cl--Dependent Neurotransmitter Transporters. Nature 2005, 437 (7056), 215– 223, DOI: 10.1038/nature03978Google Scholar21Crystal structure of a bacterial homologue of Na+/Cl--dependent neurotransmitter transportersYamashita, Atsuko; Singh, Satinder K.; Kawate, Toshimitsu; Jin, Yan; Gouaux, EricNature (London, United Kingdom) (2005), 437 (7056), 215-223CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Na+/Cl--dependent transporters terminate synaptic transmission by using electrochem. gradients to drive the uptake of neurotransmitters, including the biogenic amines, from the synapse to the cytoplasm of neurons and glia. These transporters are the targets of therapeutic and illicit compds., and their dysfunction has been implicated in multiple diseases of the nervous system. Here we present the crystal structure of a bacterial homolog of these transporters from Aquifex aeolicus, in complex with its substrate, leucine, and two sodium ions. The protein core consists of the first ten of twelve transmembrane segments, with segments 1-5 related to 6-10 by a pseudo-two-fold axis in the membrane plane. Leucine and the sodium ions are bound within the protein core, halfway across the membrane bilayer, in an occluded site devoid of water. The leucine and ion binding sites are defined by partially unwound transmembrane helixes, with main-chain atoms and helix dipoles having key roles in substrate and ion binding. The structure reveals the architecture of this important class of transporter, illuminates the determinants of substrate binding and ion selectivity, and defines the external and internal gates.
- 22Forrest, L. R.; Rudnick, G. The Rocking Bundle: A Mechanism for Ion-Coupled Solute Flux by Symmetrical Transporters. Physiology 2009, 24 (6), 377– 386, DOI: 10.1152/physiol.00030.2009Google Scholar22The rocking bundle: a mechanism for ion-coupled solute flux by symmetrical transportersForrest, Lucy R.; Rudnick, GaryPhysiology (2009), 24 (Dec.), 377-386CODEN: PHYSCI; ISSN:1548-9213. (International Union of Physiological Sciences)A review. Crystal structures of the bacterial amino acid transporter LeuT have provided the basis for understanding the conformational changes assocd. with substrate translocation by a multitude of transport proteins with the same fold. Biochem. and modeling studies led to a "rocking bundle" mechanism for LeuT that was validated by subsequent transporter structures. These advances suggest how coupled solute transport might be defined by the internal symmetry of proteins contg. inverted structural repeats.
- 23Masrati, G.; Mondal, R.; Rimon, A.; Kessel, A.; Padan, E.; Lindahl, E.; Ben-Tal, N. An Angular Motion of a Conserved Four-Helix Bundle Facilitates Alternating Access Transport in the TtNapA and EcNhaA Transporters. Proc. Natl. Acad. Sci. U. S. A. 2020, 117 (50), 31850– 31860, DOI: 10.1073/pnas.2002710117Google Scholar23An angular motion of a conserved four-helix bundle facilitates alternating access transport in the TtNapA and EcNhaA transportersMasrati, Gal; Mondal, Ramakanta; Rimon, Abraham; Kessel, Amit; Padan, Etana; Lindahl, Erik; Ben-Tal, NirProceedings of the National Academy of Sciences of the United States of America (2020), 117 (50), 31850-31860CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)There is ongoing debate regarding the mechanism through which cation/proton antiporters (CPAs), like Thermus thermophilus NapA (TtNapA) and Escherichia coli NapA (EcNhaA), alternate between their outward- and inward-facing conformations in the membrane. CPAs comprise two domains, and it is unclear whether the transition is driven by their rocking-bundle or elevator motion with respect to each other. Here we address this question using metadynamics simulations of TtNapA, where we bias conformational sampling along two axes characterizing the two proposed mechanisms: angular and translational motions, resp. By applying the bias potential for the two axes simultaneously, as well as to the angular, but not the translational, axis alone, we manage to reproduce each of the two known states of TtNapA when starting from the opposite state, in support of the rocking-bundle mechanism as the driver of conformational change. Next, starting from the inward-facing conformation of EcNhaA, we sample what could be its long-sought-after outward-facing conformation and verify it using crosslinking expts.
- 24Borgogno, M.; Savardi, A.; Manigrasso, J.; Turci, A.; Portioli, C.; Ottonello, G.; Bertozzi, S. M.; Armirotti, A.; Contestabile, A.; Cancedda, L.; De Vivo, M. Design, Synthesis, In Vitro and In Vivo Characterization of Selective NKCC1 Inhibitors for the Treatment of Core Symptoms in Down Syndrome. J. Med. Chem. 2021, 64 (14), 10203– 10229, DOI: 10.1021/acs.jmedchem.1c00603Google Scholar24Design, Synthesis, In Vitro and In Vivo Characterization of Selective NKCC1 Inhibitors for the Treatment of Core Symptoms in Down SyndromeBorgogno, Marco; Savardi, Annalisa; Manigrasso, Jacopo; Turci, Alessandra; Portioli, Corinne; Ottonello, Giuliana; Bertozzi, Sine Mandrup; Armirotti, Andrea; Contestabile, Andrea; Cancedda, Laura; De Vivo, MarcoJournal of Medicinal Chemistry (2021), 64 (14), 10203-10229CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Intracellular chloride concn. [Cl-]i is defective in several neurol. disorders. In neurons, [Cl-]i is mainly regulated by the action of the Na+-K+-Cl- importer NKCC1 and the K+-Cl- exporter KCC2. Recently, we have reported the discovery of ARN23746(I) as the lead candidate of a novel class of selective inhibitors of NKCC1. Importantly, ARN23746 is able to rescue core symptoms of Down syndrome (DS) and autism in mouse models. Here, we describe the discovery and extensive characterization of this chem. class of selective NKCC1 inhibitors, with focus on ARN23746 and other promising derivs. In particular, we present compd. 40 (ARN24092)(II) as a backup/follow-up lead with in vivo efficacy in a mouse model of DS. These results further strengthen the potential of this new class of compds. for the treatment of core symptoms of brain disorders characterized by the defective NKCC1/KCC2 expression ratio.
- 25Savardi, A.; Borgogno, M.; Narducci, R.; La Sala, G.; Ortega, J. A.; Summa, M.; Armirotti, A.; Bertorelli, R.; Contestabile, A.; De Vivo, M.; Cancedda, L. Discovery of a Small Molecule Drug Candidate for Selective NKCC1 Inhibition in Brain Disorders. Chem. 2020, 6 (8), 2073– 2096, DOI: 10.1016/j.chempr.2020.06.017Google Scholar25Discovery of a Small Molecule Drug Candidate for Selective NKCC1 Inhibition in Brain DisordersSavardi, Annalisa; Borgogno, Marco; Narducci, Roberto; La Sala, Giuseppina; Ortega, Jose Antonio; Summa, Maria; Armirotti, Andrea; Bertorelli, Rosalia; Contestabile, Andrea; De Vivo, Marco; Cancedda, LauraChem (2020), 6 (8), 2073-2096CODEN: CHEMVE; ISSN:2451-9294. (Cell Press)Aberrant expression ratio of Cl- transporters, NKCC1 and KCC2, is implicated in several brain conditions. NKCC1 inhibition by the FDA-approved diuretic drug, bumetanide, rescues core symptoms in rodent models and/or clin. trials with patients. However, bumetanide has a strong diuretic effect due to inhibition of the kidney Cl- transporter NKCC2, creating crit. drug compliance issues and health concerns. Here, we report the discovery of a new chem. class of selective NKCC1 inhibitors and the lead drug candidate ARN23746. ARN23746 restores the physiol. intracellular Cl- in murine Down syndrome neuronal cultures, has excellent soly. and metabolic stability, and displays no issues with off-target activity in vitro. ARN23746 recovers core symptoms in mouse models of Down syndrome and autism, with no diuretic effect, nor overt toxicity upon chronic treatment in adulthood. ARN23746 is ready for advanced preclin./manufg. studies toward the first sustainable therapeutics for the neurol. conditions characterized by impaired Cl- homeostasis.
- 26Savardi, A.; Patricelli Malizia, A.; De Vivo, M.; Cancedda, L.; Borgogno, M. Preclinical Development of the Na-K-2Cl Co-Transporter-1 (NKCC1) Inhibitor ARN23746 for the Treatment of Neurodevelopmental Disorders. ACS Pharmacol. Transl. Sci. 2023, 6 (1), 1– 11, DOI: 10.1021/acsptsci.2c00197Google Scholar26Preclinical Development of the Na-K-2Cl Co-transporter-1 (NKCC1) Inhibitor ARN23746 for the Treatment of Neurodevelopmental DisordersSavardi, Annalisa; Patricelli Malizia, Andrea; De Vivo, Marco; Cancedda, Laura; Borgogno, MarcoACS Pharmacology & Translational Science (2023), 6 (1), 1-11CODEN: APTSFN; ISSN:2575-9108. (American Chemical Society)Alterations in the expression of the Cl- importer Na-K-2Cl co-transporter-1 (NKCC1) and the exporter K-Cl co-transporter 2 (KCC2) lead to impaired intracellular chloride concn. in neurons and imbalanced excitation/inhibition in the brain. These alterations have been obsd. in several neurol. disorders (e.g., Down syndrome and autism). Recently, we have reported the discovery of the selective NKCC1 inhibitor "compd. ARN23746" for the treatment of Down syndrome and autism in mouse models. Here, we report on an extensive preclin. characterization of ARN23746 toward its development as a clin. candidate. ARN23746 shows an overall excellent metab. profile and good brain penetration. Moreover, ARN23746 is effective in rescuing cognitive impairment in Down syndrome mice upon per os administration, in line with oral treatment of neurodevelopmental disorders. Notably, ARN23746 does not present signs of toxicity or diuresis even if administered up to 50 times the ED. These results further support ARN23746 as a solid candidate for clin. trial-enabling studies.
- 27Chew, T. A.; Zhang, J.; Feng, L. High-Resolution Views and Transport Mechanisms of the NKCC1 and KCC Transporters. J. Mol. Biol. 2021, 433 (16), 167056 DOI: 10.1016/j.jmb.2021.167056Google Scholar27High-Resolution Views and Transport Mechanisms of the NKCC1 and KCC TransportersChew, Thomas A.; Zhang, Jinru; Feng, LiangJournal of Molecular Biology (2021), 433 (16), 167056CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)A review. Cation-chloride cotransporters (CCCs) are responsible for the coupled co-transport of Cl- with K+ and/or Na+ in an electroneutral manner. They play important roles in myriad fundamental physiol. processes--from cell vol. regulation to transepithelial solute transport and intracellular ion homeostasis--and are targeted by medicines commonly prescribed to treat hypertension and edema. After several decades of studies into the functions and pharmacol. of these transporters, there have been several breakthroughs in the structural detn. of CCC transporters. The insights provided by these new structures for the Na+/K+/Cl- cotransporter NKCC1 and the K+/Cl- cotransporters KCC1, KCC2, KCC3 and KCC4 have deepened our understanding of their mol. basis and transport function. This focused discusses recent advances in the structural and mechanistic understanding of CCC transporters, including architecture, dimerization, functional roles of regulatory domains, ion binding sites, and coupled ion transport.
- 28Lytle, C.; McManus, T. J.; Haas, M. A Model of Na-K-2Cl Cotransport Based on Ordered Ion Binding and Glide Symmetry. Am. J. Physiol.-Cell Physiol. 1998, 274 (2), C299– C309, DOI: 10.1152/ajpcell.1998.274.2.C299Google ScholarThere is no corresponding record for this reference.
- 29Delpire, E.; Gagnon, K. B. Na + -K + −2Cl – Cotransporter (NKCC) Physiological Function in Nonpolarized Cells and Transporting Epithelia. In Comprehensive Physiology; Terjung, R., Ed.; Wiley, 2018; pp. 871– 901.Google ScholarThere is no corresponding record for this reference.
- 30Shimamura, T.; Weyand, S.; Beckstein, O.; Rutherford, N. G.; Hadden, J. M.; Sharples, D.; Sansom, M. S. P.; Iwata, S.; Henderson, P. J. F.; Cameron, A. D. Molecular Basis of Alternating Access Membrane Transport by the Sodium-Hydantoin Transporter Mhp1. Science 2010, 328 (5977), 470– 473, DOI: 10.1126/science.1186303Google Scholar30Molecular Basis of Alternating Access Membrane Transport by the Sodium-Hydantoin Transporter Mhp1Shimamura, Tatsuro; Weyand, Simone; Beckstein, Oliver; Rutherford, Nicholas G.; Hadden, Jonathan M.; Sharples, David; Sansom, Mark S. P.; Iwata, So; Henderson, Peter J. F.; Cameron, Alexander D.Science (Washington, DC, United States) (2010), 328 (5977), 470-473CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The structure of the sodium-benzylhydantoin transport protein Mhp1 from Microbacterium liquefaciens is comprised of a five-helix inverted repeat, a conformation widespread among secondary transporters. Here, we report the crystal structure of an inward-facing conformation of Mhp1 at 3.8 angstroms resoln., which complements previously described structures of Mhp1 in outward-facing and occluded states. From analyses of the three structures and mol. dynamics (MD) simulations, we propose a mechanism for the transport cycle in Mhp1. Switching from the outward- to the inward-facing state, to effect the inward release of sodium and benzylhydantoin, is primarily achieved by a rigid body movement of transmembrane helixes 3, 4, 8, and 9 relative to the rest of the protein. This forms the basis of an alternating access mechanism applicable to many transporters of this emerging superfamily.
- 31Hamann, S.; Herrera-Perez, J. J.; Zeuthen, T.; Alvarez-Leefmans, F. J. Cotransport of Water by the Na + -K + −2Cl – Cotransporter NKCC1 in Mammalian Epithelial Cells: Cotransport of Water by NKCC1. J. Physiol. 2010, 588 (21), 4089– 4101, DOI: 10.1113/jphysiol.2010.194738Google Scholar31Cotransport of water by the Na+-K+-2Cl- cotransporter NKCC1 in mammalian epithelial cellsHamann, Steffen; Herrera-Perez, Jose J.; Zeuthen, Thomas; Alvarez-Leefmans, Francisco J.Journal of Physiology (Oxford, United Kingdom) (2010), 588 (21), 4089-4101CODEN: JPHYA7; ISSN:0022-3751. (Wiley-Blackwell)Water transport by the Na+-K+-2Cl- cotransporter (NKCC1) was studied in confluent cultures of pigmented epithelial (PE) cells from the ciliary body of the fetal human eye. Interdependence among water, Na+ and Cl- fluxes mediated by NKCC1 was inferred from changes in cell water vol., monitored by intracellular self-quenching of the fluorescent dye calcein. Isosmotic removal of external Cl- or Na+ caused a rapid efflux of water from the cells, which was inhibited by bumetanide (10 μM). When returned to the control soln. there was a rapid water influx that required the simultaneous presence of external Na+ and Cl-. The water influx could proceed uphill, against a transmembrane osmotic gradient, suggesting that energy contained in the ion fluxes can be transferred to the water flux. The influx of water induced by changes in external [Cl-] satd. in a sigmoidal fashion with a Km of 60 mM, while that induced by changes in external [Na+] followed first order kinetics with a Km of about 40 mM. These parameters are consistent with ion transport mediated by NKCC1. Our findings support a previous investigation, in which we showed water transport by NKCC1 to be a result of a balance between ionic and osmotic gradients. The coupling between salt and water transport in NKCC1 represents a novel aspect of cellular water homeostasis where cells can change their vol. independently of the direction of an osmotic gradient across the membrane. This has relevance for both epithelial and sym. cells.
- 32Zeuthen, T.; MacAulay, N. Cotransport of Water by Na + -K + −2Cl – Cotransporters Expressed in Xenopus Oocytes: NKCC1 versus NKCC2: Water Transport in NKCC. J. Physiol. 2012, 590 (5), 1139– 1154, DOI: 10.1113/jphysiol.2011.226316Google Scholar32Cotransport of water by Na+-K+-2Cl- cotransporters expressed in Xenopus oocytes: NKCC1 versus NKCC2Zeuthen, Thomas; MacAulay, NannaJournal of Physiology (Oxford, United Kingdom) (2012), 590 (5), 1139-1154CODEN: JPHYA7; ISSN:0022-3751. (Wiley-Blackwell)The NKCC1 and NKCC2 isoforms of the mammalian Na+-K+-2Cl- cotransporter were expressed in Xenopus oocytes and the relation between external ion concn. and water fluxes detd. Water fluxes were detd. from changes in the oocytes vol. and ion fluxes from 86Rb+ uptake. Isotonic increases in external K+ concn. elicited abrupt inward water fluxes in NKCC1; the K+ dependence obeyed one-site kinetics with a K0.5 of 7.5 mM. The water fluxes were blocked by bumetanide, had steep temp. dependence and could proceed uphill against an osmotic gradient of 20 mosmol L-1. A comparison between ion and water fluxes indicates that 460 water mols. are cotransported for each turnover of the protein. In contrast, NKCC2 did not support water fluxes. Water transport in NKCC1 induced by increases in the external osmolarity had high activation energy and was blocked by bumetanide. The osmotic effects of NaCl were smaller than those of urea and mannitol. This supports the notion of interaction between ions and water in NKCC1 and allows for an est. of around 600 water mols. transported per turnover of the protein. Osmotic gradients did not induce water transport in NKCC2. We conclude that NKCC1 plays a direct role for water balance in most cell types, while NKCC2 fulfills its role in the kidney of transporting ions but not water. The different behavior of NKCC1 and NKCC2 is discussed on the basis of recent mol. models based on studies of structural and mol. dynamics.
- 33Sadegh, C.; Xu, H.; Sutin, J.; Fatou, B.; Gupta, S.; Pragana, A.; Taylor, M.; Kalugin, P. N.; Zawadzki, M. E.; Alturkistani, O.; Shipley, F. B.; Dani, N.; Fame, R. M.; Wurie, Z.; Talati, P.; Schleicher, R. L.; Klein, E. M.; Zhang, Y.; Holtzman, M. J.; Moore, C. I.; Lin, P.-Y.; Patel, A. B.; Warf, B. C.; Kimberly, W. T.; Steen, H.; Andermann, M. L.; Lehtinen, M. K. Choroid Plexus-Targeted NKCC1 Overexpression to Treat Post-Hemorrhagic Hydrocephalus. Neuron 2023, 111 (10), 1591– 1608, DOI: 10.1016/j.neuron.2023.02.020Google Scholar33Choroid plexus-targeted NKCC1 overexpression to treat post-hemorrhagic hydrocephalusSadegh, Cameron; Xu, Huixin; Sutin, Jason; Fatou, Benoit; Gupta, Suhasini; Pragana, Aja; Taylor, Milo; Kalugin, Peter N.; Zawadzki, Miriam E.; Alturkistani, Osama; Shipley, Frederick B.; Dani, Neil; Fame, Ryann M.; Wurie, Zainab; Talati, Pratik; Schleicher, Riana L.; Klein, Eric M.; Zhang, Yong; Holtzman, Michael J.; Moore, Christopher I.; Lin, Pei-Yi; Patel, Aman B.; Warf, Benjamin C.; Kimberly, W. Taylor; Steen, Hanno; Andermann, Mark L.; Lehtinen, Maria K.Neuron (2023), 111 (10), 1591-1608.e4CODEN: NERNET; ISSN:0896-6273. (Cell Press)Post-hemorrhagic hydrocephalus (PHH) refers to a life-threatening accumulation of cerebrospinal fluid (CSF) that occurs following intraventricular hemorrhage (IVH). An incomplete understanding of this variably progressive condition has hampered the development of new therapies beyond serial neurosurgical interventions. Here, we show a key role for the bidirectional Na-K-Cl cotransporter, NKCC1, in the choroid plexus (ChP) to mitigate PHH. Mimicking IVH with intraventricular blood led to increased CSF [K+] and triggered cytosolic calcium activity in ChP epithelial cells, which was followed by NKCC1 activation. ChP-targeted adeno-assocd. viral (AAV)-NKCC1 prevented blood-induced ventriculomegaly and led to persistently increased CSF clearance capacity. These data demonstrate that intraventricular blood triggered a trans-choroidal, NKCC1-dependent CSF clearance mechanism. Inactive, phosphodeficient AAV-NKCC1-NT51 failed to mitigate ventriculomegaly. Excessive CSF [K+] fluctuations correlated with permanent shunting outcome in humans following hemorrhagic stroke, suggesting targeted gene therapy as a potential treatment to mitigate intracranial fluid accumulation following hemorrhage.
- 34Steffensen, A. B.; Oernbo, E. K.; Stoica, A.; Gerkau, N. J.; Barbuskaite, D.; Tritsaris, K.; Rose, C. R.; MacAulay, N. Cotransporter-Mediated Water Transport Underlying Cerebrospinal Fluid Formation. Nat. Commun. 2018, 9 (1), 2167, DOI: 10.1038/s41467-018-04677-9Google Scholar34Cotransporter-mediated water transport underlying cerebrospinal fluid formationSteffensen Annette B; Oernbo Eva K; Stoica Anca; MacAulay Nanna; Gerkau Niklas J; Rose Christine R; Barbuskaite Dagne; Tritsaris KaterinaNature communications (2018), 9 (1), 2167 ISSN:.Cerebrospinal fluid (CSF) production occurs at a rate of 500 ml per day in the adult human. Conventional osmotic forces do not suffice to support such production rate and the molecular mechanisms underlying this fluid production remain elusive. Using ex vivo choroid plexus live imaging and isotope flux in combination with in vivo CSF production determination in mice, we identify a key component in the CSF production machinery. The Na(+)/K(+)/2Cl(-) cotransporter (NKCC1) expressed in the luminal membrane of choroid plexus contributes approximately half of the CSF production, via its unusual outward transport direction and its unique ability to directly couple water transport to ion translocation. We thereby establish the concept of cotransport of water as a missing link in the search for molecular pathways sustaining CSF production and redefine the current model of this pivotal physiological process. Our results provide a rational pharmacological target for pathologies involving disturbed brain fluid dynamics.
- 35Li, J.; Shaikh, S. A.; Enkavi, G.; Wen, P.-C.; Huang, Z.; Tajkhorshid, E. Transient Formation of Water-Conducting States in Membrane Transporters. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (19), 7696– 7701, DOI: 10.1073/pnas.1218986110Google Scholar35Transient formation of water-conducting states in membrane transportersLi, Jing; Shaikh, Saher A.; Enkavi, Giray; Wen, Po-Chao; Huang, Zhijian; Tajkhorshid, EmadProceedings of the National Academy of Sciences of the United States of America (2013), 110 (19), 7696-7701, S7696/1-S7696/7CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Membrane transporters rely on highly coordinated structural transitions between major conformational states for their function, to prevent simultaneous access of the substrate binding site to both sides of the membrane - a mode of operation known as the alternating access model. Although this mechanism successfully accounts for the efficient exchange of the primary substrate across the membrane, accruing evidence on significant water transport and even uncoupled ion transport mediated by transporters has challenged the concept of perfect mech. coupling and coordination of the gating mechanism in transporters, which might be expected from the alternating access model. Here, we present a large set of extended equil. mol. dynamics simulations performed on several classes of membrane transporters in different conformational states, to test the presence of the phenomenon in diverse transporter classes and to investigate the underlying mol. mechanism of water transport through membrane transporters. The simulations reveal spontaneous formation of transient water-conducting (channel-like) states allowing passive water diffusion through the lumen of the transporters. These channel-like states are permeable to water but occluded to substrate, thereby not hindering the uphill transport of the primary substrate, i.e., the alternating access model remains applicable to the substrate. The rise of such water-conducting states during the large-scale structural transitions of the transporter protein is indicative of imperfections in the coordinated closing and opening motions of the cytoplasmic and extracellular gates. We propose that the obsd. water-conducting states likely represent a universal phenomenon in membrane transporters, which is consistent with their reliance on large-scale motion for function.
- 36Okazaki, K.; Wöhlert, D.; Warnau, J.; Jung, H.; Yildiz, Ö.; Kühlbrandt, W.; Hummer, G. Mechanism of the Electroneutral Sodium/Proton Antiporter PaNhaP from Transition-Path Shooting. Nat. Commun. 2019, 10 (1), 1742, DOI: 10.1038/s41467-019-09739-0Google Scholar36Mechanism of the electroneutral sodium/proton antiporter PaNhaP from transition-path shootingOkazaki Kei-Ichi; Okazaki Kei-Ichi; Warnau Judith; Jung Hendrik; Hummer Gerhard; Wohlert David; Yildiz Ozkan; Kuhlbrandt Werner; Hummer GerhardNature communications (2019), 10 (1), 1742 ISSN:.Na(+)/H(+) antiporters exchange sodium ions and protons on opposite sides of lipid membranes. The electroneutral Na(+)/H(+) antiporter NhaP from archaea Pyrococcus abyssi (PaNhaP) is a functional homolog of the human Na(+)/H(+) exchanger NHE1, which is an important drug target. Here we resolve the Na(+) and H(+) transport cycle of PaNhaP by transition-path sampling. The resulting molecular dynamics trajectories of repeated ion transport events proceed without bias force, and overcome the enormous time-scale gap between seconds-scale ion exchange and microseconds simulations. The simulations reveal a hydrophobic gate to the extracellular side that opens and closes in response to the transporter domain motion. Weakening the gate by mutagenesis makes the transporter faster, suggesting that the gate balances competing demands of fidelity and efficiency. Transition-path sampling and a committor-based reaction coordinate optimization identify the essential motions and interactions that realize conformational alternation between the two access states in transporter function.
- 37Monette, M. Y.; Somasekharan, S.; Forbush, B. Molecular Motions Involved in Na-K-Cl Cotransporter-Mediated Ion Transport and Transporter Activation Revealed by Internal Cross-Linking between Transmembrane Domains 10 and 11/12. J. Biol. Chem. 2014, 289 (11), 7569– 7579, DOI: 10.1074/jbc.M113.542258Google Scholar37Molecular Motions Involved in Na-K-Cl Cotransporter-mediated Ion Transport and Transporter Activation Revealed by Internal Cross-linking between Transmembrane Domains 10 and 11/12Monette, Michelle Y.; Somasekharan, Suma; Forbush, BiffJournal of Biological Chemistry (2014), 289 (11), 7569-7579CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)We examd. the relationship between transmembrane domain (TM) 10 and TM11/12 in NKCC1, testing homol. models based on the structure of AdiC in the same transporter superfamily. We hypothesized that introduced cysteine pairs would be close enough for disulfide formation and would alter transport function; indeed, evidence for cross-link formation with low micromolar concns. of copper phenanthroline or iodine was found in 3 of 8 initially tested pairs and in 1 of 26 addnl. tested pairs. Inhibition of transport was obsd. with copper phenanthroline and iodine treatment of P676C/A734C and I677C/A734C, consistent with the proximity of these residues and with movement of TM10 during the occlusion step of ion transport. We also found Cu2+ inhibition of the single-cysteine mutant A675C, suggesting that this residue and Met382 of TM3 are involved in a Cu2+-binding site. Surprisingly, crosslinking of P676C/I730C was found to prevent rapid deactivation of the transporter while not affecting the dephosphorylation rate, thus uncoupling the phosphorylation and activation steps. Consistent with this, (a) crosslinking of P676C/I730C was dependent on activation state, and (b) mutants lacking the phosphoregulatory domain could still be activated by crosslinking. These results suggest a model of NKCC activation that involves movement of TM12 relative to TM10, which is likely tied to movement of the large C terminus, a process somehow triggered by phosphorylation of the regulatory domain in the N terminus.
- 38McNeill, A.; Iovino, E.; Mansard, L.; Vache, C.; Baux, D.; Bedoukian, E.; Cox, H.; Dean, J.; Goudie, D.; Kumar, A.; Newbury-Ecob, R.; Fallerini, C.; Renieri, A.; Lopergolo, D.; Mari, F.; Blanchet, C.; Willems, M.; Roux, A.-F.; Pippucci, T.; Delpire, E. SLC12A2 Variants Cause a Neurodevelopmental Disorder or Cochleovestibular Defect. Brain 2020, 143 (8), 2380– 2387, DOI: 10.1093/brain/awaa176Google Scholar38SLC12A2 variants cause a neurodevelopmental disorder or cochleovestibular defectMcNeill Alisdair; McNeill Alisdair; McNeill Alisdair; Iovino Emanuela; Mansard Luke; Vache Christel; Baux David; Roux Anne-Francoise; Bedoukian Emma; Cox Helen; Dean John; Goudie David; Kumar Ajith; Newbury-Ecob Ruth; Fallerini Chiara; Renieri Alessandra; Lopergolo Diego; Mari Francesca; Fallerini Chiara; Renieri Alessandra; Lopergolo Diego; Mari Francesca; Blanchet Catherine; Willems Marjolaine; Pippucci Tommaso; Delpire EricBrain : a journal of neurology (2020), 143 (8), 2380-2387 ISSN:.The SLC12 gene family consists of SLC12A1-SLC12A9, encoding electroneutral cation-coupled chloride co-transporters. SCL12A2 has been shown to play a role in corticogenesis and therefore represents a strong candidate neurodevelopmental disorder gene. Through trio exome sequencing we identified de novo mutations in SLC12A2 in six children with neurodevelopmental disorders. All had developmental delay or intellectual disability ranging from mild to severe. Two had sensorineural deafness. We also identified SLC12A2 variants in three individuals with non-syndromic bilateral sensorineural hearing loss and vestibular areflexia. The SLC12A2 de novo mutation rate was demonstrated to be significantly elevated in the deciphering developmental disorders cohort. All tested variants were shown to reduce co-transporter function in Xenopus laevis oocytes. Analysis of SLC12A2 expression in foetal brain at 16-18 weeks post-conception revealed high expression in radial glial cells, compatible with a role in neurogenesis. Gene co-expression analysis in cells robustly expressing SLC12A2 at 16-18 weeks post-conception identified a transcriptomic programme associated with active neurogenesis. We identify SLC12A2 de novo mutations as the cause of a novel neurodevelopmental disorder and bilateral non-syndromic sensorineural hearing loss and provide further data supporting a role for this gene in human neurodevelopment.
- 39Janoš, P.; Magistrato, A. All-Atom Simulations Uncover the Molecular Terms of the NKCC1 Transport Mechanism. J. Chem. Inf. Model. 2021, 61 (7), 3649– 3658, DOI: 10.1021/acs.jcim.1c00551Google Scholar39All-Atom Simulations Uncover the Molecular Terms of the NKCC1 Transport MechanismJanos, Pavel; Magistrato, AlessandraJournal of Chemical Information and Modeling (2021), 61 (7), 3649-3658CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)The secondary-active Na-K-Cl cotransporter 1 (NKCC1), member of the cation-chloride cotransporter (CCC) family, ensures the electroneutral movement of Cl-, Na+, and K+ ions across cellular membranes. NKCC1 regulates Cl- homeostasis and cell vol., handling a pivotal role in transepithelial water transport and neuronal excitability. Aberrant NKCC1 transport is hence implicated in a variety of human diseases (hypertension, renal disorders, neuropathies, and cancer). Building on the newly resolved NKCC1 cryo-EM structure, all-atom enhanced sampling simulations unprecedentedly unlock the mechanism of NKCC1-mediated ion transport, assessing the order and the mol. basis of its interdependent ion translocation. The authors' outcomes strikingly advance the understanding of the physiol. mechanism of CCCs and disclose a key role of CCC-conserved asparagine residues, whose side-chain promiscuity ensures the transport of both neg. and pos. charged ions along the same translocation route. This study sets a conceptual basis to devise NKCC-selective inhibitors to treat diseases linked to Cl- dishomeostasis.
- 40Portioli, C.; Ruiz Munevar, M. J.; De Vivo, M.; Cancedda, L. Cation-Coupled Chloride Cotransporters: Chemical Insights and Disease Implications. Trends Chem. 2021, 3 (10), 832– 849, DOI: 10.1016/j.trechm.2021.05.004Google Scholar40Cation-coupled chloride cotransporters: chemical insights and disease implicationsPortioli, Corinne; Ruiz Munevar, Manuel Jose; De Vivo, Marco; Cancedda, LauraTrends in Chemistry (2021), 3 (10), 832-849CODEN: TCRHBQ; ISSN:2589-5974. (Cell Press)A review. Cation-coupled chloride cotransporters (CCCs) modulate the transport of sodium and/or potassium cations coupled with chloride anions across the cell membrane. CCCs thus help regulate intracellular ionic concn. and consequent cell vol. homeostasis. This has been largely exploited in the past to develop diuretic drugs that act on CCCs expressed in the kidney. However, a growing wealth of evidence has demonstrated that CCCs are also critically involved in a great variety of other pathologies, motivating most recent drug discovery programs targeting CCCs. Here, we examine the structure-function relationship of CCCs. By linking recent high-resoln. cryogenic electron microscopy (cryo-EM) data with older biochem./functional studies on CCCs, we discuss the mechanistic insights and opportunities to design selective CCC modulators to treat diverse pathologies.
- 41Zhao, Y.; Shen, J.; Wang, Q.; Ruiz Munevar, M. J.; Vidossich, P.; De Vivo, M.; Zhou, M.; Cao, E. Structure of the Human Cation–Chloride Cotransport KCC1 in an Outward-Open State. Proc. Natl. Acad. Sci. U. S. A. 2022, 119 (27), e2109083119 DOI: 10.1073/pnas.2109083119Google Scholar41Structure of the human cation-chloride cotransport KCC1 in an outward-open stateZhao, Yongxiang; Shen, Jiemin; Wang, Qinzhe; Munevar, Manuel Jose Ruiz; Vidossich, Pietro; De Vivo, Marco; Zhou, Ming; Cao, ErhuProceedings of the National Academy of Sciences of the United States of America (2022), 119 (27), e2109083119CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Cation-chloride cotransporters (CCCs) catalyze electroneutral symport of Cl- with Na+ and/or K+ across membranes. CCCs are fundamental in cell vol. homeostasis, transepithelia ion movement, maintenance of intracellular Cl- concn., and neuronal excitability. Here, we present a cryoelectron microscopy structure of human K+-Cl- cotransporter (KCC)1 bound with the VU0463271 inhibitor in an outward-open state. In contrast to many other amino acid-polyamine-organocation transporter cousins, our first outward-open CCC structure reveals that opening the KCC1 extracellular ion permeation path does not involve hinge-bending motions of the transmembrane (TM) 1 and TM6 half-helixes. Instead, rocking of TM3 and TM8, together with displacements of TM4, TM9, and a conserved intracellular loop 1 helix, underlie alternate opening and closing of extracellular and cytoplasmic vestibules. We show that KCC1 intriguingly exists in one of two distinct dimeric states via different intersubunit interfaces. Our studies provide a blueprint for understanding the mechanisms of CCCs and their inhibition by small mol. compds.
- 42Saitsu, H.; Watanabe, M.; Akita, T.; Ohba, C.; Sugai, K.; Ong, W. P.; Shiraishi, H.; Yuasa, S.; Matsumoto, H.; Beng, K. T.; Saitoh, S.; Miyatake, S.; Nakashima, M.; Miyake, N.; Kato, M.; Fukuda, A.; Matsumoto, N. Impaired Neuronal KCC2 Function by Biallelic SLC12A5Mutations in Migrating Focal Seizures and Severe Developmental Delay. Sci. Rep. 2016, 6 (1), 30072, DOI: 10.1038/srep30072Google Scholar42Impaired neuronal KCC2 function by biallelic SLC12A5 mutations in migrating focal seizures and severe developmental delaySaitsu, Hirotomo; Watanabe, Miho; Akita, Tenpei; Ohba, Chihiro; Sugai, Kenji; Ong, Winnie Peitee; Shiraishi, Hideaki; Yuasa, Shota; Matsumoto, Hiroshi; Beng, Khoo Teik; Saitoh, Shinji; Miyatake, Satoko; Nakashima, Mitsuko; Miyake, Noriko; Kato, Mitsuhiro; Fukuda, Atsuo; Matsumoto, NaomichiScientific Reports (2016), 6 (), 30072CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)Epilepsy of infancy with migrating focal seizures (EIMFS) is one of the early-onset epileptic syndromes characterized by migrating polymorphous focal seizures. Whole exome sequencing (WES) in ten sporadic and one familial case of EIMFS revealed compd. heterozygous SLC12A5 (encoding the neuronal K+-Cl- co-transporter KCC2) mutations in two families: c.279 + 1G > C causing skipping of exon 3 in the transcript (p.E50_Q93del) and c.572 C >T (p.A191V) in individuals 1 and 2, and c.967T > C (p.S323P) and c.1243 A > G (p.M415V) in individual 3. Another patient (individual 4) with migrating multifocal seizures and compd. heterozygous mutations [c.953G > C (p.W318S) and c.2242_2244del (p.S748del)] was identified by searching WES data from 526 patients and SLC12A5-targeted resequencing data from 141 patients with infantile epilepsy. Gramicidin-perforated patch-clamp anal. demonstrated strongly suppressed Cl- extrusion function of E50_Q93del and M415V mutants, with mildly impaired function of A191V and S323P mutants. Cell surface expression levels of these KCC2 mutants were similar to wildtype KCC2. Heterologous expression of two KCC2 mutants, mimicking the patient status, produced a significantly greater intracellular Cl- level than with wildtype KCC2, but less than without KCC2. These data clearly demonstrated that partially disrupted neuronal Cl- extrusion, mediated by two types of differentially impaired KCC2 mutant in an individual, causes EIMFS.
- 43Uyanik, G.; Elcioglu, N.; Penzien, J.; Gross, C.; Yilmaz, Y.; Olmez, A.; Demir, E.; Wahl, D.; Scheglmann, K.; Winner, B.; Bogdahn, U.; Topaloglu, H.; Hehr, U.; Winkler, J. Novel Truncating and Missense Mutations of the KCC3 Gene Associated with Andermann Syndrome. Neurology 2006, 67 (7), 1044, DOI: 10.1212/01.wnl.0000250608.09509.edGoogle ScholarThere is no corresponding record for this reference.
- 44Olsson, M. H. M.; So̷ndergaard, C. R.; Rostkowski, M.; Jensen, J. H. PROPKA3: Consistent Treatment of Internal and Surface Residues in Empirical p Ka Predictions. J. Chem. Theory Comput. 2011, 7 (2), 525– 537, DOI: 10.1021/ct100578zGoogle Scholar44PROPKA3: Consistent Treatment of Internal and Surface Residues in Empirical pKa PredictionsOlsson, Mats H. M.; Sondergaard, Chresten R.; Rostkowski, Michal; Jensen, Jan H.Journal of Chemical Theory and Computation (2011), 7 (2), 525-537CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The authors have revised the rules and parameters for one of the most commonly used empirical pKa predictors, PROPKA, based on better phys. description of the desolvation and dielec. response for the protein. The authors have introduced a new and consistent approach to interpolate the description between the previously distinct classifications into internal and surface residues, which otherwise is found to give rise to an erratic and discontinuous behavior. Since the goal of this study is to lay out the framework and validate the concept, it focuses on Asp and Glu residues where the protein pKa values and structures are assumed to be more reliable. The new and improved implementation is evaluated and discussed; it is found to agree better with expt. than the previous implementation (in parentheses): rmsd = 0.79 (0.91) for Asp and Glu, 0.75 (0.97) for Tyr, 0.65 (0.72) for Lys, and 1.00 (1.37) for His residues. The most significant advance, however, is in reducing the no. of outliers and removing unreasonable sensitivity to small structural changes that arise from classifying residues as either internal or surface.
- 45Schott-Verdugo, S.; Gohlke, H. PACKMOL-Memgen: A Simple-To-Use, Generalized Workflow for Membrane-Protein–Lipid-Bilayer System Building. J. Chem. Inf. Model. 2019, 59 (6), 2522– 2528, DOI: 10.1021/acs.jcim.9b00269Google Scholar45PACKMOL-Memgen: A Simple-To-Use, Generalized Workflow for Membrane-Protein-Lipid-Bilayer System BuildingSchott-Verdugo, Stephan; Gohlke, HolgerJournal of Chemical Information and Modeling (2019), 59 (6), 2522-2528CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)We present PACKMOL-Memgen, a simple-to-use, generalized workflow for automated building of membrane-protein-lipid-bilayer systems based on open-source tools including Packmol, memembed, pdbremix, and AmberTools. Compared with web-interface-based related tools, PACKMOL-Memgen allows setup of multiple configurations of a system in a user-friendly and efficient manner within minutes. The generated systems are well-packed and thus well-suited as starting configurations in MD simulations under periodic boundary conditions, requiring only moderate equilibration times. PACKMOL-Memgen is distributed with AmberTools and runs on most computing platforms, and its output can also be used for CHARMM or adapted to other mol.-simulation packages.
- 46Le Grand, S.; Götz, A. W.; Walker, R. C. SPFP: Speed without Compromise─A Mixed Precision Model for GPU Accelerated Molecular Dynamics Simulations. Comput. Phys. Commun. 2013, 184 (2), 374– 380, DOI: 10.1016/j.cpc.2012.09.022Google Scholar46SPFP: Speed without compromise-A mixed precision model for GPU accelerated molecular dynamics simulationsLe Grand, Scott; Gotz, Andreas W.; Walker, Ross C.Computer Physics Communications (2013), 184 (2), 374-380CODEN: CPHCBZ; ISSN:0010-4655. (Elsevier B.V.)A new precision model is proposed for the acceleration of all-atom classical mol. dynamics (MD) simulations on graphics processing units (GPUs). This precision model replaces double precision arithmetic with fixed point integer arithmetic for the accumulation of force components as compared to a previously introduced model that uses mixed single/double precision arithmetic. This significantly boosts performance on modern GPU hardware without sacrificing numerical accuracy. We present an implementation for NVIDIA GPUs of both generalized Born implicit solvent simulations as well as explicit solvent simulations using the particle mesh Ewald (PME) algorithm for long-range electrostatics using this precision model. Tests demonstrate both the performance of this implementation as well as its numerical stability for const. energy and const. temp. biomol. MD as compared to a double precision CPU implementation and double and mixed single/double precision GPU implementations.
- 47Salomon-Ferrer, R.; Case, D. A.; Walker, R. C. An Overview of the Amber Biomolecular Simulation Package: Amber Biomolecular Simulation Package. Wiley Interdiscip. Rev. Comput. Mol. Sci. 2013, 3 (2), 198– 210, DOI: 10.1002/wcms.1121Google Scholar47An overview of the amber biomolecular simulation packageSalomon-Ferrer, Romelia; Case, David A.; Walker, Ross C.Wiley Interdisciplinary Reviews: Computational Molecular Science (2013), 3 (2), 198-210CODEN: WIRCAH; ISSN:1759-0884. (Wiley-Blackwell)A review. Mol. dynamics (MD) allows the study of biol. and chem. systems at the atomistic level on timescales from femtoseconds to milliseconds. It complements expt. while also offering a way to follow processes difficult to discern with exptl. techniques. Numerous software packages exist for conducting MD simulations of which one of the widest used is termed Amber. Here, we outline the most recent developments, since version 9 was released in Apr. 2006, of the Amber and AmberTools MD software packages, referred to here as simply the Amber package. The latest release represents six years of continued development, since version 9, by multiple research groups and the culmination of over 33 years of work beginning with the first version in 1979. The latest release of the Amber package, version 12 released in Apr. 2012, includes a substantial no. of important developments in both the scientific and computer science arenas. We present here a condensed vision of what Amber currently supports and where things are likely to head over the coming years. Figure 1 shows the performance in ns/day of the Amber package version 12 on a single-core AMD FX-8120 8-Core 3.6GHz CPU, the Cray XT5 system, and a single GPU GTX680.
- 48Maier, J. A.; Martinez, C.; Kasavajhala, K.; Wickstrom, L.; Hauser, K. E.; Simmerling, C. ff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameters from ff99SB. J. Chem. Theory Comput. 2015, 11 (8), 3696– 3713, DOI: 10.1021/acs.jctc.5b00255Google Scholar48ff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameters from ff99SBMaier, James A.; Martinez, Carmenza; Kasavajhala, Koushik; Wickstrom, Lauren; Hauser, Kevin E.; Simmerling, CarlosJournal of Chemical Theory and Computation (2015), 11 (8), 3696-3713CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Mol. mechanics is powerful for its speed in atomistic simulations, but an accurate force field is required. The Amber ff99SB force field improved protein secondary structure balance and dynamics from earlier force fields like ff99, but weaknesses in side chain rotamer and backbone secondary structure preferences have been identified. Here, we performed a complete refit of all amino acid side chain dihedral parameters, which had been carried over from ff94. The training set of conformations included multidimensional dihedral scans designed to improve transferability of the parameters. Improvement in all amino acids was obtained as compared to ff99SB. Parameters were also generated for alternate protonation states of ionizable side chains. Av. errors in relative energies of pairs of conformations were under 1.0 kcal/mol as compared to QM, reduced 35% from ff99SB. We also took the opportunity to make empirical adjustments to the protein backbone dihedral parameters as compared to ff99SB. Multiple small adjustments of φ and ψ parameters were tested against NMR scalar coupling data and secondary structure content for short peptides. The best results were obtained from a phys. motivated adjustment to the φ rotational profile that compensates for lack of ff99SB QM training data in the β-ppII transition region. Together, these backbone and side chain modifications (hereafter called ff14SB) not only better reproduced their benchmarks, but also improved secondary structure content in small peptides and reprodn. of NMR χ1 scalar coupling measurements for proteins in soln. We also discuss the Amber ff12SB parameter set, a preliminary version of ff14SB that includes most of its improvements.
- 49Dickson, C. J.; Madej, B. D.; Skjevik, Å. A.; Betz, R. M.; Teigen, K.; Gould, I. R.; Walker, R. C. Lipid14: The Amber Lipid Force Field. J. Chem. Theory Comput. 2014, 10 (2), 865– 879, DOI: 10.1021/ct4010307Google Scholar49Lipid14: The Amber Lipid Force FieldDickson, Callum J.; Madej, Benjamin D.; Skjevik, Age A.; Betz, Robin M.; Teigen, Knut; Gould, Ian R.; Walker, Ross C.Journal of Chemical Theory and Computation (2014), 10 (2), 865-879CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The AMBER lipid force field has been updated to create Lipid14, allowing tensionless simulation of a no. of lipid types with the AMBER MD package. The modular nature of this force field allows numerous combinations of head and tail groups to create different lipid types, enabling the easy insertion of new lipid species. The Lennard-Jones and torsion parameters of both the head and tail groups have been revised and updated partial charges calcd. The force field has been validated by simulating bilayers of six different lipid types for a total of 0.5 μs each without applying a surface tension; with favorable comparison to expt. for properties such as area per lipid, vol. per lipid, bilayer thickness, NMR order parameters, scattering data, and lipid lateral diffusion. As the derivation of this force field is consistent with the AMBER development philosophy, Lipid14 is compatible with the AMBER protein, nucleic acid, carbohydrate, and small mol. force fields.
- 50Jorgensen, W. L.; Chandrasekhar, J.; Madura, J. D.; Impey, R. W.; Klein, M. L. Comparison of Simple Potential Functions for Simulating Liquid Water. J. Chem. Phys. 1983, 79 (2), 926– 935, DOI: 10.1063/1.445869Google Scholar50Comparison of simple potential functions for simulating liquid waterJorgensen, William L.; Chandrasekhar, Jayaraman; Madura, Jeffry D.; Impey, Roger W.; Klein, Michael L.Journal of Chemical Physics (1983), 79 (2), 926-35CODEN: JCPSA6; ISSN:0021-9606.Classical Monte Carlo simulations were carried out for liq. H2O in the NPT ensemble at 25° and 1 atm using 6 of the simpler intermol. potential functions for the dimer. Comparisons were made with exptl. thermodn. and structural data including the neutron diffraction results of Thiessen and Narten (1982). The computed densities and potential energies agree with expt. except for the original Bernal-Fowler model, which yields an 18% overest. of the d. and poor structural results. The discrepancy may be due to the correction terms needed in processing the neutron data or to an effect uniformly neglected in the computations. Comparisons were made for the self-diffusion coeffs. obtained from mol. dynamics simulations.
- 51Joung, I. S.; Cheatham, T. E. Determination of Alkali and Halide Monovalent Ion Parameters for Use in Explicitly Solvated Biomolecular Simulations. J. Phys. Chem. B 2008, 112 (30), 9020– 9041, DOI: 10.1021/jp8001614Google Scholar51Determination of Alkali and Halide Monovalent Ion Parameters for Use in Explicitly Solvated Biomolecular SimulationsJoung, In Suk; Cheatham, Thomas E.Journal of Physical Chemistry B (2008), 112 (30), 9020-9041CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)Alkali (Li+, Na+, K+, Rb+, and Cs+) and halide (F-, Cl-, Br-, and I-) ions play an important role in many biol. phenomena, roles that range from stabilization of biomol. structure, to influence on biomol. dynamics, to key physiol. influence on homeostasis and signaling. To properly model ionic interaction and stability in atomistic simulations of biomol. structure, dynamics, folding, catalysis, and function, an accurate model or representation of the monovalent ions is critically necessary. A good model needs to simultaneously reproduce many properties of ions, including their structure, dynamics, solvation, and moreover both the interactions of these ions with each other in the crystal and in soln. and the interactions of ions with other mols. At present, the best force fields for biomols. employ a simple additive, nonpolarizable, and pairwise potential for at. interaction. In this work, the authors describe their efforts to build better models of the monovalent ions within the pairwise Coulombic and 6-12 Lennard-Jones framework, where the models are tuned to balance crystal and soln. properties in Ewald simulations with specific choices of well-known water models. Although it has been clearly demonstrated that truly accurate treatments of ions will require inclusion of nonadditivity and polarizability (particularly with the anions) and ultimately even a quantum mech. treatment, the authors' goal was to simply push the limits of the additive treatments to see if a balanced model could be created. The applied methodol. is general and can be extended to other ions and to polarizable force-field models. The authors' starting point centered on observations from long simulations of biomols. in salt soln. with the AMBER force fields where salt crystals formed well below their soly. limit. The likely cause of the artifact in the AMBER parameters relates to the naive mixing of the Smith and Dang chloride parameters with AMBER-adapted Aqvist cation parameters. To provide a more appropriate balance, the authors reoptimized the parameters of the Lennard-Jones potential for the ions and specific choices of water models. To validate and optimize the parameters, the authors calcd. hydration free energies of the solvated ions and also lattice energies (LE) and lattice consts. (LC) of alkali halide salt crystals. This is the first effort that systematically scans across the Lennard-Jones space (well depth and radius) while balancing ion properties like LE and LC across all pair combinations of the alkali ions and halide ions. The optimization across the entire monovalent series avoids systematic deviations. The ion parameters developed, optimized, and characterized were targeted for use with some of the most commonly used rigid and nonpolarizable water models, specifically TIP3P, TIP4PEW, and SPC/E. In addn. to well reproducing the soln. and crystal properties, the new ion parameters well reproduce binding energies of the ions to water and the radii of the first hydration shells.
- 52Darden, T.; York, D.; Pedersen, L. Particle Mesh Ewald: An N ·log(N) Method for Ewald Sums in Large Systems. J. Chem. Phys. 1993, 98 (12), 10089– 10092, DOI: 10.1063/1.464397Google Scholar52Particle mesh Ewald: an N·log(N) method for Ewald sums in large systemsDarden, Tom; York, Darrin; Pedersen, LeeJournal of Chemical Physics (1993), 98 (12), 10089-92CODEN: JCPSA6; ISSN:0021-9606.An N·log(N) method for evaluating electrostatic energies and forces of large periodic systems is presented. The method is based on interpolation of the reciprocal space Ewald sums and evaluation of the resulting convolution using fast Fourier transforms. Timings and accuracies are presented for three large cryst. ionic systems.
- 53Ryckaert, J.-P.; Ciccotti, G.; Berendsen, H. J. C. Numerical Integration of the Cartesian Equations of Motion of a System with Constraints: Molecular Dynamics of n-Alkanes. J. Comput. Phys. 1977, 23 (3), 327– 341, DOI: 10.1016/0021-9991(77)90098-5Google Scholar53Numerical integration of the Cartesian equations of motion of a system with constraints: molecular dynamics of n-alkanesRyckaert, Jean Paul; Ciccotti, Giovanni; Berendsen, Herman J. C.Journal of Computational Physics (1977), 23 (3), 327-41CODEN: JCTPAH; ISSN:0021-9991.A numerical algorithm integrating the 3N Cartesian equation of motion of a system of N points subject to holonomic constraints is applied to mol. dynamics simulation of a liq. of 64 butane mols.
- 54Ansari, N.; Rizzi, V.; Parrinello, M. Water Regulates the Residence Time of Benzamidine in Trypsin. Nat. Commun. 2022, 13 (1), 5438, DOI: 10.1038/s41467-022-33104-3Google Scholar54Water regulates the residence time of Benzamidine in TrypsinAnsari, Narjes; Rizzi, Valerio; Parrinello, MicheleNature Communications (2022), 13 (1), 5438CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)Abstr.: The process of ligand-protein unbinding is crucial in biophysics. Water is an essential part of any biol. system and yet, many aspects of its role remain elusive. Here, we simulate with state-of-the-art enhanced sampling techniques the binding of Benzamidine to Trypsin which is a much studied and paradigmatic ligand-protein system. We use machine learning methods to det. efficient collective coordinates for the complex non-local network of water. These coordinates are used to perform On-the-fly Probability Enhanced Sampling simulations, which we adapt to calc. also the ligand residence time. Our results, both static and dynamic, are in good agreement with expts. We find that the presence of a water mol. located at the bottom of the binding pocket allows via a network of hydrogen bonds the ligand to be released into the soln. On a finer scale, even when unbinding is allowed, another water mol. further modulates the exit time.
- 55Rizzi, V.; Bonati, L.; Ansari, N.; Parrinello, M. The Role of Water in Host-Guest Interaction. Nat. Commun. 2021, 12 (1), 93, DOI: 10.1038/s41467-020-20310-0Google Scholar55The role of water in host-guest interactionRizzi, Valerio; Bonati, Luigi; Ansari, Narjes; Parrinello, MicheleNature Communications (2021), 12 (1), 93CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)One of the main applications of atomistic computer simulations is the calcn. of ligand binding free energies. The accuracy of these calcns. depends on the force field quality and on the thoroughness of configuration sampling. Sampling is an obstacle in simulations due to the frequent appearance of kinetic bottlenecks in the free energy landscape. Very often this difficulty is circumvented by enhanced sampling techniques. Typically, these techniques depend on the introduction of appropriate collective variables that are meant to capture the system's degrees of freedom. In ligand binding, water has long been known to play a key role, but its complex behavior has proven difficult to fully capture. In this paper we combine machine learning with phys. intuition to build a non-local and highly efficient water-describing collective variable. We use it to study a set of host-guest systems from the SAMPL5 challenge. We obtain highly accurate binding free energies and good agreement with expts. The role of water during the binding process is then analyzed in some detail.
- 56Tribello, G. A.; Bonomi, M.; Branduardi, D.; Camilloni, C.; Bussi, G. PLUMED 2: New Feathers for an Old Bird. Comput. Phys. Commun. 2014, 185 (2), 604– 613, DOI: 10.1016/j.cpc.2013.09.018Google Scholar56PLUMED 2: New feathers for an old birdTribello, Gareth A.; Bonomi, Massimiliano; Branduardi, Davide; Camilloni, Carlo; Bussi, GiovanniComputer Physics Communications (2014), 185 (2), 604-613CODEN: CPHCBZ; ISSN:0010-4655. (Elsevier B.V.)Enhancing sampling and analyzing simulations are central issues in mol. simulation. Recently, we introduced PLUMED, an open-source plug-in that provides some of the most popular mol. dynamics (MD) codes with implementations of a variety of different enhanced sampling algorithms and collective variables (CVs). The rapid changes in this field, in particular new directions in enhanced sampling and dimensionality redn. together with new hardware, require a code that is more flexible and more efficient. We therefore present PLUMED 2 here-a complete rewrite of the code in an object-oriented programming language (C++). This new version introduces greater flexibility and greater modularity, which both extends its core capabilities and makes it far easier to add new methods and CVs. It also has a simpler interface with the MD engines and provides a single software library contg. both tools and core facilities. Ultimately, the new code better serves the ever-growing community of users and contributors in coping with the new challenges arising in the field.
- 57Michaud-Agrawal, N.; Denning, E. J.; Woolf, T. B.; Beckstein, O. MDAnalysis: A Toolkit for the Analysis of Molecular Dynamics Simulations. J. Comput. Chem. 2011, 32 (10), 2319– 2327, DOI: 10.1002/jcc.21787Google Scholar57MDAnalysis: A toolkit for the analysis of molecular dynamics simulationsMichaud-Agrawal, Naveen; Denning, Elizabeth J.; Woolf, Thomas B.; Beckstein, OliverJournal of Computational Chemistry (2011), 32 (10), 2319-2327CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)MDAnal. is an object-oriented library for structural and temporal anal. of mol. dynamics (MD) simulation trajectories and individual protein structures. It is written in the Python language with some performance-crit. code in C. It uses the powerful NumPy package to expose trajectory data as fast and efficient NumPy arrays. It has been tested on systems of millions of particles. Many common file formats of simulation packages including CHARMM, Gromacs, Amber, and NAMD and the Protein Data Bank format can be read and written. Atoms can be selected with a syntax similar to CHARMM's powerful selection commands. MDAnal. enables both novice and experienced programmers to rapidly write their own anal. tools and access data stored in trajectories in an easily accessible manner that facilitates interactive explorative anal. MDAnal. has been tested on and works for most Unix-based platforms such as Linux and Mac OS X. It is freely available under the GNU General Public License from http://mdanal.googlecode.com. © 2011 Wiley Periodicals, Inc. J Comput Chem 2011.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 3 publications.
- Ayan Majumder, John E. Straub. Machine Learning Derived Collective Variables for the Study of Protein Homodimerization in Membrane. Journal of Chemical Theory and Computation 2024, 20
(13)
, 5774-5783. https://doi.org/10.1021/acs.jctc.4c00454
- Yongxiang Zhao, Pietro Vidossich, Biff Forbush, Junfeng Ma, Jesse Rinehart, Marco De Vivo, Erhu Cao. Structural basis for human NKCC1 inhibition by loop diuretic drugs. The EMBO Journal 2025, 44
(5)
, 1540-1562. https://doi.org/10.1038/s44318-025-00368-6
- Sam W. Henderson, Saeed Nourmohammadi, Maria Hrmova. Protein Structural Modeling and Transport Thermodynamics Reveal That Plant Cation–Chloride Cotransporters Mediate Potassium–Chloride Symport. International Journal of Molecular Sciences 2024, 25
(23)
, 12955. https://doi.org/10.3390/ijms252312955
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. NKCC1’s structure embedded in the membrane. (A) 3D representation of full-length human NKCC1 (PDB 7MXO), with each monomer represented in dark and light gray. Each monomer is constructed by three main components: (i) conserved TM domain, composed of 12 helices, which contains all four ion binding sites, shown in green, blue and orange for Cl–, Na+, and K+ ions, respectively; (ii) disordered amino-terminal; and (iii) large carboxy-terminal domains. (13) All structures of NKCC1, and related CCC’s of the same family, (40) revealed that the TM helices are organized in two inverted repeats of five-helix bundles─also known as LeuT-fold. (2,16,17,21) (B) Schematic representation of NKCC1, showing its homodimeric structure with one monomer represented to highlight the transmembrane domains (TMs, dark gray); the other monomer represented to highlight the channel arrangement across the cell membrane (light brown); the two five-helix bundle inverted repeats (TM 1 to TM 5 and TM 6 to TM 10) and the dimeric interface (TM 11 and TM 12). The amino and carboxy-terminal domains are also highlighted.
Figure 2
Figure 2. Essential conformational transition for alternating accessibility of ion binding sites allows NKCC1 ion transport. (A) Schematic representation of outward open (left) and inward open (right) NKCC1 conformation. Ionic binding sites are exposed to the extra and intracellular side of the membrane, respectively. (B) In gray, atomic surface representation of outward open (left) and inward open (right) NKCC1 conformation. Ionic binding sites for Na+, K+, and Cl– are shown as colored surfaces (blue, orange, and green, respectively), and ions are represented as colored spheres. Outer (left) and inner (right) vestibules, open to the extra and intracellular sides of the membrane, are highlighted with discontinuous black lines.
Figure 3
Figure 3. Rocking-bundle angular motion of specific NKCC1 TMs facilitates alternate accessibility of ion binding sites. (A) Schematic representation of NKCC1’s angular motion, defined as the change of the angle (α) between TM 4 and TM 9 (in green) and TM 2 and TM 7 (in red), during the conformational transition between the outward open state (bright green) and inward open state (dim green), calculated from the centers of mass of the backbone atoms from the extracellular and intracellular tip of TM 4 and TM 9 and the intracellular tip of TM 2 and TM 7 (in red). (B) Quantification of TM 4 and TM 9 angular motion represented by the angle α through 16 conformational transitions from OPES Explore simulations. The light brown horizontal bars represent the outward open and inward open average angle α ± 1SD calculated from 1 μs of equilibrium molecular dynamics (MD). Circles represent the starting point of each transition, whereas the triangles represent the end point of the same transition and its direction. Circles and triangles are colored depending on the NKCC1 conformation they represent (bright green for outward open and dim green for inward open).
Figure 4
Figure 4. Stabilization of the hydrophobic interface between TM 4 and TM 9 allows for their cooperative action. Representation of human NKCC1 embedded in the cell membrane, with TM 4 and TM 9 highlighted in bright yellow. Inset on the right: Higher magnification of the hydrophobic interface between TM 4 and TM 9 (highlighted by the oval), which allows for their cooperative angular motion. Relevant residues are shown as sticks with their atomic surfaces pictured in red (oxygen), blue (nitrogen), gray (carbon), and white (hydrogen).
Figure 5
Figure 5. NKCC1 TM 10's corking motion modulates ion/water access to the outer vestibule. (A) Schematic representation of NKCC1 TM 10's corking motion, defined as the change of TM 10's (in green) intrahelical angle (θ), during the conformational transition between the outward open state (bright green) and inward open state (dim green), calculated from the centers of mass of the backbone atoms from TM 10’s intra and extracellular tips, and the backbone atoms where TM 10 bends. (B) Quantification of TM 10's corking motion represented by the angle θ through 16 conformational transitions from OPES Explore simulations. The light brown horizontal bars represent the outward open and inward open average angle θ ± 1SD calculated from 1 μs of equilibrium molecular dynamics (MD). Circles represent the starting point of each transition, whereas the triangles represent the end point of the same transition and its direction. Circles and triangles are colored depending on the NKCC1 conformation they represent (bright green for outward open and dim green for inward open).
Figure 6
Figure 6. Interface between TM 10 and TM 6 highlights crucial interactions that determine accessibility of extracellular binding sites. Representation of human NKCC1 embedded in the cell membrane, with TM 10 and TM 6 highlighted in bright yellow. Inset on the top right: In the IO state, as shown by the light green schematic representation, interacting residues at the TM 10 and TM 6 interface are shown as sticks with their atomic surfaces (pictured in red─oxygen, blue─nitrogen, gray─carbon, and white─hydrogen), blocking solvent access to the outer vestibule. Inset on the bottom right: In the OO state, as shown by the light orange schematic representation, previously interacting residues at the TM 10 and TM 6 interface are now shown to be too far apart to form bonds.
Figure 7
Figure 7. Mutagenesis targeting residues from TM 10 and TM 6 highlight their functional relevance. (A) Representation of human NKCC1 embedded in the cell membrane, with TM 10 and TM 6 highlighted in bright yellow. Inset on the right: The interface between TM 10 and TM 6 where homologous mutated residues are shown as sticks and are highlighted in green surface. Namely, these residues are mouse A490W (human Ala 497), mouse L664A (human Leu 671), mouse N665A (human Asn 672), and mouse A668W (human Ala 675). Gray surface represents the position of residues that mainly form/break interactions throughout TM 10's corking motion. (B) Example traces obtained in the Cl– influx assay on HEK293 cells transfected with the WT NKCC1 transporter or NKCC1 mutated at different residues. The arrow indicates the addition of NaCl (74 mM) to initiate the NKCC1-mediated Cl– influx. (C) Quantification of the mouse NKCC1 inhibitory activity using the Cl– influx fluorescence assay in HEK293 cells. A fluorescence signal decrease, corresponding to a decrease in NKCC1 transporter activity, was observed for all the cells transfected with NKCC1 mutants. Data are normalized and the average of the last 10 s of kinetics is plotted (ΔF/F0). Data are presented as a percentage of the WT. Data represent mean ± SEM from 3 to 4 independent experiments (Kruskal–Wallis one way ANOVA, H = 216, DF = 6, followed by Dunn’s post hoc test on multiple comparisons, *** P = 0.0002, **** P < 0.0001).
Figure 8
Figure 8. Free energy surface identifies relevant NKCC1 conformations for the inward open ↔ outward open transition. (A) Representation of the Free Energy Surface of the conformational transition between human NKCC1 IO and OO states computed by OPES Explore over the DeepLDA collective variable and the outer vestibule water coordination collective variable. Energetical basins are highlighted with a schematic representation of the conformation they identify. These are, namely: the IO state, the occluded state (labeled Occ), the OOd state (outward open “dry” – lower outer vestibule hydration) and OOw (outward open “wet” – higher outer vestibule hydration. (B) Higher magnification of the hexagon in A representing the main gating interactions that occlude the outer vestibule and block solvent access to the ionic binding sites. Relevant residues are shown as sticks with their atomic surfaces pictured in red (oxygen), blue (nitrogen), gray (carbon) and white (hydrogen). (C) Higher magnification of the hexagon in A representing of the main gating interactions that occlude the inner vestibule and block solvent access to the ionic binding sites. Relevant residues are shown as sticks with their atomic surfaces pictured in red (oxygen), blue (nitrogen), yellow (sulfur), gray (carbon) and white (hydrogen).
Figure 9
Figure 9. Pore profile confirms distinct binding-site accessibility of NKCC1 states along the inward open ↔ outward open transition. (A–C) Pore profile of the IO state (A), the occluded state (B), and OO state (C), schematically represented at the bottom. These profiles were obtained by calculating the radius of the largest sphere along the Z-axis of the ion translocation cavity, and then plotting the pore profile of several snapshots from their corresponding equilibrium MD simulations, computed by the software HOLE. Pore profiles were mirrored around radius 0 for visual clarity. The blue lines represent the outer vestibule, and the orange lines represent the inner vestibule of NKCC1.
Figure 10
Figure 10. NKCC1’s outward open conformations are permeable to water. (A) Representation of NKCC1 (gray cartoon) embedded in the membrane (black horizontal bars) in a water-permeable state. Water molecules are shown as red and white lines with a red atomic surface representation. A chain of water molecules whose oxygen atoms are within 4.0 Å of each other connecting the extracellular and intracellular solvent is present. (B, C) Schematic representation of both states that present permeability to water (outward open without ions bound, B, or fully loaded, C), and their respective plot, which tracks the appearance of permeable states during each state’s equilibrium MD simulation. Vertical red bars represent snapshots from the respective simulation where a chain of water molecules whose oxygen atoms are within 4.0 Å of each other connecting the extracellular and intracellular solvent through NKCC1 is observed.
Figure 11
Figure 11. NKCC1 passively transports water. Histogram of all transport events detected in the state equilibrium MD simulation of NKCC1 outward open conformation with no ions bound, organized by the length of each transport event. Bars show the frequency of efflux/influx (orange/green) events per transport event duration. Vertical continuous lines show the mean, and discontinuous lines show the standard deviation (efflux, orange; influx, green). Some longer transport events were excluded from the histogram for clarity.
References
This article references 57 other publications.
- 1Deidda, G.; Parrini, M.; Naskar, S.; Bozarth, I. F.; Contestabile, A.; Cancedda, L. Reversing Excitatory GABAAR Signaling Restores Synaptic Plasticity and Memory in a Mouse Model of Down Syndrome. Nat. Med. 2015, 21 (4), 318– 326, DOI: 10.1038/nm.38271Reversing excitatory GABAAR signaling restores synaptic plasticity and memory in a mouse model of Down syndromeDeidda, Gabriele; Parrini, Martina; Naskar, Shovan; Bozarth, Ignacio F.; Contestabile, Andrea; Cancedda, LauraNature Medicine (New York, NY, United States) (2015), 21 (4), 318-326CODEN: NAMEFI; ISSN:1078-8956. (Nature Publishing Group)Down syndrome (DS) is the most frequent genetic cause of intellectual disability, and altered GABAergic transmission through Cl--permeable GABAA receptors (GABAARs) contributes considerably to learning and memory deficits in DS mouse models. However, the efficacy of GABAergic transmission has never been directly assessed in DS. Here GABAAR signaling was found to be excitatory rather than inhibitory, and the reversal potential for GABAAR-driven Cl- currents (ECl) was shifted toward more pos. potentials in the hippocampi of adult DS mice. Accordingly, hippocampal expression of the cation Cl- cotransporter NKCC1 was increased in both trisomic mice and individuals with DS. Notably, NKCC1 inhibition by the FDA-approved drug bumetanide restored ECl, synaptic plasticity and hippocampus-dependent memory in adult DS mice. Our findings demonstrate that GABA is excitatory in adult DS mice and identify a new therapeutic approach for the potential rescue of cognitive disabilities in individuals with DS.
- 2Zhao, Y.; Cao, E. Structural Pharmacology of Cation-Chloride Cotransporters. Membranes 2022, 12 (12), 1206, DOI: 10.3390/membranes121212062Structural Pharmacology of Cation-Chloride CotransportersZhao, Yongxiang; Cao, ErhuMembranes (Basel, Switzerland) (2022), 12 (12), 1206CODEN: MBSEB6; ISSN:2077-0375. (MDPI AG)A review. Loop and thiazide diuretics have been cornerstones of clin. management of hypertension and fluid overload conditions for more than five decades. The hunt for their mol. targets led to the discovery of cation-chloride cotransporters (CCCs) that catalyze electroneutral movement of Cl- together with Na+ and/or K+. CCCs consist of two 1 Na+-1 K+-2 Cl- (NKCC1-2), one 1 Na+-1 Cl- (NCC), and four 1 K+-1 Cl- (KCC1-4) transporters in human. CCCs are fundamental in trans-epithelia ion secretion and absorption, homeostasis of intracellular Cl- concn. and cell vol., and regulation of neuronal excitability. Malfunction of NKCC2 and NCC leads to abnormal salt and water retention in the kidney and, consequently, imbalance in electrolytes and blood pressure. Mutations in KCC2 and KCC3 are assocd. with brain disorders due to impairments in regulation of excitability and possibly cell vol. of neurons. A recent surge of structures of CCCs have defined their dimeric architecture, their ion binding sites, their conformational changes assocd. with ion translocation, and the mechanisms of action of loop diuretics and small mol. inhibitors. These breakthroughs now set the stage to expand CCC pharmacol. beyond loop and thiazide diuretics, developing the next generation of diuretics with improved potency and specificity. Beyond drugging renal-specific CCCs, brain-penetrable therapeutics are sorely needed to target CCCs in the nervous system for the treatment of neurol. disorders and psychiatric conditions.
- 3Ben-Ari, Y. NKCC1 Chloride Importer Antagonists Attenuate Many Neurological and Psychiatric Disorders. Trends Neurosci. 2017, 40 (9), 536– 554, DOI: 10.1016/j.tins.2017.07.0013NKCC1 Chloride Importer Antagonists Attenuate Many Neurological and Psychiatric DisordersBen-Ari, YehezkelTrends in Neurosciences (2017), 40 (9), 536-554CODEN: TNSCDR; ISSN:0166-2236. (Elsevier Ltd.)In physiol. conditions, adult neurons have low intracellular Cl- [(Cl-)I] levels underlying the γ-aminobutyric acid (GABA)ergic inhibitory drive. In contrast, neurons have high (Cl-)I levels and excitatory GABA actions in a wide range of pathol. conditions including spinal cord lesions, chronic pain, brain trauma, cerebrovascular infarcts, autism, Rett and Down syndrome, various types of epilepsies, and other genetic or environmental insults. The diuretic highly specific NKCC1 chloride importer antagonist bumetanide (PubChem CID: 2461) efficiently restores low (Cl-)I levels and attenuates many disorders in exptl. conditions and in some clin. trials. Here, I review the mechanisms of action, therapeutic effects, promises, and pitfalls of bumetanide.
- 4Delpire, E.; Gagnon, K. B. Elusive Role of the Na-K-2Cl Cotransporter in the Choroid Plexus. Am. J. Physiol.-Cell Physiol. 2019, 316 (4), C522– C524, DOI: 10.1152/ajpcell.00490.2018There is no corresponding record for this reference.
- 5Kaila, K.; Price, T. J.; Payne, J. A.; Puskarjov, M.; Voipio, J. Cation-Chloride Cotransporters in Neuronal Development, Plasticity and Disease. Nat. Rev. Neurosci. 2014, 15 (10), 637– 654, DOI: 10.1038/nrn38195Cation-chloride cotransporters in neuronal development, plasticity and diseaseKaila, Kai; Price, Theodore J.; Payne, John A.; Puskarjov, Martin; Voipio, JuhaNature Reviews Neuroscience (2014), 15 (10), 637-654CODEN: NRNAAN; ISSN:1471-003X. (Nature Publishing Group)Elec. activity in neurons requires a seamless functional coupling between plasmalemmal ion channels and ion transporters. Although ion channels have been studied intensively for several decades, research on ion transporters is in its infancy. In recent years, it has become evident that one family of ion transporters, cation-chloride cotransporters (CCCs), and in particular K+-Cl- cotransporter 2 (KCC2), have seminal roles in shaping GABAergic signalling and neuronal connectivity. Studying the functions of these transporters may lead to major paradigm shifts in our understanding of the mechanisms underlying brain development and plasticity in health and disease.
- 6Pressey, J. C.; de Saint-Rome, M.; Raveendran, V. A.; Woodin, M. A. Chloride Transporters Controlling Neuronal Excitability. Physiol. Rev. 2023, 103 (2), 1095– 1135, DOI: 10.1152/physrev.00025.20216Chloride transporters controlling neuronal excitabilityPressey, Jessica C.; de Saint-Rome, Miranda; Raveendran, Vineeth A.; Woodin, Melanie A.Physiological Reviews (2023), 103 (2), 1095-1135CODEN: PHREA7; ISSN:1522-1210. (American Physiological Society)A review. Synaptic inhibition plays a crucial role in regulating neuronal excitability, which is the foundation of nervous system function. This inhibition is largely mediated by the neurotransmitters GABA and glycine that activate Cl--permeable ion channels, which means that the strength of inhibition depends on the Cl- gradient across the membrane. In neurons, the Cl- gradient is primarily mediated by two secondarily active cation-chloride cotransporters (CCCs), NKCC1 and KCC2. CCC-mediated regulation of the neuronal Cl- gradient is crit. for healthy brain function, as dysregulation of CCCs has emerged as a key mechanism underlying neurol. disorders including epilepsy, neuropathic pain, and autism spectrum disorder. This review begins with an overview of neuronal chloride transporters before explaining the dependent relationship between these CCCs, Cl- regulation, and inhibitory synaptic transmission. We then discuss the evidence for how CCCs can be regulated, including by activity and their protein interactions, which underlie inhibitory synaptic plasticity. For readers who may be interested in conducting expts. on CCCs and neuronal excitability, we have included a section on techniques for estg. and recording intracellular Cl-, including their advantages and limitations. Although the focus of this review is on neurons, we also examine how Cl- is regulated in glial cells, which in turn regulate neuronal excitability through the tight relationship between this nonneuronal cell type and synapses. Finally, we discuss the relatively extensive and growing literature on how CCC-mediated neuronal excitability contributes to neurol. disorders.
- 7Savardi, A.; Borgogno, M.; De Vivo, M.; Cancedda, L. Pharmacological Tools to Target NKCC1 in Brain Disorders. Trends Pharmacol. Sci. 2021, 42 (12), 1009– 1034, DOI: 10.1016/j.tips.2021.09.0057Pharmacological tools to target NKCC1 in brain disordersSavardi, Annalisa; Borgogno, Marco; De Vivo, Marco; Cancedda, LauraTrends in Pharmacological Sciences (2021), 42 (12), 1009-1034CODEN: TPHSDY; ISSN:0165-6147. (Elsevier Ltd.)A review. The chloride importer NKCC1 and the chloride exporter KCC2 are key regulators of neuronal chloride concn. A defective NKCC1/KCC2 expression ratio is assocd. with several brain disorders. Preclin./clin. studies have shown that NKCC1 inhibition by the United States FDA-approved diuretic bumetanide is a potential therapeutic strategy in preclin./clin. studies of multiple neurol. conditions. However, bumetanide has poor brain penetration and causes unwanted diuresis by inhibiting NKCC2 in the kidney. To overcome these issues, a growing no. of studies have reported more brain-penetrating and/or selective bumetanide prodrugs, analogs, and new mol. entities. Here, we review the evidence for NKCC1 pharmacol. inhibition as an effective strategy to manage neurol. disorders. We also discuss the advantages and limitations of bumetanide repurposing and the benefits and risks of new NKCC1 inhibitors as therapeutic agents for brain disorders.
- 8Karimy, J. K.; Zhang, J.; Kurland, D. B.; Theriault, B. C.; Duran, D.; Stokum, J. A.; Furey, C. G.; Zhou, X.; Mansuri, M. S.; Montejo, J.; Vera, A.; DiLuna, M. L.; Delpire, E.; Alper, S. L.; Gunel, M.; Gerzanich, V.; Medzhitov, R.; Simard, J. M.; Kahle, K. T. Inflammation-Dependent Cerebrospinal Fluid Hypersecretion by the Choroid Plexus Epithelium in Posthemorrhagic Hydrocephalus. Nat. Med. 2017, 23 (8), 997– 1003, DOI: 10.1038/nm.43618Inflammation-dependent cerebrospinal fluid hypersecretion by the choroid plexus epithelium in posthemorrhagic hydrocephalusKarimy, Jason K.; Zhang, Jinwei; Kurland, David B.; Theriault, Brianna Carusillo; Duran, Daniel; Stokum, Jesse A.; Furey, Charuta Gavankar; Zhou, Xu; Mansuri, M. Shahid; Montejo, Julio; Vera, Alberto; Di Luna, Michael L.; Delpire, Eric; Alper, Seth L.; Gunel, Murat; Gerzanich, Volodymyr; Medzhitov, Ruslan; Simard, J. Marc; Kahle, Kristopher T.Nature Medicine (New York, NY, United States) (2017), 23 (8), 997-1003CODEN: NAMEFI; ISSN:1078-8956. (Nature Publishing Group)The choroid plexus epithelium (CPE) secretes higher vols. of fluid (cerebrospinal fluid, CSF) than any other epithelium and simultaneously functions as the blood-CSF barrier to gate immune cell entry into the central nervous system. Posthemorrhagic hydrocephalus (PHH), an expansion of the cerebral ventricles due to CSF accumulation following intraventricular hemorrhage (IVH), is a common disease usually treated by suboptimal CSF shunting techniques. PHH is classically attributed to primary impairments in CSF reabsorption, but little exptl. evidence supports this concept. In contrast, the potential contribution of CSF secretion to PHH has received little attention. In a rat model of PHH, we demonstrate that IVH causes a Toll-like receptor 4 (TLR4)- and NF-κB-dependent inflammatory response in the CPE that is assocd. with a ∼3-fold increase in bumetanide-sensitive CSF secretion. IVH-induced hypersecretion of CSF is mediated by TLR4-dependent activation of the Ste20-type stress kinase SPAK, which binds, phosphorylates, and stimulates the NKCC1 co-transporter at the CPE apical membrane. Genetic depletion of TLR4 or SPAK normalizes hyperactive CSF secretion rates and reduces PHH symptoms, as does treatment with drugs that antagonize TLR4-NF-κB signaling or the SPAK-NKCC1 co-transporter complex. These data uncover a previously unrecognized contribution of CSF hypersecretion to the pathogenesis of PHH, demonstrate a new role for TLRs in regulation of the internal brain milieu, and identify a kinase-regulated mechanism of CSF secretion that could be targeted by repurposed US Food and Drug Administration (FDA)-approved drugs to treat hydrocephalus.
- 9Kharod, S. C.; Kang, S. K.; Kadam, S. D. Off-Label Use of Bumetanide for Brain Disorders: An Overview. Front. Neurosci. 2019, 13, 310, DOI: 10.3389/fnins.2019.003109Off-Label Use of Bumetanide for Brain Disorders: An OverviewKharod Shivani C; Kang Seok Kyu; Kadam Shilpa D; Kadam Shilpa DFrontiers in neuroscience (2019), 13 (), 310 ISSN:1662-4548.Bumetanide (BTN or BUM) is a FDA-approved potent loop diuretic (LD) that acts by antagonizing sodium-potassium-chloride (Na-K-Cl) cotransporters, NKCC1 (SLc12a2) and NKCC2. While NKCC1 is expressed both in the CNS and in systemic organs, NKCC2 is kidney-specific. The off-label use of BTN to modulate neuronal transmembrane Cl(-) gradients by blocking NKCC1 in the CNS has now been tested as an anti-seizure agent and as an intervention for neurological disorders in pre-clinical studies with varying results. BTN safety and efficacy for its off-label use has also been tested in several clinical trials for neonates, children, adolescents, and adults. It failed to meet efficacy criteria for hypoxic-ischemic encephalopathy (HIE) neonatal seizures. In contrast, positive outcomes in temporal lobe epilepsy (TLE), autism, and schizophrenia trials have been attributed to BTN in studies evaluating its off-label use. NKCC1 is an electroneutral neuronal Cl(-) importer and the dominance of NKCC1 function has been proposed as the common pathology for HIE seizures, TLE, autism, and schizophrenia. Therefore, the use of BTN to antagonize neuronal NKCC1 with the goal to lower internal Cl(-) levels and promote GABAergic mediated hyperpolarization has been proposed. In this review, we summarize the data and results for pre-clinical and clinical studies that have tested off-label BTN interventions and report variable outcomes. We also compare the data underlying the developmental expression profile of NKCC1 and KCC2, highlight the limitations of BTN's brain-availability and consider its actions on non-neuronal cells.
- 10Wang, J.; Liu, R.; Hasan, M. N.; Fischer, S.; Chen, Y.; Como, M.; Fiesler, V. M.; Bhuiyan, M. I. H.; Dong, S.; Li, E.; Kahle, K. T.; Zhang, J.; Deng, X.; Subramanya, A. R.; Begum, G.; Yin, Y.; Sun, D. Role of SPAK–NKCC1 Signaling Cascade in the Choroid Plexus Blood–CSF Barrier Damage after Stroke. J. Neuroinflammation 2022, 19 (1), 91, DOI: 10.1186/s12974-022-02456-410Role of SPAK-NKCC1 signaling cascade in the choroid plexus blood-CSF barrier damage after strokeWang, Jun; Liu, Ruijia; Hasan, Md Nabiul; Fischer, Sydney; Chen, Yang; Como, Matt; Fiesler, Victoria M.; Bhuiyan, Mohammad Iqbal H.; Dong, Shuying; Li, Eric; Kahle, Kristopher T.; Zhang, Jinwei; Deng, Xianming; Subramanya, Arohan R.; Begum, Gulnaz; Yin, Yan; Sun, DandanJournal of Neuroinflammation (2022), 19 (1), 91CODEN: JNOEB3; ISSN:1742-2094. (BioMed Central Ltd.)The mechanisms underlying dysfunction of choroid plexus (ChP) blood-cerebrospinal fluid (CSF) barrier and lymphocyte invasion in neuroinflammatory responses to stroke are not well understood. In this study, we investigated whether stroke damaged the blood-CSF barrier integrity due to dysregulation of major ChP ion transport system, Na+-K+-Cl- cotransporter 1 (NKCC1), and regulatory Ste20-related proline-alanine-rich kinase (SPAK). Sham or ischemic stroke was induced in C57Bl/6J mice. Changes on the SPAK-NKCC1 complex and tight junction proteins (TJs) in the ChP were quantified by immunofluorescence staining and immunoblotting. Immune cell infiltration in the ChP was assessed by flow cytometry and immunostaining. Cultured ChP epithelium cells (CPECs) and cortical neurons were used to evaluate H2O2-mediated oxidative stress in stimulating the SPAK-NKCC1 complex and cellular damage. In vivo or in vitro pharmacol. blockade of the ChP SPAK-NKCC1 cascade with SPAK inhibitor ZT-1a or NKCC1 inhibitor bumetanide were examd. Ischemic stroke stimulated activation of the CPECs apical membrane SPAK-NKCC1 complex, NF-κB, and MMP9, which was assocd. with loss of the blood-CSF barrier integrity and increased immune cell infiltration into the ChP. Oxidative stress directly activated the SPAK-NKCC1 pathway and resulted in apoptosis, neurodegeneration, and NKCC1-mediated ion influx. Pharmacol. blockade of the SPAK-NKCC1 pathway protected the ChP barrier integrity, attenuated ChP immune cell infiltration or neuronal death. Stroke-induced patholol. stimulation of the SPAK-NKCC1 cascade caused CPECs damage and disruption of TJs at the blood-CSF barrier. The ChP SPAK-NKCC1 complex emerged as a therapeutic target for attenuating ChP dysfunction and lymphocyte invasion after stroke.
- 11Chew, T. A.; Orlando, B. J.; Zhang, J.; Latorraca, N. R.; Wang, A.; Hollingsworth, S. A.; Chen, D.-H.; Dror, R. O.; Liao, M.; Feng, L. Structure and Mechanism of the Cation–Chloride Cotransporter NKCC1. Nature 2019, 572 (7770), 488– 492, DOI: 10.1038/s41586-019-1438-211Structure and mechanism of the cation-chloride cotransporter NKCC1Chew, Thomas A.; Orlando, Benjamin J.; Zhang, Jinru; Latorraca, Naomi R.; Wang, Amy; Hollingsworth, Scott A.; Chen, Dong-Hua; Dror, Ron O.; Liao, Maofu; Feng, LiangNature (London, United Kingdom) (2019), 572 (7770), 488-492CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Cation-chloride cotransporters (CCCs) mediate the electroneutral transport of chloride, potassium and/or sodium across the membrane. They have crit. roles in regulating cell vol., controlling ion absorption and secretion across epithelia, and maintaining intracellular chloride homeostasis. These transporters are primary targets for some of the most commonly prescribed drugs. Here we detd. the cryo-electron microscopy structure of the Na-K-Cl cotransporter NKCC1, an extensively studied member of the CCC family, from Danio rerio. The structure defines the architecture of this protein family and reveals how cytosolic and transmembrane domains are strategically positioned for communication. Structural analyses, functional characterizations and computational studies reveal the ion-translocation pathway, ion-binding sites and key residues for transport activity. These results provide insights into ion selectivity, coupling and translocation, and establish a framework for understanding the physiol. functions of CCCs and interpreting disease-related mutations.
- 12Moseng, M. A.; Su, C.-C.; Rios, K.; Cui, M.; Lyu, M.; Glaza, P.; Klenotic, P. A.; Delpire, E.; Yu, E. W. Inhibition Mechanism of NKCC1 Involves the Carboxyl Terminus and Long-Range Conformational Coupling. Sci. Adv. 2022, 8 (43), eabq0952 DOI: 10.1126/sciadv.abq0952There is no corresponding record for this reference.
- 13Yang, X.; Wang, Q.; Cao, E. Structure of the Human Cation–Chloride Cotransporter NKCC1 Determined by Single-Particle Electron Cryo-Microscopy. Nat. Commun. 2020, 11 (1), 1016, DOI: 10.1038/s41467-020-14790-313Structure of the human cation-chloride cotransporter NKCC1 determined by single-particle electron cryo-microscopyYang, Xiaoyong; Wang, Qinzhe; Cao, ErhuNature Communications (2020), 11 (1), 1016CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)The secondary active cation-chloride cotransporters (CCCs) utilize the existing Na+ and/or K+ gradients to move Cl- into or out of cells. NKCC1 is an intensively studied member of the CCC family and plays fundamental roles in regulating trans-epithelial ion movement, cell vol., chloride homeostasis and neuronal excitability. Here, we report a cryo-EM structure of human NKCC1 captured in a partially loaded, inward-open state. NKCC1 assembles into a dimer, with the first ten transmembrane (TM) helixes harboring the transport core and TM11-TM12 helixes lining the dimer interface. TM1 and TM6 helixes break α-helical geometry halfway across the lipid bilayer where ion binding sites are organized around these discontinuous regions. NKCC1 may harbor multiple extracellular entryways and intracellular exits, raising the possibility that K+, Na+, and Cl- ions may traverse along their own routes for translocation. NKCC1 structure provides a blueprint for further probing structure-function relationships of NKCC1 and other CCCs.
- 14Zhang, S.; Zhou, J.; Zhang, Y.; Liu, T.; Friedel, P.; Zhuo, W.; Somasekharan, S.; Roy, K.; Zhang, L.; Liu, Y.; Meng, X.; Deng, H.; Zeng, W.; Li, G.; Forbush, B.; Yang, M. The Structural Basis of Function and Regulation of Neuronal Cotransporters NKCC1 and KCC2. Commun. Biol. 2021, 4 (1), 226, DOI: 10.1038/s42003-021-01750-w14The structural basis of function and regulation of neuronal cotransporters NKCC1 and KCC2Zhang, Sensen; Zhou, Jun; Zhang, Yuebin; Liu, Tianya; Friedel, Perrine; Zhuo, Wei; Somasekharan, Suma; Roy, Kasturi; Zhang, Laixing; Liu, Yang; Meng, Xianbin; Deng, Haiteng; Zeng, Wenwen; Li, Guohui; Forbush, Biff; Yang, MaojunCommunications Biology (2021), 4 (1), 226CODEN: CBOIDQ; ISSN:2399-3642. (Nature Research)NKCC and KCC transporters mediate coupled transport of Na++K++Cl- and K++Cl- across the plasma membrane, thus regulating cell Cl- concn. and cell vol. and playing crit. roles in transepithelial salt and water transport and in neuronal excitability. The function of these transporters has been intensively studied, but a mechanistic understanding has awaited structural studies of the transporters. Here, we present the cryo-electron microscopy (cryo-EM) structures of the two neuronal cation-chloride cotransporters human NKCC1 (SLC12A2) and mouse KCC2 (SLC12A5), along with computational anal. and functional characterization. These structures highlight essential residues in ion transport and allow us to propose mechanisms by which phosphorylation regulates transport activity.
- 15Zhao, Y.; Roy, K.; Vidossich, P.; Cancedda, L.; De Vivo, M.; Forbush, B.; Cao, E. Structural Basis for Inhibition of the Cation-Chloride Cotransporter NKCC1 by the Diuretic Drug Bumetanide. Nat. Commun. 2022, 13 (1), 2747, DOI: 10.1038/s41467-022-30407-315Structural basis for inhibition of the Cation-chloride cotransporter NKCC1 by the diuretic drug bumetanideZhao, Yongxiang; Roy, Kasturi; Vidossich, Pietro; Cancedda, Laura; De Vivo, Marco; Forbush, Biff; Cao, ErhuNature Communications (2022), 13 (1), 2747CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)Cation-chloride cotransporters (CCCs) NKCC1 and NKCC2 catalyze electroneutral symport of 1 Na+, 1 K+, and 2 Cl- across cell membranes. NKCC1 mediates trans-epithelial Cl- secretion and regulates excitability of some neurons and NKCC2 is crit. to renal salt reabsorption. Both transporters are inhibited by the so-called loop diuretics including bumetanide, and these drugs are a mainstay for treating edema and hypertension. Here, our single-particle electron cryo-microscopy structures supported by functional studies reveal an outward-facing conformation of NKCC1, showing bumetanide wedged into a pocket in the extracellular ion translocation pathway. Based on these and the previously published inward-facing structures, we define the translocation pathway and the conformational changes necessary for ion translocation. We also identify an NKCC1 dimer with sepd. transmembrane domains and extensive transmembrane and C-terminal domain interactions. We further define an N-terminal phosphoregulatory domain that interacts with the C-terminal domain, suggesting a mechanism whereby (de)phosphorylation regulates NKCC1 by tuning the strength of this domain assocn.
- 16Krishnamurthy, H.; Gouaux, E. X-Ray Structures of LeuT in Substrate-Free Outward-Open and Apo Inward-Open States. Nature 2012, 481 (7382), 469– 474, DOI: 10.1038/nature1073716X-ray structures of LeuT in substrate-free outward-open and apo inward-open statesKrishnamurthy, Harini; Gouaux, EricNature (London, United Kingdom) (2012), 481 (7382), 469-474CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Neurotransmitter sodium symporters are integral membrane proteins that remove chem. transmitters from the synapse and terminate neurotransmission mediated by serotonin, dopamine, noradrenaline, glycine and GABA (γ-aminobutyric acid). Crystal structures of the bacterial homolog, LeuT, in substrate-bound outward-occluded and competitive inhibitor-bound outward-facing states have advanced our mechanistic understanding of neurotransmitter sodium symporters but have left fundamental questions unanswered. Here we report crystal structures of LeuT mutants in complexes with conformation-specific antibody fragments in the outward-open and inward-open states. In the absence of substrate but in the presence of sodium the transporter is outward-open, illustrating how the binding of substrate closes the extracellular gate through local conformational changes: hinge-bending movements of the extracellular halves of transmembrane domains 1, 2 and 6, together with translation of extracellular loop 4. The inward-open conformation, by contrast, involves large-scale conformational changes, including a reorientation of transmembrane domains 1, 2, 5, 6 and 7, a marked hinge bending of transmembrane domain 1a and occlusion of the extracellular vestibule by extracellular loop 4. These changes close the extracellular gate, open an intracellular vestibule, and largely disrupt the two sodium sites, thus providing a mechanism by which ions and substrate are released to the cytoplasm. The new structures establish a structural framework for the mechanism of neurotransmitter sodium symporters and their modulation by therapeutic and illicit substances.
- 17del Alamo, D.; Meiler, J.; Mchaourab, H. S. Principles of Alternating Access in LeuT-Fold Transporters: Commonalities and Divergences. J. Mol. Biol. 2022, 434 (19), 167746 DOI: 10.1016/j.jmb.2022.16774617Principles of Alternating Access in LeuT-fold Transporters: Commonalities and Divergencesdel Alamo, Diego; Meiler, Jens; Mchaourab, Hassane S.Journal of Molecular Biology (2022), 434 (19), 167746CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)A review. Found in all domains of life, transporters belonging to the LeuT-fold class mediate the import and exchange of hydrophilic and charged compds. such as amino acids, metals, and sugar mols. Nearly two decades of studies on the eponymous bacterial transporter LeuT have yielded a library of high-resoln. snapshots of its conformational cycle linked by soln.-state exptl. data obtained from multiple techniques. In parallel, its topol. was obsd. in symporters and antiporters characterized by a spectrum of substrate specificities and coupled to gradients of distinct ions. Here the authors review and compare mechanistic models of transport for LeuT, its well-studied homologs, as well as functionally distant members of the fold, emphasizing the commonalities and divergences in alternating access and the corresponding energy landscapes. The authors' integrated summary illustrates how fold conservation, a hallmark of the LeuT fold, coincides with divergent choreogs. of alternating access that nevertheless capitalize on recurrent structural motifs. In addn., it highlights the knowledge gap that hinders the leveraging of the current body of research into detailed mechanisms of transport for this important class of membrane proteins.
- 18Bonati, L.; Rizzi, V.; Parrinello, M. Data-Driven Collective Variables for Enhanced Sampling. J. Phys. Chem. Lett. 2020, 11 (8), 2998– 3004, DOI: 10.1021/acs.jpclett.0c0053518Data-Driven Collective Variables for Enhanced SamplingBonati, Luigi; Rizzi, Valerio; Parrinello, MicheleJournal of Physical Chemistry Letters (2020), 11 (8), 2998-3004CODEN: JPCLCD; ISSN:1948-7185. (American Chemical Society)Designing an appropriate set of collective variables is crucial to the success of several enhanced sampling methods. Here we focus on how to obtain such variables from information limited to the metastable states. We characterize these states by a large set of descriptors and employ neural networks to compress this information in a lower-dimensional space, using Fisher's linear discriminant as an objective function to maximize the discriminative power of the network. We test this method on alanine dipeptide, using the nonlinearly separable data set composed by at. distances. We then study an intermol. aldol reaction characterized by a concerted mechanism. The resulting variables are able to promote sampling by drawing nonlinear paths in the phys. space connecting the fluctuations between metastable basins. Lastly, we interpret the behavior of the neural network by studying its relation to the phys. variables. Through the identification of its most relevant features, we are able to gain chem. insight into the process.
- 19Invernizzi, M.; Parrinello, M. Exploration vs Convergence Speed in Adaptive-Bias Enhanced Sampling. J. Chem. Theory Comput. 2022, 18 (6), 3988– 3996, DOI: 10.1021/acs.jctc.2c0015219Exploration vs Convergence Speed in Adaptive-Bias Enhanced SamplingInvernizzi, Michele; Parrinello, MicheleJournal of Chemical Theory and Computation (2022), 18 (6), 3988-3996CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)In adaptive-bias enhanced sampling methods, a bias potential is added to the system to drive transitions between metastable states. The bias potential is a function of a few collective variables and is gradually modified according to the underlying free energy surface. We show that when the collective variables are suboptimal, there is an exploration-convergence tradeoff, and one must choose between a quickly converging bias that will lead to fewer transitions or a slower to converge bias that can explore the phase space more efficiently but might require a much longer time to produce an accurate free energy est. The recently proposed on-the-fly probability enhanced sampling (OPES) method focuses on fast convergence, but there are cases where fast exploration is preferred instead. For this reason, we introduce a new variant of the OPES method that focuses on quickly escaping metastable states at the expense of convergence speed. We illustrate the benefits of this approach in prototypical systems and show that it outperforms the popular metadynamics method.
- 20Invernizzi, M.; Parrinello, M. Rethinking Metadynamics: From Bias Potentials to Probability Distributions. J. Phys. Chem. Lett. 2020, 11 (7), 2731– 2736, DOI: 10.1021/acs.jpclett.0c0049720Rethinking Metadynamics: From Bias Potentials to Probability DistributionsInvernizzi, Michele; Parrinello, MicheleJournal of Physical Chemistry Letters (2020), 11 (7), 2731-2736CODEN: JPCLCD; ISSN:1948-7185. (American Chemical Society)Metadynamics is an enhanced sampling method of great popularity, based on the on-the-fly construction of a bias potential that is a function of a selected no. of collective variables. We propose here a change in perspective that shifts the focus from the bias to the probability distribution reconstruction while retaining some of the key characteristics of metadynamics, such as flexible on-the-fly adjustments to the free energy est. The result is an enhanced sampling method that presents a drastic improvement in convergence speed, esp. when dealing with suboptimal and/or multidimensional sets of collective variables. The method is esp. robust and easy to use and in fact requires only a few simple parameters to be set, and it has a straightforward reweighting scheme to recover the statistics of the unbiased ensemble. Furthermore, it gives more control of the desired exploration of the phase space since the deposited bias is not allowed to grow indefinitely and it does not push the simulation to uninteresting high free energy regions. We demonstrate the performance of the method in a no. of representative examples.
- 21Yamashita, A.; Singh, S. K.; Kawate, T.; Jin, Y.; Gouaux, E. Crystal Structure of a Bacterial Homologue of Na+/Cl--Dependent Neurotransmitter Transporters. Nature 2005, 437 (7056), 215– 223, DOI: 10.1038/nature0397821Crystal structure of a bacterial homologue of Na+/Cl--dependent neurotransmitter transportersYamashita, Atsuko; Singh, Satinder K.; Kawate, Toshimitsu; Jin, Yan; Gouaux, EricNature (London, United Kingdom) (2005), 437 (7056), 215-223CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Na+/Cl--dependent transporters terminate synaptic transmission by using electrochem. gradients to drive the uptake of neurotransmitters, including the biogenic amines, from the synapse to the cytoplasm of neurons and glia. These transporters are the targets of therapeutic and illicit compds., and their dysfunction has been implicated in multiple diseases of the nervous system. Here we present the crystal structure of a bacterial homolog of these transporters from Aquifex aeolicus, in complex with its substrate, leucine, and two sodium ions. The protein core consists of the first ten of twelve transmembrane segments, with segments 1-5 related to 6-10 by a pseudo-two-fold axis in the membrane plane. Leucine and the sodium ions are bound within the protein core, halfway across the membrane bilayer, in an occluded site devoid of water. The leucine and ion binding sites are defined by partially unwound transmembrane helixes, with main-chain atoms and helix dipoles having key roles in substrate and ion binding. The structure reveals the architecture of this important class of transporter, illuminates the determinants of substrate binding and ion selectivity, and defines the external and internal gates.
- 22Forrest, L. R.; Rudnick, G. The Rocking Bundle: A Mechanism for Ion-Coupled Solute Flux by Symmetrical Transporters. Physiology 2009, 24 (6), 377– 386, DOI: 10.1152/physiol.00030.200922The rocking bundle: a mechanism for ion-coupled solute flux by symmetrical transportersForrest, Lucy R.; Rudnick, GaryPhysiology (2009), 24 (Dec.), 377-386CODEN: PHYSCI; ISSN:1548-9213. (International Union of Physiological Sciences)A review. Crystal structures of the bacterial amino acid transporter LeuT have provided the basis for understanding the conformational changes assocd. with substrate translocation by a multitude of transport proteins with the same fold. Biochem. and modeling studies led to a "rocking bundle" mechanism for LeuT that was validated by subsequent transporter structures. These advances suggest how coupled solute transport might be defined by the internal symmetry of proteins contg. inverted structural repeats.
- 23Masrati, G.; Mondal, R.; Rimon, A.; Kessel, A.; Padan, E.; Lindahl, E.; Ben-Tal, N. An Angular Motion of a Conserved Four-Helix Bundle Facilitates Alternating Access Transport in the TtNapA and EcNhaA Transporters. Proc. Natl. Acad. Sci. U. S. A. 2020, 117 (50), 31850– 31860, DOI: 10.1073/pnas.200271011723An angular motion of a conserved four-helix bundle facilitates alternating access transport in the TtNapA and EcNhaA transportersMasrati, Gal; Mondal, Ramakanta; Rimon, Abraham; Kessel, Amit; Padan, Etana; Lindahl, Erik; Ben-Tal, NirProceedings of the National Academy of Sciences of the United States of America (2020), 117 (50), 31850-31860CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)There is ongoing debate regarding the mechanism through which cation/proton antiporters (CPAs), like Thermus thermophilus NapA (TtNapA) and Escherichia coli NapA (EcNhaA), alternate between their outward- and inward-facing conformations in the membrane. CPAs comprise two domains, and it is unclear whether the transition is driven by their rocking-bundle or elevator motion with respect to each other. Here we address this question using metadynamics simulations of TtNapA, where we bias conformational sampling along two axes characterizing the two proposed mechanisms: angular and translational motions, resp. By applying the bias potential for the two axes simultaneously, as well as to the angular, but not the translational, axis alone, we manage to reproduce each of the two known states of TtNapA when starting from the opposite state, in support of the rocking-bundle mechanism as the driver of conformational change. Next, starting from the inward-facing conformation of EcNhaA, we sample what could be its long-sought-after outward-facing conformation and verify it using crosslinking expts.
- 24Borgogno, M.; Savardi, A.; Manigrasso, J.; Turci, A.; Portioli, C.; Ottonello, G.; Bertozzi, S. M.; Armirotti, A.; Contestabile, A.; Cancedda, L.; De Vivo, M. Design, Synthesis, In Vitro and In Vivo Characterization of Selective NKCC1 Inhibitors for the Treatment of Core Symptoms in Down Syndrome. J. Med. Chem. 2021, 64 (14), 10203– 10229, DOI: 10.1021/acs.jmedchem.1c0060324Design, Synthesis, In Vitro and In Vivo Characterization of Selective NKCC1 Inhibitors for the Treatment of Core Symptoms in Down SyndromeBorgogno, Marco; Savardi, Annalisa; Manigrasso, Jacopo; Turci, Alessandra; Portioli, Corinne; Ottonello, Giuliana; Bertozzi, Sine Mandrup; Armirotti, Andrea; Contestabile, Andrea; Cancedda, Laura; De Vivo, MarcoJournal of Medicinal Chemistry (2021), 64 (14), 10203-10229CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Intracellular chloride concn. [Cl-]i is defective in several neurol. disorders. In neurons, [Cl-]i is mainly regulated by the action of the Na+-K+-Cl- importer NKCC1 and the K+-Cl- exporter KCC2. Recently, we have reported the discovery of ARN23746(I) as the lead candidate of a novel class of selective inhibitors of NKCC1. Importantly, ARN23746 is able to rescue core symptoms of Down syndrome (DS) and autism in mouse models. Here, we describe the discovery and extensive characterization of this chem. class of selective NKCC1 inhibitors, with focus on ARN23746 and other promising derivs. In particular, we present compd. 40 (ARN24092)(II) as a backup/follow-up lead with in vivo efficacy in a mouse model of DS. These results further strengthen the potential of this new class of compds. for the treatment of core symptoms of brain disorders characterized by the defective NKCC1/KCC2 expression ratio.
- 25Savardi, A.; Borgogno, M.; Narducci, R.; La Sala, G.; Ortega, J. A.; Summa, M.; Armirotti, A.; Bertorelli, R.; Contestabile, A.; De Vivo, M.; Cancedda, L. Discovery of a Small Molecule Drug Candidate for Selective NKCC1 Inhibition in Brain Disorders. Chem. 2020, 6 (8), 2073– 2096, DOI: 10.1016/j.chempr.2020.06.01725Discovery of a Small Molecule Drug Candidate for Selective NKCC1 Inhibition in Brain DisordersSavardi, Annalisa; Borgogno, Marco; Narducci, Roberto; La Sala, Giuseppina; Ortega, Jose Antonio; Summa, Maria; Armirotti, Andrea; Bertorelli, Rosalia; Contestabile, Andrea; De Vivo, Marco; Cancedda, LauraChem (2020), 6 (8), 2073-2096CODEN: CHEMVE; ISSN:2451-9294. (Cell Press)Aberrant expression ratio of Cl- transporters, NKCC1 and KCC2, is implicated in several brain conditions. NKCC1 inhibition by the FDA-approved diuretic drug, bumetanide, rescues core symptoms in rodent models and/or clin. trials with patients. However, bumetanide has a strong diuretic effect due to inhibition of the kidney Cl- transporter NKCC2, creating crit. drug compliance issues and health concerns. Here, we report the discovery of a new chem. class of selective NKCC1 inhibitors and the lead drug candidate ARN23746. ARN23746 restores the physiol. intracellular Cl- in murine Down syndrome neuronal cultures, has excellent soly. and metabolic stability, and displays no issues with off-target activity in vitro. ARN23746 recovers core symptoms in mouse models of Down syndrome and autism, with no diuretic effect, nor overt toxicity upon chronic treatment in adulthood. ARN23746 is ready for advanced preclin./manufg. studies toward the first sustainable therapeutics for the neurol. conditions characterized by impaired Cl- homeostasis.
- 26Savardi, A.; Patricelli Malizia, A.; De Vivo, M.; Cancedda, L.; Borgogno, M. Preclinical Development of the Na-K-2Cl Co-Transporter-1 (NKCC1) Inhibitor ARN23746 for the Treatment of Neurodevelopmental Disorders. ACS Pharmacol. Transl. Sci. 2023, 6 (1), 1– 11, DOI: 10.1021/acsptsci.2c0019726Preclinical Development of the Na-K-2Cl Co-transporter-1 (NKCC1) Inhibitor ARN23746 for the Treatment of Neurodevelopmental DisordersSavardi, Annalisa; Patricelli Malizia, Andrea; De Vivo, Marco; Cancedda, Laura; Borgogno, MarcoACS Pharmacology & Translational Science (2023), 6 (1), 1-11CODEN: APTSFN; ISSN:2575-9108. (American Chemical Society)Alterations in the expression of the Cl- importer Na-K-2Cl co-transporter-1 (NKCC1) and the exporter K-Cl co-transporter 2 (KCC2) lead to impaired intracellular chloride concn. in neurons and imbalanced excitation/inhibition in the brain. These alterations have been obsd. in several neurol. disorders (e.g., Down syndrome and autism). Recently, we have reported the discovery of the selective NKCC1 inhibitor "compd. ARN23746" for the treatment of Down syndrome and autism in mouse models. Here, we report on an extensive preclin. characterization of ARN23746 toward its development as a clin. candidate. ARN23746 shows an overall excellent metab. profile and good brain penetration. Moreover, ARN23746 is effective in rescuing cognitive impairment in Down syndrome mice upon per os administration, in line with oral treatment of neurodevelopmental disorders. Notably, ARN23746 does not present signs of toxicity or diuresis even if administered up to 50 times the ED. These results further support ARN23746 as a solid candidate for clin. trial-enabling studies.
- 27Chew, T. A.; Zhang, J.; Feng, L. High-Resolution Views and Transport Mechanisms of the NKCC1 and KCC Transporters. J. Mol. Biol. 2021, 433 (16), 167056 DOI: 10.1016/j.jmb.2021.16705627High-Resolution Views and Transport Mechanisms of the NKCC1 and KCC TransportersChew, Thomas A.; Zhang, Jinru; Feng, LiangJournal of Molecular Biology (2021), 433 (16), 167056CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)A review. Cation-chloride cotransporters (CCCs) are responsible for the coupled co-transport of Cl- with K+ and/or Na+ in an electroneutral manner. They play important roles in myriad fundamental physiol. processes--from cell vol. regulation to transepithelial solute transport and intracellular ion homeostasis--and are targeted by medicines commonly prescribed to treat hypertension and edema. After several decades of studies into the functions and pharmacol. of these transporters, there have been several breakthroughs in the structural detn. of CCC transporters. The insights provided by these new structures for the Na+/K+/Cl- cotransporter NKCC1 and the K+/Cl- cotransporters KCC1, KCC2, KCC3 and KCC4 have deepened our understanding of their mol. basis and transport function. This focused discusses recent advances in the structural and mechanistic understanding of CCC transporters, including architecture, dimerization, functional roles of regulatory domains, ion binding sites, and coupled ion transport.
- 28Lytle, C.; McManus, T. J.; Haas, M. A Model of Na-K-2Cl Cotransport Based on Ordered Ion Binding and Glide Symmetry. Am. J. Physiol.-Cell Physiol. 1998, 274 (2), C299– C309, DOI: 10.1152/ajpcell.1998.274.2.C299There is no corresponding record for this reference.
- 29Delpire, E.; Gagnon, K. B. Na + -K + −2Cl – Cotransporter (NKCC) Physiological Function in Nonpolarized Cells and Transporting Epithelia. In Comprehensive Physiology; Terjung, R., Ed.; Wiley, 2018; pp. 871– 901.There is no corresponding record for this reference.
- 30Shimamura, T.; Weyand, S.; Beckstein, O.; Rutherford, N. G.; Hadden, J. M.; Sharples, D.; Sansom, M. S. P.; Iwata, S.; Henderson, P. J. F.; Cameron, A. D. Molecular Basis of Alternating Access Membrane Transport by the Sodium-Hydantoin Transporter Mhp1. Science 2010, 328 (5977), 470– 473, DOI: 10.1126/science.118630330Molecular Basis of Alternating Access Membrane Transport by the Sodium-Hydantoin Transporter Mhp1Shimamura, Tatsuro; Weyand, Simone; Beckstein, Oliver; Rutherford, Nicholas G.; Hadden, Jonathan M.; Sharples, David; Sansom, Mark S. P.; Iwata, So; Henderson, Peter J. F.; Cameron, Alexander D.Science (Washington, DC, United States) (2010), 328 (5977), 470-473CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The structure of the sodium-benzylhydantoin transport protein Mhp1 from Microbacterium liquefaciens is comprised of a five-helix inverted repeat, a conformation widespread among secondary transporters. Here, we report the crystal structure of an inward-facing conformation of Mhp1 at 3.8 angstroms resoln., which complements previously described structures of Mhp1 in outward-facing and occluded states. From analyses of the three structures and mol. dynamics (MD) simulations, we propose a mechanism for the transport cycle in Mhp1. Switching from the outward- to the inward-facing state, to effect the inward release of sodium and benzylhydantoin, is primarily achieved by a rigid body movement of transmembrane helixes 3, 4, 8, and 9 relative to the rest of the protein. This forms the basis of an alternating access mechanism applicable to many transporters of this emerging superfamily.
- 31Hamann, S.; Herrera-Perez, J. J.; Zeuthen, T.; Alvarez-Leefmans, F. J. Cotransport of Water by the Na + -K + −2Cl – Cotransporter NKCC1 in Mammalian Epithelial Cells: Cotransport of Water by NKCC1. J. Physiol. 2010, 588 (21), 4089– 4101, DOI: 10.1113/jphysiol.2010.19473831Cotransport of water by the Na+-K+-2Cl- cotransporter NKCC1 in mammalian epithelial cellsHamann, Steffen; Herrera-Perez, Jose J.; Zeuthen, Thomas; Alvarez-Leefmans, Francisco J.Journal of Physiology (Oxford, United Kingdom) (2010), 588 (21), 4089-4101CODEN: JPHYA7; ISSN:0022-3751. (Wiley-Blackwell)Water transport by the Na+-K+-2Cl- cotransporter (NKCC1) was studied in confluent cultures of pigmented epithelial (PE) cells from the ciliary body of the fetal human eye. Interdependence among water, Na+ and Cl- fluxes mediated by NKCC1 was inferred from changes in cell water vol., monitored by intracellular self-quenching of the fluorescent dye calcein. Isosmotic removal of external Cl- or Na+ caused a rapid efflux of water from the cells, which was inhibited by bumetanide (10 μM). When returned to the control soln. there was a rapid water influx that required the simultaneous presence of external Na+ and Cl-. The water influx could proceed uphill, against a transmembrane osmotic gradient, suggesting that energy contained in the ion fluxes can be transferred to the water flux. The influx of water induced by changes in external [Cl-] satd. in a sigmoidal fashion with a Km of 60 mM, while that induced by changes in external [Na+] followed first order kinetics with a Km of about 40 mM. These parameters are consistent with ion transport mediated by NKCC1. Our findings support a previous investigation, in which we showed water transport by NKCC1 to be a result of a balance between ionic and osmotic gradients. The coupling between salt and water transport in NKCC1 represents a novel aspect of cellular water homeostasis where cells can change their vol. independently of the direction of an osmotic gradient across the membrane. This has relevance for both epithelial and sym. cells.
- 32Zeuthen, T.; MacAulay, N. Cotransport of Water by Na + -K + −2Cl – Cotransporters Expressed in Xenopus Oocytes: NKCC1 versus NKCC2: Water Transport in NKCC. J. Physiol. 2012, 590 (5), 1139– 1154, DOI: 10.1113/jphysiol.2011.22631632Cotransport of water by Na+-K+-2Cl- cotransporters expressed in Xenopus oocytes: NKCC1 versus NKCC2Zeuthen, Thomas; MacAulay, NannaJournal of Physiology (Oxford, United Kingdom) (2012), 590 (5), 1139-1154CODEN: JPHYA7; ISSN:0022-3751. (Wiley-Blackwell)The NKCC1 and NKCC2 isoforms of the mammalian Na+-K+-2Cl- cotransporter were expressed in Xenopus oocytes and the relation between external ion concn. and water fluxes detd. Water fluxes were detd. from changes in the oocytes vol. and ion fluxes from 86Rb+ uptake. Isotonic increases in external K+ concn. elicited abrupt inward water fluxes in NKCC1; the K+ dependence obeyed one-site kinetics with a K0.5 of 7.5 mM. The water fluxes were blocked by bumetanide, had steep temp. dependence and could proceed uphill against an osmotic gradient of 20 mosmol L-1. A comparison between ion and water fluxes indicates that 460 water mols. are cotransported for each turnover of the protein. In contrast, NKCC2 did not support water fluxes. Water transport in NKCC1 induced by increases in the external osmolarity had high activation energy and was blocked by bumetanide. The osmotic effects of NaCl were smaller than those of urea and mannitol. This supports the notion of interaction between ions and water in NKCC1 and allows for an est. of around 600 water mols. transported per turnover of the protein. Osmotic gradients did not induce water transport in NKCC2. We conclude that NKCC1 plays a direct role for water balance in most cell types, while NKCC2 fulfills its role in the kidney of transporting ions but not water. The different behavior of NKCC1 and NKCC2 is discussed on the basis of recent mol. models based on studies of structural and mol. dynamics.
- 33Sadegh, C.; Xu, H.; Sutin, J.; Fatou, B.; Gupta, S.; Pragana, A.; Taylor, M.; Kalugin, P. N.; Zawadzki, M. E.; Alturkistani, O.; Shipley, F. B.; Dani, N.; Fame, R. M.; Wurie, Z.; Talati, P.; Schleicher, R. L.; Klein, E. M.; Zhang, Y.; Holtzman, M. J.; Moore, C. I.; Lin, P.-Y.; Patel, A. B.; Warf, B. C.; Kimberly, W. T.; Steen, H.; Andermann, M. L.; Lehtinen, M. K. Choroid Plexus-Targeted NKCC1 Overexpression to Treat Post-Hemorrhagic Hydrocephalus. Neuron 2023, 111 (10), 1591– 1608, DOI: 10.1016/j.neuron.2023.02.02033Choroid plexus-targeted NKCC1 overexpression to treat post-hemorrhagic hydrocephalusSadegh, Cameron; Xu, Huixin; Sutin, Jason; Fatou, Benoit; Gupta, Suhasini; Pragana, Aja; Taylor, Milo; Kalugin, Peter N.; Zawadzki, Miriam E.; Alturkistani, Osama; Shipley, Frederick B.; Dani, Neil; Fame, Ryann M.; Wurie, Zainab; Talati, Pratik; Schleicher, Riana L.; Klein, Eric M.; Zhang, Yong; Holtzman, Michael J.; Moore, Christopher I.; Lin, Pei-Yi; Patel, Aman B.; Warf, Benjamin C.; Kimberly, W. Taylor; Steen, Hanno; Andermann, Mark L.; Lehtinen, Maria K.Neuron (2023), 111 (10), 1591-1608.e4CODEN: NERNET; ISSN:0896-6273. (Cell Press)Post-hemorrhagic hydrocephalus (PHH) refers to a life-threatening accumulation of cerebrospinal fluid (CSF) that occurs following intraventricular hemorrhage (IVH). An incomplete understanding of this variably progressive condition has hampered the development of new therapies beyond serial neurosurgical interventions. Here, we show a key role for the bidirectional Na-K-Cl cotransporter, NKCC1, in the choroid plexus (ChP) to mitigate PHH. Mimicking IVH with intraventricular blood led to increased CSF [K+] and triggered cytosolic calcium activity in ChP epithelial cells, which was followed by NKCC1 activation. ChP-targeted adeno-assocd. viral (AAV)-NKCC1 prevented blood-induced ventriculomegaly and led to persistently increased CSF clearance capacity. These data demonstrate that intraventricular blood triggered a trans-choroidal, NKCC1-dependent CSF clearance mechanism. Inactive, phosphodeficient AAV-NKCC1-NT51 failed to mitigate ventriculomegaly. Excessive CSF [K+] fluctuations correlated with permanent shunting outcome in humans following hemorrhagic stroke, suggesting targeted gene therapy as a potential treatment to mitigate intracranial fluid accumulation following hemorrhage.
- 34Steffensen, A. B.; Oernbo, E. K.; Stoica, A.; Gerkau, N. J.; Barbuskaite, D.; Tritsaris, K.; Rose, C. R.; MacAulay, N. Cotransporter-Mediated Water Transport Underlying Cerebrospinal Fluid Formation. Nat. Commun. 2018, 9 (1), 2167, DOI: 10.1038/s41467-018-04677-934Cotransporter-mediated water transport underlying cerebrospinal fluid formationSteffensen Annette B; Oernbo Eva K; Stoica Anca; MacAulay Nanna; Gerkau Niklas J; Rose Christine R; Barbuskaite Dagne; Tritsaris KaterinaNature communications (2018), 9 (1), 2167 ISSN:.Cerebrospinal fluid (CSF) production occurs at a rate of 500 ml per day in the adult human. Conventional osmotic forces do not suffice to support such production rate and the molecular mechanisms underlying this fluid production remain elusive. Using ex vivo choroid plexus live imaging and isotope flux in combination with in vivo CSF production determination in mice, we identify a key component in the CSF production machinery. The Na(+)/K(+)/2Cl(-) cotransporter (NKCC1) expressed in the luminal membrane of choroid plexus contributes approximately half of the CSF production, via its unusual outward transport direction and its unique ability to directly couple water transport to ion translocation. We thereby establish the concept of cotransport of water as a missing link in the search for molecular pathways sustaining CSF production and redefine the current model of this pivotal physiological process. Our results provide a rational pharmacological target for pathologies involving disturbed brain fluid dynamics.
- 35Li, J.; Shaikh, S. A.; Enkavi, G.; Wen, P.-C.; Huang, Z.; Tajkhorshid, E. Transient Formation of Water-Conducting States in Membrane Transporters. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (19), 7696– 7701, DOI: 10.1073/pnas.121898611035Transient formation of water-conducting states in membrane transportersLi, Jing; Shaikh, Saher A.; Enkavi, Giray; Wen, Po-Chao; Huang, Zhijian; Tajkhorshid, EmadProceedings of the National Academy of Sciences of the United States of America (2013), 110 (19), 7696-7701, S7696/1-S7696/7CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Membrane transporters rely on highly coordinated structural transitions between major conformational states for their function, to prevent simultaneous access of the substrate binding site to both sides of the membrane - a mode of operation known as the alternating access model. Although this mechanism successfully accounts for the efficient exchange of the primary substrate across the membrane, accruing evidence on significant water transport and even uncoupled ion transport mediated by transporters has challenged the concept of perfect mech. coupling and coordination of the gating mechanism in transporters, which might be expected from the alternating access model. Here, we present a large set of extended equil. mol. dynamics simulations performed on several classes of membrane transporters in different conformational states, to test the presence of the phenomenon in diverse transporter classes and to investigate the underlying mol. mechanism of water transport through membrane transporters. The simulations reveal spontaneous formation of transient water-conducting (channel-like) states allowing passive water diffusion through the lumen of the transporters. These channel-like states are permeable to water but occluded to substrate, thereby not hindering the uphill transport of the primary substrate, i.e., the alternating access model remains applicable to the substrate. The rise of such water-conducting states during the large-scale structural transitions of the transporter protein is indicative of imperfections in the coordinated closing and opening motions of the cytoplasmic and extracellular gates. We propose that the obsd. water-conducting states likely represent a universal phenomenon in membrane transporters, which is consistent with their reliance on large-scale motion for function.
- 36Okazaki, K.; Wöhlert, D.; Warnau, J.; Jung, H.; Yildiz, Ö.; Kühlbrandt, W.; Hummer, G. Mechanism of the Electroneutral Sodium/Proton Antiporter PaNhaP from Transition-Path Shooting. Nat. Commun. 2019, 10 (1), 1742, DOI: 10.1038/s41467-019-09739-036Mechanism of the electroneutral sodium/proton antiporter PaNhaP from transition-path shootingOkazaki Kei-Ichi; Okazaki Kei-Ichi; Warnau Judith; Jung Hendrik; Hummer Gerhard; Wohlert David; Yildiz Ozkan; Kuhlbrandt Werner; Hummer GerhardNature communications (2019), 10 (1), 1742 ISSN:.Na(+)/H(+) antiporters exchange sodium ions and protons on opposite sides of lipid membranes. The electroneutral Na(+)/H(+) antiporter NhaP from archaea Pyrococcus abyssi (PaNhaP) is a functional homolog of the human Na(+)/H(+) exchanger NHE1, which is an important drug target. Here we resolve the Na(+) and H(+) transport cycle of PaNhaP by transition-path sampling. The resulting molecular dynamics trajectories of repeated ion transport events proceed without bias force, and overcome the enormous time-scale gap between seconds-scale ion exchange and microseconds simulations. The simulations reveal a hydrophobic gate to the extracellular side that opens and closes in response to the transporter domain motion. Weakening the gate by mutagenesis makes the transporter faster, suggesting that the gate balances competing demands of fidelity and efficiency. Transition-path sampling and a committor-based reaction coordinate optimization identify the essential motions and interactions that realize conformational alternation between the two access states in transporter function.
- 37Monette, M. Y.; Somasekharan, S.; Forbush, B. Molecular Motions Involved in Na-K-Cl Cotransporter-Mediated Ion Transport and Transporter Activation Revealed by Internal Cross-Linking between Transmembrane Domains 10 and 11/12. J. Biol. Chem. 2014, 289 (11), 7569– 7579, DOI: 10.1074/jbc.M113.54225837Molecular Motions Involved in Na-K-Cl Cotransporter-mediated Ion Transport and Transporter Activation Revealed by Internal Cross-linking between Transmembrane Domains 10 and 11/12Monette, Michelle Y.; Somasekharan, Suma; Forbush, BiffJournal of Biological Chemistry (2014), 289 (11), 7569-7579CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)We examd. the relationship between transmembrane domain (TM) 10 and TM11/12 in NKCC1, testing homol. models based on the structure of AdiC in the same transporter superfamily. We hypothesized that introduced cysteine pairs would be close enough for disulfide formation and would alter transport function; indeed, evidence for cross-link formation with low micromolar concns. of copper phenanthroline or iodine was found in 3 of 8 initially tested pairs and in 1 of 26 addnl. tested pairs. Inhibition of transport was obsd. with copper phenanthroline and iodine treatment of P676C/A734C and I677C/A734C, consistent with the proximity of these residues and with movement of TM10 during the occlusion step of ion transport. We also found Cu2+ inhibition of the single-cysteine mutant A675C, suggesting that this residue and Met382 of TM3 are involved in a Cu2+-binding site. Surprisingly, crosslinking of P676C/I730C was found to prevent rapid deactivation of the transporter while not affecting the dephosphorylation rate, thus uncoupling the phosphorylation and activation steps. Consistent with this, (a) crosslinking of P676C/I730C was dependent on activation state, and (b) mutants lacking the phosphoregulatory domain could still be activated by crosslinking. These results suggest a model of NKCC activation that involves movement of TM12 relative to TM10, which is likely tied to movement of the large C terminus, a process somehow triggered by phosphorylation of the regulatory domain in the N terminus.
- 38McNeill, A.; Iovino, E.; Mansard, L.; Vache, C.; Baux, D.; Bedoukian, E.; Cox, H.; Dean, J.; Goudie, D.; Kumar, A.; Newbury-Ecob, R.; Fallerini, C.; Renieri, A.; Lopergolo, D.; Mari, F.; Blanchet, C.; Willems, M.; Roux, A.-F.; Pippucci, T.; Delpire, E. SLC12A2 Variants Cause a Neurodevelopmental Disorder or Cochleovestibular Defect. Brain 2020, 143 (8), 2380– 2387, DOI: 10.1093/brain/awaa17638SLC12A2 variants cause a neurodevelopmental disorder or cochleovestibular defectMcNeill Alisdair; McNeill Alisdair; McNeill Alisdair; Iovino Emanuela; Mansard Luke; Vache Christel; Baux David; Roux Anne-Francoise; Bedoukian Emma; Cox Helen; Dean John; Goudie David; Kumar Ajith; Newbury-Ecob Ruth; Fallerini Chiara; Renieri Alessandra; Lopergolo Diego; Mari Francesca; Fallerini Chiara; Renieri Alessandra; Lopergolo Diego; Mari Francesca; Blanchet Catherine; Willems Marjolaine; Pippucci Tommaso; Delpire EricBrain : a journal of neurology (2020), 143 (8), 2380-2387 ISSN:.The SLC12 gene family consists of SLC12A1-SLC12A9, encoding electroneutral cation-coupled chloride co-transporters. SCL12A2 has been shown to play a role in corticogenesis and therefore represents a strong candidate neurodevelopmental disorder gene. Through trio exome sequencing we identified de novo mutations in SLC12A2 in six children with neurodevelopmental disorders. All had developmental delay or intellectual disability ranging from mild to severe. Two had sensorineural deafness. We also identified SLC12A2 variants in three individuals with non-syndromic bilateral sensorineural hearing loss and vestibular areflexia. The SLC12A2 de novo mutation rate was demonstrated to be significantly elevated in the deciphering developmental disorders cohort. All tested variants were shown to reduce co-transporter function in Xenopus laevis oocytes. Analysis of SLC12A2 expression in foetal brain at 16-18 weeks post-conception revealed high expression in radial glial cells, compatible with a role in neurogenesis. Gene co-expression analysis in cells robustly expressing SLC12A2 at 16-18 weeks post-conception identified a transcriptomic programme associated with active neurogenesis. We identify SLC12A2 de novo mutations as the cause of a novel neurodevelopmental disorder and bilateral non-syndromic sensorineural hearing loss and provide further data supporting a role for this gene in human neurodevelopment.
- 39Janoš, P.; Magistrato, A. All-Atom Simulations Uncover the Molecular Terms of the NKCC1 Transport Mechanism. J. Chem. Inf. Model. 2021, 61 (7), 3649– 3658, DOI: 10.1021/acs.jcim.1c0055139All-Atom Simulations Uncover the Molecular Terms of the NKCC1 Transport MechanismJanos, Pavel; Magistrato, AlessandraJournal of Chemical Information and Modeling (2021), 61 (7), 3649-3658CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)The secondary-active Na-K-Cl cotransporter 1 (NKCC1), member of the cation-chloride cotransporter (CCC) family, ensures the electroneutral movement of Cl-, Na+, and K+ ions across cellular membranes. NKCC1 regulates Cl- homeostasis and cell vol., handling a pivotal role in transepithelial water transport and neuronal excitability. Aberrant NKCC1 transport is hence implicated in a variety of human diseases (hypertension, renal disorders, neuropathies, and cancer). Building on the newly resolved NKCC1 cryo-EM structure, all-atom enhanced sampling simulations unprecedentedly unlock the mechanism of NKCC1-mediated ion transport, assessing the order and the mol. basis of its interdependent ion translocation. The authors' outcomes strikingly advance the understanding of the physiol. mechanism of CCCs and disclose a key role of CCC-conserved asparagine residues, whose side-chain promiscuity ensures the transport of both neg. and pos. charged ions along the same translocation route. This study sets a conceptual basis to devise NKCC-selective inhibitors to treat diseases linked to Cl- dishomeostasis.
- 40Portioli, C.; Ruiz Munevar, M. J.; De Vivo, M.; Cancedda, L. Cation-Coupled Chloride Cotransporters: Chemical Insights and Disease Implications. Trends Chem. 2021, 3 (10), 832– 849, DOI: 10.1016/j.trechm.2021.05.00440Cation-coupled chloride cotransporters: chemical insights and disease implicationsPortioli, Corinne; Ruiz Munevar, Manuel Jose; De Vivo, Marco; Cancedda, LauraTrends in Chemistry (2021), 3 (10), 832-849CODEN: TCRHBQ; ISSN:2589-5974. (Cell Press)A review. Cation-coupled chloride cotransporters (CCCs) modulate the transport of sodium and/or potassium cations coupled with chloride anions across the cell membrane. CCCs thus help regulate intracellular ionic concn. and consequent cell vol. homeostasis. This has been largely exploited in the past to develop diuretic drugs that act on CCCs expressed in the kidney. However, a growing wealth of evidence has demonstrated that CCCs are also critically involved in a great variety of other pathologies, motivating most recent drug discovery programs targeting CCCs. Here, we examine the structure-function relationship of CCCs. By linking recent high-resoln. cryogenic electron microscopy (cryo-EM) data with older biochem./functional studies on CCCs, we discuss the mechanistic insights and opportunities to design selective CCC modulators to treat diverse pathologies.
- 41Zhao, Y.; Shen, J.; Wang, Q.; Ruiz Munevar, M. J.; Vidossich, P.; De Vivo, M.; Zhou, M.; Cao, E. Structure of the Human Cation–Chloride Cotransport KCC1 in an Outward-Open State. Proc. Natl. Acad. Sci. U. S. A. 2022, 119 (27), e2109083119 DOI: 10.1073/pnas.210908311941Structure of the human cation-chloride cotransport KCC1 in an outward-open stateZhao, Yongxiang; Shen, Jiemin; Wang, Qinzhe; Munevar, Manuel Jose Ruiz; Vidossich, Pietro; De Vivo, Marco; Zhou, Ming; Cao, ErhuProceedings of the National Academy of Sciences of the United States of America (2022), 119 (27), e2109083119CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Cation-chloride cotransporters (CCCs) catalyze electroneutral symport of Cl- with Na+ and/or K+ across membranes. CCCs are fundamental in cell vol. homeostasis, transepithelia ion movement, maintenance of intracellular Cl- concn., and neuronal excitability. Here, we present a cryoelectron microscopy structure of human K+-Cl- cotransporter (KCC)1 bound with the VU0463271 inhibitor in an outward-open state. In contrast to many other amino acid-polyamine-organocation transporter cousins, our first outward-open CCC structure reveals that opening the KCC1 extracellular ion permeation path does not involve hinge-bending motions of the transmembrane (TM) 1 and TM6 half-helixes. Instead, rocking of TM3 and TM8, together with displacements of TM4, TM9, and a conserved intracellular loop 1 helix, underlie alternate opening and closing of extracellular and cytoplasmic vestibules. We show that KCC1 intriguingly exists in one of two distinct dimeric states via different intersubunit interfaces. Our studies provide a blueprint for understanding the mechanisms of CCCs and their inhibition by small mol. compds.
- 42Saitsu, H.; Watanabe, M.; Akita, T.; Ohba, C.; Sugai, K.; Ong, W. P.; Shiraishi, H.; Yuasa, S.; Matsumoto, H.; Beng, K. T.; Saitoh, S.; Miyatake, S.; Nakashima, M.; Miyake, N.; Kato, M.; Fukuda, A.; Matsumoto, N. Impaired Neuronal KCC2 Function by Biallelic SLC12A5Mutations in Migrating Focal Seizures and Severe Developmental Delay. Sci. Rep. 2016, 6 (1), 30072, DOI: 10.1038/srep3007242Impaired neuronal KCC2 function by biallelic SLC12A5 mutations in migrating focal seizures and severe developmental delaySaitsu, Hirotomo; Watanabe, Miho; Akita, Tenpei; Ohba, Chihiro; Sugai, Kenji; Ong, Winnie Peitee; Shiraishi, Hideaki; Yuasa, Shota; Matsumoto, Hiroshi; Beng, Khoo Teik; Saitoh, Shinji; Miyatake, Satoko; Nakashima, Mitsuko; Miyake, Noriko; Kato, Mitsuhiro; Fukuda, Atsuo; Matsumoto, NaomichiScientific Reports (2016), 6 (), 30072CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)Epilepsy of infancy with migrating focal seizures (EIMFS) is one of the early-onset epileptic syndromes characterized by migrating polymorphous focal seizures. Whole exome sequencing (WES) in ten sporadic and one familial case of EIMFS revealed compd. heterozygous SLC12A5 (encoding the neuronal K+-Cl- co-transporter KCC2) mutations in two families: c.279 + 1G > C causing skipping of exon 3 in the transcript (p.E50_Q93del) and c.572 C >T (p.A191V) in individuals 1 and 2, and c.967T > C (p.S323P) and c.1243 A > G (p.M415V) in individual 3. Another patient (individual 4) with migrating multifocal seizures and compd. heterozygous mutations [c.953G > C (p.W318S) and c.2242_2244del (p.S748del)] was identified by searching WES data from 526 patients and SLC12A5-targeted resequencing data from 141 patients with infantile epilepsy. Gramicidin-perforated patch-clamp anal. demonstrated strongly suppressed Cl- extrusion function of E50_Q93del and M415V mutants, with mildly impaired function of A191V and S323P mutants. Cell surface expression levels of these KCC2 mutants were similar to wildtype KCC2. Heterologous expression of two KCC2 mutants, mimicking the patient status, produced a significantly greater intracellular Cl- level than with wildtype KCC2, but less than without KCC2. These data clearly demonstrated that partially disrupted neuronal Cl- extrusion, mediated by two types of differentially impaired KCC2 mutant in an individual, causes EIMFS.
- 43Uyanik, G.; Elcioglu, N.; Penzien, J.; Gross, C.; Yilmaz, Y.; Olmez, A.; Demir, E.; Wahl, D.; Scheglmann, K.; Winner, B.; Bogdahn, U.; Topaloglu, H.; Hehr, U.; Winkler, J. Novel Truncating and Missense Mutations of the KCC3 Gene Associated with Andermann Syndrome. Neurology 2006, 67 (7), 1044, DOI: 10.1212/01.wnl.0000250608.09509.edThere is no corresponding record for this reference.
- 44Olsson, M. H. M.; So̷ndergaard, C. R.; Rostkowski, M.; Jensen, J. H. PROPKA3: Consistent Treatment of Internal and Surface Residues in Empirical p Ka Predictions. J. Chem. Theory Comput. 2011, 7 (2), 525– 537, DOI: 10.1021/ct100578z44PROPKA3: Consistent Treatment of Internal and Surface Residues in Empirical pKa PredictionsOlsson, Mats H. M.; Sondergaard, Chresten R.; Rostkowski, Michal; Jensen, Jan H.Journal of Chemical Theory and Computation (2011), 7 (2), 525-537CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The authors have revised the rules and parameters for one of the most commonly used empirical pKa predictors, PROPKA, based on better phys. description of the desolvation and dielec. response for the protein. The authors have introduced a new and consistent approach to interpolate the description between the previously distinct classifications into internal and surface residues, which otherwise is found to give rise to an erratic and discontinuous behavior. Since the goal of this study is to lay out the framework and validate the concept, it focuses on Asp and Glu residues where the protein pKa values and structures are assumed to be more reliable. The new and improved implementation is evaluated and discussed; it is found to agree better with expt. than the previous implementation (in parentheses): rmsd = 0.79 (0.91) for Asp and Glu, 0.75 (0.97) for Tyr, 0.65 (0.72) for Lys, and 1.00 (1.37) for His residues. The most significant advance, however, is in reducing the no. of outliers and removing unreasonable sensitivity to small structural changes that arise from classifying residues as either internal or surface.
- 45Schott-Verdugo, S.; Gohlke, H. PACKMOL-Memgen: A Simple-To-Use, Generalized Workflow for Membrane-Protein–Lipid-Bilayer System Building. J. Chem. Inf. Model. 2019, 59 (6), 2522– 2528, DOI: 10.1021/acs.jcim.9b0026945PACKMOL-Memgen: A Simple-To-Use, Generalized Workflow for Membrane-Protein-Lipid-Bilayer System BuildingSchott-Verdugo, Stephan; Gohlke, HolgerJournal of Chemical Information and Modeling (2019), 59 (6), 2522-2528CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)We present PACKMOL-Memgen, a simple-to-use, generalized workflow for automated building of membrane-protein-lipid-bilayer systems based on open-source tools including Packmol, memembed, pdbremix, and AmberTools. Compared with web-interface-based related tools, PACKMOL-Memgen allows setup of multiple configurations of a system in a user-friendly and efficient manner within minutes. The generated systems are well-packed and thus well-suited as starting configurations in MD simulations under periodic boundary conditions, requiring only moderate equilibration times. PACKMOL-Memgen is distributed with AmberTools and runs on most computing platforms, and its output can also be used for CHARMM or adapted to other mol.-simulation packages.
- 46Le Grand, S.; Götz, A. W.; Walker, R. C. SPFP: Speed without Compromise─A Mixed Precision Model for GPU Accelerated Molecular Dynamics Simulations. Comput. Phys. Commun. 2013, 184 (2), 374– 380, DOI: 10.1016/j.cpc.2012.09.02246SPFP: Speed without compromise-A mixed precision model for GPU accelerated molecular dynamics simulationsLe Grand, Scott; Gotz, Andreas W.; Walker, Ross C.Computer Physics Communications (2013), 184 (2), 374-380CODEN: CPHCBZ; ISSN:0010-4655. (Elsevier B.V.)A new precision model is proposed for the acceleration of all-atom classical mol. dynamics (MD) simulations on graphics processing units (GPUs). This precision model replaces double precision arithmetic with fixed point integer arithmetic for the accumulation of force components as compared to a previously introduced model that uses mixed single/double precision arithmetic. This significantly boosts performance on modern GPU hardware without sacrificing numerical accuracy. We present an implementation for NVIDIA GPUs of both generalized Born implicit solvent simulations as well as explicit solvent simulations using the particle mesh Ewald (PME) algorithm for long-range electrostatics using this precision model. Tests demonstrate both the performance of this implementation as well as its numerical stability for const. energy and const. temp. biomol. MD as compared to a double precision CPU implementation and double and mixed single/double precision GPU implementations.
- 47Salomon-Ferrer, R.; Case, D. A.; Walker, R. C. An Overview of the Amber Biomolecular Simulation Package: Amber Biomolecular Simulation Package. Wiley Interdiscip. Rev. Comput. Mol. Sci. 2013, 3 (2), 198– 210, DOI: 10.1002/wcms.112147An overview of the amber biomolecular simulation packageSalomon-Ferrer, Romelia; Case, David A.; Walker, Ross C.Wiley Interdisciplinary Reviews: Computational Molecular Science (2013), 3 (2), 198-210CODEN: WIRCAH; ISSN:1759-0884. (Wiley-Blackwell)A review. Mol. dynamics (MD) allows the study of biol. and chem. systems at the atomistic level on timescales from femtoseconds to milliseconds. It complements expt. while also offering a way to follow processes difficult to discern with exptl. techniques. Numerous software packages exist for conducting MD simulations of which one of the widest used is termed Amber. Here, we outline the most recent developments, since version 9 was released in Apr. 2006, of the Amber and AmberTools MD software packages, referred to here as simply the Amber package. The latest release represents six years of continued development, since version 9, by multiple research groups and the culmination of over 33 years of work beginning with the first version in 1979. The latest release of the Amber package, version 12 released in Apr. 2012, includes a substantial no. of important developments in both the scientific and computer science arenas. We present here a condensed vision of what Amber currently supports and where things are likely to head over the coming years. Figure 1 shows the performance in ns/day of the Amber package version 12 on a single-core AMD FX-8120 8-Core 3.6GHz CPU, the Cray XT5 system, and a single GPU GTX680.
- 48Maier, J. A.; Martinez, C.; Kasavajhala, K.; Wickstrom, L.; Hauser, K. E.; Simmerling, C. ff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameters from ff99SB. J. Chem. Theory Comput. 2015, 11 (8), 3696– 3713, DOI: 10.1021/acs.jctc.5b0025548ff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameters from ff99SBMaier, James A.; Martinez, Carmenza; Kasavajhala, Koushik; Wickstrom, Lauren; Hauser, Kevin E.; Simmerling, CarlosJournal of Chemical Theory and Computation (2015), 11 (8), 3696-3713CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Mol. mechanics is powerful for its speed in atomistic simulations, but an accurate force field is required. The Amber ff99SB force field improved protein secondary structure balance and dynamics from earlier force fields like ff99, but weaknesses in side chain rotamer and backbone secondary structure preferences have been identified. Here, we performed a complete refit of all amino acid side chain dihedral parameters, which had been carried over from ff94. The training set of conformations included multidimensional dihedral scans designed to improve transferability of the parameters. Improvement in all amino acids was obtained as compared to ff99SB. Parameters were also generated for alternate protonation states of ionizable side chains. Av. errors in relative energies of pairs of conformations were under 1.0 kcal/mol as compared to QM, reduced 35% from ff99SB. We also took the opportunity to make empirical adjustments to the protein backbone dihedral parameters as compared to ff99SB. Multiple small adjustments of φ and ψ parameters were tested against NMR scalar coupling data and secondary structure content for short peptides. The best results were obtained from a phys. motivated adjustment to the φ rotational profile that compensates for lack of ff99SB QM training data in the β-ppII transition region. Together, these backbone and side chain modifications (hereafter called ff14SB) not only better reproduced their benchmarks, but also improved secondary structure content in small peptides and reprodn. of NMR χ1 scalar coupling measurements for proteins in soln. We also discuss the Amber ff12SB parameter set, a preliminary version of ff14SB that includes most of its improvements.
- 49Dickson, C. J.; Madej, B. D.; Skjevik, Å. A.; Betz, R. M.; Teigen, K.; Gould, I. R.; Walker, R. C. Lipid14: The Amber Lipid Force Field. J. Chem. Theory Comput. 2014, 10 (2), 865– 879, DOI: 10.1021/ct401030749Lipid14: The Amber Lipid Force FieldDickson, Callum J.; Madej, Benjamin D.; Skjevik, Age A.; Betz, Robin M.; Teigen, Knut; Gould, Ian R.; Walker, Ross C.Journal of Chemical Theory and Computation (2014), 10 (2), 865-879CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The AMBER lipid force field has been updated to create Lipid14, allowing tensionless simulation of a no. of lipid types with the AMBER MD package. The modular nature of this force field allows numerous combinations of head and tail groups to create different lipid types, enabling the easy insertion of new lipid species. The Lennard-Jones and torsion parameters of both the head and tail groups have been revised and updated partial charges calcd. The force field has been validated by simulating bilayers of six different lipid types for a total of 0.5 μs each without applying a surface tension; with favorable comparison to expt. for properties such as area per lipid, vol. per lipid, bilayer thickness, NMR order parameters, scattering data, and lipid lateral diffusion. As the derivation of this force field is consistent with the AMBER development philosophy, Lipid14 is compatible with the AMBER protein, nucleic acid, carbohydrate, and small mol. force fields.
- 50Jorgensen, W. L.; Chandrasekhar, J.; Madura, J. D.; Impey, R. W.; Klein, M. L. Comparison of Simple Potential Functions for Simulating Liquid Water. J. Chem. Phys. 1983, 79 (2), 926– 935, DOI: 10.1063/1.44586950Comparison of simple potential functions for simulating liquid waterJorgensen, William L.; Chandrasekhar, Jayaraman; Madura, Jeffry D.; Impey, Roger W.; Klein, Michael L.Journal of Chemical Physics (1983), 79 (2), 926-35CODEN: JCPSA6; ISSN:0021-9606.Classical Monte Carlo simulations were carried out for liq. H2O in the NPT ensemble at 25° and 1 atm using 6 of the simpler intermol. potential functions for the dimer. Comparisons were made with exptl. thermodn. and structural data including the neutron diffraction results of Thiessen and Narten (1982). The computed densities and potential energies agree with expt. except for the original Bernal-Fowler model, which yields an 18% overest. of the d. and poor structural results. The discrepancy may be due to the correction terms needed in processing the neutron data or to an effect uniformly neglected in the computations. Comparisons were made for the self-diffusion coeffs. obtained from mol. dynamics simulations.
- 51Joung, I. S.; Cheatham, T. E. Determination of Alkali and Halide Monovalent Ion Parameters for Use in Explicitly Solvated Biomolecular Simulations. J. Phys. Chem. B 2008, 112 (30), 9020– 9041, DOI: 10.1021/jp800161451Determination of Alkali and Halide Monovalent Ion Parameters for Use in Explicitly Solvated Biomolecular SimulationsJoung, In Suk; Cheatham, Thomas E.Journal of Physical Chemistry B (2008), 112 (30), 9020-9041CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)Alkali (Li+, Na+, K+, Rb+, and Cs+) and halide (F-, Cl-, Br-, and I-) ions play an important role in many biol. phenomena, roles that range from stabilization of biomol. structure, to influence on biomol. dynamics, to key physiol. influence on homeostasis and signaling. To properly model ionic interaction and stability in atomistic simulations of biomol. structure, dynamics, folding, catalysis, and function, an accurate model or representation of the monovalent ions is critically necessary. A good model needs to simultaneously reproduce many properties of ions, including their structure, dynamics, solvation, and moreover both the interactions of these ions with each other in the crystal and in soln. and the interactions of ions with other mols. At present, the best force fields for biomols. employ a simple additive, nonpolarizable, and pairwise potential for at. interaction. In this work, the authors describe their efforts to build better models of the monovalent ions within the pairwise Coulombic and 6-12 Lennard-Jones framework, where the models are tuned to balance crystal and soln. properties in Ewald simulations with specific choices of well-known water models. Although it has been clearly demonstrated that truly accurate treatments of ions will require inclusion of nonadditivity and polarizability (particularly with the anions) and ultimately even a quantum mech. treatment, the authors' goal was to simply push the limits of the additive treatments to see if a balanced model could be created. The applied methodol. is general and can be extended to other ions and to polarizable force-field models. The authors' starting point centered on observations from long simulations of biomols. in salt soln. with the AMBER force fields where salt crystals formed well below their soly. limit. The likely cause of the artifact in the AMBER parameters relates to the naive mixing of the Smith and Dang chloride parameters with AMBER-adapted Aqvist cation parameters. To provide a more appropriate balance, the authors reoptimized the parameters of the Lennard-Jones potential for the ions and specific choices of water models. To validate and optimize the parameters, the authors calcd. hydration free energies of the solvated ions and also lattice energies (LE) and lattice consts. (LC) of alkali halide salt crystals. This is the first effort that systematically scans across the Lennard-Jones space (well depth and radius) while balancing ion properties like LE and LC across all pair combinations of the alkali ions and halide ions. The optimization across the entire monovalent series avoids systematic deviations. The ion parameters developed, optimized, and characterized were targeted for use with some of the most commonly used rigid and nonpolarizable water models, specifically TIP3P, TIP4PEW, and SPC/E. In addn. to well reproducing the soln. and crystal properties, the new ion parameters well reproduce binding energies of the ions to water and the radii of the first hydration shells.
- 52Darden, T.; York, D.; Pedersen, L. Particle Mesh Ewald: An N ·log(N) Method for Ewald Sums in Large Systems. J. Chem. Phys. 1993, 98 (12), 10089– 10092, DOI: 10.1063/1.46439752Particle mesh Ewald: an N·log(N) method for Ewald sums in large systemsDarden, Tom; York, Darrin; Pedersen, LeeJournal of Chemical Physics (1993), 98 (12), 10089-92CODEN: JCPSA6; ISSN:0021-9606.An N·log(N) method for evaluating electrostatic energies and forces of large periodic systems is presented. The method is based on interpolation of the reciprocal space Ewald sums and evaluation of the resulting convolution using fast Fourier transforms. Timings and accuracies are presented for three large cryst. ionic systems.
- 53Ryckaert, J.-P.; Ciccotti, G.; Berendsen, H. J. C. Numerical Integration of the Cartesian Equations of Motion of a System with Constraints: Molecular Dynamics of n-Alkanes. J. Comput. Phys. 1977, 23 (3), 327– 341, DOI: 10.1016/0021-9991(77)90098-553Numerical integration of the Cartesian equations of motion of a system with constraints: molecular dynamics of n-alkanesRyckaert, Jean Paul; Ciccotti, Giovanni; Berendsen, Herman J. C.Journal of Computational Physics (1977), 23 (3), 327-41CODEN: JCTPAH; ISSN:0021-9991.A numerical algorithm integrating the 3N Cartesian equation of motion of a system of N points subject to holonomic constraints is applied to mol. dynamics simulation of a liq. of 64 butane mols.
- 54Ansari, N.; Rizzi, V.; Parrinello, M. Water Regulates the Residence Time of Benzamidine in Trypsin. Nat. Commun. 2022, 13 (1), 5438, DOI: 10.1038/s41467-022-33104-354Water regulates the residence time of Benzamidine in TrypsinAnsari, Narjes; Rizzi, Valerio; Parrinello, MicheleNature Communications (2022), 13 (1), 5438CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)Abstr.: The process of ligand-protein unbinding is crucial in biophysics. Water is an essential part of any biol. system and yet, many aspects of its role remain elusive. Here, we simulate with state-of-the-art enhanced sampling techniques the binding of Benzamidine to Trypsin which is a much studied and paradigmatic ligand-protein system. We use machine learning methods to det. efficient collective coordinates for the complex non-local network of water. These coordinates are used to perform On-the-fly Probability Enhanced Sampling simulations, which we adapt to calc. also the ligand residence time. Our results, both static and dynamic, are in good agreement with expts. We find that the presence of a water mol. located at the bottom of the binding pocket allows via a network of hydrogen bonds the ligand to be released into the soln. On a finer scale, even when unbinding is allowed, another water mol. further modulates the exit time.
- 55Rizzi, V.; Bonati, L.; Ansari, N.; Parrinello, M. The Role of Water in Host-Guest Interaction. Nat. Commun. 2021, 12 (1), 93, DOI: 10.1038/s41467-020-20310-055The role of water in host-guest interactionRizzi, Valerio; Bonati, Luigi; Ansari, Narjes; Parrinello, MicheleNature Communications (2021), 12 (1), 93CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)One of the main applications of atomistic computer simulations is the calcn. of ligand binding free energies. The accuracy of these calcns. depends on the force field quality and on the thoroughness of configuration sampling. Sampling is an obstacle in simulations due to the frequent appearance of kinetic bottlenecks in the free energy landscape. Very often this difficulty is circumvented by enhanced sampling techniques. Typically, these techniques depend on the introduction of appropriate collective variables that are meant to capture the system's degrees of freedom. In ligand binding, water has long been known to play a key role, but its complex behavior has proven difficult to fully capture. In this paper we combine machine learning with phys. intuition to build a non-local and highly efficient water-describing collective variable. We use it to study a set of host-guest systems from the SAMPL5 challenge. We obtain highly accurate binding free energies and good agreement with expts. The role of water during the binding process is then analyzed in some detail.
- 56Tribello, G. A.; Bonomi, M.; Branduardi, D.; Camilloni, C.; Bussi, G. PLUMED 2: New Feathers for an Old Bird. Comput. Phys. Commun. 2014, 185 (2), 604– 613, DOI: 10.1016/j.cpc.2013.09.01856PLUMED 2: New feathers for an old birdTribello, Gareth A.; Bonomi, Massimiliano; Branduardi, Davide; Camilloni, Carlo; Bussi, GiovanniComputer Physics Communications (2014), 185 (2), 604-613CODEN: CPHCBZ; ISSN:0010-4655. (Elsevier B.V.)Enhancing sampling and analyzing simulations are central issues in mol. simulation. Recently, we introduced PLUMED, an open-source plug-in that provides some of the most popular mol. dynamics (MD) codes with implementations of a variety of different enhanced sampling algorithms and collective variables (CVs). The rapid changes in this field, in particular new directions in enhanced sampling and dimensionality redn. together with new hardware, require a code that is more flexible and more efficient. We therefore present PLUMED 2 here-a complete rewrite of the code in an object-oriented programming language (C++). This new version introduces greater flexibility and greater modularity, which both extends its core capabilities and makes it far easier to add new methods and CVs. It also has a simpler interface with the MD engines and provides a single software library contg. both tools and core facilities. Ultimately, the new code better serves the ever-growing community of users and contributors in coping with the new challenges arising in the field.
- 57Michaud-Agrawal, N.; Denning, E. J.; Woolf, T. B.; Beckstein, O. MDAnalysis: A Toolkit for the Analysis of Molecular Dynamics Simulations. J. Comput. Chem. 2011, 32 (10), 2319– 2327, DOI: 10.1002/jcc.2178757MDAnalysis: A toolkit for the analysis of molecular dynamics simulationsMichaud-Agrawal, Naveen; Denning, Elizabeth J.; Woolf, Thomas B.; Beckstein, OliverJournal of Computational Chemistry (2011), 32 (10), 2319-2327CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)MDAnal. is an object-oriented library for structural and temporal anal. of mol. dynamics (MD) simulation trajectories and individual protein structures. It is written in the Python language with some performance-crit. code in C. It uses the powerful NumPy package to expose trajectory data as fast and efficient NumPy arrays. It has been tested on systems of millions of particles. Many common file formats of simulation packages including CHARMM, Gromacs, Amber, and NAMD and the Protein Data Bank format can be read and written. Atoms can be selected with a syntax similar to CHARMM's powerful selection commands. MDAnal. enables both novice and experienced programmers to rapidly write their own anal. tools and access data stored in trajectories in an easily accessible manner that facilitates interactive explorative anal. MDAnal. has been tested on and works for most Unix-based platforms such as Linux and Mac OS X. It is freely available under the GNU General Public License from http://mdanal.googlecode.com. © 2011 Wiley Periodicals, Inc. J Comput Chem 2011.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/jacs.3c10258.
Alternating access mechanisms, collective variable design, and relevant TM helices sequence alignment, and further analysis of the equilibrium and enhanced sampling MD simulations (state stability, salt-bridge formation and water content, and others) (PDF)
Angular motion for the rocking-bundle mechanism (MP4)
Presence of water-permeable states (MP4)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.