1
|
Su Z, Chen T, Liu X, Kang X. Size-tunable transmembrane nanopores assembled from decomposable molecular templates. Biosens Bioelectron 2025; 267:116780. [PMID: 39277918 DOI: 10.1016/j.bios.2024.116780] [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: 05/18/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Transmembrane nanopores, as key elements in molecular transport and single-molecule sensors, are assembled naturally from multiple monomers in the presence of lipid bilayers. The nanopore size, especially the precise diameter of the inner space, determines its sensing targets and further biological application. In this paper, we introduce a template molecule-aided assembly strategy for constructing size-tunable transmembrane nanopores. Inspired by the barrel-like structure, similar to many transmembrane proteins, cyclodextrin molecules of different sizes are utilized as templates and modulators to assemble the α-helical barreled peptide of polysaccharide transporters (Wza). The functional nanopores assembled by this strategy possess high biological and chemical activity and can be inserted into lipid bilayers, forming stable single channels for single-molecule sensing. After enzyme digestion, the cyclodextrins on protein nanopores can be degraded, and the remaining nontemplate transmembrane protein nanopores can also preserve the integrity of their structure and function. The template molecule-aided assembly strategy employed a simple and convenient method for fully artificially synthesizing transmembrane protein nanopores; the pore size is completely dependent on the size of the template molecule and controllable, ranging from 1.1 to 1.8 nm. Furthermore, by chemically synthesized peptides and modifications, the pore function is easily modulated and does not involve the cumbersome genetic mutations of other biological techniques.
Collapse
Affiliation(s)
- Zhuoqun Su
- College of Chemistry & Materials Science, Key Laboratory of Synthetic and Natural Functional Molecular Chemistry, Northwest University, Xi'an, 710127, China; School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, China
| | - Tingting Chen
- College of Chemistry & Materials Science, Key Laboratory of Synthetic and Natural Functional Molecular Chemistry, Northwest University, Xi'an, 710127, China
| | - Xingtong Liu
- College of Chemistry & Materials Science, Key Laboratory of Synthetic and Natural Functional Molecular Chemistry, Northwest University, Xi'an, 710127, China
| | - Xiaofeng Kang
- College of Chemistry & Materials Science, Key Laboratory of Synthetic and Natural Functional Molecular Chemistry, Northwest University, Xi'an, 710127, China.
| |
Collapse
|
2
|
Wei R, Liu X, Cao L, Chen C, Chen IC, Li Z, Miao J, Lai Z. Zeolite membrane with sub-nanofluidic channels for superior blue energy harvesting. Nat Commun 2024; 15:10489. [PMID: 39622835 PMCID: PMC11612162 DOI: 10.1038/s41467-024-54755-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/21/2024] [Indexed: 12/06/2024] Open
Abstract
Blue energy, a clean energy source derived from salinity gradients, has recently drawn increased research attention. It can be harvested using charged membranes, typically composed of amorphous materials that suffer from low power density due to their disordered structure and low charge density. Crystalline materials, with inherently ordered porous structures, offer a promising alternative for overcoming these limitations. Zeolite, a crystalline material with ordered sub-nanofluidic channels and tunable charge density, is particularly well-suited for this purpose. Here, we demonstrate that NaX zeolite functions as a high-performance membrane for blue energy generation. The NaX zeolite membrane achieves a power density of 21.27 W m⁻² under a 50-fold NaCl concentration gradient, exceeding the performance of state-of-the-art membranes under similar conditions. When tested under practical scenarios, it yields power densities of 29.1 W m⁻², 81.0 W m⁻², and 380.1 W m⁻² in the Red Sea/River, Dead Sea/River, and Qinghai Brine/River configurations, respectively. Notably, the membrane operates effectively in high alkaline conditions (~0.5 M NaOH) and selectively separates CO₃²⁻ from OH⁻ ions with a selectivity of 25. These results underscore zeolite membranes' potential for blue energy, opening further opportunities in this field.
Collapse
Affiliation(s)
- Ruicong Wei
- Advanced Membranes and Porous Materials Center, Division of Physical Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Xiaowei Liu
- Advanced Membranes and Porous Materials Center, Division of Physical Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Li Cao
- Advanced Membranes and Porous Materials Center, Division of Physical Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Cailing Chen
- Advanced Membranes and Porous Materials Center, Division of Physical Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - I-Chun Chen
- Advanced Membranes and Porous Materials Center, Division of Physical Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Zhen Li
- Advanced Membranes and Porous Materials Center, Division of Physical Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Jun Miao
- Advanced Membranes and Porous Materials Center, Division of Physical Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Zhiping Lai
- Advanced Membranes and Porous Materials Center, Division of Physical Science and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
3
|
Abe K, McDermott J, Valia Madapally H, Marimuthu P, Gopalasingam CC, Gerle C, Shigematsu H, Khandelia H, Blanco G. Molecular Structure of the Na +,K +-ATPase α4β1 Isoform in Its Ouabain-Bound Conformation. Int J Mol Sci 2024; 25:12397. [PMID: 39596464 PMCID: PMC11594824 DOI: 10.3390/ijms252212397] [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: 10/17/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Na+,K+-ATPase is the active ion transport system that maintains the electrochemical gradients for Na+ and K+ across the plasma membrane of most animal cells. Na+,K+-ATPase is constituted by the association of two major subunits, a catalytic α and a glycosylated β subunit, both of which exist as different isoforms (in mammals known as α1, α2, α3, α4, β1, β2 and β3). Na+,K+-ATPase α and β isoforms assemble in different combinations to produce various isozymes with tissue specific expression and distinct biochemical properties. Na+,K+-ATPase α4β1 is only found in male germ cells of the testis and is mainly expressed in the sperm flagellum, where it plays a critical role in sperm motility and male fertility. Here, we report the molecular structure of Na+,K+-ATPase α4β1 at 2.37 Å resolution in the ouabain-bound state and in the presence of beryllium fluoride. Overall, Na+,K+-ATPase α4 structure exhibits the basic major domains of a P-Type ATPase, resembling Na+,K+-ATPase α1, but has differences specific to its distinct sequence. Dissimilarities include the site where the inhibitor ouabain binds. Molecular simulations indicate that glycosphingolipids can bind to a putative glycosphingolipid binding site, which could potentially modulate Na+,K+-ATPase α4 activity. This is the first experimental evidence for the structure of Na+,K+-ATPase α4β1. These data provide a template that will aid in better understanding the function Na+,K+-ATPase α4β1 and will be important for the design and development of compounds that can modulate Na+,K+-ATPase α4 activity for the purpose of improving male fertility or to achieve male contraception.
Collapse
Affiliation(s)
- Kazuhiro Abe
- Department of Chemistry, Faculty of Science, Hokkaido University, Hokkaido 060-0808, Japan
| | - Jeff McDermott
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, KS 66103, USA;
| | - Hridya Valia Madapally
- PhyLife: Physical Life Science, Department of Physics Chemistry and Pharmacy, University of Southern Denmark, 5230 Odense, Denmark; (H.V.M.); (H.K.)
| | - Parthiban Marimuthu
- Pharmaceutical Science Laboratory (PSL—Pharmacy) and Structural Bioinformatics Laboratory (SBL—Biochemistry), Faculty of Science and Engineering, Åbo Akademi University, FI-20520 Turku, Finland;
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai 602 105, India
| | | | - Christoph Gerle
- RIKEN SPring-8 Center, Kouto, Sayo-gun, Hyogo 679-5148, Japan; (C.C.G.); (C.G.)
| | - Hideki Shigematsu
- Japan Synchrotron Radiation Research Institute (JASRI), SPring-8, 1-1-1 Kouto, Sayo, Hyogo 679-5148, Japan;
| | - Himanshu Khandelia
- PhyLife: Physical Life Science, Department of Physics Chemistry and Pharmacy, University of Southern Denmark, 5230 Odense, Denmark; (H.V.M.); (H.K.)
| | - Gustavo Blanco
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, KS 66103, USA;
| |
Collapse
|
4
|
Yan T, Liu J. Transmembrane Ion Channels: From Natural to Artificial Systems. Angew Chem Int Ed Engl 2024:e202416200. [PMID: 39545394 DOI: 10.1002/anie.202416200] [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: 08/23/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/17/2024]
Abstract
Natural channel proteins allow the selective permeation of ions, water or other nutritious entities across bilayer membranes, facilitating various essential physiological functions in living systems. Inspired by nature, chemists endeavor to simulate the structural features and transport behaviors of channel proteins through biomimetic strategies. In this review, we start from introducing the inherent traits of channel proteins such as their crystal structures, functions and mechanisms. Subsequently, different kind of synthetic ion channels including their design principles, dynamic regulations and therapeutic applications were carefully reviewed. Finally, the potential challenges and opportunities in this research field were also carefully discussed. It is anticipated that this review could provide some inspiring ideas and future directions towards the construction of novel bionic ion channels with higher-level structures, properties, functions and practical applications.
Collapse
Affiliation(s)
- Tengfei Yan
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, P. R. China
| | - Junqiu Liu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, P. R. China
| |
Collapse
|
5
|
Palmgren M, Morth JP, Nissen P. TMEM94 cannot be called a P-type ATPase. Cell Calcium 2024; 123:102911. [PMID: 38879951 DOI: 10.1016/j.ceca.2024.102911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/18/2024]
Affiliation(s)
- Michael Palmgren
- Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark.
| | - Jens Preben Morth
- Department of Biotechnology & Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark.
| | - Poul Nissen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
6
|
Castro de Jesus L, Gonçalves-de-Albuquerque CF, Burth P. Onset of bipolar disorder by COVID-19: The roles of endogenous ouabain and the Na,K-ATPase. J Psychiatr Res 2024; 179:60-68. [PMID: 39260109 DOI: 10.1016/j.jpsychires.2024.08.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/27/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Bipolar Disorder (BD) is a psychiatric disorder marked by mood swings between manic and depressive episodes. The reduction in the Na,K-ATPase (NKA) enzyme activity and the inability of individuals with BD to produce endogenous ouabain (EO) at sufficient levels to stimulate this enzyme during stressful events are factors proposed for BD etiology. According to these hypotheses, reduction in NKA activity would result in altered neuronal resting potential, leading to BD symptoms. Recently, damage to the adrenals (EO synthesis site) in coronavirus disease (COVID-19) patients has been reported, however studies pointing to the pathophysiological mechanisms shared by these two diseases are scarce. Through a literature review, this study aims to correlate COVID-19 and BD, focusing on the role of NKA and EO to identify possible mechanisms for the worsening of BD due to COVID-19. The search in the PubMed database for the descriptors ("bipolar disorder" AND "Na,K-ATPase"), ("bipolar disorder" AND "endogenous ouabain"), ("covid-19" AND "bipolar disorder") and ("covid-19" AND "adrenal gland") resulted in 390 articles. The studies identified the adrenals as a vulnerable organ to SARS-CoV-2 infection. Cases of adrenal damage in patients with COVID-19 showing lower levels of adrenal hormones were reported. Cases of COVID-19 patients with symptoms of mania were reported worldwide. Given these results, we propose that adrenal cortical cell damage could lead to EO deficiency following neuronal NKA activity impairment, with small reductions in activity leading to mania and greater reductions leading to depression.
Collapse
Affiliation(s)
- Louise Castro de Jesus
- Laboratory of Enzymology and Cell Signaling, Department of Cellular and Molecular Biology, Institute of Biology, Universidade Federal Fluminense, Niterói, RJ, 24020-141, Brazil.
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, RJ, 21040-900, Brazil; Laboratory Immunopharmacology, Department of Physiological Sciences, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, RJ, 20211-010, Brazil.
| | - Patrícia Burth
- Laboratory of Enzymology and Cell Signaling, Department of Cellular and Molecular Biology, Institute of Biology, Universidade Federal Fluminense, Niterói, RJ, 24020-141, Brazil.
| |
Collapse
|
7
|
Moraes CM, Fabri LM, Garçon DP, Augusto A, Faria SC, McNamara JC, Leone FA. Kinetic properties of gill (Na +, K +)-ATPase in the Pacific whiteleg shrimp Penaeus vannamei (Decapoda, Penaeidae). Comp Biochem Physiol B Biochem Mol Biol 2024; 275:111038. [PMID: 39374865 DOI: 10.1016/j.cbpb.2024.111038] [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: 06/19/2024] [Revised: 09/21/2024] [Accepted: 10/02/2024] [Indexed: 10/09/2024]
Abstract
The whiteleg marine shrimp Penaeus vannamei, originally from the Eastern Pacific Ocean, now inhabits tropical waters across Asia and Central and Southern America. This benthic species exhibits rapid growth, wide salinity and temperature tolerance, and disease resistance. These physiological traits have led to extensive research on its osmoregulatory mechanisms, including next-generation sequencing, transcriptomic analyses, and lipidomic responses. In crustaceans, osmotic and ionic homeostasis is primarily maintained by the membrane-bound metalloenzyme (Na+, K+)-ATPase. However, little is known about how various ligands modulate this enzyme in P. vannamei. Here, we examined the kinetic characteristics of the gill (Na+, K+)-ATPase to get biochemical insights into its modulation. A prominent immunoreactive band of ~120 kDa, corresponding to the (Na+, K+)-ATPase alpha-subunit, was identified. The enzyme exhibited two ATP hydrolyzing sites with K0.5 = 0.0003 ± 0.00002 and 0.05 ± 0.003 mmol L-1 and was stimulated by low sodium ion concentrations. Potassium and ammonium ions also stimulated enzyme activity with similar K0.5 values of 0.08 ± 0.004 and 0.06 ± 0.003 mmol L-1, respectively. Ouabain inhibition profile suggested a single enzyme isoform with a KI value of 2.10 ± 0.16 mmol L-1. Our findings showed significant kinetic differences in the (Na+, K+)-ATPase in Penaeus vannamei compared to marine and freshwater crustaceans. We expect our results to enhance understanding of the modulation of gill (Na+, K+)-ATPase in Penaeus vannamei and to provide a valuable tool for studying the shrimp's biochemical acclimation to varying salinity conditions.
Collapse
Affiliation(s)
- Cintya M Moraes
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Brazil
| | - Leonardo M Fabri
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Brazil
| | | | - Alessandra Augusto
- lnstituto de Biociências, Universidade Estadual Paulista, Campus Experimental do Litoral Paulista, São Vicente, Brazil
| | - Samuel C Faria
- Centro de Biologia Marinha, Universidade de São Paulo, São Sebastião, Brazil
| | - John C McNamara
- Departamento de Biologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil; Centro de Biologia Marinha, Universidade de São Paulo, São Sebastião, Brazil
| | - Francisco A Leone
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
8
|
Fabri LM, Moraes CM, Garçon DP, McNamara JC, Faria SC, Leone FA. Primary amino acid sequences of decapod (Na +, K +)-ATPase provide evolutionary insights into osmoregulatory mechanisms. Comp Biochem Physiol A Mol Integr Physiol 2024; 296:111696. [PMID: 39004301 DOI: 10.1016/j.cbpa.2024.111696] [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: 04/29/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
Decapod Crustacea exhibit a marine origin, but many taxa have occupied environments ranging from brackish to fresh water and terrestrial habitats, overcoming their inherent osmotic challenges. Osmotic and ionic regulation is achieved by the gill epithelia, driven by two active ATP-hydrolyzing ion transporters, the basal (Na+, K+)-ATPase and the apical V(H+)-ATPase. The kinetic characteristic of gill (Na+, K+)-ATPase and the mRNA expression of its α subunit have been widely studied in various decapod species under different salinity challenges. However, the evolution of the primary structure has not been explored, especially considering the functional modifications associated with decapod phylogeny. Here, we proposed a model for the topology of the decapod α subunit, identifying the sites and motifs involved in its function and regulation, as well as the patterns of its evolution assuming a decapod phylogeny. We also examined both the amino acid substitutions and their functional implications within the context of biochemical and physiological adaptation. The α-subunit of decapod crustaceans shows greater conservation (∼94% identity) compared to the β-subunit (∼40%). While the binding sites for ATP and modulators are conserved in the decapod enzyme, the residues involved in the α-β interaction are only partially conserved. In the phylogenetic context of the complete sequence of (Na+, K+)-ATPase α-subunit, most substitutions appear to be characteristic of the entire group, with specific changes for different subgroups, especially among brachyuran crabs. Interestingly, there was no consistent separation of α-subunit partial sequences related to habitat, suggesting that the convergent evolution for freshwater or terrestrial modes of life is not correlated with similar changes in the enzyme's primary amino acid sequence.
Collapse
Affiliation(s)
- Leonardo M Fabri
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Cintya M Moraes
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - John C McNamara
- Departamento de Biologia Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil; Centro de Biologia Marinha, Universidade de São Paulo, São Sebastião, São Paulo, Brazil
| | - Samuel C Faria
- Centro de Biologia Marinha, Universidade de São Paulo, São Sebastião, São Paulo, Brazil
| | - Francisco A Leone
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
9
|
Haj Mohammad Hassani B, Malekzadeh K. The lethal homozygous variant in the ATP1A2 gene is associated with FARIMPD syndrome phenotypes in newborns. Neurogenetics 2024; 25:417-424. [PMID: 39046620 DOI: 10.1007/s10048-024-00775-7] [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: 05/31/2024] [Accepted: 07/19/2024] [Indexed: 07/25/2024]
Abstract
FARIMPD (Fetal akinesia, respiratory insufficiency, microcephaly, polymicrogyria, and dysmorphic facies) syndrome is a severe condition caused by ATP1A2 gene variants. The syndrome's novelty and rarity have limited its clinical and molecular knowledge. This research tries to provide new insight by investigating the cause of the early deaths due to FARIMPD syndrome in a particular family and reviewing previous studies. DNA and RNA were extracted from the blood samples of newborns and their parents, followed by whole exome sequencing and segregation analysis. A pathogenic homozygous nonsense variant (c.1234C > T: p.Arg412*) in the ATP1A2 gene was found in newborns. This variant is reported as homozygous for the first time. The migraine symptoms were the result of the heterozygous state of this particular variant, which supported the dominant inheritance pattern of this disease. Real-time PCR was used to analyze ATP1A2 gene expression in the newborns compared to parents and control subjects. The expression analysis also showed significant mRNA degradation in the newborns compared to heterozygous and healthy individuals, due to Nonsense-mediated mRNA Decay phenomena. Our study describes an ATP1A2 nonsense variant (c.1234C > T) that appears compatible with infant survival in the heterozygous and compound heterozygous states but is lethal in the homozygous state.
Collapse
Affiliation(s)
- Behzad Haj Mohammad Hassani
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Kianoosh Malekzadeh
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| |
Collapse
|
10
|
Cao DQ, Lei SC, Liu H, Jin Y, Wu YF, Cui Y, Wu R. Reverse Solute Diffusion Enhances Sludge Dewatering in Dead-End Forward Osmosis. MEMBRANES 2024; 14:196. [PMID: 39330537 PMCID: PMC11433884 DOI: 10.3390/membranes14090196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Wastewater treatment plants produce high quantities of excess sludge. However, traditional sludge dewatering technology has high energy consumption and occupies a large area. Dead-end forward osmosis (DEFO) is an efficient and energy-saving deep dewatering technology for sludge. In this study, the reverse osmosis of salt ions in the draw solution was used to change the sludge cake structure and further reduce its moisture content in cake by releasing the bound water in cell. Three salts, NaCl, KCl, and CaCl2, were added to the excess sludge feed solution to explore the roles of the reverse osmosis of draw solutes in DEFO. When the added quantities of NaCl and CaCl2 were 15 and 10 mM, respectively, the moisture content of the sludge after dewatering decreased from 98.1% to 79.7% and 67.3%, respectively. However, KCl did not improve the sludge dewatering performance because of the "high K and low Na" phenomenon in biological cells. The water flux increased significantly for the binary draw solute involving NaCl and CaCl2 compared to the single draw solute. The extracellular polymer substances in the sludge changed the structure of the filter cake to improve the formation of water channels and decrease osmosis resistance, resulting in an increase in sludge dewatering efficiency. These findings provide support for improving the sludge dewatering performance of DEFO.
Collapse
Affiliation(s)
- Da-Qi Cao
- Sino-Dutch R&D Centre for Future Wastewater Treatment Technologies/Key Laboratory of Urban Stormwater System and Water Environment, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
- Department of Statistics and Probability, Michigan State University, East Lansing, MI 48824, USA
- Yau Mathematical Sciences Center, Tsinghua University, Beijing 100084, China
| | - Shi-Cheng Lei
- Sino-Dutch R&D Centre for Future Wastewater Treatment Technologies/Key Laboratory of Urban Stormwater System and Water Environment, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
| | - Hui Liu
- Sino-Dutch R&D Centre for Future Wastewater Treatment Technologies/Key Laboratory of Urban Stormwater System and Water Environment, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
| | - Yan Jin
- Sino-Dutch R&D Centre for Future Wastewater Treatment Technologies/Key Laboratory of Urban Stormwater System and Water Environment, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
| | - Yun-Feng Wu
- Sino-Dutch R&D Centre for Future Wastewater Treatment Technologies/Key Laboratory of Urban Stormwater System and Water Environment, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
| | - Yuehua Cui
- Department of Statistics and Probability, Michigan State University, East Lansing, MI 48824, USA
| | - Rongling Wu
- Yau Mathematical Sciences Center, Tsinghua University, Beijing 100084, China
- Beijing Institute of Mathematical Sciences and Applications, Beijing 101408, China
| |
Collapse
|
11
|
Sultana OF, Bandaru M, Islam MA, Reddy PH. Unraveling the complexity of human brain: Structure, function in healthy and disease states. Ageing Res Rev 2024; 100:102414. [PMID: 39002647 PMCID: PMC11384519 DOI: 10.1016/j.arr.2024.102414] [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: 05/23/2024] [Revised: 06/29/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
The human brain stands as an intricate organ, embodying a nexus of structure, function, development, and diversity. This review delves into the multifaceted landscape of the brain, spanning its anatomical intricacies, diverse functional capacities, dynamic developmental trajectories, and inherent variability across individuals. The dynamic process of brain development, from early embryonic stages to adulthood, highlights the nuanced changes that occur throughout the lifespan. The brain, a remarkably complex organ, is composed of various anatomical regions, each contributing uniquely to its overall functionality. Through an exploration of neuroanatomy, neurophysiology, and electrophysiology, this review elucidates how different brain structures interact to support a wide array of cognitive processes, sensory perception, motor control, and emotional regulation. Moreover, it addresses the impact of age, sex, and ethnic background on brain structure and function, and gender differences profoundly influence the onset, progression, and manifestation of brain disorders shaped by genetic, hormonal, environmental, and social factors. Delving into the complexities of the human brain, it investigates how variations in anatomical configuration correspond to diverse functional capacities across individuals. Furthermore, it examines the impact of neurodegenerative diseases on the structural and functional integrity of the brain. Specifically, our article explores the pathological processes underlying neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Huntington's diseases, shedding light on the structural alterations and functional impairments that accompany these conditions. We will also explore the current research trends in neurodegenerative diseases and identify the existing gaps in the literature. Overall, this article deepens our understanding of the fundamental principles governing brain structure and function and paves the way for a deeper understanding of individual differences and tailored approaches in neuroscience and clinical practice-additionally, a comprehensive understanding of structural and functional changes that manifest in neurodegenerative diseases.
Collapse
Affiliation(s)
- Omme Fatema Sultana
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Madhuri Bandaru
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA 5. Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
12
|
Vidović A, Dolinar K, Chibalin AV, Pirkmajer S. AMPK and glucose deprivation exert an isoform-specific effect on the expression of Na +,K +-ATPase subunits in cultured myotubes. J Muscle Res Cell Motil 2024; 45:139-154. [PMID: 38709429 DOI: 10.1007/s10974-024-09673-9] [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: 01/31/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
In skeletal muscle, Na+,K+-ATPase (NKA), a heterodimeric (α/β) P-type ATPase, has an essential role in maintenance of Na+ and K+ homeostasis, excitability, and contractility. AMP-activated protein kinase (AMPK), an energy sensor, increases the membrane abundance and activity of NKA in L6 myotubes, but its potential role in regulation of NKA content in skeletal muscle, which determines maximum capacity for Na+ and K+ transport, has not been clearly delineated. We examined whether energy stress and/or AMPK affect expression of NKA subunits in rat L6 and primary human myotubes. Energy stress, induced by glucose deprivation, increased protein content of NKAα1 and NKAα2 in L6 myotubes, while decreasing the content of NKAα1 in human myotubes. Pharmacological AMPK activators (AICAR, A-769662, and diflunisal) modulated expression of NKA subunits, but their effects only partially mimicked those that occurred in response to glucose deprivation, indicating that AMPK does not mediate all effects of energy stress on NKA expression. Gene silencing of AMPKα1/α2 increased protein levels of NKAα1 in L6 myotubes and NKAα1 mRNA levels in human myotubes, while decreasing NKAα2 protein levels in L6 myotubes. Collectively, our results suggest a role for energy stress and AMPK in modulation of NKA expression in skeletal muscle. However, their modulatory effects were not conserved between L6 myotubes and primary human myotubes, which suggests that coupling between energy stress, AMPK, and regulation of NKA expression in vitro depends on skeletal muscle cell model.
Collapse
Affiliation(s)
- Anja Vidović
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | - Klemen Dolinar
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
- National Research Tomsk State University, Tomsk, Russia
| | - Sergej Pirkmajer
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
13
|
Arystarkhova E, Sweadner KJ. Na,K-ATPase Expression Can Be Limited Post-Transcriptionally: A Test of the Role of the Beta Subunit, and a Review of Evidence. Int J Mol Sci 2024; 25:7414. [PMID: 39000521 PMCID: PMC11242325 DOI: 10.3390/ijms25137414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
The Na,K-ATPase is an α-β heterodimer. It is well known that the Na,K-ATPase β subunit is required for the biosynthesis and trafficking of the α subunit to the plasma membrane. During investigation of properties of human ATP1A3 mutations in 293 cells, we observed a reciprocal loss of endogenous ATP1A1 when expressing ATP1A3. Scattered reports going back as far as 1991 have shown that experimental expression of one subunit can result in reduction in another, suggesting that the total amount is strictly limited. It seems logical that either α or β subunit should be rate-limiting for assembly and functional expression. Here, we present evidence that neither α nor β may be limiting and that there is another level of control that limits the amount of Na,K-ATPase to physiological levels. We propose that α subunits compete for something specific, like a private chaperone, required to finalize their biosynthesis or to prevent their degradation in the endoplasmic reticulum.
Collapse
Affiliation(s)
- Elena Arystarkhova
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Kathleen J. Sweadner
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
14
|
Hammes UZ, Pedersen BP. Structure and Function of Auxin Transporters. ANNUAL REVIEW OF PLANT BIOLOGY 2024; 75:185-209. [PMID: 38211951 DOI: 10.1146/annurev-arplant-070523-034109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Auxins, a group of central hormones in plant growth and development, are transported by a diverse range of transporters with distinct biochemical and structural properties. This review summarizes the current knowledge on all known auxin transporters with respect to their biochemical and biophysical properties and the methods used to characterize them. In particular, we focus on the recent advances that were made concerning the PIN-FORMED family of auxin exporters. Insights derived from solving their structures have improved our understanding of the auxin export process, and we discuss the current state of the art on PIN-mediated auxin transport, including the use of biophysical methods to examine their properties. Understanding the mechanisms of auxin transport is crucial for understanding plant growth and development, as well as for the development of more effective strategies for crop production and plant biotechnology.
Collapse
Affiliation(s)
- Ulrich Z Hammes
- School of Life Sciences, Plant Systems Biology, Technical University of Munich, Freising, Germany;
| | | |
Collapse
|
15
|
Hodeify R, Kreydiyyeh S, Zaid LMJ. Identified and potential internalization signals involved in trafficking and regulation of Na +/K + ATPase activity. Mol Cell Biochem 2024; 479:1583-1598. [PMID: 37634170 PMCID: PMC11254989 DOI: 10.1007/s11010-023-04831-y] [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: 05/04/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023]
Abstract
The sodium-potassium pump (NKA) or Na+/K+ ATPase consumes around 30-40% of the total energy expenditure of the animal cell on the generation of the sodium and potassium electrochemical gradients that regulate various electrolyte and nutrient transport processes. The vital role of this protein entails proper spatial and temporal regulation of its activity through modulatory mechanisms involving its expression, localization, enzymatic activity, and protein-protein interactions. The residence of the NKA at the plasma membrane is compulsory for its action as an antiporter. Despite the huge body of literature reporting on its trafficking between the cell membrane and intracellular compartments, the mechanisms controlling the trafficking process are by far the least understood. Among the molecular determinants of the plasma membrane proteins trafficking are intrinsic sequence-based endocytic motifs. In this review, we (i) summarize previous reports linking the regulation of Na+/K+ ATPase trafficking and/or plasma membrane residence to its activity, with particular emphasis on the endocytic signals in the Na+/K+ ATPase alpha-subunit, (ii) map additional potential internalization signals within Na+/K+ ATPase catalytic alpha-subunit, based on canonical and noncanonical endocytic motifs reported in the literature, (iii) pinpoint known and potential phosphorylation sites associated with NKA trafficking, (iv) highlight our recent studies on Na+/K+ ATPase trafficking and PGE2-mediated Na+/K+ ATPase modulation in intestine, liver, and kidney cells.
Collapse
Affiliation(s)
- Rawad Hodeify
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, United Arab Emirates.
| | - Sawsan Kreydiyyeh
- Department of Biology, Faculty of Arts & Sciences, American University of Beirut, Beirut, Lebanon
| | - Leen Mohammad Jamal Zaid
- Department of Biotechnology, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, United Arab Emirates
| |
Collapse
|
16
|
Zhang R, Xiang Y, Yang Y. Passing Behavior of Oligonucleotides through a Stacked DNA Nanochannel with Featured Path Design. J Am Chem Soc 2024; 146:17122-17130. [PMID: 38861703 DOI: 10.1021/jacs.4c02734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
DNA nanotechnology has emerged as a useful tool for constructing artificial channels penetrating the lipid bilayer. In this work, we introduce a stacked DNA origami nanochannel device characterized by a width-variable pathway, consisting of narrow entrance and exit channels coupled with a wide, modifiable lumen. This design modulates the translocation behavior of oligonucleotides, revealing distinct stages of signal patterns in the recorded current traces. The observed prolonged dwell times indicate oligonucleotide retention, specifically due to the transition from the wide lumen to the narrower exit channel, while variations in current recovery between events suggested intermediate channel states between conducting and blocking. Further, by incorporating sequence-specific overhangs within the channel lumen, we achieved unique asymmetric current profiles during ATP aptamer translocation events. Featured stages also highlighted the aptamer binding dynamics and ATP-induced release. The distinguished oligonucleotide passing behaviors afforded by the stacked DNA origami channel with interior decoration demonstrated the strategic and profitable attempts at DNA nanochannel engineering for nanodevice development and applications.
Collapse
Affiliation(s)
- Rui Zhang
- State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yaozu Xiang
- State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yang Yang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
17
|
Chkadua G, Nozadze E, Tsakadze L, Shioshvili L, Arutinova N, Leladze M, Dzneladze S, Javakhishvili M, Jariashvili T, Petriashvili E. The effect of cytochrome c on Na,K-ATPase. J Bioenerg Biomembr 2024; 56:221-234. [PMID: 38517564 DOI: 10.1007/s10863-024-10012-3] [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: 09/05/2023] [Accepted: 03/09/2024] [Indexed: 03/24/2024]
Abstract
Na,K-ATPase is a crucial enzyme responsible for maintaining Na+, K+-gradients across the cell membrane, which is essential for numerous physiological processes within various organs and tissues. Due to its significance in cellular physiology, inhibiting Na,K-ATPase can have profound physiological consequences. This characteristic makes it a target for various pharmacological applications, and drugs that modulate the pump's activity are thus used in the treatment of several medical conditions. Cytochrome c (Cytc) is a protein with dual functions in the cell. In the mitochondria, it is essential for ATP synthesis and energy production. However, in response to apoptotic stimuli, it is released into the cytosol, where it triggers programmed cell death through the intrinsic apoptosis pathway. Aside from its role in canonical intrinsic apoptosis, Cytc also plays additional roles. For instance, Cytc participates in certain non-apoptotic functions -those which are less well-understood in comparison to its role in apoptosis. Within this in vitro study, we have shown the impact of Cytc on Na,K-ATPase for the first time. Cytc has a biphasic action on Na,K-ATPase, with activation at low concentrations (0.06 ng/ml; 6 ng/ml) and inhibition at high concentration (120 ng/ml). Cytc moreover displays isoform/subunit specificity and regulates the Na+ form of the enzyme, while having no effect on the activity or kinetic parameters of the K+-dependent form of the enzyme. Changing the affinity of p-chloromercuribenzoic acid (PCMB) by Cytc is therefore both a required and sufficient condition for confirming that PCMB and Cytc share the same target, namely the thiol groups of cysteine in Na,K-ATPase.
Collapse
Affiliation(s)
- Gvantsa Chkadua
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia.
| | - Eka Nozadze
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | - Leila Tsakadze
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | - Lia Shioshvili
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | - Nana Arutinova
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | - Marine Leladze
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | - Sopio Dzneladze
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | - Maia Javakhishvili
- Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Str, 0160, Tbilisi, Georgia
| | | | | |
Collapse
|
18
|
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
|
19
|
Dmitriev OY, Patry J. Structure and mechanism of the human copper transporting ATPases: Fitting the pieces into a moving puzzle. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184306. [PMID: 38408697 DOI: 10.1016/j.bbamem.2024.184306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/07/2024] [Accepted: 02/18/2024] [Indexed: 02/28/2024]
Abstract
Human copper transporters ATP7B and ATP7A deliver copper to biosynthetic pathways and maintain copper homeostasis in the cell. These enzymes combine several challenges for structural biology because they are large low abundance membrane proteins with many highly mobile domains and long disordered loops. No method has yet succeeded in solving the structure of the complete fully functional protein. Still, X-ray crystallography, Cryo-EM and NMR helped to piece together a structure based model of the enzyme activity and regulation by copper. We review the structures of ATP7B and ATP7A with an emphasis on the mechanistic insights into the unique aspects of the transport function and regulation of the human copper ATPases that have emerged from more than twenty years of research.
Collapse
Affiliation(s)
- Oleg Y Dmitriev
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada.
| | - Jaala Patry
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
20
|
Pietrobon D, Conti F. Astrocytic Na +, K + ATPases in physiology and pathophysiology. Cell Calcium 2024; 118:102851. [PMID: 38308916 DOI: 10.1016/j.ceca.2024.102851] [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: 12/13/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/05/2024]
Abstract
The Na+, K+ ATPases play a fundamental role in the homeostatic functions of astrocytes. After a brief historic prologue and discussion of the subunit composition and localization of the astrocytic Na+, K+ ATPases, the review focuses on the role of the astrocytic Na+, K+ pumps in extracellular K+ and glutamate homeostasis, intracellular Na+ and Ca2+ homeostasis and signaling, regulation of synaptic transmission and neurometabolic coupling between astrocytes and neurons. Loss-of-function mutations in the gene encoding the astrocytic α2 Na+, K+ ATPase cause a rare monogenic form of migraine with aura (familial hemiplegic migraine type 2). On the other hand, the α2 Na+, K+ ATPase is upregulated in spinal cord and brain samples from amyotrophic lateral sclerosis and Alzheimer disease patients, respectively. In the last part, the review focuses on i) the migraine relevant phenotypes shown by familial hemiplegic migraine type 2 knock-in mice with 50 % reduced expression of the astrocytic α2 Na+, K+ ATPase and the insights into the pathophysiology of migraine obtained from these genetic mouse models, and ii) the evidence that upregulation of the astrocytic α2 Na+, K+ ATPase in mouse models of amyotrophic lateral sclerosis and Alzheimer disease promotes neuroinflammation and contributes to progressive neurodegeneration.
Collapse
Affiliation(s)
- Daniela Pietrobon
- Department of Biomedical Sciences and Padova Neuroscience Center (PNC), University of Padova, Padova 35131, Italy.
| | - Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy.
| |
Collapse
|
21
|
McKenna MJ, Renaud JM, Ørtenblad N, Overgaard K. A century of exercise physiology: effects of muscle contraction and exercise on skeletal muscle Na +,K +-ATPase, Na + and K + ions, and on plasma K + concentration-historical developments. Eur J Appl Physiol 2024; 124:681-751. [PMID: 38206444 PMCID: PMC10879387 DOI: 10.1007/s00421-023-05335-9] [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: 02/02/2023] [Accepted: 09/27/2023] [Indexed: 01/12/2024]
Abstract
This historical review traces key discoveries regarding K+ and Na+ ions in skeletal muscle at rest and with exercise, including contents and concentrations, Na+,K+-ATPase (NKA) and exercise effects on plasma [K+] in humans. Following initial measures in 1896 of muscle contents in various species, including humans, electrical stimulation of animal muscle showed K+ loss and gains in Na+, Cl- and H20, then subsequently bidirectional muscle K+ and Na+ fluxes. After NKA discovery in 1957, methods were developed to quantify muscle NKA activity via rates of ATP hydrolysis, Na+/K+ radioisotope fluxes, [3H]-ouabain binding and phosphatase activity. Since then, it became clear that NKA plays a central role in Na+/K+ homeostasis and that NKA content and activity are regulated by muscle contractions and numerous hormones. During intense exercise in humans, muscle intracellular [K+] falls by 21 mM (range - 13 to - 39 mM), interstitial [K+] increases to 12-13 mM, and plasma [K+] rises to 6-8 mM, whilst post-exercise plasma [K+] falls rapidly, reflecting increased muscle NKA activity. Contractions were shown to increase NKA activity in proportion to activation frequency in animal intact muscle preparations. In human muscle, [3H]-ouabain-binding content fully quantifies NKA content, whilst the method mainly detects α2 isoforms in rats. Acute or chronic exercise affects human muscle K+, NKA content, activity, isoforms and phospholemman (FXYD1). Numerous hormones, pharmacological and dietary interventions, altered acid-base or redox states, exercise training and physical inactivity modulate plasma [K+] during exercise. Finally, historical research approaches largely excluded female participants and typically used very small sample sizes.
Collapse
Affiliation(s)
- Michael J McKenna
- Institute for Health and Sport, Victoria University, Melbourne, VIC, 8001, Australia.
- College of Physical Education, Southwest University, Chongqing, China.
- College of Sport Science, Zhuhai College of Science and Technology, Zhuhai, China.
| | - Jean-Marc Renaud
- Department of Cellular and Molecular Medicine, Neuromuscular Research Center, University of Ottawa, Ottawa, ON, Canada
| | - Niels Ørtenblad
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Kristian Overgaard
- Exercise Biology, Department of Public Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
22
|
Parker KS, El N, Buldo EC, MacCormack TJ. Mechanisms of PVP-functionalized silver nanoparticle toxicity in fish: Intravascular exposure disrupts cardiac pacemaker function and inhibits Na +/K +-ATPase activity in heart, but not gill. Comp Biochem Physiol C Toxicol Pharmacol 2024; 277:109837. [PMID: 38218567 DOI: 10.1016/j.cbpc.2024.109837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
Polyvinylpyrrolidone-functionalized silver nanoparticles (nAgPVP) are popular in consumer products for their colloidal stability and antimicrobial activity. Whole lake additions of nAgPVP cause long term, ecosystem-scale changes in fish populations but the mechanisms underlying this effect are unclear. We have previously shown that in fish, nAgPVP impairs cardiac contractility and Na+/K+-ATPase (NKA) activity in vitro, raising the possibility that heart dysfunction could underlie population-level exposure effects. The goal of this study was to determine if nAgPVP influences the control of heart rate (fh), blood pressure, or cardiac NKA activity in vivo. First, a dose-response curve for the effects of 5 nm nAgPVP on contractility was completed on isometrically contracting ventricular muscle preparations from Arctic char (Salvelinus alpinus) and showed that force production was lowest at 500 μg L-1 and maximum pacing frequency increased with nAgPVP concentration. Stroke volume, cardiac output, and power output were maintained in isolated working heart preparations from brook char (Salvelinus fontinalis) exposed to 700 μg L-1 nAgPVP. Both fh and blood pressure were elevated after 24 h in brook char injected with 700 μg kg body mass-1 nAgPVP and fh was insensitive to modulation with blockers of β-adrenergic and muscarinic cholinergic receptors. Na+/K+-ATPase activity was significantly lower in heart, but not gill of nAgPVP injected fish. The results indicate that nAgPVP influences cardiac function in vivo by disrupting regulation of the pacemaker and cardiomyocyte ionoregulation. Impaired fh regulation may prevent fish from appropriately responding to environmental or social stressors and affect their ability to survive.
Collapse
Affiliation(s)
- K S Parker
- Department of Chemistry and Biochemistry, Mount Allison University, Sackville, NB, Canada
| | - N El
- Department of Chemistry and Biochemistry, Mount Allison University, Sackville, NB, Canada
| | - E C Buldo
- Department of Chemistry and Biochemistry, Mount Allison University, Sackville, NB, Canada
| | - T J MacCormack
- Department of Chemistry and Biochemistry, Mount Allison University, Sackville, NB, Canada.
| |
Collapse
|
23
|
Savoj R, Agnew H, Zhou R, Paesani F. Molecular Insights into the Influence of Ions on the Water Structure. I. Alkali Metal Ions in Solution. J Phys Chem B 2024; 128:1953-1962. [PMID: 38373140 DOI: 10.1021/acs.jpcb.3c08150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
In this study, we explore the impact of alkali metal ions (Li+, Na+, K+, Rb+, and Cs+) on the hydration structure of water using molecular dynamics simulations carried out with MB-nrg potential energy functions (PEFs). Our analyses include radial distribution functions, coordination numbers, dipole moments, and infrared spectra of water molecules, calculated as a function of solvation shells. The results collectively indicate a highly local influence of all of the alkali metal ions on the hydrogen-bond network established by the surrounding water molecules, with the smallest and most densely charged Li+ ion exerting the most pronounced effect. Remarkably, the MB-nrg PEFs demonstrate excellent agreement with available experimental data for the position and size of the first solvation shells, underscoring their potential as predictive models for realistic simulations of ionic aqueous solutions across various thermodynamic conditions and environments.
Collapse
Affiliation(s)
- Roya Savoj
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
| | - Henry Agnew
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
| | - Ruihan Zhou
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
| | - Francesco Paesani
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
- Materials Science and Engineering, University of California San Diego, La Jolla, California 92093, United States
- Halicioğlu Data Science Institute, University of California San Diego, La Jolla, California 92093, United States
- San Diego Supercomputer Center, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
24
|
Zeinert R, Zhou F, Franco P, Zöller J, Lessen HJ, Aravind L, Langer JD, Sodt AJ, Storz G, Matthies D. Magnesium Transporter MgtA revealed as a Dimeric P-type ATPase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582502. [PMID: 38464158 PMCID: PMC10925321 DOI: 10.1101/2024.02.28.582502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Magnesium (Mg2+) uptake systems are present in all domains of life given the vital role of this ion. Bacteria acquire Mg2+ via conserved Mg2+ channels and transporters. The transporters are required for growth when Mg2+ is limiting or during bacterial pathogenesis, but, despite their significance, there are no known structures for these transporters. Here we report the first structure of the Mg2+ transporter MgtA solved by single particle cryo-electron microscopy (cryo-EM). Using mild membrane extraction, we obtained high resolution structures of both a homodimeric form (2.9 Å), the first for a P-type ATPase, and a monomeric form (3.6 Å). Each monomer unit of MgtA displays a structural architecture that is similar to other P-type ATPases with a transmembrane domain and two soluble domains. The dimer interface consists of contacts between residues in adjacent soluble nucleotide binding and phosphotransfer regions of the haloacid dehalogenase (HAD) domain. We suggest oligomerization is a conserved structural feature of the diverse family of P-type ATPase transporters. The ATP binding site and conformational dynamics upon nucleotide binding to MgtA were characterized using a combination of cryo-EM, molecular dynamics simulations, hydrogen-deuterium exchange mass spectrometry, and mutagenesis. Our structure also revealed a Mg2+ ion in the transmembrane segments, which, when combined with sequence conservation and mutagenesis studies, allowed us to propose a model for Mg2+ transport across the lipid bilayer. Finally, our work revealed the N-terminal domain structure and cytoplasmic Mg2+ binding sites, which have implications for related P-type ATPases defective in human disease.
Collapse
Affiliation(s)
- Rilee Zeinert
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda MD 20892, USA
| | - Fei Zhou
- Unit on Structural Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda MD 20892, USA
| | - Pedro Franco
- Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Jonathan Zöller
- Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Henry J. Lessen
- Unit on Membrane Chemical Physics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda MD 20892, USA
| | - L. Aravind
- National Center for Biotechnology Information, National Institutes of Health, Bethesda MD 20892, USA
| | - Julian D. Langer
- Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Alexander J. Sodt
- Unit on Membrane Chemical Physics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda MD 20892, USA
| | - Gisela Storz
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda MD 20892, USA
| | - Doreen Matthies
- Unit on Structural Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda MD 20892, USA
| |
Collapse
|
25
|
Fan L, Su W, Zhang X, Yang S, Zhu Y, Liu X. Self-assembly of sophorolipid and eugenol into stable nanoemulsions for synergetic antibacterial properties through alerting membrane integrity. Colloids Surf B Biointerfaces 2024; 234:113749. [PMID: 38241893 DOI: 10.1016/j.colsurfb.2024.113749] [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: 11/15/2023] [Revised: 12/27/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024]
Abstract
Exploring the natural, safe, and effective antimicrobial is one of the preferable ways to control foodborne bacteria. In this work, novel oil-in-water nanoemulsions were formulated with sophorolipids and eugenol without any co-surfactant using a self-assembling strategy. These nanoemulsions showed high stability with sizes less than 200 nm when exposure to low concentrations of salt ions, various pH values (5.0, 7.0, 10.0), storage temperature and time. The synergistic antibacterial effects against both Gram-negative Escherichia coli and Gram-positive Bacillus cereus were determined with a minimum inhibitory concentration (MIC) value of 0.5 mg/mL and 0.125 mg/mL, respectively. Further microscopy (SEM, TEM, LCSM) examination and ATP/Na+-K+-ATPase assay results showed that the morphological changes, intensive cell membrane permeability, leakage of ATP, and decreased Na+-K+-ATPase contributed to the antibacterial effects. Moreover, the bonding mechanism between nanoemulsions and cell membranes were further evaluated by FTIR and ITC using a DPPC vesicle model, which demonstrated that the nanoemulsions adsorbed on the surface of bilayer, interacted with the hydrophobic chains of DPPC membrane mainly through the hydrophobic interaction, and altered the structural integrity of the lipid bilayer. These results not only provide a facile green strategy for fabricating stable nanoemulsions, but also highlight a new perspective for stabilizing essential oils for their widely application in food industry.
Collapse
Affiliation(s)
- Linlin Fan
- Institute of Agro-product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; Key Laboratory of Cold Chain Logistics Technology for Agro-product, Ministry of Agriculture and Rural Affairs, Nanjing 210014, China
| | - Wei Su
- Institute of Agro-product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Xiaoqian Zhang
- Institute of Agro-product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Suqun Yang
- Institute of Agro-product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Yongsheng Zhu
- Institute of Agro-product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; Key Laboratory of Cold Chain Logistics Technology for Agro-product, Ministry of Agriculture and Rural Affairs, Nanjing 210014, China
| | - Xiaoli Liu
- Institute of Agro-product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; Key Laboratory of Cold Chain Logistics Technology for Agro-product, Ministry of Agriculture and Rural Affairs, Nanjing 210014, China.
| |
Collapse
|
26
|
Nielsen HN, Holm R, Sweazey R, Andersen JP, Artigas P, Vilsen B. Na +,K +-ATPase with Disrupted Na + Binding Sites I and III Binds Na + with Increased Affinity at Site II and Undergoes Na +-Activated Phosphorylation with ATP. Biomolecules 2024; 14:135. [PMID: 38275764 PMCID: PMC10812997 DOI: 10.3390/biom14010135] [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: 12/12/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/27/2024] Open
Abstract
Na+,K+-ATPase actively extrudes three cytoplasmic Na+ ions in exchange for two extracellular K+ ions for each ATP hydrolyzed. The atomic structure with bound Na+ identifies three Na+ sites, named I, II, and III. It has been proposed that site III is the first to be occupied and site II last, when Na+ binds from the cytoplasmic side. It is usually assumed that the occupation of all three Na+ sites is obligatory for the activation of phosphoryl transfer from ATP. To obtain more insight into the individual roles of the ion-binding sites, we have analyzed a series of seven mutants with substitution of the critical ion-binding residue Ser777, which is a shared ligand between Na+ sites I and III. Surprisingly, mutants with large and bulky substituents expected to prevent or profoundly disturb Na+ access to sites I and III retain the ability to form a phosphoenzyme from ATP, even with increased apparent Na+ affinity. This indicates that Na+ binding solely at site II is sufficient to promote phosphorylation. These mutations appear to lock the membrane sector into an E1-like configuration, allowing Na+ but not K+ to bind at site II, while the cytoplasmic sector undergoes conformational changes uncoupled from the membrane sector.
Collapse
Affiliation(s)
- Hang N. Nielsen
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| | - Rikke Holm
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| | - Ryan Sweazey
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA (P.A.)
| | | | - Pablo Artigas
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA (P.A.)
| | - Bente Vilsen
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| |
Collapse
|
27
|
Fairuz S, Ang CW, Mraiche F, Goh JK. Current Targets and Future Directions of Positive Inotropes for Heart Failure. Curr Med Chem 2024; 31:6971-6991. [PMID: 37909442 DOI: 10.2174/0109298673262360231018193823] [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: 05/23/2023] [Revised: 06/23/2023] [Accepted: 09/11/2023] [Indexed: 11/03/2023]
Abstract
While a congestive heart failure patient will ultimately need an assist device or even a replacement heart as the disease progresses, not every patient is qualified for such advanced therapy. Such patients awaiting better circulatory support benefit from positive inotropes in the meantime as palliative care. These agents are often prescribed in patients with acute decompensated heart failure, with reduced left ventricular ejection fraction and symptoms of organ dysfunction. Although positive inotropes, for example, digoxin, dobutamine, milrinone, levosimendan, etc., are successfully marketed and in use, a lot of their adverse effects, like arrhythmias, hypotension, and even sudden cardiac death, are rather encouraging further research on the development of novel positive inotropes. This review has investigated the molecular mechanisms of some of these adverse effects in terms of the proteins they target, followed by research on newer targets. Studies from 2013-2023 that have reported new small molecules with positive inotropic effects have been revisited in order to determine the progress made so far in drug discovery.
Collapse
Affiliation(s)
- Shadreen Fairuz
- School of Science, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Chee Wei Ang
- School of Science, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Fatima Mraiche
- Department of Pharmacology, University of Alberta, 116 St & 85 Ave, Edmonton, ABT6G 2R3, Canada
| | - Joo Kheng Goh
- School of Science, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| |
Collapse
|
28
|
Madapally HV, Abe K, Dubey V, Khandelia H. Specific protonation of acidic residues confers K + selectivity to the gastric proton pump. J Biol Chem 2024; 300:105542. [PMID: 38072058 PMCID: PMC10825007 DOI: 10.1016/j.jbc.2023.105542] [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: 10/22/2023] [Revised: 11/20/2023] [Accepted: 11/29/2023] [Indexed: 01/11/2024] Open
Abstract
The gastric proton pump (H+,K+-ATPase) transports a proton into the stomach lumen for every K+ ion exchanged in the opposite direction. In the lumen-facing state of the pump (E2), the pump selectively binds K+ despite the presence of a 10-fold higher concentration of Na+. The molecular basis for the ion selectivity of the pump is unknown. Using molecular dynamics simulations, free energy calculations, and Na+ and K+-dependent ATPase activity assays, we demonstrate that the K+ selectivity of the pump depends upon the simultaneous protonation of the acidic residues E343 and E795 in the ion-binding site. We also show that when E936 is protonated, the pump becomes Na+ sensitive. The protonation-mimetic mutant E936Q exhibits weak Na+-activated ATPase activity. A 2.5-Å resolution cryo-EM structure of the E936Q mutant in the K+-occluded E2-Pi form shows, however, no significant structural difference compared with wildtype except less-than-ideal coordination of K+ in the mutant. The selectivity toward a specific ion correlates with a more rigid and less fluctuating ion-binding site. Despite being exposed to a pH of 1, the fundamental principle driving the K+ ion selectivity of H+,K+-ATPase is similar to that of Na+,K+-ATPase: the ionization states of the acidic residues in the ion-binding sites determine ion selectivity. Unlike the Na+,K+-ATPase, however, protonation of an ion-binding glutamate residue (E936) confers Na+ sensitivity.
Collapse
Affiliation(s)
- Hridya Valia Madapally
- PHYLIFE, Physical Life Science, Department of Physics Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark
| | - Kazuhiro Abe
- Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan; Cellular and Structural Physiology Institute, Nagoya University, Nagoya, Japan
| | - Vikas Dubey
- PHYLIFE, Physical Life Science, Department of Physics Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark
| | - Himanshu Khandelia
- PHYLIFE, Physical Life Science, Department of Physics Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
29
|
Weikum J, van Dyck JF, Subramani S, Klebl DP, Storflor M, Muench SP, Abel S, Sobott F, Morth JP. The bacterial magnesium transporter MgtA reveals highly selective interaction with specific cardiolipin species. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119614. [PMID: 37879515 DOI: 10.1016/j.bbamcr.2023.119614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/13/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023]
Abstract
The bacterial magnesium transporter A (MgtA) is a specialized P-type ATPase important for Mg2+ import into the cytoplasm; disrupted magnesium homeostasis is linked to intrinsic ribosome instability and antibacterial resistance in Salmonella strains. Here, we show that MgtA has functional specificity for cardiolipin 18:1. Still, it reaches maximum activity only in combination with cardiolipin 16:0, equivalent to the major components of native cardiolipin found in E. coli membranes. Native mass spectrometry indicates the presence of two binding sites for cardiolipin, agreeing with the kinetic studies revealing that a cooperative relationship likely exists between the two cardiolipin variants. This is the first experimental evidence of cooperative effects between lipids of the same class, with only minor variations in their acyl chain composition, acting on a membrane protein. In summary, our results reveal that MgtA exhibits a highly complex interaction with one cardiolipin 18:1 and one cardiolipin 16:0, affecting protein activity and stability, contributing to our understanding of the particular interactions between lipid environment and membrane proteins. Further, a better understanding of Mg2+ homeostasis in bacteria, due to its role as a virulence regulator, will provide further insights into the regulation and mechanism of bacterial infections.
Collapse
Affiliation(s)
- Julia Weikum
- Membrane Transport Group, Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, P.O. Box 1137, Blindern, 0318 Oslo, Norway; Enzyme and Protein Chemistry, Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, 2800 Kgs. Lyngby, Denmark
| | - Jeroen F van Dyck
- Department of Chemistry, University of Antwerp, Campus Groenenborger, Groenenborgerlaan 171, G.V. 418, 2020 Antwerpen, Belgium
| | - Saranya Subramani
- Membrane Transport Group, Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, P.O. Box 1137, Blindern, 0318 Oslo, Norway
| | - David P Klebl
- School of Biomedical Sciences & The Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, United Kingdom
| | - Merete Storflor
- Infections Biology Lab, Department of Pharmacy, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Stephen P Muench
- School of Biomedical Sciences & The Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, United Kingdom
| | - Sören Abel
- Infections Biology Lab, Department of Pharmacy, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Frank Sobott
- Department of Chemistry, University of Antwerp, Campus Groenenborger, Groenenborgerlaan 171, G.V. 418, 2020 Antwerpen, Belgium; School of Molecular and Cellular Biology & The Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, United Kingdom.
| | - J Preben Morth
- Membrane Transport Group, Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, P.O. Box 1137, Blindern, 0318 Oslo, Norway; Enzyme and Protein Chemistry, Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, 2800 Kgs. Lyngby, Denmark; Institute for Experimental Medical Research (IEMR), Oslo University Hospital, Ullevål PB 4956 Nydalen, NO-0424 Oslo, Norway.
| |
Collapse
|
30
|
Young MR, Heit S, Bublitz M. Structure, function and biogenesis of the fungal proton pump Pma1. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119600. [PMID: 37741574 DOI: 10.1016/j.bbamcr.2023.119600] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/19/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023]
Abstract
The fungal plasma membrane proton pump Pma1 is an integral plasma membrane protein of the P-type ATPase family. It is an essential enzyme responsible for maintaining a constant cytosolic pH and for energising the plasma membrane to secondary transport processes. Due to its importance for fungal survival and absence from animals, Pma1 is also a highly sought-after drug target. Until recently, its characterisation has been limited to functional, mutational and localisation studies, due to a lack of high-resolution structural information. The determination of three cryo-EM structures of Pma1 in its unique hexameric state offers a new level of understanding the molecular mechanisms underlying the protein's stability, regulated activity and druggability. In light of this context, this article aims to review what we currently know about the structure, function and biogenesis of fungal Pma1.
Collapse
Affiliation(s)
- Margaret R Young
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, United Kingdom
| | - Sabine Heit
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, United Kingdom
| | - Maike Bublitz
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, United Kingdom.
| |
Collapse
|
31
|
Clarke RJ. Electrostatic switch mechanisms of membrane protein trafficking and regulation. Biophys Rev 2023; 15:1967-1985. [PMID: 38192346 PMCID: PMC10771482 DOI: 10.1007/s12551-023-01166-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/17/2023] [Indexed: 01/10/2024] Open
Abstract
Lipid-protein interactions are normally classified as either specific or general. Specific interactions refer to lipid binding to specific binding sites within a membrane protein, thereby modulating the protein's thermal stability or kinetics. General interactions refer to indirect effects whereby lipids affect membrane proteins by modulating the membrane's physical properties, e.g., its fluidity, thickness, or dipole potential. It is not widely recognized that there is a third distinct type of lipid-protein interaction. Intrinsically disordered N- or C-termini of membrane proteins can interact directly but nonspecifically with the surrounding membrane. Many peripheral membrane proteins are held to the cytoplasmic surface of the plasma membrane via a cooperative combination of two forces: hydrophobic anchoring and electrostatic attraction. An acyl chain, e.g., myristoyl, added post-translationally to one of the protein's termini inserts itself into the lipid matrix and helps hold peripheral membrane proteins onto the membrane. Electrostatic attraction occurs between positively charged basic amino acid residues (lysine and arginine) on one of the protein's terminal tails and negatively charged phospholipid head groups, such as phosphatidylserine. Phosphorylation of either serine or tyrosine residues on the terminal tails via regulatory protein kinases allows for an electrostatic switch mechanism to control trafficking of the protein. Kinase action reduces the positive charge on the protein's tail, weakening the electrostatic attraction and releasing the protein from the membrane. A similar mechanism regulates many integral membrane proteins, but here only electrostatic interactions are involved, and the electrostatic switch modulates protein activity by altering the stabilities of different protein conformational states.
Collapse
Affiliation(s)
- Ronald J. Clarke
- School of Chemistry, University of Sydney, Sydney, NSW 2006 Australia
- The University of Sydney Nano Institute, Sydney, NSW 2006 Australia
| |
Collapse
|
32
|
Anand U, Anand P, Sodergren MH. Terpenes in Cannabis sativa Inhibit Capsaicin Responses in Rat DRG Neurons via Na +/K + ATPase Activation. Int J Mol Sci 2023; 24:16340. [PMID: 38003528 PMCID: PMC10671062 DOI: 10.3390/ijms242216340] [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: 10/17/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Terpenes in Cannabis sativa exert analgesic effects, but the mechanisms are uncertain. We examined the effects of 10 terpenes on capsaicin responses in an established model of neuronal hypersensitivity. Adult rat DRG neurons cultured with neurotrophic factors NGF and GDNF were loaded with Fura2AM for calcium imaging, and treated with individual terpenes or vehicle for 5 min, followed by 1 µMol capsaicin. In vehicle treated control experiments, capsaicin elicited immediate and sustained calcium influx. Most neurons treated with terpenes responded to capsaicin after 6-8 min. Few neurons showed immediate capsaicin responses that were transient or normal. The delayed responses were found to be due to calcium released from the endoplasmic reticulum, as they were maintained in calcium/magnesium free media, but not after thapsigargin pre-treatment. Terpene inhibition of calcium influx was reversed after washout of medium, in the absence of terpenes, and in the presence of the Na+/K+ ATPase inhibitor ouabain, but not CB1 or CB2 receptor antagonists. Thus, terpenes inhibit capsaicin evoked calcium influx by Na+/K+ ATPase activation. Immunofluorescence showed TRPV1 co-expression with α1β1 Na+/K+ ATPase in most neurons while others were either TRPV1 or α1β1 Na+/K+ ATPase positive.
Collapse
Affiliation(s)
- Uma Anand
- Faculty of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Rd, London W12 ONN, UK; (P.A.); (M.H.S.)
| | - Praveen Anand
- Faculty of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Rd, London W12 ONN, UK; (P.A.); (M.H.S.)
| | - Mikael Hans Sodergren
- Faculty of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Rd, London W12 ONN, UK; (P.A.); (M.H.S.)
- Curaleaf International Ltd., 179 Great Portland Street, London W1W 5PL, UK
| |
Collapse
|
33
|
Staehr C, Aalkjaer C, Matchkov V. The vascular Na,K-ATPase: clinical implications in stroke, migraine, and hypertension. Clin Sci (Lond) 2023; 137:1595-1618. [PMID: 37877226 PMCID: PMC10600256 DOI: 10.1042/cs20220796] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
In the vascular wall, the Na,K-ATPase plays an important role in the control of arterial tone. Through cSrc signaling, it contributes to the modulation of Ca2+ sensitivity in vascular smooth muscle cells. This review focuses on the potential implication of Na,K-ATPase-dependent intracellular signaling pathways in severe vascular disorders; ischemic stroke, familial migraine, and arterial hypertension. We propose similarity in the detrimental Na,K-ATPase-dependent signaling seen in these pathological conditions. The review includes a retrospective proteomics analysis investigating temporal changes after ischemic stroke. The analysis revealed that the expression of Na,K-ATPase α isoforms is down-regulated in the days and weeks following reperfusion, while downstream Na,K-ATPase-dependent cSrc kinase is up-regulated. These results are important since previous studies have linked the Na,K-ATPase-dependent cSrc signaling to futile recanalization and vasospasm after stroke. The review also explores a link between the Na,K-ATPase and migraine with aura, as reduced expression or pharmacological inhibition of the Na,K-ATPase leads to cSrc kinase signaling up-regulation and cerebral hypoperfusion. The review discusses the role of an endogenous cardiotonic steroid-like compound, ouabain, which binds to the Na,K-ATPase and initiates the intracellular cSrc signaling, in the pathophysiology of arterial hypertension. Currently, our understanding of the precise control mechanisms governing the Na,K-ATPase/cSrc kinase regulation in the vascular wall is limited. Understanding the role of vascular Na,K-ATPase signaling is essential for developing targeted treatments for cerebrovascular disorders and hypertension, as the Na,K-ATPase is implicated in the pathogenesis of these conditions and may contribute to their comorbidity.
Collapse
Affiliation(s)
- Christian Staehr
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, Aarhus, Denmark
| | - Christian Aalkjaer
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
- Danish Cardiovascular Academy, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
| | - Vladimir V. Matchkov
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
| |
Collapse
|
34
|
Baloglu E. HIF-2α Controls Expression and Intracellular Trafficking of the α2-Subunit of Na,K-ATPase in Hypoxic H9c2 Cardiomyocytes. Biomedicines 2023; 11:2879. [PMID: 38001879 PMCID: PMC10669276 DOI: 10.3390/biomedicines11112879] [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: 09/07/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 11/26/2023] Open
Abstract
The Na,K-ATPase (NKA) pump plays essential roles for optimal function of the heart. NKA activity decreases in necropsy materials from ischemic heart disease, heart failure and in experimental models. Cellular adaptation to hypoxia is regulated by hypoxia-induced transcription factors (HIF); we tested whether HIFs are involved in regulating the expression and intracellular dynamics of the α2-isoform of NKA (α2-NKA). HIF-1α and HIF-2α expression was suppressed in H9c2 cardiomyocytes by adenoviral infection, where cells were kept in 1% O2 for 24 h. The silencing efficiency of HIFs was tested on the mRNA and protein expression. We measured the mRNA expression of α2-NKA in HIF-silenced and hypoxia-exposed cells. The membrane and intracellular expression of α2-NKA was measured after labelling the cell surface with NHS-SS-biotin, immunoprecipitation and Western blotting. Hypoxia increased the mRNA expression of α2-NKA 5-fold compared to normoxic cells in an HIF-2α-sensitive manner. The plasma membrane expression of α2-NKA increased in hypoxia by 2-fold and was fully prevented by HIF-2α silencing. Intracellular expression of α2-NKA was not affected. These results showed for the first time that in hypoxic cardiomyocytes α2-NKA is transcriptionally and translationally regulated by HIF-2α. The molecular mechanism behind this regulation needs further investigation.
Collapse
Affiliation(s)
- Emel Baloglu
- Department of Medical Pharmacology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey
| |
Collapse
|
35
|
Gurler B, Gencay G, Baloglu E. Hypoxia and HIF-1α Regulate the Activity and Expression of Na,K-ATPase Subunits in H9c2 Cardiomyoblasts. Curr Issues Mol Biol 2023; 45:8277-8288. [PMID: 37886965 PMCID: PMC10605391 DOI: 10.3390/cimb45100522] [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: 09/23/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
The optimal function of the Na,K-ATPase (NKA) pump is essential for the heart. In ischemic heart disease, NKA activity decreases due to the decreased expression of the pump subunits. Here, we tested whether the hypoxia-inducible transcription factor (HIF-1α), the key signaling molecule regulating the adaptation of cells to hypoxia, is involved in controlling the expression and cellular dynamics of α1- and β1-NKA isoforms and of NKA activity in in-vitro hypoxic H9c2 cardiomyoblasts. HIF-1α was silenced through adenoviral infection, and cells were kept in normoxia (19% O2) or hypoxia (1% O2) for 24 h. We investigated the mRNA and protein expression of α1-, β1-NKA using RT-qPCR and Western blot in whole-cell lysates, cell membranes, and cytoplasmic fractions after labeling the cell surface with NHS-SS-biotin and immunoprecipitation. NKA activity and intracellular ATP levels were also measured. We found that in hypoxia, silencing HIF-1α prevented the decreased mRNA expression of α1-NKA but not of β1-NKA. Hypoxia decreased the plasma membrane expression of α1-NKA and β1- NKA compared to normoxic cells. In hypoxic cells, HIF-1α silencing prevented this effect by inhibiting the internalization of α1-NKA. Total protein expression was not affected. The decreased activity of NKA in hypoxic cells was fully prevented by silencing HIF-1α independent of cellular ATP levels. This study is the first to show that in hypoxic H9c2 cardiomyoblasts, HIF-1α controls the internalization and membrane insertion of α1-NKA subunit and of NKA activity. The mechanism behind this regulation needs further investigation.
Collapse
Affiliation(s)
- Beyza Gurler
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| | - Gizem Gencay
- Department of Molecular and Translational Biomedicine, Institute of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| | - Emel Baloglu
- Department of Medical Pharmacology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| |
Collapse
|
36
|
Abe K, Nishizawa T, Artigas P. An unusual conformation from Na +-sensitive non-gastric proton pump mutants reveals molecular mechanisms of cooperative Na +-binding. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119543. [PMID: 37482134 DOI: 10.1016/j.bbamcr.2023.119543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/29/2023] [Accepted: 07/14/2023] [Indexed: 07/25/2023]
Abstract
The Na+,K+-ATPase (NKA) and non-gastric H+,K+- ATPase (ngHKA) share ~65 % sequence identity, and nearly identical catalytic cycles. These pumps alternate between inward-facing (E1) and outward-facing (E2) conformations and differ in their exported substrate (Na+ or H+) and stoichiometries (3 Na+:2 K+ or 1 H+:1 K+). We reported that structures of the NKA-mimetic ngHKA mutant K794S/A797P/W940/R949C (SPWC) with 2 K+ occluded in E2-Pi and 3 Na+-bound in E1·ATP states were nearly identical to NKA structures in equivalent states. Here we report the cryo-EM structures of K794A and K794S, two poorly-selective ngHKA mutants, under conditions to stabilize the E1·ATP state. Unexpectedly, the structures show a hybrid with both E1- and E2-like structural features. While transmembrane segments TM1-TM3 and TM4's extracellular half adopted an E2-like conformation, the rest of the protein assumed an E1 configuration. Two spherical densities, likely bound Na+, were observed at cation-binding sites I and III, without density at site II. This explains the E2-like conformation of TM4's exoplasmic half. In NKA, oxygen atoms derived from the unwound portion of TM4 coordinated Na+ at site II. Thus, the lack of Na+ at site II of K794A/S prevents the luminal portion of TM4 from taking an E1-like position. The K794A structure also suggests that incomplete coordination of Na+ at site III induces the halfway rotation of TM6, which impairs Na+-binding at the site II. Thus, our observations provide insight into the molecular mechanism of E2-E1 transition and cooperative Na+-binding in the NKA and other related cation pumps.
Collapse
Affiliation(s)
- Kazuhiro Abe
- Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya 464-8601, Japan; Cellular and Structural Physiology Institute, Nagoya University, Nagoya 464-8601, Japan; Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Japan.
| | - Tomohiro Nishizawa
- Graduate School of Medical Life Sciences, Yokohama City University, Tsurumi, Yokohama 230-0045, Japan
| | - Pablo Artigas
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
37
|
Mahato DR, Andersson M. Dynamic lipid interactions in the plasma membrane Na +,K +-ATPase. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119545. [PMID: 37481079 DOI: 10.1016/j.bbamcr.2023.119545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/12/2023] [Accepted: 07/13/2023] [Indexed: 07/24/2023]
Abstract
The function of ion-transporting Na+,K+-ATPases depends on the surrounding lipid environment in biological membranes. Two established lipid-interaction sites A and B within the transmembrane domain have been observed to induce protein activation and stabilization, respectively. In addition, lipid-mediated inhibition has been assigned to a site C, but with the exact location not experimentally confirmed. Also, possible effects on lipid interactions by disease mutants dwelling in the membrane-protein interface remain relatively uncharacterized. We simulated human Na+,K+-ATPase α1β1FXYD homology models in E1 and E2 states in an asymmetric, multicomponent plasma membrane to determine both wild-type and disease mutant lipid-protein interactions. The simulated wild-type lipid interactions at the established sites A and B were in agreement with experimental results thereby confirming the membrane-protein model system. The less well-characterized, proposed inhibitory site C was dominated by lipids lacking inhibitory properties. Instead, two sites hosting inhibitory lipids were identified at the extracellular side and also a cytoplasmic CHL-binding site that provide putative alternative locations of Na+,K+-ATPase inhibition. Three disease mutations, Leu302Arg, Glu840Arg and Met859Arg resided in the lipid-protein interface and caused drastic changes in the lipid interactions. The simulation results show that lipid interactions to the human Na+,K+-ATPase α1β1FXYD protein in the plasma membrane are highly state-dependent and can be disturbed by disease mutations located in the lipid interface, which can open up for new venues to understand genetic disorders.
Collapse
Affiliation(s)
- Dhani Ram Mahato
- Department of Chemistry, Umeå University, Umeå, Sweden; Institut de Química Computacional i Catàlisi, Universitat de Girona, Girona, 17003, Spain
| | | |
Collapse
|
38
|
Lev B, Chennath M, Cranfield CG, Cornelius F, Allen TW, Clarke RJ. Involvement of the alpha-subunit N-terminus in the mechanism of the Na +,K +-ATPase. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119539. [PMID: 37479188 DOI: 10.1016/j.bbamcr.2023.119539] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 07/10/2023] [Indexed: 07/23/2023]
Abstract
Previous studies have shown that cytoplasmic K+ release and the associated E2 → E1 conformational change of the Na+,K+-ATPase is a major rate-determining step of the enzyme's ion pumping cycle and hence a prime site of acute regulatory intervention. From the ionic strength dependence of the enzyme's distribution between the E2 and E1 states, it has also been found that E2 is stabilized by an electrostatic attraction. Any disruption of this electrostatic attraction would, thus, have profound effects on the rate of ion pumping. The aim of this paper is to identify the location of this interaction. Using enhanced-sampling molecular dynamics simulations with a predicted N-terminal structure added to the X-ray crystal structure of the Na+,K+-ATPase, a previously postulated salt bridge between Lys32 and Glu233 (rat sequence numbering) of the enzyme's α-subunit can be excluded. The residues never approach closely enough to form a salt bridge. In contrast, strong interactions with anionic lipid head groups were seen. To investigate the possibility of a protein-lipid interaction experimentally, the surface charge density of Na+,K+-ATPase-containing membrane fragments was estimated from zeta potential measurements to be 0.019 (± 0.001) C m-2. This is in good agreement with the charge density previously determined to be responsible for stabilization of the E2 state of 0.023 (± 0.009) C m-2 and the membrane charge density estimated here from published electron-microscopic images of 0.018C m-2. The results are, therefore, consistent with an interaction of the Na+,K+-ATPase α-subunit N-terminus with negatively-charged lipid head groups of the neighbouring cytoplasmic membrane surface as the origin of the electrostatic interaction stabilising the E2 state.
Collapse
Affiliation(s)
- B Lev
- School of Science, RMIT University, Melbourne, Vic, 3001, Australia
| | - M Chennath
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - C G Cranfield
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - F Cornelius
- Department of Biomedicine, University of Aarhus, DK-8000 Aarhus, C, Denmark
| | - T W Allen
- School of Science, RMIT University, Melbourne, Vic, 3001, Australia
| | - R J Clarke
- School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia; The University of Sydney Nano Institute, Sydney, NSW 2006, Australia.
| |
Collapse
|
39
|
Palmgren M. Evolution of the sodium pump. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119511. [PMID: 37301269 DOI: 10.1016/j.bbamcr.2023.119511] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/16/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
Eukaryotic plasma membranes (PMs) are energized by electrogenic P-type ATPases that generate either Na+ or H+ motive forces to drive Na+ and H+ dependent transport processes, respectively. For this purpose, animal rely on Na+/K+-ATPases whereas fungi and plants employ PM H+-ATPases. Prokaryotes, on the other hand, depend on H+ or Na+-motive electron transport complexes to energize their cell membranes. This raises the question as to why and when electrogenic Na+ and H+ pumps evolved? Here it is shown that prokaryotic Na+/K+-ATPases have near perfect conservation of binding sites involved in coordination of three Na+ and two K+ ions. Such pumps are rare in Eubacteria but are common in methanogenic Archaea where they often are found together with P-type putative PM H+-ATPases. With some exceptions, Na+/K+-ATPases and PM H+-ATPases are found everywhere in the eukaryotic tree of life, but never together in animals, fungi and land plants. It is hypothesized that Na+/K+-ATPases and PM H+-ATPases evolved in methanogenic Archaea to support the bioenergetics of these ancestral organisms, which can utilize both H+ and Na+ as energy currencies. Both pumps must have been simultaneously present in the first eukaryotic cell, but during diversification of the major eukaryotic kingdoms, and at the time animals diverged from fungi, animals kept Na+/K+-ATPases but lost PM H+-ATPases. At the same evolutionary branch point, fungi did loose Na+/K+-ATPases, and their role was taken over by PM H+-ATPases. An independent but similar scenery emerged during terrestrialization of plants: they lost Na+/K+-ATPases but kept PM H+-ATPases.
Collapse
Affiliation(s)
- Michael Palmgren
- Department of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark.
| |
Collapse
|
40
|
Paulson OB, Schousboe A, Hultborn H. The history of Danish neuroscience. Eur J Neurosci 2023; 58:2893-2960. [PMID: 37477973 DOI: 10.1111/ejn.16062] [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: 11/22/2022] [Revised: 05/04/2023] [Accepted: 05/29/2023] [Indexed: 07/22/2023]
Abstract
The history of Danish neuroscience starts with an account of impressive contributions made at the 17th century. Thomas Bartholin was the first Danish neuroscientist, and his disciple Nicolaus Steno became internationally one of the most prominent neuroscientists in this period. From the start, Danish neuroscience was linked to clinical disciplines. This continued in the 19th and first half of the 20th centuries with new initiatives linking basic neuroscience to clinical neurology and psychiatry in the same scientific environment. Subsequently, from the middle of the 20th century, basic neuroscience was developing rapidly within the preclinical university sector. Clinical neuroscience continued and was even reinforced during this period with important translational research and a close co-operation between basic and clinical neuroscience. To distinguish 'history' from 'present time' is not easy, as many historical events continue in present time. Therefore, we decided to consider 'History' as new major scientific developments in Denmark, which were launched before the end of the 20th century. With this aim, scientists mentioned will have been born, with a few exceptions, no later than the early 1960s. However, we often refer to more recent publications in documenting the developments of initiatives launched before the end of the last century. In addition, several scientists have moved to Denmark after the beginning of the present century, and they certainly are contributing to the present status of Danish neuroscience-but, again, this is not the History of Danish neuroscience.
Collapse
Affiliation(s)
- Olaf B Paulson
- Neurobiology Research Unit, Department of Neurology, Rigshospitalet, 9 Blegdamsvej, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans Hultborn
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
41
|
Baloglu E. Hypoxic Stress-Dependent Regulation of Na,K-ATPase in Ischemic Heart Disease. Int J Mol Sci 2023; 24:ijms24097855. [PMID: 37175562 PMCID: PMC10177966 DOI: 10.3390/ijms24097855] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
In cardiomyocytes, regular activity of the Na,K-ATPase (NKA) and its Na/K pump activity is essential for maintaining ion gradients, excitability, propagation of action potentials, electro-mechanical coupling, trans-membrane Na+ and Ca2+ gradients and, thus, contractility. The activity of NKA is impaired in ischemic heart disease and heart failure, which has been attributed to decreased expression of the NKA subunits. Decreased NKA activity leads to intracellular Na+ and Ca2+ overload, diastolic dysfunction and arrhythmias. One signal likely related to these events is hypoxia, where hypoxia-inducible factors (HIF) play a critical role in the adaptation of cells to low oxygen tension. HIF activity increases in ischemic heart, hypertension, heart failure and cardiac fibrosis; thus, it might contribute to the impaired function of NKA. This review will mainly focus on the regulation of NKA in ischemic heart disease in the context of stressed myocardium and the hypoxia-HIF axis and argue on possible consequences of treatment.
Collapse
Affiliation(s)
- Emel Baloglu
- Department of Medical Pharmacology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey
| |
Collapse
|
42
|
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: 3.0] [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
|
43
|
Ruan X, Zhang C, Zhu Y, Cai F, Yang Y, Feng J, Ma X, Zheng Y, Li H, Yuan Y, Zhu G. Constructing Mechanical Shuttles in a Three-dimensional (3D) Porous Architecture for Selective Transport of Lithium Ions. Angew Chem Int Ed Engl 2023; 62:e202216549. [PMID: 36482169 DOI: 10.1002/anie.202216549] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022]
Abstract
Lithium (Li) extraction from brines is a major barrier to the sustainable development of batteries and alloys; however, current separation technology suffers from a trade-off between ion selectivity and permeability. Herein, a crown ether mechanically interlocked 3D porous organic framework (Crown-POF) was prepared as the porous filler of thin-film nanocomposite membranes. Crown-POF with penta-coordinated (four Ocrown atoms and one Ntert-amine atom) adsorption sites enables a special recognition for Li+ ion. Moreover, the four Ntert-amine atoms on each POF branch facilitate the flipping motion of Li+ ion along the skeletal thread, while retaining the specified binding pattern. Accordingly, the crown ether interlocked POF network displays an ultrafast ion transfer rate, over 10 times that of the conventional porous materials. Notably, the nanocomposite membrane gives high speed and selectivity for Li+ ion transport as compared with other porous solid-based mixed-matrix membranes.
Collapse
Affiliation(s)
- Xianghui Ruan
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Northeast Normal University, Renmin Avenue, Changchun, 130024, China
| | - Cheng Zhang
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Northeast Normal University, Renmin Avenue, Changchun, 130024, China
| | - Yuzhang Zhu
- Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Fuli Cai
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Northeast Normal University, Renmin Avenue, Changchun, 130024, China
| | - Yajie Yang
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Northeast Normal University, Renmin Avenue, Changchun, 130024, China
| | - Jiahui Feng
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Northeast Normal University, Renmin Avenue, Changchun, 130024, China
| | - Xujiao Ma
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Northeast Normal University, Renmin Avenue, Changchun, 130024, China
| | - Yue Zheng
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Northeast Normal University, Renmin Avenue, Changchun, 130024, China
| | - Huanhuan Li
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Northeast Normal University, Renmin Avenue, Changchun, 130024, China
| | - Ye Yuan
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Northeast Normal University, Renmin Avenue, Changchun, 130024, China
| | - Guangshan Zhu
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education, Northeast Normal University, Renmin Avenue, Changchun, 130024, China
| |
Collapse
|
44
|
Huang D, Song X, Ma J, Li X, Guo Y, Li M, Luo H, Fang Z, Yang C, Xie L, Jiang L. ATP1A3-related phenotypes in Chinese children: AHC, CAPOS, and RECA. Eur J Pediatr 2023; 182:825-836. [PMID: 36484864 DOI: 10.1007/s00431-022-04744-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 11/13/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022]
Abstract
UNLABELLED The aim of this research is to study the phenotype, genotype, treatment strategies, and short-term prognosis of Chinese children with ATP1A3 (Na+/K+-ATPase alpha 3 gene)-related disorders in Southwest China. Patients with pathogenic ATP1A3 variants identified using next-generation sequencing were registered at the Children's Hospital of Chongqing Medical University from December 2015 to May 2019. We followed them as a cohort and analyzed their clinical data. Eleven patients were identified with de novo pathogenic ATP1A3 heterozygous variants. One (c.2542 + 1G > T, splicing) has not been reported. Eight patients with alternating hemiplegia of childhood (AHC), one with cerebellar ataxia, areflexia, pes cavus, optic atrophy, and sensorineural hearing loss (CAPOS), and two with relapsing encephalopathy with cerebellar ataxia (RECA) were included. The initial manifestations of AHC included hemiplegia, oculomotor abnormalities, and seizures, and the most common trigger was an upper respiratory tract infection without fever. All patients had paroxysmal hemiplegic attacks during their disease course. The brain MRI showed no abnormalities. Six out of eight AHC cases reached a stable disease state after treatment. The initial symptom of the patient with CAPOS was ataxia followed by developmental regression, seizures, deafness, visual impairment, and dysarthria, and the brain MRI indicated mild cerebellar atrophy. No fluctuation was noted after using Acetazolamide. The initial manifestations of the two RECA cases were dystonia and encephalopathy, respectively. One manifested a rapid-onset course of dystonia triggered by a fever followed by dysarthria and action tremors, and independent walking was impossible. The brain MRI image was normal. The other one presented with disturbance of consciousness, seizures, sleep disturbance, tremor, and dyskinesias. The EEG revealed a slow background (δ activity), and the brain MRI result was normal. No response to Flunarizine was noted for them, and it took 61 and 60 months for them to reach a stable disease state, respectively. CONCLUSION Pathogenic ATP1A3 variants play an essential role in the pathogenesis of Sodium-Potassium pump disorders, and AHC is the most common phenotype. The treatment strategies and prognosis depend on the phenotype categories caused by different variation sites and types. The correlation between the genotype and phenotype requires further exploration. WHAT IS KNOWN • Pathogenic heterozygous ATP1A3 variants cause a spectrum of neurological phenotypes, and ATP1A3-disorders are viewed as a phenotypic continuum presenting with atypical and overlapping features. • The genotype-phenotype correlation of ATP1A3-disorders remains unclear. WHAT IS NEW • In this study, the genotypes and phenotypes of ATP1A3-related disorders from Southwest of China were described. The splice-site variation c.2542+1G>T was detected for the first time in ATP1A3-related disorders. • The prognosis of twins with AHC p. Gly947Arg was more serious than AHC cases with other variants, which was inconsistent with previous reports. The phenomenon indicated the diversity of the correlation between the genotype and phenotype.
Collapse
Affiliation(s)
- Dishu Huang
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Xiaojie Song
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Jiannan Ma
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Xiujuan Li
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Yi Guo
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Mei Li
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Hanyu Luo
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Zhixu Fang
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Chen Yang
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Lingling Xie
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China. .,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China. .,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, People's Republic of China. .,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China. .,Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China.
| | - Li Jiang
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China. .,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China. .,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, People's Republic of China. .,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China. .,Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China.
| |
Collapse
|
45
|
Manoj KM, Gideon DA, Bazhin NM, Tamagawa H, Nirusimhan V, Kavdia M, Jaeken L. Na,K-ATPase: A murzyme facilitating thermodynamic equilibriums at the membrane-interface. J Cell Physiol 2023; 238:109-136. [PMID: 36502470 DOI: 10.1002/jcp.30925] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 12/14/2022]
Abstract
The redox metabolic paradigm of murburn concept advocates that diffusible reactive species (DRS, particularly oxygen-centric radicals) are mainstays of physiology, and not mere pathological manifestations. The murburn purview of cellular function also integrates the essential principles of bioenergetics, thermogenesis, homeostasis, electrophysiology, and coherence. In this context, any enzyme that generates/modulates/utilizes/sustains DRS functionality is called a murzyme. We have demonstrated that several water-soluble (peroxidases, lactate dehydrogenase, hemogoblin, etc.) and membrane-embedded (Complexes I-V in mitochondria, Photosystems I/II in chloroplasts, rhodopsin/transducin in rod cells, etc.) proteins serve as murzymes. The membrane protein of Na,K-ATPase (NKA, also known as sodium-potassium pump) is the focus of this article, owing to its centrality in neuro-cardio-musculo electrophysiology. Herein, via a series of critical queries starting from the geometric/spatio-temporal considerations of diffusion/mass transfer of solutes in cells to an update on structural/distributional features of NKA in diverse cellular systems, and from various mechanistic aspects of ion-transport (thermodynamics, osmoregulation, evolutionary dictates, etc.) to assays/explanations of inhibitory principles like cardiotonic steroids (CTS), we first highlight some unresolved problems in the field. Thereafter, we propose and apply a minimalist murburn model of trans-membrane ion-differentiation by NKA to address the physiological inhibitory effects of trans-dermal peptide, lithium ion, volatile anesthetics, confirmed interfacial DRS + proton modulators like nitrophenolics and unsaturated fatty acid, and the diverse classes of molecules like CTS, arginine, oximes, etc. These explanations find a pan-systemic connectivity with the inhibitions/uncouplings of other membrane proteins in cells.
Collapse
Affiliation(s)
- Kelath Murali Manoj
- Satyamjayatu: The Science & Ethics Foundation, Kulappully, Shoranur-2, Kerala, India
| | - Daniel A Gideon
- Satyamjayatu: The Science & Ethics Foundation, Kulappully, Shoranur-2, Kerala, India
| | - Nikolai M Bazhin
- Institute of Chemical Kinetics and Combustion, Russian Academy of Sciences, Novosibirsk, Russia
| | - Hirohisa Tamagawa
- Department of Mechanical Engineering, Gifu University, Gifu City, Japan
| | - Vijay Nirusimhan
- Satyamjayatu: The Science & Ethics Foundation, Kulappully, Shoranur-2, Kerala, India
| | - Mahendra Kavdia
- Department of Biomedical Engineering, Wayne State University, Detroit, Michigan, USA
| | - Laurent Jaeken
- Department of Industrial Sciences and Technology, Karel de Grote-Hogeschool, Antwerp University Association, Antwerp, Belgium
| |
Collapse
|
46
|
Arystarkhova E, Toustrup-Jensen MS, Holm R, Ko JK, Lee KE, Feschenko P, Ozelius LJ, Brashear A, Vilsen B, Sweadner KJ. Temperature instability of a mutation at a multidomain junction in Na,K-ATPase isoform ATP1A3 (p.Arg756His) produces a fever-induced neurological syndrome. J Biol Chem 2023; 299:102758. [PMID: 36462665 PMCID: PMC9860391 DOI: 10.1016/j.jbc.2022.102758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 12/02/2022] Open
Abstract
ATP1A3 encodes the α3 isoform of Na,K-ATPase. In the brain, it is expressed only in neurons. Human ATP1A3 mutations produce a wide spectrum of phenotypes, but particular syndromes are associated with unique substitutions. For arginine 756, at the junction of membrane and cytoplasmic domains, mutations produce encephalopathy during febrile infections. Here we tested the pathogenicity of p.Arg756His (R756H) in isogenic mammalian cells. R756H protein had sufficient transport activity to support cells when endogenous ATP1A1 was inhibited. It had half the turnover rate of wildtype, reduced affinity for Na+, and increased affinity for K+. There was modest endoplasmic reticulum retention during biosynthesis at 37 °C but little benefit from the folding drug phenylbutyrate (4-PBA), suggesting a tolerated level of misfolding. When cells were incubated at just 39 °C, however, α3 protein level dropped without loss of β subunit, paralleled by an increase of endogenous α1. Elevated temperature resulted in internalization of α3 from the surface along with some β subunit, accompanied by cytoplasmic redistribution of a marker of lysosomes and endosomes, lysosomal-associated membrane protein 1. After return to 37 °C, α3 protein levels recovered with cycloheximide-sensitive new protein synthesis. Heating in vitro showed activity loss at a rate 20- to 30-fold faster than wildtype, indicating a temperature-dependent destabilization of protein structure. Arg756 appears to confer thermal resistance as an anchor, forming hydrogen bonds among four linearly distant parts of the Na,K-ATPase structure. Taken together, our observations are consistent with fever-induced symptoms in patients.
Collapse
Affiliation(s)
- Elena Arystarkhova
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA.
| | | | - Rikke Holm
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Jae-Kyun Ko
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Kyung Eun Lee
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Polina Feschenko
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Laurie J Ozelius
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Allison Brashear
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Bente Vilsen
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Kathleen J Sweadner
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
47
|
Bioinformatic Analysis of Na +, K +-ATPase Regulation through Phosphorylation of the Alpha-Subunit N-Terminus. Int J Mol Sci 2022; 24:ijms24010067. [PMID: 36613508 PMCID: PMC9820343 DOI: 10.3390/ijms24010067] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/01/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
The Na+, K+-ATPase is an integral membrane protein which uses the energy of ATP hydrolysis to pump Na+ and K+ ions across the plasma membrane of all animal cells. It plays crucial roles in numerous physiological processes, such as cell volume regulation, nutrient reabsorption in the kidneys, nerve impulse transmission, and muscle contraction. Recent data suggest that it is regulated via an electrostatic switch mechanism involving the interaction of its lysine-rich N-terminus with the cytoplasmic surface of its surrounding lipid membrane, which can be modulated through the regulatory phosphorylation of the conserved serine and tyrosine residues on the protein's N-terminal tail. Prior data indicate that the kinases responsible for phosphorylation belong to the protein kinase C (PKC) and Src kinase families. To provide indications of which particular enzyme of these families might be responsible, we analysed them for evidence of coevolution via the mirror tree method, utilising coevolution as a marker for a functional interaction. The results obtained showed that the most likely kinase isoforms to interact with the Na+, K+-ATPase were the θ and η isoforms of PKC and the Src kinase itself. These theoretical results will guide the direction of future experimental studies.
Collapse
|
48
|
Forzano I, Mone P, Mottola G, Kansakar U, Salemme L, De Luca A, Tesorio T, Varzideh F, Santulli G. Efficacy of the New Inotropic Agent Istaroxime in Acute Heart Failure. J Clin Med 2022; 11:7503. [PMID: 36556120 PMCID: PMC9786901 DOI: 10.3390/jcm11247503] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Current therapeutic strategies for acute heart failure (AHF) are based on traditional inotropic agents that are often associated with untoward effects; therefore, finding new effective approaches with a safer profile is dramatically needed. Istaroxime is a novel compound, chemically unrelated to cardiac glycosides, that is currently being studied for the treatment of AHF. Its effects are essentially related to its inotropic and lusitropic positive properties exerted through a dual mechanism of action: activation of the sarcoplasmic reticulum Ca2+ ATPase isoform 2a (SERCA2a) and inhibition of the Na+/K+-ATPase (NKA) activity. The advantages of istaroxime over the available inotropic agents include its lower arrhythmogenic action combined with its capability of increasing systolic blood pressure without augmenting heart rate. However, it has a limited half-life (1 hour) and is associated with adverse effects including pain at the injection site and gastrointestinal issues. Herein, we describe the main mechanism of action of istaroxime and we present a systematic overview of both clinical and preclinical trials testing this drug, underlining the latest insights regarding its adoption in clinical practice for AHF.
Collapse
Affiliation(s)
- Imma Forzano
- Division of Cardiology, Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
| | - Pasquale Mone
- Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Gaetano Mottola
- Casa di Cura “Montevergine”, Mercogliano, 83013 Avellino, Italy
| | - Urna Kansakar
- Division of Cardiology, Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
| | - Luigi Salemme
- Casa di Cura “Montevergine”, Mercogliano, 83013 Avellino, Italy
| | - Antonio De Luca
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Vanvitelli”, 81100 Caserta, Italy
| | - Tullio Tesorio
- Casa di Cura “Montevergine”, Mercogliano, 83013 Avellino, Italy
| | - Fahimeh Varzideh
- Division of Cardiology, Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
| | - Gaetano Santulli
- Division of Cardiology, Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
- Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
49
|
Faraj SE, Valsecchi WM, Ferreira-Gomes M, Centeno M, Saint Martin EM, Fedosova NU, Rossi JPFC, Montes MR, Rossi RC. Measurements of Na +-occluded intermediates during the catalytic cycle of the Na +/K +-ATPase provide novel insights into the mechanism of Na + transport. J Biol Chem 2022; 299:102811. [PMID: 36539036 PMCID: PMC9860123 DOI: 10.1016/j.jbc.2022.102811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
The Na+/K+-ATPase is an integral plasma membrane glycoprotein of all animal cells that couples the exchange of intracellular Na+ for extracellular K+ to the hydrolysis of ATP. The asymmetric distribution of Na+ and K+ is essential for cellular life and constitutes the physical basis of a series of fundamental biological phenomena. The pumping mechanism is explained by the Albers-Post model. It involves the presence of gates alternatively exposing Na+/K+-ATPase transport sites to the intracellular and extracellular sides and includes occluded states in which both gates are simultaneously closed. Unlike for K+, information is lacking about Na+-occluded intermediates, as occluded Na+ was only detected in states incapable of performing a catalytic cycle, including two Na+-containing crystallographic structures. The current knowledge is that intracellular Na+ must bind to the transport sites and become occluded upon phosphorylation by ATP to be transported to the extracellular medium. Here, taking advantage of epigallocatechin-3-gallate to instantaneously stabilize native Na+-occluded intermediates, we isolated species with tightly bound Na+ in an enzyme able to perform a catalytic cycle, consistent with a genuine occluded state. We found that Na+ becomes spontaneously occluded in the E1 dephosphorylated form of the Na+/K+-ATPase, exhibiting positive interactions between binding sites. In fact, the addition of ATP does not produce an increase in Na+ occlusion as it would have been expected; on the contrary, occluded Na+ transiently decreases, whereas ATP lasts. These results reveal new properties of E1 intermediates of the Albers-Post model for explaining the Na+ transport pathway.
Collapse
Affiliation(s)
- Santiago E. Faraj
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) – Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas "Prof. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina
| | - Wanda M. Valsecchi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) – Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas "Prof. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina
| | - Mariela Ferreira-Gomes
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) – Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas "Prof. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina
| | - Mercedes Centeno
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) – Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas "Prof. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina
| | - Elina Malén Saint Martin
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina
| | | | - Juan Pablo FC. Rossi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) – Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas "Prof. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina
| | - Mónica R. Montes
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) – Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas "Prof. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina
| | - Rolando C. Rossi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) – Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas "Prof. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina,For correspondence: Rolando C. Rossi
| |
Collapse
|
50
|
Bentsen S, Bang LE, Hasbak P, Kjaer A, Ripa RS. Amiodarone attenuates cardiac Rubidium-82 in consecutive PET/CT scans in a rodent model. J Nucl Cardiol 2022; 29:2853-2862. [PMID: 34611850 DOI: 10.1007/s12350-021-02785-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/23/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND Risk stratification and diagnosis using Rubidium-82 (82Rb) positron emission tomography (PET) is a routine clinical approach in coronary artery disease (CAD). Various drugs are used to treat CAD; however, whether any of them change the uptake of 82Rb in the heart has not been investigated. The aim of this study is to determine whether drugs used in treatment of CAD affect the uptake of 82Rb in the heart in healthy rats. METHODS Seventy-seven Sprague-Dawley rats were included in the cross-sectional study. All rats underwent baseline 82Rb PET/CT and divided into eleven groups treated with different drugs. One group was control group (no treatment), eight groups were treated with monotherapy (amiodarone, acetylsalicylic acid (ASA), clopidogrel, ticagrelor, atorvastatin, enalapril, amlodipine, metoprolol succinate), and two groups were treated with polypharmacy (ASA, ticagrelor, atorvastatin, amlodipine or ASA, clopidogrel, atorvastatin, amlodipine). Once a day, they were administered pharmacological therapy through oral gavage, and on day seven, follow-up scanned with 82Rb PET/CT. RESULTS In the control group without pharmacological treatment, no difference in the standard uptake value (SUV) ratio between heart and muscle from baseline to follow-up (5.8 vs 7.0, P = .3) was found. The group treated with amiodarone had a significantly reduced SUV ratio from baseline to follow-up (5.8 vs 5.1, P = .008). All other drugs investigated had no difference in SUV ratio from baseline to follow-up. CONCLUSION In this study, we showed that drugs normally used to treat CAD do not affect the uptake of 82Rb. However, amiodarone result in a significantly lowered 82Rb uptake, compared to control. This information about amiodarone would probably not change the size assessment of a myocardial perfusion defect in a clinical setting. However, it could change the kinetic parameters when assessing absolute myocardial blood flow in patients treated with amiodarone.
Collapse
Affiliation(s)
- Simon Bentsen
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Copenhagen, Denmark
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lia E Bang
- Department of Cardiology, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Philip Hasbak
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Copenhagen, Denmark.
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Rasmus S Ripa
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Copenhagen, Denmark
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|