1
|
Viskupicova J, Michel Espinoza-Fonseca L. Allosteric modulation of SERCA pumps in health and disease: structural dynamics, posttranslational modifications, and therapeutic potential. J Mol Biol 2025:169200. [PMID: 40349954 DOI: 10.1016/j.jmb.2025.169200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/29/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Sarco/endoplasmic reticulum (SR/ER) Ca2+-ATPase (SERCA) pumps are ubiquitous membrane proteins in all eukaryotic cells, playing a central role in maintaining intracellular calcium homeostasis by re-sequestering Ca2+ ions from the cytosol into the SR/ER at the expense of ATP hydrolysis. SERCA pumps are well-characterized components of the calcium transport machinery in the cell, playing a role in various physiological processes, including muscle contraction, energy metabolism, secretion exocytosis, gene expression, synaptic transmission, cell survival, and fertilization. Allosteric regulation of SERCA pumps plays a key role in health and disease, and modulation of the SERCA pumps has emerged as a therapeutic approach for the treatment of cardiovascular, muscular, metabolic, and neurodegenerative disorders. In this review, we provide a comprehensive overview of the structural dynamics underlying allosteric modulation of SERCA, focusing on the effects of endogenous regulatory proteins, Ca2+ ions, ATP, and small-molecule effectors on the dynamics and function of the pump. We also examine in detail the role of posttranslational modifications as allosteric modulators of SERCA function, focusing on the oxidative modifications S-glutathionylation, S-nitrosylation, tyrosine nitration, and carbonylation, and non-oxidative modifications that include SUMOylation, acetylation, O-GlcNAcylation, phosphorylation, and ubiquitination. Finally, we discuss the therapeutic potential and challenges of allosteric modulation of SERCA pumps, including the design of small-molecule effectors, microRNA-based interventions, and targeted strategies that modulate SERCA posttranslational regulation. Overall, this review aims to bridge the gap between the mechanisms underlying allosteric modulation of SERCA and the translation of basic science discoveries into effective therapies targeting SERCA pumps.
Collapse
Affiliation(s)
- Jana Viskupicova
- Centre of Experimental Medicine, Institute of Experimental Pharmacology & Toxicology, Slovak Academy of Sciences, 84104 Bratislava, Slovakia.
| | - L Michel Espinoza-Fonseca
- Center for Arrhythmia Research, Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
2
|
Khare D, Pimple MV, Acharya C. A novel Zn (II)/Cd (II)/Pb (II)-translocating P IB-type ATPase mediates metal resistance in Chryseobacterium sp. strain PMSZPI in metal-enriched soil of uranium ore deposit. Int J Biol Macromol 2025; 305:141189. [PMID: 39978524 DOI: 10.1016/j.ijbiomac.2025.141189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/31/2025] [Accepted: 02/15/2025] [Indexed: 02/22/2025]
Abstract
Transition metals at higher concentrations are toxic to the cells. Membrane bound, ATP-driven efflux pumps belonging to the P-type ATPase superfamily maintain metal homeostasis by transporting metals/ions across the biological membranes. A soil bacterium, Chryseobacterium sp. strain PMSZPI, residing in metal enriched environment of uranium ore deposit exhibited high tolerance to multiple heavy metals. In an attempt to unveil one of the molecular determinants of metal resistance in PMSZPI, we characterized an unannotated, novel metal exporting PIB-2-ATPase that was categorized as Zn (II)/Cd (II)/Pb(II) PIB-2-ATPase based on amino-acid sequence alignment and the substrate specificities. The heterologously expressed and purified PIB-2-ATPase exhibited zinc/cadmium/lead dependent ATP hydrolysis activity, ATP dependent phosphorylation and activity inhibition in the presence of vanadate. In-vivo metal tolerance assays and analysis of intracellular metal contents indicated involvement of PIB-2-ATPase in metal efflux. The disordered N-terminal metal binding domain of PIB-2-ATPase was found to be inconsequential for its function. Mutagenesis studies revealed the role of the conserved transmembrane (TM) residues (cysteine, aspartate and lysine) in metal translocation. Overall, our data establishes the vital role of Zn(II)/Cd(II)/Pb(II) PIB-2-ATPase in conferring metal resistance in PMSZPI.
Collapse
Affiliation(s)
- Devanshi Khare
- Molecular Biology Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai 400094, India
| | - Mehzabin Vivek Pimple
- Analytical Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Celin Acharya
- Molecular Biology Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai 400094, India.
| |
Collapse
|
3
|
Eisele D, Blatzer M, Dietl AM, Binder U, Müller C, Hagen F, Sae-Ong T, Schäuble S, Panagiotou G, Vahedi-Shahandashti R, Lass-Flörl C. Aspergillus terreus sectorization: a morphological phenomenon shedding light on amphotericin B resistance mechanism. mBio 2025; 16:e0392624. [PMID: 39998230 PMCID: PMC11980562 DOI: 10.1128/mbio.03926-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
Prolonged cultivation of certain filamentous fungi, including Aspergillus terreus, on drug-free medium leads to degeneration and morphological heterogeneity, marked by the emergence of fluffy mycelium-type sectors. This phenomenon may indicate alterations in antifungal susceptibility profiles (particularly to amphotericin B (AmB) in A. terreus), as well as reductions or losses in conidiation, sexuality, secondary metabolite production, and/or virulence. In the present study, various characteristics of an AmB-resistant wild-type (WT) strain and its AmB-susceptible sectorized derivative (ATSec) were characterized. Compared to WT, ATSec exhibited increased susceptibility to AmB, reduced sporulation, and comparable sterol contents and virulence in Galleria mellonella. To elucidate the genes involved in AmB resistance, gene expression levels were compared between WT and ATSec with and without AmB treatment. The expression of P-type ATPase-related genes, which are implicated in membrane composition changes and consequently in AmB resistance, was significantly higher in the WT strain compared to ATSec. Moreover, the up-regulation of genes involved in the biosynthesis of polyketides-a diverse group of secondary metabolites-was higher in WT compared to ATSec, with a significant number of these genes also carrying at least one mutation. The findings of this study indicate that P-type ATPases may significantly be involved in AmB susceptibility and resistance observed in ATSec and WT strains. Additionally, mutations in polyketide synthase genes in ATSec may contribute to the phenotypic alterations associated with the sectorized phenotype. IMPORTANCE Prolonged cultivation of certain filamentous fungi, including Aspergillus terreus, on drug-free medium leads to degeneration and morphological heterogeneity, marked by the emergence of fluffy mycelium-type sectors. This phenomenon may indicate alterations in antifungal susceptibility profiles (particularly to amphotericin B (AmB) in A. terreus), as well as reductions or losses in conidiation, sexuality, secondary metabolite production, and/or virulence. In the present study, various characteristics of an AmB-resistant wild-type strain (WT) and its AmB-susceptible sectorized derivative (ATSec) were characterized. Compared to WT, ATSec exhibited increased susceptibility to AmB, reduced sporulation, and comparable sterol contents and virulence in Galleria mellonella. To elucidate the genes involved in AmB resistance, gene expression levels were compared between WT and ATSec with and without AmB treatment. The expression of P-type ATPase-related genes, which are implicated in membrane composition changes and consequently in AmB resistance, was significantly higher in the WT strain compared to ATSec. Moreover, the up-regulation of genes involved in the biosynthesis of polyketides - a diverse group of secondary metabolites - was higher in WT compared to ATSec, with a significant number of these genes also carrying at least one mutation. The findings of this study indicate that P-type ATPases may significantly be involved in AmB susceptibility and resistance observed in ATSec and WT strains. Additionally, mutations in polyketide synthase genes in ATSec may contribute to the phenotypic alterations associated with the sectorized phenotype.
Collapse
Affiliation(s)
- David Eisele
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Blatzer
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
- Experimental Neuropathy Unit, Institute Pasteur, Paris, France
- Global Health Department, Institute Pasteur, Paris, France
| | - Anna Maria Dietl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ulrike Binder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Müller
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ferry Hagen
- Department of Medical Mycology, Westerdijk Fungal Biodiversity Institute (WI-KNAW), Utrecht, the Netherlands
- Institute of Biodiversity and Ecosystem Dynamics (IBED), University of Amsterdam, Amsterdam, the Netherlands
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Tongta Sae-Ong
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Sascha Schäuble
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), Jena, Germany
- Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
- Jena University Hospital, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University, Jena, Germany
| | - Roya Vahedi-Shahandashti
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Cornelia Lass-Flörl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
4
|
Beyer T, Caliebe J, Kähler L, Beitz E. A Soluble Expression Construct of the Isolated Catalytic Domain of Plasmodium falciparum ATP4 Exhibits ATPase Activity Independent of a γ-Phosphate Receiving Aspartate. Mol Microbiol 2025. [PMID: 40091859 DOI: 10.1111/mmi.15358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
The sodium/proton-exchanging ATPase of Plasmodium falciparum malaria parasites, PfATP4, is an emerging drug target. Inhibition results in detrimental cell swelling due to cytosolic accumulation of sodium and alkalization. PfATP4 is a sodium-releasing type II P-type ATPase restricted to apicomplexan parasites. Experimental data on structure-function relationships of the isolated protein are absent. Here, we produced and purified the soluble catalytic domain of PfATP4 and evaluated kinetic properties by in vitro phosphate colorimetry. The protein exhibited Mg2+-dependent ATPase activity at the same order of magnitude as the native cellular PfATP4 and was insensitive to the presence of sodium. AlphaFold 3-based structure and ATP/Mg2+ interaction predictions identified key residues of the nucleotide binding domain (Lys619, Lys652, Arg703). Replacement of the lysines by methionine decreased the enzymatic activity to one quarter. Individual mutation of the putative Mg2+-coordinating Asp865 of the phosphorylation domain was tolerated, while a joint replacement with Asp869 decreased ATPase again to one quarter. Mutation of the putative γ-phosphate receiving Asp451 maintained the rate of Pi release. Our data attribute typical functional roles for P-type ATPases to the basic and acidic residues of the soluble PfATP4 catalytic domain and show that its ATP hydrolysis is independent of phosphorylation of Asp451.
Collapse
Affiliation(s)
- Timo Beyer
- Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Jesko Caliebe
- Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Lara Kähler
- Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Eric Beitz
- Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University of Kiel, Kiel, Germany
| |
Collapse
|
5
|
Mohammadi A, Shafiee A. Quantum effects in ion transport: A thermodynamic resource theory approach. Biosystems 2025; 249:105412. [PMID: 39971150 DOI: 10.1016/j.biosystems.2025.105412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/21/2025]
Abstract
In recent years, understanding thermodynamics in the quantum regime has garnered significant attention, driven by advances in nanoscale physics and experimental techniques. In parallel, growing evidence supports the importance of quantum effects in various biological processes, making them increasingly relevant to quantum thermodynamics. In this study, we apply resource theory formulations of thermodynamics to investigate the role of quantum properties in ion transport across cell membranes. Within this framework, quantum properties are treated as resources under generalized thermodynamic constraints in the quantum regime. Specifically, our findings reveal that non-Markovianity, which reflects memory effects in ion transport dynamics, is a key quantum resource that enhances the yield and efficiency of the ion transport process. In contrast, quantum coherence, manifested as the superposition of energy states in ion-transport proteins, reduces these metrics but plays a crucial role in distinguishing between ion channels and ion pumps-two distinct types of ion-transport proteins in cell membranes. Finally, we demonstrate that introducing an additional coherent system allows coherence to facilitate the transformation of an ion pump into an ion channel.
Collapse
Affiliation(s)
- Amin Mohammadi
- Research Group on Foundations of Quantum Theory and Information, Department of Chemistry, Sharif University of Technology, P.O. Box 11365-9516, Tehran, Iran.
| | - Afshin Shafiee
- Research Group on Foundations of Quantum Theory and Information, Department of Chemistry, Sharif University of Technology, P.O. Box 11365-9516, Tehran, Iran
| |
Collapse
|
6
|
Allegrini B, Mignotet M, Rapetti-Mauss R, Borgese F, Soriani O, Guizouarn H. A new regulation mechanism for KCNN4, the Ca 2+-dependent K + channel, by molecular interactions with the Ca 2+pump PMCA4b. J Biol Chem 2025; 301:108114. [PMID: 39716493 PMCID: PMC11787511 DOI: 10.1016/j.jbc.2024.108114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/20/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024] Open
Abstract
KCNN4, a Ca2+-activated K+ channel, is involved in various physiological and pathological processes. It is essential for epithelial transport, immune system, and other physiological mechanisms, but its activation is also involved in cancer pathophysiology as well as red blood cell (RBC) disorders. The activation of KCNN4 in RBC leads to loss of KCl and water, a mechanism known as the "Gardos effect" described 70 years ago. This Ca2+-induced dehydration is irreversible in human RBC and must be tightly controlled to prevent not only hemolysis but also alterations in RBC rheological properties. In this study, we have investigated the regulation of KCNN4 activity after changes in RBC Ca2+ concentration. Using electrophysiology, immunoprecipitation, and proximity ligation assay in human embryonic kidney 293-transfected cells, K562 cells, or RBCs, we have found that KCNN4 and the Ca2+ pump PMCA4b (plasma membrane calcium-transporting ATPase 4b) interact tightly with each other, such that the C-terminal domain of PMCA4b regulates KCNN4 activity, independently of the Ca2+ extrusion activity of the pump. This regulation was not restricted to KCNN4: the small-conductance Ca2+-activated K+ channel KCNN2 was similarly regulated by the calcium pump. We propose a new mechanism that could control KCNN4 activity by a molecular inhibitory interaction with PMCA4b. It is suggested that this mechanism could attenuate erythrocyte dehydration in response to an increase in intracellular Ca2+.
Collapse
Affiliation(s)
- Benoit Allegrini
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France
| | - Morgane Mignotet
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France
| | | | - Franck Borgese
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France
| | - Olivier Soriani
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France
| | - Hélène Guizouarn
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France; Laboratory of Excellence for RBC, LABEX GR-Ex, Paris, France.
| |
Collapse
|
7
|
Sun Y, Tao H, Han H, Zou Y, Xue Y, Chen S, Tao F. Identification and expression analysis of P-type ATPase IIIA subfamily in Puccinia Striiformis f. sp. tritici. BMC Genomics 2025; 26:68. [PMID: 39856561 PMCID: PMC11759449 DOI: 10.1186/s12864-025-11219-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Puccinia striiformis f. sp. tritici (Pst) causes wheat stripe (yellow) rust disease, which is one of the most destructive diseases affecting wheat worldwide. ATPases, a class of membrane proteins, play an important role in material exchange and signal transduction both within and outside biological cells by transporting ions and phospholipids. In plant pathogens, P-type ATPases primarily participate in pathogen development and virulence regulation. However, the P-type ATPase of subfamily IIIA (PMA) has not yet been identified in Pst. To investigate the potential functions of the PMA gene family in Pst, we conducted a genome-wide bioinformatics analysis and examined the expression profiles of the PMA gene family. RESULTS Six PMA genes were identified in the genome of P. striiformis f. sp. tritici (CYR34 race). The PMA proteins encoded by these genes ranged in length from 811 to 960 amino acids (aa). Each of the six PMA genes contained a typical ATPase IIIA H superfamily domain and was distributed across four chromosomes. Thirty-six major cis-regulatory elements were detected within the PMA gene family members. Elements such as the CGTCA-motif and TGACG-motif play significant roles in responding to environmental stresses and hormone signals. Quantitative PCR analysis revealed that the expression of the PMA04 gene was generally higher at 9 °C under various temperature stresses. The PMA06 gene typically exhibited higher expression levels at 16 °C. During the infection of Pst, the expression levels of PMA04, PMA05, and PMA06 were elevated at 72 h post treatment. CONCLUSIONS Our results indicate that the PMA gene family in the CYR34 strain comprises six PMA genes, which are crucial for managing temperature stress and pathogen infection, and exhibit a distinctive splicing pattern. This study not only identifies a target and direction for the development of new, efficient, and environmentally friendly control agents for wheat stripe rust but also establishes a foundation for analyzing its pathogenic mechanisms.
Collapse
Affiliation(s)
- Yingjie Sun
- College of Plant Protection, Gansu Agricultural University, Lanzhou, 730070, China
| | - Hong Tao
- Forest Seedling Service Station of Linxia Hui Autonomous Prefecture, Linxia, 731100, China
| | - Hong Han
- Academy of Agricultural Sciences of Linxia Hui Autonomous Prefecture, Linxia, 731100, China
| | - Yiping Zou
- College of Life Sciences, Shandong Normal University, Jinan, 250358, China
| | - Yingyu Xue
- College of Plant Protection, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Shiwen Chen
- College of Plant Protection, Gansu Agricultural University, Lanzhou, 730070, China
| | - Fei Tao
- College of Plant Protection, Gansu Agricultural University, Lanzhou, 730070, China.
| |
Collapse
|
8
|
Rachuri S, Nepal B, Shukla A, Ramanathan A, Morrisey JM, Daly T, Mather MW, Bergman LW, Kortagere S, Vaidya AB. Mutational analysis of an antimalarial drug target, PfATP4. Proc Natl Acad Sci U S A 2025; 122:e2403689122. [PMID: 39773028 PMCID: PMC11745376 DOI: 10.1073/pnas.2403689122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 11/24/2024] [Indexed: 01/11/2025] Open
Abstract
Among new antimalarials discovered over the past decade are multiple chemical scaffolds that target Plasmodium falciparum P-type ATPase (PfATP4). This essential protein is a Na+ pump responsible for the maintenance of Na+ homeostasis. PfATP4 belongs to the type two-dimensional (2D) subfamily of P-type ATPases, for which no structures have been determined. To gain better insight into the structure/function relationship of this validated drug target, we generated a homology model of PfATP4 based on sarco/endoplasmic reticulum Ca2+ ATPase, a P2A-type ATPase, and refined the model using molecular dynamics in its explicit membrane environment. This model predicted several residues in PfATP4 critical for its function, as well as those that impart resistance to various PfATP4 inhibitors. To validate our model, we developed a genetic system involving merodiploid states of PfATP4 in which the endogenous gene was conditionally expressed, and the second allele was mutated to assess its effect on the parasite. Our model predicted residues involved in Na+ coordination as well as the phosphorylation cycle of PfATP4. Phenotypic characterization of these mutants involved assessment of parasite growth, localization of mutated PfATP4, response to treatment with known PfATP4 inhibitors, and evaluation of the downstream consequences of Na+ influx. Our results were consistent with modeled predictions of the essentiality of the critical residues. Additionally, our approach confirmed the phenotypic consequences of resistance-associated mutations as well as a potential structural basis for the fitness cost associated with some mutations. Taken together, our approach provides a means to explore the structure/function relationship of essential genes in haploid organisms.
Collapse
Affiliation(s)
- Swaksha Rachuri
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, PA19129
| | - Binod Nepal
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, PA19129
| | - Anurag Shukla
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, PA19129
| | - Aarti Ramanathan
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, PA19129
| | - Joanne M. Morrisey
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, PA19129
| | - Thomas Daly
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, PA19129
| | - Michael W. Mather
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, PA19129
| | - Lawrence W. Bergman
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, PA19129
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, PA19129
| | - Akhil B. Vaidya
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, PA19129
| |
Collapse
|
9
|
Rayevsky A, Platonov M, Elijah B, Volochnyuk D, Veklich T, Cherenok S, Rodik R, Kalchenko V, Kosterin S. Structural Insight on the Selectivity of Calyx[4]Arene-Based Inhibitors of Mg 2+-Dependent Atp-Hydrolases. Mol Inform 2025; 44:e202400200. [PMID: 39635768 DOI: 10.1002/minf.202400200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024]
Abstract
Located in plasma membranes, ATP hydrolases are involved in several dynamic transport processes, helping to control the movement of ions across cell membranes. ATP hydrolase acts as a transport protein, converting energy from ATP hydrolysis into transport molecules against their concentration gradients. In addition to energy metabolism and active transport, ATP hydrolase is essential for maintaining cellular homeostasis and cell function. This study focused on the domain architecture model of P-type ATPases, which participate in the reaction cycles of ATP hydrolysis carried out by membrane transport systems - Na+, K+-ATPase and Ca2+, Mg2+-ATPase. Targeted modulation of Na+, K+-ATPase and Ca2+, Mg2+-ATPase by unnatural drugs is of greatest interest due to the lack of known effectors. This new discovery presents a convenient model based on our recent experimental studies of the membrane structures and myocytes of the uterine smooth muscle, the myometrium. This current study strongly supports the fact that nanosized calix[4]arenes functionalised on the upper rings of the macrocycle with biologically active phosphonic acid fragments can serve as selective and potent inhibitors of cation-transporting electroenzymes. This is how we discovered that calix[4]arene of methylenebisphosphonic acid C-97 and calix[4]arene of bis-aminophosphonic acid C-107 selectively and effectively (I0.5 <100 nM) inhibit the activity of Mg2+, ATP-dependent electrogenic Na+ K+ plasma membrane pump. As drug discovery in the field of Mg2+-ATPase inhibitors is uncharted territory, basic research holds the key to explaining and predicting the mechanism of interaction and action of different classes of compounds. In light of the presented results, new calix[4]arene compounds can be used as potent inhibitors of Mg2+, ATP-dependent electrogenic ion pumps.
Collapse
Affiliation(s)
- Alexey Rayevsky
- Institute of Food Biotechnology and Genomics, Natl. Academy of Sciences of Ukraine, Osypovskoho Str., 2 A, Kyiv, 04123, Ukraine
- Institute of Molecular Biology and Genetics, Natl. Academy of Sciences of Ukraine, Zabolotnogo Str., 150, Kyiv, 03143, Ukraine
- Enamine Ltd., 78 Chervonotkatska Str., Kyiv, 02660, Ukraine
| | - Maksym Platonov
- Institute of Molecular Biology and Genetics, Natl. Academy of Sciences of Ukraine, Zabolotnogo Str., 150, Kyiv, 03143, Ukraine
- Enamine Ltd., 78 Chervonotkatska Str., Kyiv, 02660, Ukraine
| | - Bulgakov Elijah
- Institute of Food Biotechnology and Genomics, Natl. Academy of Sciences of Ukraine, Osypovskoho Str., 2 A, Kyiv, 04123, Ukraine
- Enamine Ltd., 78 Chervonotkatska Str., Kyiv, 02660, Ukraine
| | - Dmytro Volochnyuk
- Enamine Ltd., 78 Chervonotkatska Str., Kyiv, 02660, Ukraine
- Institute of High Technologies, Taras Shevchenko National University of Kyiv, Glushkova Ave, Kyiv, 03022, Ukraine
- Institute of Organic Chemistry NAS of Ukraine, 5 Murmanska Str., Kyiv, 02660, Ukraine
| | - Tetyana Veklich
- Palladin Institute of Biochemistry NAS of Ukraine, 9 Leontovich str., Kyiv, 01054, Ukraine
| | - Sergiy Cherenok
- Institute of Organic Chemistry NAS of Ukraine, 5 Murmanska Str., Kyiv, 02660, Ukraine
| | - Roman Rodik
- Institute of Organic Chemistry NAS of Ukraine, 5 Murmanska Str., Kyiv, 02660, Ukraine
| | - Vitaliy Kalchenko
- Institute of Organic Chemistry NAS of Ukraine, 5 Murmanska Str., Kyiv, 02660, Ukraine
| | - Sergiy Kosterin
- Palladin Institute of Biochemistry NAS of Ukraine, 9 Leontovich str., Kyiv, 01054, Ukraine
| |
Collapse
|
10
|
Yang W, Yang Y, Wang H, Wang J, Zhang S. Clinical and genetic characterization of patients with late onset Wilson's disease. NPJ Genom Med 2024; 9:71. [PMID: 39719440 DOI: 10.1038/s41525-024-00459-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 12/17/2024] [Indexed: 12/26/2024] Open
Abstract
Wilson's disease (WD) typically manifests in children and young adults, with little knowledge of its late-onset forms. In this study, we performed a retrospective cohort study of 105 WD patients (99 index cases, 6 siblings) with an onset age ≥35 years. We compared 99 index late-onset patients with 1237 early-onset patients and analyzed the ATP7B variant penetrance referring to the Genome Aggregation Database (gnomAD). Sixty-two ATP7B variants were identified in the late-onset patients, among which A874V, V1106I, R919G, and T935M were correlated with late presentation of WD. Regarding gnomAD, V1106I and T935M exhibited significantly low penetrance, and there is a lack of patients carrying a genotype of V1106I/V1106I, R919G/R919G, T935M/T935M, V1106I/T935M, V1106I/R919G, or T935M/R919G. Our data revealed that patients carrying a combination of two late-onset variants may be overlooked due to atypical or lack of WD symptoms, which may provide valuable insights into the genetic basis and diagnosis of WD.
Collapse
Affiliation(s)
- Wenming Yang
- Department of Neurology, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Yue Yang
- Department of Neurology, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Han Wang
- Department of Neurology, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Jiuxiang Wang
- Experimental Center of Clinical Research, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Shijie Zhang
- Experimental Center of Clinical Research, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
11
|
Li ZH, Asady B, Chang L, Triana MAH, Li C, Coppens I, Moreno SN. Calcium transfer from the ER to other organelles for optimal signaling in Toxoplasma gondii. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608087. [PMID: 39185237 PMCID: PMC11343207 DOI: 10.1101/2024.08.15.608087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Ca2+ signaling in cells begins with the opening of Ca2+ channels in either the plasma membrane (PM) or the endoplasmic reticulum (ER) and results in a dramatic increase in the physiologically low (<100 nM) cytosolic Ca2+ level. The temporal and spatial Ca2+ levels are well regulated to enable precise and specific activation of critical biological processes. Ca2+ signaling regulates pathogenic features of apicomplexan parasites like Toxoplasma gondii which infects approximately one-third of the world's population. T. gondii relies on Ca2+ signals to stimulate traits of its infection cycle and several Ca2+ signaling elements play essential roles in its parasitic cycle. Active egress, an essential step for the infection cycle of T. gondii is preceded by a large increase in cytosolic Ca2+ most likely by release from intracellular stores. Intracellular parasites take up Ca2+ from the host cell during host Ca2+ signaling events to replenish intracellular stores. In this work, we investigated the mechanism by which intracellular stores are replenished with Ca2+ and demonstrated a central role for the SERCA-Ca2+-ATPase in keeping not only the ER filled with Ca2+ but also other stores. We show mitochondrial Ca2+ uptake, by transfer of Ca2+ from the ER likely through membrane contact sites. We propose a central role for the ER in sequestering and redistributing calcium to other intracellular organelles following influx at the PM.
Collapse
Affiliation(s)
- Zhu-Hong Li
- Center for Tropical and Emerging Global Diseases, Department of Computes Science, University of Georgia, Athens, Georgia 30602
| | - Beejan Asady
- Center for Tropical and Emerging Global Diseases, Department of Computes Science, University of Georgia, Athens, Georgia 30602
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, MD 21205
| | - Le Chang
- Center for Tropical and Emerging Global Diseases, Department of Computes Science, University of Georgia, Athens, Georgia 30602
| | - Miryam Andrea Hortua Triana
- Center for Tropical and Emerging Global Diseases, Department of Computes Science, University of Georgia, Athens, Georgia 30602
| | - Catherine Li
- Center for Tropical and Emerging Global Diseases, Department of Computes Science, University of Georgia, Athens, Georgia 30602
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, MD 21205
| | - Silvia N.J. Moreno
- Center for Tropical and Emerging Global Diseases, Department of Computes Science, University of Georgia, Athens, Georgia 30602
- Department of Cellular Biology, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
12
|
Große C, Grau J, Herzberg M, Nies DH. Antisense transcription is associated with expression of metal resistance determinants in Cupriavidus metallidurans CH34. Metallomics 2024; 16:mfae057. [PMID: 39562278 DOI: 10.1093/mtomcs/mfae057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 11/16/2024] [Indexed: 11/21/2024]
Abstract
Cupriavidus metallidurans is able to thrive in metal-rich environments but also survives metal starvation. Expression of metal resistance determinants in C. metallidurans was investigated on a global scale. Cupriavidus metallidurans was challenged with a MultiTox metal mix specifically designed for the wildtype strain CH34 and its plasmid-free derivative AE104, including treatment with ethylenediamintetraacetate (EDTA), or without challenge. The sense and antisense transcripts were analyzed in both strains and under all three conditions by RNASeq. A total of 10 757 antisense transcripts (ASTs) were assigned to sense signals from genes and untranslated regions, and 1 319 of these ASTs were expressed and were longer than 50 bases. Most of these (82%) were dual-use transcripts that contained antisense and sense regions, but ASTs (16%) were also observed that had no sense regions. Especially in metal-treated cells of strains CH34 and AE104, up- or down-regulated sense transcripts were accompanied by antisense transcription activities that were also regulated. The presence of selected asRNAs was verified by reverse transcription polymerase chain reaction (RT-PCR). Following metal stress, expression of genes encoding components of the respiratory chain, motility, transcription, translation, and protein export were down-regulated. This should also affect the integration of the metal efflux pumps into the membrane and the supply of the energy required to operate them. To solve this dilemma, transcripts for the metal efflux pumps may be stabilized by interactions with ASTs to allow their translation and import into the membrane. Alternatively, metal stress possibly causes recruitment of RNA polymerase from housekeeping genes for preferential expression of metal resistance determinants.
Collapse
Affiliation(s)
- Cornelia Große
- Molecular Microbiology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Jan Grau
- Computer Sciences, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Martin Herzberg
- Molecular Microbiology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Dietrich H Nies
- Molecular Microbiology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| |
Collapse
|
13
|
Makio T, Chen J, Simmen T. ER stress as a sentinel mechanism for ER Ca 2+ homeostasis. Cell Calcium 2024; 124:102961. [PMID: 39471738 DOI: 10.1016/j.ceca.2024.102961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 11/01/2024]
Abstract
Endoplasmic reticulum (ER) stress is triggered upon the interference with oxidative protein folding that aims to produce fully folded, disulfide-bonded and glycosylated proteins, which are then competent to exit the ER. Many of the enzymes catalyzing this process require the binding of Ca2+ ions, including the chaperones BiP/GRP78, calnexin and calreticulin. The induction of ER stress with a variety of drugs interferes with chaperone Ca2+ binding, increases cytosolic Ca2+through the opening of ER Ca2+ channels, and activates store-operated Ca2+ entry (SOCE). Posttranslational modifications (PTMs) of the ER Ca2+ handling proteins through ER stress-dependent phosphorylation or oxidation control these mechanisms, as demonstrated in the case of the sarco/endoplasmic reticulum ATPase (SERCA), inositol 1,4,5 trisphosphate receptors (IP3Rs) or stromal interaction molecule 1 (STIM1). Their aim is to restore ER Ca2+ homeostasis but also to increase Ca2+ transfer from the ER to mitochondria during ER stress. This latter function boosts ER bioenergetics, but also triggers apoptosis if ER Ca2+ signaling persists. ER Ca2+ toolkit oxidative modifications upon ER stress can occur within the ER lumen or in the adjacent cytosol. Enzymes involved in this redox control include ER oxidoreductin 1 (ERO1) or the thioredoxin-family protein disulfide isomerases (PDI) and ERp57. A tight, but adaptive connection between ER Ca2+ content, ER stress and mitochondrial readouts allows for the proper functioning of many tissues, including skeletal muscle, the liver, and the pancreas, where ER stress either maintains or compromises their function, depending on its extent and context. Upon mutation of key regulators of ER Ca2+ signaling, diseases such as muscular defects (e.g., from mutated selenoprotein N, SEPN1/SELENON), or diabetes (e.g., from mutated PERK) are the result.
Collapse
Affiliation(s)
- Tadashi Makio
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada
| | - Junsheng Chen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada.
| |
Collapse
|
14
|
Li P, Bågenholm V, Hägglund P, Lindkvist-Petersson K, Wang K, Gourdon P. The structure and function of P5A-ATPases. Nat Commun 2024; 15:9605. [PMID: 39505844 PMCID: PMC11541931 DOI: 10.1038/s41467-024-53757-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Endoplasmic reticulum (ER) membrane resident P5A-ATPases broadly affect protein biogenesis and quality control, and yet their molecular function remains debated. Here, we report cryo-EM structures of a P5A-ATPase, CtSpf1, covering multiple transport intermediates of the E1 → E1-ATP → E1P-ADP → E1P → E2P → E2.Pi → E2 → E1 cycle. In the E2P and E2.Pi states a cleft spans the entire membrane, holding a polypeptide cargo molecule. The cargo includes an ER luminal extension, pinpointed as the C-terminus in the E2.Pi state, which reenters the membrane in E2P. The E1 structure harbors a cytosol-facing cavity that is blocked by an insertion we refer to as the Plug-domain. The Plug-domain is nestled to key ATPase features and is displaced in the E1P-ADP and E1P states. Collectively, our findings are compatible with a broad range of proteins as cargo, with the P5A-ATPases serving a role in membrane removal of helices, although insertion/secretion cannot be excluded, as well as with a mechanistic role of the Plug-domain.
Collapse
Affiliation(s)
- Ping Li
- Department of Experimental Medical Science, Lund University, Sölvegatan 19, SE-221 84, Lund, Sweden.
| | - Viktoria Bågenholm
- Department of Biomedical Sciences, University of Copenhagen, Nørre Allé 14, DK-2200, Copenhagen N, Denmark
| | - Per Hägglund
- Department of Biomedical Sciences, University of Copenhagen, Nørre Allé 14, DK-2200, Copenhagen N, Denmark
| | | | - Kaituo Wang
- Department of Biomedical Sciences, University of Copenhagen, Nørre Allé 14, DK-2200, Copenhagen N, Denmark
- State Key Laboratory of Plant Diversity and Specialty Crops, Institute of Botany, Chinese Academy of Sciences, Beijing, China
| | - Pontus Gourdon
- Department of Experimental Medical Science, Lund University, Sölvegatan 19, SE-221 84, Lund, Sweden.
- Department of Biomedical Sciences, University of Copenhagen, Nørre Allé 14, DK-2200, Copenhagen N, Denmark.
| |
Collapse
|
15
|
Liu H, Lu C, Liu XQ, Zhuo CJ, Luo RJ, Huang QT, Tang Z, Zhao CQ, Guerinot ML, Salt DE, Zhao FJ, Huang XY. A chloroplast localized heavy metal-associated domain containing protein regulates grain calcium accumulation in rice. Nat Commun 2024; 15:9265. [PMID: 39462135 PMCID: PMC11513116 DOI: 10.1038/s41467-024-53648-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Calcium (Ca) is an essential mineral nutrient and plays a crucial signaling role in all living organisms. Increasing Ca content in staple foods such as rice is vital for improving Ca nutrition of humans. Here we map a quantitative trait locus that controls Ca concentration in rice grains and identify the causal gene as GCSC1 (Grain Ca and Sr Concentrations 1), which encodes a chloroplast vesicle localized homo-oligomeric protein. GCSC1 exhibits Ca2+ transport activity in heterologous assays in yeast and Xenopus laevis oocytes and is involved in the efflux of Ca2+ from the chloroplast to the cytosol. Knockout of GCSC1 results in increased chloroplast Ca concentration, lower stomatal conductance in leaves and enhanced Ca allocation to grains. Natural variation in grain Ca concentration is attributed to the variable expression of GCSC1 resulting from its promoter sequence variation. Our study identifies a chloroplast localized heavy metal-associated domain containing protein that regulates chloroplast Ca2+ efflux and provides a way to biofortify Ca in rice to benefit human nutrition.
Collapse
Affiliation(s)
- Huan Liu
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, China
| | - Cun Lu
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, China
| | - Xiang-Qian Liu
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, China
| | - Chen-Jin Zhuo
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, China
- Sanya Institute of Nanjing Agricultural University, Sanya, China
| | - Rong-Jian Luo
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, China
| | - Qiu-Tang Huang
- Key Laboratory of Integrated Pest Management on Crops in East China, Ministry of Agriculture, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Zhong Tang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, China
| | - Chun-Qing Zhao
- Key Laboratory of Integrated Pest Management on Crops in East China, Ministry of Agriculture, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Mary Lou Guerinot
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - David E Salt
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, UK
| | - Fang-Jie Zhao
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, China
| | - Xin-Yuan Huang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, China.
- Sanya Institute of Nanjing Agricultural University, Sanya, China.
| |
Collapse
|
16
|
Binti S, Edeen PT, Fay DS. Loss of the Na+/K+ cation pump CATP-1 suppresses nekl-associated molting defects. G3 (BETHESDA, MD.) 2024; 14:jkae244. [PMID: 39428996 PMCID: PMC11631496 DOI: 10.1093/g3journal/jkae244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
The conserved Caenorhabditis elegans protein kinases NEKL-2 and NEKL-3 regulate membrane trafficking and are required for larval molting. Through a forward genetic screen we identified a mutation in catp-1 as a suppressor of molting defects in synthetically lethal nekl-2; nekl-3 double mutants. catp-1 encodes a membrane-associated P4-type ATPase involved in Na+-K+ exchange. A previous study found that wild-type worms exposed to the nicotinic agonist dimethylphenylpiperazinium (DMPP) exhibited larval arrest and molting-associated defects, which were suppressed by inhibition of catp-1. By testing a spectrum catp-1 alleles, we found that resistance to DMPP toxicity and the suppression of nekl defects did not strongly correlate, suggesting key differences in the mechanism of catp-1-mediated suppression. Through whole genome sequencing of additional nekl-2; nekl-3 suppressor strains, we identified two additional coding-altering mutations in catp-1. However, neither mutation, when introduced into nekl-2; nekl-3 mutants using CRISPR, was sufficient to elicit robust suppression of molting defects, suggesting the involvement of other loci. Endogenously tagged CATP-1 was primarily expressed in epidermal cells within punctate structures located near the apical plasma membrane, consistent with a role in regulating cellular processes within the epidermis. Based on previous studies, we tested the hypothesis that catp-1 inhibition induces entry into the pre-dauer L2d stage, potentially accounting for the ability of catp-1 mutants to suppress nekl molting defects. However, we found no evidence that loss of catp-1 leads to entry into L2d. As such, loss of catp-1 may suppress nekl-associated and DMPP-induced defects by altering electrochemical gradients within membrane-bound compartments.
Collapse
Affiliation(s)
- Shaonil Binti
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, WY 82071, USA
| | - Philip T Edeen
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, WY 82071, USA
| | - David S Fay
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
17
|
Prabudiansyah I, Orädd F, Magkakis K, Pounot K, Levantino M, Andersson M. Dephosphorylation and ion binding in prokaryotic calcium transport. SCIENCE ADVANCES 2024; 10:eadp2916. [PMID: 39908574 PMCID: PMC11468904 DOI: 10.1126/sciadv.adp2916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/11/2024] [Indexed: 02/07/2025]
Abstract
Calcium (Ca2+) signaling is fundamental to cellular processes in both eukaryotic and prokaryotic organisms. While the mechanisms underlying eukaryotic Ca2+ transport are well documented, an understanding of prokaryotic transport remains nascent. LMCA1, a Ca2+ adenosine triphosphatase (ATPase) from Listeria monocytogenes, has emerged as a prototype for elucidating structure and dynamics in prokaryotic Ca2+ transport. Here, we used a multidisciplinary approach integrating kinetics, structure, and dynamics to unravel the intricacies of LMCA1 function. A cryo-electron microscopy (cryo-EM) structure of a Ca2+-bound E1 state showed ion coordination by Asp720, Asn716, and Glu292. Time-resolved x-ray solution scattering experiments identified phosphorylation as the rate-determining step. A cryo-EM E2P state structure exhibited remarkable similarities to a SERCA1a E2-P* state, which highlights the essential role of the unique P-A domain interface in enhancing dephosphorylation rates and reconciles earlier proposed mechanisms. Our study underscores the distinctiveness between eukaryotic and prokaryotic Ca2+ ATPase transport systems and positions LMCA1 as a promising drug target for developing antimicrobial strategies.
Collapse
Affiliation(s)
| | - Fredrik Orädd
- Department of Chemistry, Umeå University, Umeå, Sweden
| | | | - Kevin Pounot
- ESRF, The European Synchrotron CS40220, 38043 Grenoble Cedex 9, France
| | - Matteo Levantino
- ESRF, The European Synchrotron CS40220, 38043 Grenoble Cedex 9, France
| | | |
Collapse
|
18
|
Fitisemanu FM, Padilla-Benavides T. Emerging perspectives of copper-mediated transcriptional regulation in mammalian cell development. Metallomics 2024; 16:mfae046. [PMID: 39375833 PMCID: PMC11503025 DOI: 10.1093/mtomcs/mfae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
Copper (Cu) is a vital micronutrient necessary for proper development and function of mammalian cells and tissues. Cu mediates the function of redox active enzymes that facilitate metabolic processes and signaling pathways. Cu levels are tightly regulated by a network of Cu-binding transporters, chaperones, and small molecule ligands. Extensive research has focused on the mammalian Cu homeostasis (cuprostasis) network and pathologies, which result from mutations and perturbations. There are roles for Cu-binding proteins as transcription factors (Cu-TFs) and regulators that mediate metal homeostasis through the activation or repression of genes associated with Cu handling. Emerging evidence suggests that Cu and some Cu-TFs may be involved in the regulation of targets related to development-expanding the biological roles of Cu-binding proteins. Cu and Cu-TFs are implicated in embryonic and tissue-specific development alongside the mediation of the cellular response to oxidative stress and hypoxia. Cu-TFs are also involved in the regulation of targets implicated in neurological disorders, providing new biomarkers and therapeutic targets for diseases such as Parkinson's disease, prion disease, and Friedreich's ataxia. This review provides a critical analysis of the current understanding of the role of Cu and cuproproteins in transcriptional regulation.
Collapse
|
19
|
Ramírez-Alonso JI, Sampedro JG. Effect of Cations on ATP Binding to the N-domain of Na +, K +-ATPase. J Fluoresc 2024:10.1007/s10895-024-03922-3. [PMID: 39298054 DOI: 10.1007/s10895-024-03922-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/04/2024] [Indexed: 09/21/2024]
Abstract
The nucleotide-binding domain (N-domain) of the Na+, K+-ATPase (NKA) is physicochemically characterized by a high content of Glu and Asp residues, resulting in a low isoelectric point (pI = 5.0). Acidic proteins are known to interact with cations. The analysis in silico revealed potential cation interaction sites in the NKA N-domain structure. The interaction with cations was tested in vitro by using a recombinant NKA N-domain. The N-domain contains two Trp residues at the protein surface, as determined by acrylamide-mediated fluorescence quenching, that are useful for structural studies through fluorescence changes. Intrinsic fluorescence of the N-domain was decreased by the presence of cations (Na+, K+, Ca2+) indicating an effect on the protein structure. ATP binding also decreased the N-domain intrinsic fluorescence, which allowed nucleotide affinity determination. In the presence of cations, the N-domain affinity for ATP was increased. Molecular docking of fluorescein isothiocyanate (FITC) with the N-domain showed two binding modes with the isothiocyanate group located 5-6 Å close to Lys485 and Lys506 in the nucleotide-binding site. The presence of ATP prevented the FITC covalent labeling of the N-domain demonstrating the competitive behavior for the binding site. It is proposed that cations interact with the N-domain structure and thereby modulate nucleotide (ATP) affinity and possibly affecting NKA catalysis.
Collapse
Affiliation(s)
- Jocelin I Ramírez-Alonso
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Av. Parque Chapultepec 1570, Privadas del Pedregal, San Luis Potosí, SLP, C.P. 78295, México
| | - José G Sampedro
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Av. Parque Chapultepec 1570, Privadas del Pedregal, San Luis Potosí, SLP, C.P. 78295, México.
| |
Collapse
|
20
|
Tan X, Xu S, Zeng Y, Qin Z, Yu F, Jiang H, Xu H, Li X, Wang X, Zhang G, Ma B, Zhang T, Fan J, Bo X, Kang P, Tang J, Fan H, Zhou Y. Identification of diagnostic signature and immune infiltration for ischemic cardiomyopathy based on cuproptosis-related genes through bioinformatics analysis and experimental validation. Int Immunopharmacol 2024; 138:112574. [PMID: 38971104 DOI: 10.1016/j.intimp.2024.112574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND Ischemic cardiomyopathy (IC) is primarily due to long-term ischemia/hypoxia of the coronary arteries, leading to impaired cardiac contractile or diastolic function. A new form of cell death induced by copper, called "cuproptosis" is related to the development and progression of multiple diseases. The cuproptosis-related gene (CuGs) plays an important role in acute myocardial infarction, while the specific mechanisms of CuGs in ischemic cardiomyopathy remain unclear. METHODS The expressions of CuGs and their immune characteristics were analyzed with the IC datasets obtained from the Gene Expression Omnibus, namely GSE5406 and GSE57338, identifying core genes associated with IC development. By comparing RF, SVM, GLM and XGB models, the optimal machine learning model was selected. The expression of marker genes was validated based on the GSE57345, GSE48166 and GSE42955 datasets. Construct a CeRNA network based on core genes. Therapeutic chemiacals targeting core genes were acquired using the CTD database, and molecular docking was performed using Autodock vina software. By ligating the left anterior descending (LAD) coronary artery, an IC mouse model is established, and core genes were experimentally validated using Western blot (WB) and immunohistochemistry (IHC) methods. RESULTS We identified 14 CuGs closely associated with the onset of IC. The SVM model exhibited superior discriminative power (AUC = 0.914), with core genes being DLST, ATP7B, FDX1, SLC31A1 and DLAT. Core genes were validated on the GSE42955, GSE48166 and GSE57345 datasets, showing excellent performance (AUC = 0.943, AUC = 0.800, and AUC = 0.932). The CeRNA network consists of 218 nodes and 264 lines, including 5 core diagnostic genes, 52 miRNAs, and 161 lncRNAs. Chemicals predictions indicated 8 chemicals have therapeutic effects on the core diagnostic genes, with benzo(a)pyrene molecular docking showing the highest affinity (-11.3 kcal/mol). Compared to the normal group, the IC group,which was established by LAD ligation, showed a significant decrease in LVEF as indicated by cardiac ultrasound, and increased fibrosis as shown by MASSON staining, WB results suggest increased expression of DLST and ATP7B, and decreased expression of FDX1, SLC31A1 and DLAT in the myocardial ischemic area (p < 0.05), which was also confirmed by IHC in tissue sections. CONCLUSION In summary, this study comprehensively revealed that DLST, ATP7B, FDX1, SLC31A1 and DLAT could be identified as potential immunological biomarkers in IC, and validated through an IC mouse model, providing valuable insights for future research into the mechanisms of CuGs and its diagnostic value to IC.
Collapse
Affiliation(s)
- Xin Tan
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215000, China; Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Shuai Xu
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215000, China; Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Yiyao Zeng
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215000, China; Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Zhen Qin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou 450052, China; Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou 450052, China
| | - Fengyi Yu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou 450052, China; Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou 450052, China
| | - Hezi Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215000, China; Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Hui Xu
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215000, China; Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Xian Li
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215000, China; Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Xiangyu Wang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215000, China; Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou 450052, China; Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou 450052, China
| | - Bin Ma
- Luoyang Central Hospital Affiliated of Zhengzhou University, Luoyang 471009, China
| | - Ting Zhang
- Department of Cardiology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Ahhui Medical University, Hefei 230011, China
| | - Jili Fan
- Department of Cardiovascular Disease, Taihe County People's Hospital, Fuyang 236600, China
| | - Xiaohong Bo
- Department of Cardiovascular Disease, Taihe County People's Hospital, Fuyang 236600, China
| | - Pinfang Kang
- Department of Cardiovascular Disease, the First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, China
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou 450052, China; Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou 450052, China
| | - Huimin Fan
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215000, China; Center of Translational Medicine and Clinical Laboratory, The Fourth Affiliated Hospital to Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215028, China
| | - Yafeng Zhou
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215000, China; Institute for Hypertension, Soochow University, Suzhou 215000, China.
| |
Collapse
|
21
|
Shih KY, Chang YT, Wang YJ, Huang JM. Ouabain, ATPase inhibitor, potentially enhances the effect of polyhexamethylene biguanide on Acanthamoeba castellanii. Int J Parasitol Drugs Drug Resist 2024; 25:100550. [PMID: 38821038 PMCID: PMC11177127 DOI: 10.1016/j.ijpddr.2024.100550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/02/2024]
Abstract
Acanthamoeba, a free-living amoeba, is commonly found in various natural environments, such as rivers and soil, as well as in public baths, swimming pools, and sewers. Acanthamoeba can cause severe illness such as granulomatous amoebic encephalitis and Acanthamoeba keratitis (AK) in humans. AK, the most recognized disease, can cause permanent visual impairment or blindness by affecting the cornea. AK commonly affects contact lens wearers who neglect proper cleaning habits. The symptoms of AK include epithelial and stromal destruction, corneal infiltrate, and intense ocular pain, occasionally necessitating surgical removal of the entire eyeball. Current AK treatment involves the hourly application of eye drops containing polyhexamethylene biocide (PHMB). However, studies have revealed their ineffectiveness against drug-resistant strains. Acanthamoeba can form cysts as a survival mechanism in adverse environments, though the exact mechanism remains unknown. Our experiments revealed that sodium P-type ATPase (ACA1_065450) is closely linked to encystation. In addition, various encystation buffers, such as MgCl2 or NaCl, induced the expression of P-type ATPase. Furthermore, we used ouabain, an ATPase inhibitor, to inhibit the Na+/K+ ion pump, consequently decreasing the encystation rate of Acanthamoeba. Our primary objective is to develop an advanced treatment for AK. We anticipate that the combination of ouabain and PHMB may serve as an effective therapeutic approach against AK in the future.
Collapse
Affiliation(s)
- Kuang-Yi Shih
- Department of Life Science, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan.
| | - Yao-Tsung Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Yu-Jen Wang
- Department of Parasitology, School of Medicine, China Medical University, Taichung, Taiwan.
| | - Jian-Ming Huang
- School of Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan; Department of Medical Science, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan; Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
22
|
Guo X, Farag M, Qian N, Yu X, Ni A, Ma Y, Yu W, King MR, Liu V, Lee J, Zare RN, Min W, Pappu RV, Dai Y. Biomolecular condensates can function as inherent catalysts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.06.602359. [PMID: 39026887 PMCID: PMC11257451 DOI: 10.1101/2024.07.06.602359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
We report the discovery that chemical reactions such as ATP hydrolysis can be catalyzed by condensates formed by intrinsically disordered proteins (IDPs), which themselves lack any intrinsic ability to function as enzymes. This inherent catalytic feature of condensates derives from the electrochemical environments and the electric fields at interfaces that are direct consequences of phase separation. The condensates we studied were capable of catalyzing diverse hydrolysis reactions, including hydrolysis and radical-dependent breakdown of ATP whereby ATP fully decomposes to adenine and multiple carbohydrates. This distinguishes condensates from naturally occurring ATPases, which can only catalyze the dephosphorylation of ATP. Interphase and interfacial properties of condensates can be tuned via sequence design, thus enabling control over catalysis through sequence-dependent electrochemical features of condensates. Incorporation of hydrolase-like synthetic condensates into live cells enables activation of transcriptional circuits that depend on products of hydrolysis reactions. Inherent catalytic functions of condensates, which are emergent consequences of phase separation, are likely to affect metabolic regulation in cells.
Collapse
Affiliation(s)
- Xiao Guo
- Department of Biomedical Engineering, Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63130
| | - Mina Farag
- Department of Biomedical Engineering, Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63130
| | - Naixin Qian
- Department of Chemistry, Columbia University, New York, NY 10027
| | - Xia Yu
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Anton Ni
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Yuefeng Ma
- Department of Biomedical Engineering, Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63130
| | - Wen Yu
- Department of Biomedical Engineering, Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63130
| | - Matthew R. King
- Department of Biomedical Engineering, Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63130
| | - Vicky Liu
- Department of Biomedical Engineering, Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63130
| | - Joonho Lee
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Richard N. Zare
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Wei Min
- Department of Chemistry, Columbia University, New York, NY 10027
| | - Rohit V. Pappu
- Department of Biomedical Engineering, Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63130
| | - Yifan Dai
- Department of Biomedical Engineering, Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63130
| |
Collapse
|
23
|
Vavallo M, Cingolani S, Cozza G, Schiavone FP, Dottori L, Palumbo C, Lahner E. Autoimmune Gastritis and Hypochlorhydria: Known Concepts from a New Perspective. Int J Mol Sci 2024; 25:6818. [PMID: 38999928 PMCID: PMC11241626 DOI: 10.3390/ijms25136818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Autoimmune atrophic gastritis is an immune-mediated disease resulting in autoimmune destruction of the specialized acid-producing gastric parietal cells. As a consequence, in autoimmune atrophic gastritis, gastric acid secretion is irreversibly impaired, and the resulting hypochlorhydria leads to the main clinical manifestations and is linked, directly or indirectly, to the long-term neoplastic complications of this disease. In the last few years, autoimmune atrophic gastritis has gained growing interest leading to the acquisition of new knowledge on different aspects of this disorder. Although reliable serological biomarkers are available and gastrointestinal endoscopy techniques have substantially evolved, the diagnosis of autoimmune atrophic gastritis is still affected by a considerable delay and relies on histopathological assessment of gastric biopsies. One of the reasons for the diagnostic delay is that the clinical presentations of autoimmune atrophic gastritis giving rise to clinical suspicion are very different, ranging from hematological to neurological-psychiatric up to gastrointestinal and less commonly to gynecological-obstetric symptoms or signs. Therefore, patients with autoimmune atrophic gastritis often seek advice from physicians of other medical specialties than gastroenterologists, thus underlining the need for increased awareness of this disease in a broad medical and scientific community.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Edith Lahner
- Gastroenterology Unit, Sant’Andrea University Hospital, Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy (G.C.); (F.P.S.)
| |
Collapse
|
24
|
Contreras RG, Torres-Carrillo A, Flores-Maldonado C, Shoshani L, Ponce A. Na +/K +-ATPase: More than an Electrogenic Pump. Int J Mol Sci 2024; 25:6122. [PMID: 38892309 PMCID: PMC11172918 DOI: 10.3390/ijms25116122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
The sodium pump, or Na+/K+-ATPase (NKA), is an essential enzyme found in the plasma membrane of all animal cells. Its primary role is to transport sodium (Na+) and potassium (K+) ions across the cell membrane, using energy from ATP hydrolysis. This transport creates and maintains an electrochemical gradient, which is crucial for various cellular processes, including cell volume regulation, electrical excitability, and secondary active transport. Although the role of NKA as a pump was discovered and demonstrated several decades ago, it remains the subject of intense research. Current studies aim to delve deeper into several aspects of this molecular entity, such as describing its structure and mode of operation in atomic detail, understanding its molecular and functional diversity, and examining the consequences of its malfunction due to structural alterations. Additionally, researchers are investigating the effects of various substances that amplify or decrease its pumping activity. Beyond its role as a pump, growing evidence indicates that in various cell types, NKA also functions as a receptor for cardiac glycosides like ouabain. This receptor activity triggers the activation of various signaling pathways, producing significant morphological and physiological effects. In this report, we present the results of a comprehensive review of the most outstanding studies of the past five years. We highlight the progress made regarding this new concept of NKA and the various cardiac glycosides that influence it. Furthermore, we emphasize NKA's role in epithelial physiology, particularly its function as a receptor for cardiac glycosides that trigger intracellular signals regulating cell-cell contacts, proliferation, differentiation, and adhesion. We also analyze the role of NKA β-subunits as cell adhesion molecules in glia and epithelial cells.
Collapse
Affiliation(s)
| | | | | | | | - Arturo Ponce
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City 07360, Mexico; (R.G.C.); (A.T.-C.); (C.F.-M.); (L.S.)
| |
Collapse
|
25
|
Duan HD, Li H. Consensus, controversies, and conundrums of P4-ATPases: The emerging face of eukaryotic lipid flippases. J Biol Chem 2024; 300:107387. [PMID: 38763336 PMCID: PMC11225554 DOI: 10.1016/j.jbc.2024.107387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/21/2024] Open
Abstract
The cryo-EM resolution revolution has heralded a new era in our understanding of eukaryotic lipid flippases with a rapidly growing number of high-resolution structures. Flippases belong to the P4 family of ATPases (type IV P-type ATPases) that largely follow the reaction cycle proposed for the more extensively studied cation-transporting P-type ATPases. However, unlike the canonical P-type ATPases, no flippase cargos are transported in the phosphorylation half-reaction. Instead of being released into the intracellular or extracellular milieu, lipid cargos are transported to their destination at the inner leaflet of the membrane. Recent flippase structures have revealed multiple conformational states during the lipid transport cycle. Nonetheless, critical conformational states capturing the lipid cargo "in transit" are still missing. In this review, we highlight the amazing structural advances of these lipid transporters, discuss various perspectives on catalytic and regulatory mechanisms in the literature, and shed light on future directions in further deciphering the detailed molecular mechanisms of lipid flipping.
Collapse
Affiliation(s)
- H Diessel Duan
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan, USA.
| | - Huilin Li
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan, USA.
| |
Collapse
|
26
|
Michaels AM, Zoccarato A, Hoare Z, Firth G, Chung YJ, Kuchel PW, Shah AM, Shattock MJ, Southworth R, Eykyn TR. Disrupting Na + ion homeostasis and Na +/K + ATPase activity in breast cancer cells directly modulates glycolysis in vitro and in vivo. Cancer Metab 2024; 12:15. [PMID: 38783368 PMCID: PMC11119389 DOI: 10.1186/s40170-024-00343-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Glycolytic flux is regulated by the energy demands of the cell. Upregulated glycolysis in cancer cells may therefore result from increased demand for adenosine triphosphate (ATP), however it is unknown what this extra ATP turnover is used for. We hypothesise that an important contribution to the increased glycolytic flux in cancer cells results from the ATP demand of Na+/K+-ATPase (NKA) due to altered sodium ion homeostasis in cancer cells. METHODS Live whole-cell measurements of intracellular sodium [Na+]i were performed in three human breast cancer cells (MDA-MB-231, HCC1954, MCF-7), in murine breast cancer cells (4T1), and control human epithelial cells MCF-10A using triple quantum filtered 23Na nuclear magnetic resonance (NMR) spectroscopy. Glycolytic flux was measured by 2H NMR to monitor conversion of [6,6-2H2]D-glucose to [2H]-labelled L-lactate at baseline and in response to NKA inhibition with ouabain. Intracellular [Na+]i was titrated using isotonic buffers with varying [Na+] and [K+] and introducing an artificial Na+ plasma membrane leak using the ionophore gramicidin-A. Experiments were carried out in parallel with cell viability assays, 1H NMR metabolomics of intracellular and extracellular metabolites, extracellular flux analyses and in vivo measurements in a MDA-MB-231 human-xenograft mouse model using 2-deoxy-2-[18F]fluoroglucose (18F-FDG) positron emission tomography (PET). RESULTS Intracellular [Na+]i was elevated in human and murine breast cancer cells compared to control MCF-10A cells. Acute inhibition of NKA by ouabain resulted in elevated [Na+]i and inhibition of glycolytic flux in all three human cancer cells which are ouabain sensitive, but not in the murine cells which are ouabain resistant. Permeabilization of cell membranes with gramicidin-A led to a titratable increase of [Na+]i in MDA-MB-231 and 4T1 cells and a Na+-dependent increase in glycolytic flux. This was attenuated with ouabain in the human cells but not in the murine cells. 18FDG PET imaging in an MDA-MB-231 human-xenograft mouse model recorded lower 18FDG tumour uptake when treated with ouabain while murine tissue uptake was unaffected. CONCLUSIONS Glycolytic flux correlates with Na+-driven NKA activity in breast cancer cells, providing evidence for the 'centrality of the [Na+]i-NKA nexus' in the mechanistic basis of the Warburg effect.
Collapse
Affiliation(s)
- Aidan M Michaels
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Anna Zoccarato
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Zoe Hoare
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - George Firth
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Yu Jin Chung
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Philip W Kuchel
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Ajay M Shah
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Michael J Shattock
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Richard Southworth
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Thomas R Eykyn
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK.
| |
Collapse
|
27
|
Zhao CR, You ZL, Bai L. Fungal Plasma Membrane H +-ATPase: Structure, Mechanism, and Drug Discovery. J Fungi (Basel) 2024; 10:273. [PMID: 38667944 PMCID: PMC11051447 DOI: 10.3390/jof10040273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
The fungal plasma membrane H+-ATPase (Pma1) pumps protons out of the cell to maintain the transmembrane electrochemical gradient and membrane potential. As an essential P-type ATPase uniquely found in fungi and plants, Pma1 is an attractive antifungal drug target. Two recent Cryo-EM studies on Pma1 have revealed its hexameric architecture, autoinhibitory and activation mechanisms, and proton transport mechanism. These structures provide new perspectives for the development of antifungal drugs targeting Pma1. In this article, we review the history of Pma1 structure determination, the latest structural insights into Pma1, and drug discoveries targeting Pma1.
Collapse
Affiliation(s)
- Chao-Ran Zhao
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
- Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing 100005, China
| | - Zi-Long You
- Department of Biophysics, School of Basic Medical Sciences, Peking University, Beijing 100083, China
| | - Lin Bai
- Department of Biophysics, School of Basic Medical Sciences, Peking University, Beijing 100083, China
| |
Collapse
|
28
|
Vishnu N, Venkatesan M, Madaris TR, Venkateswaran MK, Stanley K, Ramachandran K, Chidambaram A, Madesh AK, Yang W, Nair J, Narkunan M, Muthukumar T, Karanam V, Joseph LC, Le A, Osidele A, Aslam MI, Morrow JP, Malicdan MC, Stathopulos PB, Madesh M. ERMA (TMEM94) is a P-type ATPase transporter for Mg 2+ uptake in the endoplasmic reticulum. Mol Cell 2024; 84:1321-1337.e11. [PMID: 38513662 PMCID: PMC10997467 DOI: 10.1016/j.molcel.2024.02.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 10/16/2023] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Intracellular Mg2+ (iMg2+) is bound with phosphometabolites, nucleic acids, and proteins in eukaryotes. Little is known about the intracellular compartmentalization and molecular details of Mg2+ transport into/from cellular organelles such as the endoplasmic reticulum (ER). We found that the ER is a major iMg2+ compartment refilled by a largely uncharacterized ER-localized protein, TMEM94. Conventional and AlphaFold2 predictions suggest that ERMA (TMEM94) is a multi-pass transmembrane protein with large cytosolic headpiece actuator, nucleotide, and phosphorylation domains, analogous to P-type ATPases. However, ERMA uniquely combines a P-type ATPase domain and a GMN motif for ERMg2+ uptake. Experiments reveal that a tyrosine residue is crucial for Mg2+ binding and activity in a mechanism conserved in both prokaryotic (mgtB and mgtA) and eukaryotic Mg2+ ATPases. Cardiac dysfunction by haploinsufficiency, abnormal Ca2+ cycling in mouse Erma+/- cardiomyocytes, and ERMA mRNA silencing in human iPSC-cardiomyocytes collectively define ERMA as an essential component of ERMg2+ uptake in eukaryotes.
Collapse
Affiliation(s)
- Neelanjan Vishnu
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Manigandan Venkatesan
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Travis R Madaris
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Mridula K Venkateswaran
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Kristen Stanley
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Karthik Ramachandran
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Adhishree Chidambaram
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Abitha K Madesh
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Wenli Yang
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jyotsna Nair
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Melanie Narkunan
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Tharani Muthukumar
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Varsha Karanam
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Leroy C Joseph
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
| | - Amy Le
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Ayodeji Osidele
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - M Imran Aslam
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - John P Morrow
- Department of Medicine, College of Physicians and Surgeons of Columbia University, 650 W 168 Street, New York, NY 10032, USA
| | - May C Malicdan
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA; NIH Undiagnosed Diseases Program, Office of the Clinical Director, National Human Genome Research Institute, and the Common Fund, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Western University, London, ON N6A 5C1, Canada
| | - Muniswamy Madesh
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
29
|
Binti S, Edeen PT, Fay DS. Loss of the Na + /K + cation pump CATP-1 suppresses nekl -associated molting defects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585189. [PMID: 38559007 PMCID: PMC10979969 DOI: 10.1101/2024.03.15.585189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The conserved C. elegans protein kinases NEKL-2 and NEKL-3 regulate multiple steps of membrane trafficking and are required for larval molting. Through a forward genetic screen we identified a loss-of-function mutation in catp-1 as a suppressor of molting defects in synthetically lethal nekl-2; nekl-3 double mutants. catp-1 is predicted to encode a membrane- associated P4-type ATPase involved in Na + -K + exchange. Moreover, a mutation predicted to abolish CATP-1 ion-pump activity also suppressed nekl-2; nekl-3 mutants. Endogenously tagged CATP-1 was primarily expressed in epidermal (hypodermal) cells within punctate structures located at or near the apical plasma membrane. Through whole genome sequencing, we identified two additional nekl-2; nekl-3 suppressor strains containing coding-altering mutations in catp-1 but found that neither mutation, when introduced into nekl-2; nekl-3 mutants using CRISPR methods, was sufficient to elicit robust suppression of molting defects. Our data also suggested that the two catp-1 isoforms, catp-1a and catp-1b , may in some contexts be functionally redundant. On the basis of previously published studies, we tested the hypothesis that loss of catp-1 may suppress nekl -associated defects by inducing partial entry into the dauer pathway. Contrary to expectations, however, we failed to obtain evidence that loss of catp-1 suppresses nekl-2; nekl-3 defects through a dauer-associated mechanism or that loss of catp-1 leads to entry into the pre-dauer L2d stage. As such, loss of catp-1 may suppress nekl- associated molting and membrane trafficking defects by altering electrochemical gradients within membrane-bound compartments.
Collapse
|
30
|
Bui HB, Inaba K. Structures, Mechanisms, and Physiological Functions of Zinc Transporters in Different Biological Kingdoms. Int J Mol Sci 2024; 25:3045. [PMID: 38474291 PMCID: PMC10932157 DOI: 10.3390/ijms25053045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/14/2024] Open
Abstract
Zinc transporters take up/release zinc ions (Zn2+) across biological membranes and maintain intracellular and intra-organellar Zn2+ homeostasis. Since this process requires a series of conformational changes in the transporters, detailed information about the structures of different reaction intermediates is required for a comprehensive understanding of their Zn2+ transport mechanisms. Recently, various Zn2+ transport systems have been identified in bacteria, yeasts, plants, and humans. Based on structural analyses of human ZnT7, human ZnT8, and bacterial YiiP, we propose updated models explaining their mechanisms of action to ensure efficient Zn2+ transport. We place particular focus on the mechanistic roles of the histidine-rich loop shared by several zinc transporters, which facilitates Zn2+ recruitment to the transmembrane Zn2+-binding site. This review provides an extensive overview of the structures, mechanisms, and physiological functions of zinc transporters in different biological kingdoms.
Collapse
Affiliation(s)
- Han Ba Bui
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai 980-8577, Japan;
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
- Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai 980-8577, Japan;
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
- Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai 980-8578, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
31
|
Kim LW, Osorio-Castillo V. Quantitative Analysis of the Inactivation Process of Internalized Bacteria in Dictyostelium Cells. Methods Mol Biol 2024; 2814:89-96. [PMID: 38954199 DOI: 10.1007/978-1-0716-3894-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The understanding of the inactivation process of ingested bacteria by phagocytes is a key focus in the field of host-pathogen interactions. Dictyostelium is a model organism that has been at the forefront of uncovering the mechanisms underlying this type of interaction. In this study, we describe an assay designed to measure the inactivation of Klebsiella aerogenes in the phagosomes of Dictyostelium discoideum.
Collapse
Affiliation(s)
- Lou W Kim
- Biological Sciences, Biomolecular Sciences Institute, Florida International University, Miami, FL, USA.
| | - Victor Osorio-Castillo
- Biological Sciences, Biomolecular Sciences Institute, Florida International University, Miami, FL, USA
| |
Collapse
|
32
|
Salas-Orozco MF, Lorenzo-Leal AC, de Alba Montero I, Marín NP, Santana MAC, Bach H. Mechanism of escape from the antibacterial activity of metal-based nanoparticles in clinically relevant bacteria: A systematic review. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 55:102715. [PMID: 37907198 DOI: 10.1016/j.nano.2023.102715] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/05/2023] [Accepted: 10/05/2023] [Indexed: 11/02/2023]
Abstract
The emergency of antibiotic-resistant bacteria in severe infections is increasing, especially in nosocomial environments. The ESKAPE group is of special importance in the groups of multi-resistant bacteria due to its high capacity to generate resistance to antibiotics and bactericides. Therefore, metal-based nanomaterials are an attractive alternative to combat them because they have been demonstrated to damage biomolecules in the bacterial cells. However, there is a concern about bacteria developing resistance to NPs and their harmful effects due to environmental accumulation. Therefore, this systematic review aims to report the clinically relevant bacteria that have developed resistance to the NPs. According to the results of this systematic review, various mechanisms to counteract the antimicrobial activity of various NP types have been proposed. These mechanisms can be grouped into the following categories: production of extracellular compounds, metal efflux pumps, ROS response, genetic changes, DNA repair, adaptative morphogenesis, and changes in the plasma membrane.
Collapse
Affiliation(s)
- Marco Felipe Salas-Orozco
- Facultad de Estomatología, Doctorado en Ciencias Odontológicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.
| | - Ana Cecilia Lorenzo-Leal
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Nuria Patiño Marín
- Facultad de Estomatología, Laboratorio de Investigación Clinica, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Miguel Angel Casillas Santana
- Maestría en Estomatología con Opcion Terminal en Ortodoncia, Facultad de Estomatología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Horacio Bach
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
33
|
Ramli AH, Mohd Faudzi SM. Diarylpentanoids, the privileged scaffolds in antimalarial and anti-infectives drug discovery: A review. Arch Pharm (Weinheim) 2023; 356:e2300391. [PMID: 37806761 DOI: 10.1002/ardp.202300391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 10/10/2023]
Abstract
Asia is a hotspot for infectious diseases, including malaria, dengue fever, tuberculosis, and the pandemic COVID-19. Emerging infectious diseases have taken a heavy toll on public health and the economy and have been recognized as a major cause of morbidity and mortality, particularly in Southeast Asia. Infectious disease control is a major challenge, but many surveillance systems and control strategies have been developed and implemented. These include vector control, combination therapies, vaccine development, and the development of new anti-infectives. Numerous newly discovered agents with pharmacological anti-infective potential are being actively and extensively studied for their bioactivity, toxicity, selectivity, and mode of action, but many molecules lose their efficacy over time due to resistance developments. These facts justify the great importance of the search for new, effective, and safe anti-infectives. Diarylpentanoids, a curcumin derivative, have been developed as an alternative with better bioavailability and metabolism as a therapeutic agent. In this review, the mechanisms of action and potential targets of antimalarial drugs as well as the classes of antimalarial drugs are presented. The bioactivity of diarylpentanoids as a potential scaffold for a new class of anti-infectives and their structure-activity relationships are also discussed in detail.
Collapse
Affiliation(s)
- Amirah H Ramli
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Siti M Mohd Faudzi
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
34
|
Rivera-Morán MA, Sampedro JG. Isolation of the Sarcoplasmic Reticulum Ca 2+-ATPase from Rabbit Fast-Twitch Muscle. Methods Protoc 2023; 6:102. [PMID: 37888034 PMCID: PMC10608927 DOI: 10.3390/mps6050102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
The sarcoendoplasmic reticulum Ca2+-ATPase (SERCA) is a membrane protein that is destabilized during purification in the absence of calcium ions. The disaccharide trehalose is a protein stabilizer that accumulates in the yeast cytoplasm when under stress. In the present work, SERCA was purified by including trehalose in the purification protocol. The purified SERCA showed high protein purity (~95%) and ATPase activity. ATP hydrolysis was dependent on the presence of Ca2+ and the enzyme kinetics showed a hyperbolic dependence on ATP (Km = 12.16 ± 2.25 μM ATP). FITC labeling showed the integrity of the ATP-binding site and the identity of the isolated enzyme as a P-type ATPase. Circular dichroism (CD) spectral changes at a wavelength of 225 nm were observed upon titration with ATP, indicating α-helical rearrangements in the nucleotide-binding domain (N-domain), which correlated with ATP affinity (Km). The presence of Ca2+ did not affect FITC labeling or the ATP-mediated structural changes at the N-domain. The use of trehalose in the SERCA purification protocol stabilized the enzyme. The isolated SERCA appears to be suitable for structural and ligand binding studies, e.g., for testing newly designed or natural inhibitors. The use of trehalose is recommended for the isolation of unstable enzymes.
Collapse
Affiliation(s)
| | - José G. Sampedro
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Avenida Chapultepec 1570, Privadas del Pedregal, San Luis Potosí 78295, Mexico
| |
Collapse
|
35
|
Vig I, Benkő Z, Gila BC, Palczert Z, Jakab Á, Nagy F, Miskei M, Lee MK, Yu JH, Pócsi I, Emri T. Functional characterization of genes encoding cadmium pumping P 1B-type ATPases in Aspergillus fumigatus and Aspergillus nidulans. Microbiol Spectr 2023; 11:e0028323. [PMID: 37676031 PMCID: PMC10581124 DOI: 10.1128/spectrum.00283-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/12/2023] [Indexed: 09/08/2023] Open
Abstract
Several P1B-type ATPases are important Cd2+/Cu2+ pumps in Aspergillus species, and they are tightly associated with the heavy metal stress tolerance of these ascomycetous fungi. To better understand the roles of the two P1B-type ATPases, Aspergillus nidulans CrpA Cd2+/Cu2+ pump (orthologue of the Candida albicans Crp1 Cd2+/Cu2+ pump) and Aspergillus fumigatus PcaA Cd2+ pump (orthologue of the Saccharomyces cerevisiae Pca1 Cd2+ pump), we have generated individual mutants and characterized their heavy metal susceptibilities. The deletion of CrpA in A. nidulans has led to the increased sensitivity of the fungus to stresses induced by Zn2+, Fe2+, or the combination of oxidative-stress-inducing menadione sodium bisulfite and Fe3+. Heterologous expression of A. fumigatus PcaA in the S. cerevisiae pca1 deletion mutant has resulted in enhanced tolerance of the yeast to stresses elicited by Cd2+or Zn2+ but not by Fe2+/Fe3+ or Cu2+. Mammalian host immune defense can attack microbes by secreting Zn2+ or Cu2+, and the oxidative stress induced by host immune systems can also disturb metal (Cu2+, Fe2+, and Zn2+) homeostasis in microbes. In summary, PcaA and CrpA can protect fungal cells from these complex stresses that contribute to the virulence of the pathogenic Aspergillus species. Moreover, due to their presence on the fungal cell surface, these P1B-type ATPases may serve as a novel drug target in the future. IMPORTANCE Mammalian host immune defense disrupts heavy metal homeostasis of fungal pathogens. P1B-type ATPase of Aspergillus fumigatus and Aspergillus nidulans may help to cope with this stress and serve as virulence traits. In our experiments, both A. nidulans Cd2+/Cu2+ pump CrpA and A. fumigatus Cd2+ pump PcaA protected fungal cells from toxic Zn2+, and CrpA also decreased Fe2+ susceptibility most likely indirectly. In addition, CrpA protected cells against the combined stress induced by the oxidative stressor menadione and Fe3+. Since P1B-type ATPases are present on the fungal cell surface, these proteins may serve as a novel drug target in the future.
Collapse
Affiliation(s)
- Ildikó Vig
- Department of Molecular Biotechnology and Microbiology, Faculty of Sciences and Technology, University of Debrecen, Debrecen, Hungary
- ELRN-UD Fungal Stress Biology Research Group, Debrecen, Hungary
| | - Zsigmond Benkő
- Department of Molecular Biotechnology and Microbiology, Faculty of Sciences and Technology, University of Debrecen, Debrecen, Hungary
| | - Barnabás Cs. Gila
- Department of Molecular Biotechnology and Microbiology, Faculty of Sciences and Technology, University of Debrecen, Debrecen, Hungary
| | - Zoltán Palczert
- Department of Molecular Biotechnology and Microbiology, Faculty of Sciences and Technology, University of Debrecen, Debrecen, Hungary
| | - Ágnes Jakab
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Fruzsina Nagy
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Márton Miskei
- ELRN-UD Fungal Stress Biology Research Group, Debrecen, Hungary
| | - Mi-Kyung Lee
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, South Korea
| | - Jae-Hyuk Yu
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - István Pócsi
- Department of Molecular Biotechnology and Microbiology, Faculty of Sciences and Technology, University of Debrecen, Debrecen, Hungary
- ELRN-UD Fungal Stress Biology Research Group, Debrecen, Hungary
| | - Tamás Emri
- Department of Molecular Biotechnology and Microbiology, Faculty of Sciences and Technology, University of Debrecen, Debrecen, Hungary
- ELRN-UD Fungal Stress Biology Research Group, Debrecen, Hungary
| |
Collapse
|
36
|
Huynh U, Nguyen HN, Trinh BK, Elhaj J, Zastrow ML. A bioinformatic analysis of zinc transporters in intestinal Lactobacillaceae. Metallomics 2023; 15:mfad044. [PMID: 37463796 PMCID: PMC10391621 DOI: 10.1093/mtomcs/mfad044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/17/2023] [Indexed: 07/20/2023]
Abstract
As the second most abundant transition element and a crucial cofactor for many proteins, zinc is essential for the survival of all living organisms. To maintain required zinc levels and prevent toxic overload, cells and organisms have a collection of metal transport proteins for uptake and efflux of zinc. In bacteria, metal transport proteins are well defined for model organisms and many pathogens, but fewer studies have explored metal transport proteins, including those for zinc, in commensal bacteria from the gut microbiota. The healthy human gut microbiota comprises hundreds of species and among these, bacteria from the Lactobacillaceae family are well documented to have various beneficial effects on health. Furthermore, changes in dietary metal intake, such as for zinc and iron, are frequently correlated with changes in abundance of Lactobacillaceae. Few studies have explored zinc requirements and zinc homeostasis mechanisms in Lactobacillaceae, however. Here we applied a bioinformatics approach to identify and compare predicted zinc uptake and efflux proteins in several Lactobacillaceae genera of intestinal relevance. Few Lactobacillaceae had zinc transporters currently annotated in proteomes retrieved from the UniProt database, but protein sequence-based homology searches revealed that high-affinity ABC transporter genes are likely common, albeit with genus-specific domain features. P-type ATPase transporters are probably also common and some Lactobacillaceae genera code for predicted zinc efflux cation diffusion facilitators. This analysis confirms that Lactobacillaceae harbor genes for various zinc transporter homologs, and provides a foundation for systematic experimental studies to elucidate zinc homeostasis mechanisms in these bacteria.
Collapse
Affiliation(s)
- Uyen Huynh
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Hazel N Nguyen
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Brittany K Trinh
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Joanna Elhaj
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Melissa L Zastrow
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
37
|
Romano JD, Li H, Napolitano T, Realubit R, Karan C, Holford M, Tatonetti NP. Discovering Venom-Derived Drug Candidates Using Differential Gene Expression. Toxins (Basel) 2023; 15:451. [PMID: 37505720 PMCID: PMC10467105 DOI: 10.3390/toxins15070451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/16/2023] [Accepted: 07/07/2023] [Indexed: 07/29/2023] Open
Abstract
Venoms are a diverse and complex group of natural toxins that have been adapted to treat many types of human disease, but rigorous computational approaches for discovering new therapeutic activities are scarce. We have designed and validated a new platform-named VenomSeq-to systematically identify putative associations between venoms and drugs/diseases via high-throughput transcriptomics and perturbational differential gene expression analysis. In this study, we describe the architecture of VenomSeq and its evaluation using the crude venoms from 25 diverse animal species and 9 purified teretoxin peptides. By integrating comparisons to public repositories of differential expression, associations between regulatory networks and disease, and existing knowledge of venom activity, we provide a number of new therapeutic hypotheses linking venoms to human diseases supported by multiple layers of preliminary evidence.
Collapse
Affiliation(s)
- Joseph D. Romano
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center of Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hai Li
- Department of Systems Biology, Columbia University, New York, NY 10032, USA; (H.L.); (R.R.); (C.K.)
- Columbia Genome Center, Columbia University, New York, NY 10032, USA
| | - Tanya Napolitano
- Department of Chemistry, CUNY Hunter College, New York, NY 10032, USA (M.H.)
- The PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA
| | - Ronald Realubit
- Department of Systems Biology, Columbia University, New York, NY 10032, USA; (H.L.); (R.R.); (C.K.)
- Columbia Genome Center, Columbia University, New York, NY 10032, USA
| | - Charles Karan
- Department of Systems Biology, Columbia University, New York, NY 10032, USA; (H.L.); (R.R.); (C.K.)
- Columbia Genome Center, Columbia University, New York, NY 10032, USA
| | - Mandë Holford
- Department of Chemistry, CUNY Hunter College, New York, NY 10032, USA (M.H.)
- The PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA
- The PhD Program in Chemistry, Graduate Center of the City University of New York, New York, NY 10016, USA
- The PhD Program in Biology, Graduate Center of the City University of New York, New York, NY 10016, USA
- Department of Invertebrate Zoology, The American Museum of Natural History, New York, NY 10032, USA
| | - Nicholas P. Tatonetti
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA 90069, USA
| |
Collapse
|
38
|
Bailey MLP, Pratt SE, Hinrichsen M, Zhang Y, Bewersdorf J, Regan LJ, Mochrie SGJ. Uncovering diffusive states of the yeast membrane protein, Pma1, and how labeling method can change diffusive behavior. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2023; 46:42. [PMID: 37294385 PMCID: PMC10369454 DOI: 10.1140/epje/s10189-023-00301-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 05/15/2023] [Indexed: 06/10/2023]
Abstract
We present and analyze video-microscopy-based single-particle-tracking measurements of the budding yeast (Saccharomyces cerevisiae) membrane protein, Pma1, fluorescently labeled either by direct fusion to the switchable fluorescent protein, mEos3.2, or by a novel, light-touch, labeling scheme, in which a 5 amino acid tag is directly fused to the C-terminus of Pma1, which then binds mEos3.2. The track diffusivity distributions of these two populations of single-particle tracks differ significantly, demonstrating that labeling method can be an important determinant of diffusive behavior. We also applied perturbation expectation maximization (pEMv2) (Koo and Mochrie in Phys Rev E 94(5):052412, 2016), which sorts trajectories into the statistically optimum number of diffusive states. For both TRAP-labeled Pma1 and Pma1-mEos3.2, pEMv2 sorts the tracks into two diffusive states: an essentially immobile state and a more mobile state. However, the mobile fraction of Pma1-mEos3.2 tracks is much smaller ([Formula: see text]) than the mobile fraction of TRAP-labeled Pma1 tracks ([Formula: see text]). In addition, the diffusivity of Pma1-mEos3.2's mobile state is several times smaller than the diffusivity of TRAP-labeled Pma1's mobile state. Thus, the two different labeling methods give rise to very different overall diffusive behaviors. To critically assess pEMv2's performance, we compare the diffusivity and covariance distributions of the experimental pEMv2-sorted populations to corresponding theoretical distributions, assuming that Pma1 displacements realize a Gaussian random process. The experiment-theory comparisons for both the TRAP-labeled Pma1 and Pma1-mEos3.2 reveal good agreement, bolstering the pEMv2 approach.
Collapse
Affiliation(s)
- Mary Lou P Bailey
- Integrated Graduate Program in Physical and Engineering Biology, Yale University, New Haven, CT, 06511, USA
- Department of Applied Physics, Yale University, New Haven, CT, 06511, USA
| | - Susan E Pratt
- Integrated Graduate Program in Physical and Engineering Biology, Yale University, New Haven, CT, 06511, USA
- Department of Physics, Yale University, New Haven, CT, 06511, USA
| | | | - Yongdeng Zhang
- Department of Cell Biology, Yale University, New Haven, CT, 06511, USA
| | - Joerg Bewersdorf
- Department of Applied Physics, Yale University, New Haven, CT, 06511, USA
- Department of Physics, Yale University, New Haven, CT, 06511, USA
- Department of Cell Biology, Yale University, New Haven, CT, 06511, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Lynne J Regan
- Institute of Quantitative Biology, Biochemistry and Biotechnology, Center for Synthetic and Systems Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, 06511, UK
| | - Simon G J Mochrie
- Integrated Graduate Program in Physical and Engineering Biology, Yale University, New Haven, CT, 06511, USA.
- Department of Applied Physics, Yale University, New Haven, CT, 06511, USA.
- Department of Physics, Yale University, New Haven, CT, 06511, USA.
| |
Collapse
|
39
|
Roberts SM, Aldis M, Wright ET, Gonzales CB, Lai Z, Weintraub ST, Hardies SC, Serwer P. Siphophage 0105phi7-2 of Bacillus thuringiensis: Novel Propagation, DNA, and Genome-Implied Assembly. Int J Mol Sci 2023; 24:ijms24108941. [PMID: 37240285 DOI: 10.3390/ijms24108941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Diversity of phage propagation, physical properties, and assembly promotes the use of phages in ecological studies and biomedicine. However, observed phage diversity is incomplete. Bacillus thuringiensis siphophage, 0105phi-7-2, first described here, significantly expands known phage diversity, as seen via in-plaque propagation, electron microscopy, whole genome sequencing/annotation, protein mass spectrometry, and native gel electrophoresis (AGE). Average plaque diameter vs. plaque-supporting agarose gel concentration plots reveal unusually steep conversion to large plaques as agarose concentration decreases below 0.2%. These large plaques sometimes have small satellites and are made larger by orthovanadate, an ATPase inhibitor. Phage head-host-cell binding is observed by electron microscopy. We hypothesize that this binding causes plaque size-increase via biofilm evolved, ATP stimulated ride-hitching on motile host cells by temporarily inactive phages. Phage 0105phi7-2 does not propagate in liquid culture. Genomic sequencing/annotation reveals history as temperate phage and distant similarity, in a virion-assembly gene cluster, to prototypical siphophage SPP1 of Bacillus subtilis. Phage 0105phi7-2 is distinct in (1) absence of head-assembly scaffolding via either separate protein or classically sized, head protein-embedded peptide, (2) producing partially condensed, head-expelled DNA, and (3) having a surface relatively poor in AGE-detected net negative charges, which is possibly correlated with observed low murine blood persistence.
Collapse
Affiliation(s)
- Samantha M Roberts
- Department of Microbiology, Immunology and Molecular Genetics, UT Health, San Antonio, TX 78229, USA
| | - Miranda Aldis
- Department of Microbiology, Immunology and Molecular Genetics, UT Health, San Antonio, TX 78229, USA
| | - Elena T Wright
- Department of Biochemistry and Structural Biology, UT Health, San Antonio, TX 78229, USA
| | - Cara B Gonzales
- Department of Comprehensive Dentistry, UT Health, San Antonio, TX 78229, USA
| | - Zhao Lai
- Department of Molecular Medicine, UT Health, San Antonio, TX 78229, USA
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, UT Health, San Antonio, TX 78229, USA
| | - Stephen C Hardies
- Department of Biochemistry and Structural Biology, UT Health, San Antonio, TX 78229, USA
| | - Philip Serwer
- Department of Biochemistry and Structural Biology, UT Health, San Antonio, TX 78229, USA
| |
Collapse
|
40
|
Yang GM, Xu L, Wang RM, Tao X, Zheng ZW, Chang S, Ma D, Zhao C, Dong Y, Wu S, Guo J, Wu ZY. Structures of the human Wilson disease copper transporter ATP7B. Cell Rep 2023; 42:112417. [PMID: 37074913 DOI: 10.1016/j.celrep.2023.112417] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/22/2023] [Accepted: 04/05/2023] [Indexed: 04/20/2023] Open
Abstract
The P-type ATPase ATP7B exports cytosolic copper and plays an essential role in the regulation of cellular copper homeostasis. Mutants of ATP7B cause Wilson disease (WD), an autosomal recessive disorder of copper metabolism. Here, we present cryoelectron microscopy (cryo-EM) structures of human ATP7B in the E1 state in the apo, the putative copper-bound, and the putative cisplatin-bound forms. In ATP7B, the N-terminal sixth metal-binding domain (MBD6) binds at the cytosolic copper entry site of the transmembrane domain (TMD), facilitating the delivery of copper from the MBD6 to the TMD. The sulfur-containing residues in the TMD of ATP7B mark the copper transport pathway. By comparing structures of the E1 state human ATP7B and E2-Pi state frog ATP7B, we propose the ATP-driving copper transport model of ATP7B. These structures not only advance our understanding of the mechanisms of ATP7B-mediated copper export but can also guide the development of therapeutics for the treatment of WD.
Collapse
Affiliation(s)
- Guo-Min Yang
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Lingyi Xu
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Rou-Min Wang
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Xin Tao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Zi-Wei Zheng
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Shenghai Chang
- Department of Biophysics, and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Center of Cryo Electron Microscopy, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Demin Ma
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Cheng Zhao
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yi Dong
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Shan Wu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China.
| | - Jiangtao Guo
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; State Key Laboratory of Plant Physiology and Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Cardiology, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, Zhejiang 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China.
| | - Zhi-Ying Wu
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, Zhejiang 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China.
| |
Collapse
|
41
|
Mu J, Xue C, Fu L, Yu Z, Nie M, Wu M, Chen X, Liu K, Bu R, Huang Y, Yang B, Han J, Jiang Q, Chan KC, Zhou R, Li H, Huang A, Wang Y, Liu Z. Conformational cycle of human polyamine transporter ATP13A2. Nat Commun 2023; 14:1978. [PMID: 37031211 PMCID: PMC10082790 DOI: 10.1038/s41467-023-37741-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 03/28/2023] [Indexed: 04/10/2023] Open
Abstract
Dysregulation of polyamine homeostasis strongly associates with human diseases. ATP13A2, which is mutated in juvenile-onset Parkinson's disease and autosomal recessive spastic paraplegia 78, is a transporter with a critical role in balancing the polyamine concentration between the lysosome and the cytosol. Here, to better understand human ATP13A2-mediated polyamine transport, we use single-particle cryo-electron microscopy to solve high-resolution structures of human ATP13A2 in six intermediate states, including the putative E2 structure for the P5 subfamily of the P-type ATPases. These structures comprise a nearly complete conformational cycle spanning the polyamine transport process and capture multiple substrate binding sites distributed along the transmembrane regions, suggesting a potential polyamine transport pathway. Integration of high-resolution structures, biochemical assays, and molecular dynamics simulations allows us to obtain a better understanding of the structural basis of how hATP13A2 transports polyamines, providing a mechanistic framework for ATP13A2-related diseases.
Collapse
Affiliation(s)
- Jianqiang Mu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Chenyang Xue
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Lei Fu
- Shanghai Institute for Advanced Study, Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, 310027, Hangzhou, China
| | - Zongjun Yu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Minhan Nie
- School of Pharmaceutical Sciences, Sun Yat-sen University, No.132 Wai Huan Dong Lu, Guangzhou Higher Education Mega Center, 510006, Guangzhou, China
| | - Mengqi Wu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Xinmeng Chen
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Kun Liu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Ruiqian Bu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Ying Huang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Baisheng Yang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Jianming Han
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Qianru Jiang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Kevin C Chan
- Shanghai Institute for Advanced Study, Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, 310027, Hangzhou, China
| | - Ruhong Zhou
- Shanghai Institute for Advanced Study, Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, 310027, Hangzhou, China
| | - Huilin Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, No.132 Wai Huan Dong Lu, Guangzhou Higher Education Mega Center, 510006, Guangzhou, China
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, Guangdong, China
| | - Ancheng Huang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Yong Wang
- Shanghai Institute for Advanced Study, Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, 310027, Hangzhou, China.
- The Provincial International Science and Technology Cooperation Base on Engineering Biology, International Campus of Zhejiang University, 314400, Haining, China.
| | - Zhongmin Liu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
| |
Collapse
|
42
|
Kalavacherla T, Buschmann S, Schleker ESM, Michel H, Reinhart C. Purification and characterization of eukaryotic ATP-dependent transporters homologously expressed in Pichia pastoris for structural studies by cryo-electron microscopy. Protein Expr Purif 2023; 204:106230. [PMID: 36632890 DOI: 10.1016/j.pep.2023.106230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/22/2022] [Accepted: 01/01/2023] [Indexed: 01/09/2023]
Abstract
Membrane proteins play an essential role in all living organisms. Although there have been numerous efforts in the past to elucidate the structure and function of eukaryotic primary active transporters, knowledge about the majority of these membrane proteins is still minimal. This is often due to their low availability and complex handling. In this study, we homologously expressed three ATP-dependent transport proteins, STE6-2p, NEO1-p, and YPK9-p, in Pichia pastoris and subsequently optimized the solubilization and purification processes. Sequential use of different mild detergents and utilization of hydrophilic matrices in the purification procedure allowed us to obtain all three transporters monodisperse and in high purity, enabling initial structural analysis by cryo-electron microscopy. Using the respective substrates, we determined the specific activity of all target proteins using an ATPase assay. This study opens the door to further functional and structural studies of this pharmacologically important class of membrane proteins.
Collapse
Affiliation(s)
- Tejaswi Kalavacherla
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, D-60438, Frankfurt am Main, Germany
| | - Sabine Buschmann
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, D-60438, Frankfurt am Main, Germany
| | - E Sabine M Schleker
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, D-60438, Frankfurt am Main, Germany
| | - Hartmut Michel
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, D-60438, Frankfurt am Main, Germany
| | - Christoph Reinhart
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, D-60438, Frankfurt am Main, Germany.
| |
Collapse
|
43
|
Sim SI, Park E. P5-ATPases: Structure, substrate specificities, and transport mechanisms. Curr Opin Struct Biol 2023; 79:102531. [PMID: 36724561 DOI: 10.1016/j.sbi.2023.102531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/30/2022] [Accepted: 12/19/2022] [Indexed: 02/01/2023]
Abstract
P5A- and P5B- ATPases, or collectively P5-ATPases, are eukaryotic-specific ATP-dependent transporters that are important for the function of the endoplasmic reticulum (ER) and endo-/lysosomes. However, their substrate specificities had remained enigmatic for many years. Recent cryo-electron microscopy (cryo-EM) and biochemical studies of P5-ATPases have revealed their substrate specificities and transport mechanisms, which were found to be markedly different from other members of the P-type ATPase superfamily. The P5A-ATPase extracts mistargeted or mis-inserted transmembrane helices from the ER membrane for protein quality control, while the P5B-ATPases mediate export of polyamines from late endo-/lysosomes into the cytosol. In this review, we discuss the mechanisms of their substrate recognition and transport based on the cryo-EM structures of the yeast and human P5-ATPases. We highlight how structural diversification of the transmembrane domain has enabled the P5-ATPase subfamily to adapt for transport of atypical substrates.
Collapse
Affiliation(s)
- Sue Im Sim
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Eunyong Park
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA; California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
44
|
The physical exercise-induced oxidative/inflammatory response in peripheral blood mononuclear cells: Signaling cellular energetic stress situations. Life Sci 2023; 321:121440. [PMID: 36921686 DOI: 10.1016/j.lfs.2023.121440] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/14/2023] [Accepted: 01/22/2023] [Indexed: 03/14/2023]
Abstract
Peripheral blood mononuclear cells (PBMCs) are a variety of specialized immune cells produced in the bone marrow from hematopoietic stem cells (HSCs) that work together to protect our bodies from harmful pathogens. From a metabolic point of view, these cells can serve as sentinel tissue source for distinguishing multiple types of whole-body physiological perturbations. The significant interaction of PBMCs with systemic physiology makes these cells an attractive target for several interventions such as physical exercise. Analyses of oxidative/inflammatory and metabolic markers of PBMCs obtained from unhealthy and healthy humans have been used in monitoring immune response in different exercise conditions. It is already a common consensus that regular practice of physical exercise, that is planned, structured, and repetitive, influences personal health by altering the metabolic state and the immune system. However, the role of distinct metabolic processes responsible for maintaining metabolic balance during physical exercise in PBMCs is not fully understood. Furthermore, a complete dose-response analysis between different exercise protocols and biomarkers capable of predicting physical performance needs to be better elucidated. The absence of published reviews on this topic compromises the understanding of the crosstalk between the metabolic adaptations of PBMCs and exercise-induced changes in the immune system. Given the above, this review highlights the main findings in the literature involving the responses of PBMCs in the inflammatory/oxidative stress induced by physical exercise. The present review also highlights how distinct phenotypes and functional diversity of PBMCs make these cells an accessible alternative for assessing exercise-induced metabolic adaptations.
Collapse
|
45
|
Camponeschi F, Banci L. Metal trafficking in the cell: Combining atomic resolution with cellular dimension. FEBS Lett 2023; 597:122-133. [PMID: 36285633 DOI: 10.1002/1873-3468.14524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 01/14/2023]
Abstract
Metals are widely present in biological systems as simple ions or complex cofactors, and are involved in a variety of processes essential for life. Their transport inside cells and insertion into the binding sites of the proteins that need metals to function occur through complex and selective pathways involving dedicated multiprotein machineries specifically and transiently interacting with each other, often sharing the coordination of metal ions and/or cofactors. The understanding of these machineries requires integrated approaches, ranging from bioinformatics to experimental investigations, possibly in the cellular context. In this review, we report two case studies where the use of integrated in vitro and in cellulo approaches is necessary to clarify at atomic resolution essential aspects of metal trafficking in cells.
Collapse
Affiliation(s)
- Francesca Camponeschi
- Magnetic Resonance Center CERM, University of Florence, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Florence, Italy
| | - Lucia Banci
- Magnetic Resonance Center CERM, University of Florence, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Florence, Italy.,Department of Chemistry, University of Florence, Italy
| |
Collapse
|
46
|
Identification of potassium transport proteins in algae and determination of their role under salt and saline-alkaline stress. ALGAL RES 2023. [DOI: 10.1016/j.algal.2022.102923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
47
|
Azfar M, van Veen S, Houdou M, Hamouda NN, Eggermont J, Vangheluwe P. P5B-ATPases in the mammalian polyamine transport system and their role in disease. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119354. [PMID: 36064065 DOI: 10.1016/j.bbamcr.2022.119354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 06/15/2023]
Abstract
Polyamines (PAs) are physiologically relevant molecules that are ubiquitous in all organisms. The vitality of PAs to the healthy functioning of a cell is due to their polycationic nature causing them to interact with a vast plethora of cellular players and partake in numerous cellular pathways. Naturally, the homeostasis of such essential molecules is tightly regulated in a strictly controlled interplay between intracellular synthesis and degradation, uptake from and secretion to the extracellular compartment, as well as intracellular trafficking. Not surprisingly, dysregulated PA homeostasis and signaling are implicated in multiple disorders, ranging from cancer to neurodegeneration; leading many to propose rectifying the PA balance as a potential therapeutic strategy. Despite being well characterized in bacteria, fungi and plants, the molecular identity and properties of the PA transporters in animals are poorly understood. This review brings together the current knowledge of the cellular function of the mammalian PA transport system (PTS). We will focus on the role of P5B-ATPases ATP13A2-5 which are PA transporters in the endosomal system that have emerged as key players in cellular PA uptake and organelle homeostasis. We will discuss recent breakthroughs on their biochemical and structural properties as well as their implications for disease and therapy.
Collapse
Affiliation(s)
- Mujahid Azfar
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, KU Leuven, B-3000 Leuven, Belgium
| | - Sarah van Veen
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, KU Leuven, B-3000 Leuven, Belgium
| | - Marine Houdou
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, KU Leuven, B-3000 Leuven, Belgium
| | - Norin Nabil Hamouda
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Jan Eggermont
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, KU Leuven, B-3000 Leuven, Belgium.
| |
Collapse
|
48
|
Huang Z, Feng Z, Zou Y. New wine in old bottles: current progress on P5 ATPases. FEBS J 2022; 289:7304-7313. [PMID: 34449980 DOI: 10.1111/febs.16172] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/19/2021] [Accepted: 08/26/2021] [Indexed: 01/13/2023]
Abstract
P5 ATPases are evolutionarily conserved P-type transporters. Despite their important roles in the endoplasmic reticulum (ER) and in lysosomes, the substrate specificities and transporting mechanisms of P5 ATPases have remained mysterious. Recently, several studies have provided genetic, biochemical, and structural evidence to help elucidate the physiological functions and substrates of P5 ATPases. Here, we summarize this progress and discuss the potential transport mechanisms of the P5 ATPases-in particular, P5A ATPase-for further study.
Collapse
Affiliation(s)
- Zhiwen Huang
- School of Life Science and Technology, ShanghaiTech University, China
| | - Zhigang Feng
- School of Life Science and Technology, ShanghaiTech University, China
| | - Yan Zou
- School of Life Science and Technology, ShanghaiTech University, China
| |
Collapse
|
49
|
Hogg DW, Casatti CC, Belsham DD, Baršytė-Lovejoy D, Lovejoy DA. Distal extracellular teneurin region (teneurin C-terminal associated peptide; TCAP) possesses independent intracellular calcium regulating actions, in vitro: A potential antagonist of corticotropin-releasing factor (CRF). Biochem Biophys Rep 2022; 32:101397. [DOI: 10.1016/j.bbrep.2022.101397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
|
50
|
Cunliffe G, Lim YT, Chae W, Jung S. Alternative Pharmacological Strategies for the Treatment of Alzheimer's Disease: Focus on Neuromodulator Function. Biomedicines 2022; 10:3064. [PMID: 36551821 PMCID: PMC9776382 DOI: 10.3390/biomedicines10123064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, comprising 70% of dementia diagnoses worldwide and affecting 1 in 9 people over the age of 65. However, the majority of its treatments, which predominantly target the cholinergic system, remain insufficient at reversing pathology and act simply to slow the inevitable progression of the disease. The most recent neurotransmitter-targeting drug for AD was approved in 2003, strongly suggesting that targeting neurotransmitter systems alone is unlikely to be sufficient, and that research into alternate treatment avenues is urgently required. Neuromodulators are substances released by neurons which influence neurotransmitter release and signal transmission across synapses. Neuromodulators including neuropeptides, hormones, neurotrophins, ATP and metal ions display altered function in AD, which underlies aberrant neuronal activity and pathology. However, research into how the manipulation of neuromodulators may be useful in the treatment of AD is relatively understudied. Combining neuromodulator targeting with more novel methods of drug delivery, such as the use of multi-targeted directed ligands, combinatorial drugs and encapsulated nanoparticle delivery systems, may help to overcome limitations of conventional treatments. These include difficulty crossing the blood-brain-barrier and the exertion of effects on a single target only. This review aims to highlight the ways in which neuromodulator functions are altered in AD and investigate how future therapies targeting such substances, which act upstream to classical neurotransmitter systems, may be of potential therapeutic benefit in the sustained search for more effective treatments.
Collapse
Affiliation(s)
- Grace Cunliffe
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138667, Singapore
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Yi Tang Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138667, Singapore
- Faculty of Science, National University of Singapore, Singapore 117546, Singapore
| | - Woori Chae
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138667, Singapore
- Department of BioNano Technology, Gachon University, 1342 Seongnam-daero, Seongnam-si 13120, Republic of Korea
| | - Sangyong Jung
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138667, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| |
Collapse
|