1
|
Barrie KR, Rebowski G, Dominguez R. Mechanism of actin filament severing and capping by gelsolin. Nat Struct Mol Biol 2025; 32:237-242. [PMID: 39448849 PMCID: PMC11832341 DOI: 10.1038/s41594-024-01412-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024]
Abstract
Gelsolin is the prototypical member of a family of Ca2+-activated F-actin severing and capping proteins. Here we report structures of Ca2+-bound human gelsolin at the barbed end of F-actin. One structure reveals gelsolin's six domains (G1G6) and interdomain linkers wrapping around F-actin, while another shows domains G1G3-a fragment observed during apoptosis-binding on both sides of F-actin. Conformational changes that trigger severing occur on one side of F-actin with G1G6 and on both sides with G1G3. Gelsolin remains bound after severing, blocking subunit exchange.
Collapse
Affiliation(s)
- Kyle R Barrie
- Department of Physiology and Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Grzegorz Rebowski
- Department of Physiology and Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roberto Dominguez
- Department of Physiology and Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
McCaig CD. Electrical Forces Improve Memory in Old Age. Rev Physiol Biochem Pharmacol 2025; 187:453-520. [PMID: 39838022 DOI: 10.1007/978-3-031-68827-0_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
This penultimate chapter is based on a single paper published in Nature in 2022. I have used it specifically as an exemplar, in this case to show that memory improvement in old age may be regulated by a multiplicity of electrical forces. However, I include it because I believe that one could pick almost any other substantial single paper and show that a completely disparate set of biological mechanisms similarly depend crucially on multiple electrical forces.
Collapse
Affiliation(s)
- Colin D McCaig
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| |
Collapse
|
3
|
Kim HU, Park YH, An MY, Kim YK, Song C, Jung HS. Structural insights into calcium-induced conformational changes in human gelsolin. Biochem Biophys Res Commun 2024; 735:150826. [PMID: 39426132 DOI: 10.1016/j.bbrc.2024.150826] [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: 09/30/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Gelsolin is known as one of the actin-binding proteins capable of severing and capping filamentous actin, and of undergoing structural changes in the presence of calcium ions to interact with actin filaments. In this study, single-particle 3D reconstruction using electron microscopy (EM) revealed that, in the presence of calcium, the structure of gelsolin undergoes structural changes before interacting with actin. These differences are subtle with similarities, as confirmed by the EM map. According to the results of the molecular dynamics simulations, these nuanced structural differences primarily manifest at the domain level when calcium is present. These results provide structural evidence that, in the presence of calcium, gelsolin enters a phase of conformational preparation to transition into the active state. This process enables gelsolin to bind to actin, whereupon gelsolin undergoes more drastic structural changes upon interaction with actin filaments, which allows it to participate in binding and severing to regulate the cytoskeleton. This is the first visualization of full-length gelsolin, and helps to clarify crucial aspects of the as of yet incompletely understood interaction between gelsolin and actin.
Collapse
Affiliation(s)
- Han-Ul Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea; Kangwon Center for Systems Imaging, Chuncheon, 24341, Republic of Korea
| | - Yoon Ho Park
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Mi Young An
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Young Kwan Kim
- Kangwon Center for Systems Imaging, Chuncheon, 24341, Republic of Korea
| | - Chihong Song
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
| | - Hyun Suk Jung
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea; Kangwon Center for Systems Imaging, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
4
|
Sagar A, Peddada N, Choudhary V, Mir Y, Garg R, Ashish. Visualizing the nucleating and capped states of f-actin by Ca 2+-gelsolin: Saxs data based structures of binary and ternary complexes. Int J Biol Macromol 2024; 278:134556. [PMID: 39128762 DOI: 10.1016/j.ijbiomac.2024.134556] [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: 04/16/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Structural insight eludes on how full-length gelsolin depolymerizes and caps filamentous (F-)actin, while the same entity can nucleate polymerization of G-actins. Analyzing small angle X-ray scattering (SAXS) data, we deciphered assemblies which enable these contrasting processes. Mixing Ca2+-gelsolin with F-actin in high salt F-buffer resulted in depolymerization of ordered F-actin rods to smaller sized species which became monodispersed upon dialysis with low salt G-buffer. These entities were the ternary (GA2) and binary (GA) complexes of gelsolin and actin with radius of gyration and maximum linear dimension of 4.55 and 4.68 nm, and 15 and 16 nm, respectively. Using size exclusion chromatography in-line with SAXS, we confirmed that initially GA and GA2 species are formed as seen upon depolymerization of F-actin followed by dialysis. Interestingly, while GA2 could seed formation of native-like F-actin in both G- and F-buffer, GA failed in G-buffer. Thus, GA2 and GA are the central species formed via depolymerization or towards nucleation. SAXS profile referenced modeling revealed that: 1) in GA, actin is bound to the C-terminal half of gelsolin, and 2) in GA2, second actin binds to the open N-terminal half accompanied by dramatic rearrangements across g1-g2 and g3-g4 linkers.
Collapse
Affiliation(s)
- Amin Sagar
- Csir - Institute Of Microbial Technology, Chandigarh, India
| | - Nagesh Peddada
- Csir - Institute Of Microbial Technology, Chandigarh, India
| | | | - Yawar Mir
- Csir - Institute Of Microbial Technology, Chandigarh, India
| | - Renu Garg
- Csir - Institute Of Microbial Technology, Chandigarh, India
| | - Ashish
- Csir - Institute Of Microbial Technology, Chandigarh, India.
| |
Collapse
|
5
|
Barrie KR, Rebowski G, Dominguez R. Mechanism of Actin Filament Severing and Capping by Gelsolin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612341. [PMID: 39345426 PMCID: PMC11430012 DOI: 10.1101/2024.09.10.612341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Gelsolin is the prototypical member of a family of Ca 2+ -dependent F-actin severing and capping proteins. A structure of Ca 2+ -bound full-length gelsolin at the barbed end shows domains G1G6 and the inter-domain linkers wrapping around F-actin. Another structure shows domains G1G3, a fragment produced during apoptosis, on both sides of F-actin. Conformational changes that trigger severing occur on one side of F-actin with G1G6 and on both sides with G1G3. Gelsolin remains bound after severing, blocking subunit exchange.
Collapse
|
6
|
Cui Y, Megawati D, Lin J, Rehard DG, Grant DG, Liu P, Jurkevich A, Reid WR, Mooney BP, Franz AW. Cytoskeleton-associated gelsolin responds to the midgut distention process in saline meal-fed Aedes aegypti and affects arbovirus dissemination from the midgut. FASEB J 2024; 38:e23764. [PMID: 39042395 PMCID: PMC11268798 DOI: 10.1096/fj.202302684rr] [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: 12/27/2023] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 07/24/2024]
Abstract
The mosquito, Aedes aegypti, is the principal vector for several arboviruses. The mosquito midgut is the initial tissue that gets infected with an arbovirus acquired along with a blood meal from a vertebrate host. Blood meal ingestion leads to midgut tissue distention thereby increasing the pore size of the surrounding basal lamina. This allows newly synthesized virions to exit the midgut by traversing the distended basal lamina to infect secondary tissues of the mosquito. We conducted a quantitative label-free proteomic time course analysis with saline meal-fed Ae. aegypti females to identify host factors involved in midgut tissue distention. Around 2000 proteins were detected during each of the seven sampling time points and 164 of those were uniquely expressed. Forty-five of 97 differentially expressed proteins were upregulated during the 96-h time course and most of those were involved in cytoskeleton modulation, metabolic activity, and vesicle/vacuole formation. The F-actin-modulating Ae. aegypti (Aa)-gelsolin was selected for further functional studies. Stable knockout of Aa-gelsolin resulted in a mosquito line, which showed distorted actin filaments in midgut-associated tissues likely due to diminished F-actin processing by gelsolin. Zika virus dissemination from the midgut of these mosquitoes was diminished and delayed. The loss of Aa-gelsolin function was associated with an increased induction of apoptosis in midgut tissue indicating an involvement of Aa-gelsolin in apoptotic signaling in mosquitoes. Here, we used proteomics to discover a novel host factor, Aa-gelsolin, which affects the midgut escape barrier for arboviruses in mosquitoes and apoptotic signaling in the midgut.
Collapse
Affiliation(s)
- Yingjun Cui
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Dewi Megawati
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
- Department of Microbiology and Parasitology, School of Medicine, Warmadewa University, Bali, Indonesia
| | - Jingyi Lin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - David G. Rehard
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - DeAna G. Grant
- Electron Microscopy Core, University of Missouri, Columbia, Missouri, USA
| | - Pei Liu
- Charles W. Gehrke Proteomics Center, University of Missouri, Columbia, Missouri, USA
| | - Alexander Jurkevich
- Advanced Light Microscopy Core, University of Missouri, Columbia, Missouri, USA
| | - William R. Reid
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Brian P. Mooney
- Charles W. Gehrke Proteomics Center, University of Missouri, Columbia, Missouri, USA
| | - Alexander W.E. Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
7
|
Zhi J, Zhao KX, Liu JH, Yang D, Deng XM, Xu J, Zhang H. The therapeutic potential of gelsolin in attenuating cytokine storm, ARDS, and ALI in severe COVID-19. Front Pharmacol 2024; 15:1447403. [PMID: 39130641 PMCID: PMC11310015 DOI: 10.3389/fphar.2024.1447403] [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: 06/11/2024] [Accepted: 07/01/2024] [Indexed: 08/13/2024] Open
Abstract
Severe COVID-19 cases often progress to life-threatening conditions such as acute respiratory distress syndrome (ARDS), sepsis, and multiple organ dysfunction syndrome (MODS). Gelsolin (GSN), an actin-binding protein with anti-inflammatory and immunomodulatory properties, is a promising therapeutic target for severe COVID-19. Plasma GSN levels are significantly decreased in critical illnesses, including COVID-19, correlating with dysregulated immune responses and poor outcomes. GSN supplementation may mitigate acute lung injury, ARDS, and sepsis, which share pathophysiological features with severe COVID-19, by scavenging actin, modulating cytokine production, enhancing macrophage phagocytosis, and stabilizing the alveolar-capillary barrier. Preliminary data indicate that recombinant human plasma GSN improves oxygenation and lung function in severe COVID-19 patients with ARDS. Although further research is needed to optimize GSN therapy, current evidence supports its potential to mitigate severe consequences of COVID-19 and improve patient outcomes. This review provides a comprehensive analysis of the biological characteristics, mechanisms, and therapeutic value of GSN in severe COVID-19.
Collapse
Affiliation(s)
| | | | | | - Dong Yang
- Department of Anesthesiology at the Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | |
Collapse
|
8
|
Trofimova DN, Aeluri M, Veeranna KD, Jiang Y, Grange RL, Pipaliya BV, Subaramanian M, Craig AW, Evans PA, Allingham JS. Toward a Template for Synthetic Actin-Targeting Macrolide Analogues That Inhibit Cancer Cell Invasiveness. J Med Chem 2024; 67:5315-5332. [PMID: 38401158 DOI: 10.1021/acs.jmedchem.3c01532] [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: 02/26/2024]
Abstract
Actin barbed end-binding macrolides have been shown to inhibit cancer cell motility and invasion of extracellular matrix (ECM), evoking their potential utility as therapies for metastatic cancers. Unfortunately, the direct use of these compounds in clinical settings is impeded by their limited natural abundance, challenging total synthesis, and detrimental effects on normal tissues. To develop potent analogues of these compounds that are simpler to synthesize and compatible with cell-specific targeting systems, such as antibodies, we designed over 20 analogues of the acyclic side chain (tail) of the macrolide Mycalolide B. These analogues probed the contributions of four distinct regions of the tail towards the inhibition of actin polymerization and ECM invasion by human lung cancer A549 cells. We observed that two of these regions tolerate considerable substituent variability, and we identified a specific combination of substituents that leads to the optimal inhibition of the ECM invasion activity of A549 cells.
Collapse
Affiliation(s)
- Daria N Trofimova
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart Street, Kingston, ON K7L 3N6, Canada
| | - Madhu Aeluri
- Department of Chemistry, Queen's University, 90 Bader Lane, Kingston, ON K7L 3N6, Canada
| | - Kirana D Veeranna
- Department of Chemistry, Queen's University, 90 Bader Lane, Kingston, ON K7L 3N6, Canada
| | - Yun Jiang
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart Street, Kingston, ON K7L 3N6, Canada
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, 18 Stuart Street, Kingston, ON K7L 3N6, Canada
| | - Rebecca L Grange
- Department of Chemistry, Queen's University, 90 Bader Lane, Kingston, ON K7L 3N6, Canada
| | - Bhavin V Pipaliya
- Department of Chemistry, Queen's University, 90 Bader Lane, Kingston, ON K7L 3N6, Canada
| | - Murugan Subaramanian
- Department of Chemistry, Queen's University, 90 Bader Lane, Kingston, ON K7L 3N6, Canada
| | - Andrew W Craig
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart Street, Kingston, ON K7L 3N6, Canada
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, 18 Stuart Street, Kingston, ON K7L 3N6, Canada
| | - P Andrew Evans
- Department of Chemistry, Queen's University, 90 Bader Lane, Kingston, ON K7L 3N6, Canada
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. of China
| | - John S Allingham
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart Street, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
9
|
Vyatchin IG, Shevchenko UV. Gelsolin from mussel's catch muscle. Biochem Biophys Res Commun 2023; 688:149221. [PMID: 37976813 DOI: 10.1016/j.bbrc.2023.149221] [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: 10/26/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023]
Abstract
Proteins of the gelsolin family are Ca2+-dependent, multifunctional, actin-binding proteins containing three (S1-S3, about 40 kDa) or six (S1-S6, about 80 kDa) highly conserved repeats in the amino acid sequence. The pattern of interaction of these proteins with actin is complex: they can sever actin filaments; promote polymer nucleation after binding to two actin monomers; and cap the growing barbed end of actin filaments. In the present study, an actin polymerizing factor (46 kDa) from the adductor muscle of a bivalve mollusc has been discovered and identified for the first time. This protein has turned out to belong to the gelsolin family of actin regulatory proteins. The expression of gelsolin-like proteins in the tissues of bivalves was predicted after analyzing their proteome, but this is the first study where an actually expressed protein has been found. A primary determination of its physicochemical properties such as molecular weight, charge, resistance to urea, influence on actin polymerization by viscosity, and light scattering is carried out and the molecular structure analyzed.
Collapse
Affiliation(s)
- Ilya G Vyatchin
- Laboratory of Cell Biophysics, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, ul. Palchevskogo 17, Vladivostok, 690041, Russia.
| | - Ulyana V Shevchenko
- Laboratory of Cell Biophysics, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, ul. Palchevskogo 17, Vladivostok, 690041, Russia
| |
Collapse
|
10
|
Meenakshi S I, Rao M, Mayor S, Sowdhamini R. A census of actin-associated proteins in humans. Front Cell Dev Biol 2023; 11:1168050. [PMID: 37187613 PMCID: PMC10175787 DOI: 10.3389/fcell.2023.1168050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 03/31/2023] [Indexed: 05/17/2023] Open
Abstract
Actin filaments help in maintaining the cell structure and coordinating cellular movements and cargo transport within the cell. Actin participates in the interaction with several proteins and also with itself to form the helical filamentous actin (F-actin). Actin-binding proteins (ABPs) and actin-associated proteins (AAPs) coordinate the actin filament assembly and processing, regulate the flux between globular G-actin and F-actin in the cell, and help maintain the cellular structure and integrity. We have used protein-protein interaction data available through multiple sources (STRING, BioGRID, mentha, and a few others), functional annotation, and classical actin-binding domains to identify actin-binding and actin-associated proteins in the human proteome. Here, we report 2482 AAPs and present an analysis of their structural and sequential domains, functions, evolutionary conservation, cellular localization, abundance, and tissue-specific expression patterns. This analysis provides a base for the characterization of proteins involved in actin dynamics and turnover in the cell.
Collapse
Affiliation(s)
| | - Madan Rao
- National Centre for Biological Sciences, TIFR, Bangalore, India
| | - Satyajit Mayor
- National Centre for Biological Sciences, TIFR, Bangalore, India
| | - Ramanathan Sowdhamini
- National Centre for Biological Sciences, TIFR, Bangalore, India
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
- *Correspondence: Ramanathan Sowdhamini,
| |
Collapse
|
11
|
Gineste C, Youhanna S, Vorrink SU, Henriksson S, Hernández A, Cheng AJ, Chaillou T, Buttgereit A, Schneidereit D, Friedrich O, Hultenby K, Bruton JD, Ivarsson N, Sandblad L, Lauschke VM, Westerblad H. Enzymatically dissociated muscle fibers display rapid dedifferentiation and impaired mitochondrial calcium control. iScience 2022; 25:105654. [PMID: 36479146 PMCID: PMC9720020 DOI: 10.1016/j.isci.2022.105654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/19/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Cells rapidly lose their physiological phenotype upon disruption of their extracellular matrix (ECM)-intracellular cytoskeleton interactions. By comparing adult mouse skeletal muscle fibers, isolated either by mechanical dissection or by collagenase-induced ECM digestion, we investigated acute effects of ECM disruption on cellular and mitochondrial morphology, transcriptomic signatures, and Ca2+ handling. RNA-sequencing showed striking differences in gene expression patterns between the two isolation methods with enzymatically dissociated fibers resembling myopathic phenotypes. Mitochondrial appearance was grossly similar in the two groups, but 3D electron microscopy revealed shorter and less branched mitochondria following enzymatic dissociation. Repeated contractions resulted in a prolonged mitochondrial Ca2+ accumulation in enzymatically dissociated fibers, which was partially prevented by cyclophilin inhibitors. Of importance, muscle fibers of mice with severe mitochondrial myopathy show pathognomonic mitochondrial Ca2+ accumulation during repeated contractions and this accumulation was concealed with enzymatic dissociation, making this an ambiguous method in studies of native intracellular Ca2+ fluxes.
Collapse
Affiliation(s)
- Charlotte Gineste
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sabine U. Vorrink
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sara Henriksson
- Umeå Core Facility for Electron Microscopy, Department of Chemistry, Umeå University, 901 87 Umeå, Sweden
| | - Andrés Hernández
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Arthur J. Cheng
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Thomas Chaillou
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Andreas Buttgereit
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Dominik Schneidereit
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Kjell Hultenby
- Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Huddinge, Sweden
| | - Joseph D. Bruton
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Niklas Ivarsson
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Linda Sandblad
- Umeå Core Facility for Electron Microscopy, Department of Chemistry, Umeå University, 901 87 Umeå, Sweden
| | - Volker M. Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Håkan Westerblad
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
12
|
Actin-Binding Proteins in Cardiac Hypertrophy. Cells 2022; 11:cells11223566. [PMID: 36428995 PMCID: PMC9688942 DOI: 10.3390/cells11223566] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
The heart reacts to a large number of pathological stimuli through cardiac hypertrophy, which finally can lead to heart failure. However, the molecular mechanisms of cardiac hypertrophy remain elusive. Actin participates in the formation of highly differentiated myofibrils under the regulation of actin-binding proteins (ABPs), which provides a structural basis for the contractile function and morphological change in cardiomyocytes. Previous studies have shown that the functional abnormality of ABPs can contribute to cardiac hypertrophy. Here, we review the function of various actin-binding proteins associated with the development of cardiac hypertrophy, which provides more references for the prevention and treatment of cardiomyopathy.
Collapse
|
13
|
Structural and biochemical evidence for the emergence of a calcium-regulated actin cytoskeleton prior to eukaryogenesis. Commun Biol 2022; 5:890. [PMID: 36045281 PMCID: PMC9433394 DOI: 10.1038/s42003-022-03783-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/01/2022] [Indexed: 11/18/2022] Open
Abstract
Charting the emergence of eukaryotic traits is important for understanding the characteristics of organisms that contributed to eukaryogenesis. Asgard archaea and eukaryotes are the only organisms known to possess regulated actin cytoskeletons. Here, we determined that gelsolins (2DGels) from Lokiarchaeota (Loki) and Heimdallarchaeota (Heim) are capable of regulating eukaryotic actin dynamics in vitro and when expressed in eukaryotic cells. The actin filament severing and capping, and actin monomer sequestering, functionalities of 2DGels are strictly calcium controlled. We determined the X-ray structures of Heim and Loki 2DGels bound actin monomers. Each structure possesses common and distinct calcium-binding sites. Loki2DGel has an unusual WH2-like motif (LVDV) between its two gelsolin domains, in which the aspartic acid coordinates a calcium ion at the interface with actin. We conclude that the calcium-regulated actin cytoskeleton predates eukaryogenesis and emerged in the predecessors of the last common ancestor of Loki, Heim and Thorarchaeota. Calcium-regulated actin filament assembly predates eukaryogenesis and was present in the last common ancestor of Asgard archaea Loki, Heim, and Thorarchaeota.
Collapse
|
14
|
The p53 and Calcium Regulated Actin Rearrangement in Model Cells. Int J Mol Sci 2022; 23:ijms23169078. [PMID: 36012344 PMCID: PMC9408879 DOI: 10.3390/ijms23169078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/04/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Long-term cellular stress maintains high intracellular Ca2+ concentrations which ultimately initiates apoptosis. Our interest is focused on how the gelsolin (GSN) and junctional mediating and regulating Y protein (JMY) play important roles in stress response. Both of these proteins can bind p53 and actin. We investigated using in vitro fluorescence spectroscopy and found that the p53 competes with actin in GSN to inhibit p53–JMY complex formation. A high Ca2+ level initializes p53 dimerization; the dimer competes with actin on JMY, which can lead to p53–JMY cotransport into the nucleus. Here we investigated how the motility and division rate of HeLa cells changes due to low-voltage electroporation of GSN or JMY in scratching assays. We revealed that JMY inhibits their motion, but that it can accelerate the cell division. GSN treatment slows down cell division but does not affect cell motility. HeLa cells fully recovered the gap 20 h after the electroporation with JMY and then started to release from the glass slides. Taken together, our in vitro results indicate that GSN and JMY may play an important role in the cellular stress response.
Collapse
|
15
|
Gao J, Nakamura F. Actin-Associated Proteins and Small Molecules Targeting the Actin Cytoskeleton. Int J Mol Sci 2022; 23:2118. [PMID: 35216237 PMCID: PMC8880164 DOI: 10.3390/ijms23042118] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023] Open
Abstract
Actin-associated proteins (AAPs) act on monomeric globular actin (G-actin) and polymerized filamentous actin (F-actin) to regulate their dynamics and architectures which ultimately control cell movement, shape change, division; organelle localization and trafficking. Actin-binding proteins (ABPs) are a subset of AAPs. Since actin was discovered as a myosin-activating protein (hence named actin) in 1942, the protein has also been found to be expressed in non-muscle cells, and numerous AAPs continue to be discovered. This review article lists all of the AAPs discovered so far while also allowing readers to sort the list based on the names, sizes, functions, related human diseases, and the dates of discovery. The list also contains links to the UniProt and Protein Atlas databases for accessing further, related details such as protein structures, associated proteins, subcellular localization, the expression levels in cells and tissues, mutations, and pathology. Because the actin cytoskeleton is involved in many pathological processes such as tumorigenesis, invasion, and developmental diseases, small molecules that target actin and AAPs which hold potential to treat these diseases are also listed.
Collapse
Affiliation(s)
| | - Fumihiko Nakamura
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China;
| |
Collapse
|
16
|
Hoyer M, Crevenna AH, Correia JRC, Quezada AG, Lamb DC. Zero-mode waveguides visualize the first steps during gelsolin-mediated actin filament formation. Biophys J 2022; 121:327-335. [PMID: 34896371 PMCID: PMC8790234 DOI: 10.1016/j.bpj.2021.12.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 06/29/2021] [Accepted: 12/07/2021] [Indexed: 01/21/2023] Open
Abstract
Actin filament dynamics underlie key cellular processes. Although the elongation of actin filaments has been extensively studied, the mechanism of nucleation remains unclear. The micromolar concentrations needed for filament formation have prevented direct observation of nucleation dynamics on the single molecule level. To overcome this limitation, we have used the attoliter excitation volume of zero-mode waveguides to directly monitor the early steps of filament assembly. Immobilizing single gelsolin molecules as a nucleator at the bottom of the zero-mode waveguide, we could visualize the actin filament nucleation process. The process is surprisingly dynamic, and two distinct populations during gelsolin-mediated nucleation are observed. The two populations are defined by the stability of the actin dimers and determine whether elongation occurs. Furthermore, by using an inhibitor to block flattening, a conformational change in actin associated with filament formation, elongation was prevented. These observations indicate that a conformational transition and pathway competition determine the nucleation of gelsolin-mediated actin filament formation.
Collapse
Affiliation(s)
- Maria Hoyer
- Department of Chemistry, Center for NanoScience, Nanosystems Initiative Munich (NIM) and Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians University Munich, Munich, Germany
| | - Alvaro H. Crevenna
- Department of Chemistry, Center for NanoScience, Nanosystems Initiative Munich (NIM) and Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians University Munich, Munich, Germany,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal,Corresponding author
| | - Jose Rafael Cabral Correia
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Andrea G. Quezada
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Don C. Lamb
- Department of Chemistry, Center for NanoScience, Nanosystems Initiative Munich (NIM) and Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians University Munich, Munich, Germany,Corresponding author
| |
Collapse
|
17
|
Vemula V, Huber T, Ušaj M, Bugyi B, Månsson A. Myosin and gelsolin cooperate in actin filament severing and actomyosin motor activity. J Biol Chem 2020; 296:100181. [PMID: 33303625 PMCID: PMC7948409 DOI: 10.1074/jbc.ra120.015863] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/05/2020] [Accepted: 12/10/2020] [Indexed: 01/06/2023] Open
Abstract
Actin is a major intracellular protein with key functions in cellular motility, signaling, and structural rearrangements. Its dynamic behavior, such as polymerization and depolymerization of actin filaments in response to intracellular and extracellular cues, is regulated by an abundance of actin binding proteins. Out of these, gelsolin is one of the most potent for filament severing. However, myosin motor activity also fragments actin filaments through motor-induced forces, suggesting that these two proteins could cooperate to regulate filament dynamics and motility. To test this idea, we used an in vitro motility assay, where actin filaments are propelled by surface-adsorbed heavy meromyosin (HMM) motor fragments. This allows studies of both motility and filament dynamics using isolated proteins. Gelsolin, at both nanomolar and micromolar Ca2+ concentration, appreciably enhanced actin filament severing caused by HMM-induced forces at 1 mM MgATP, an effect that was increased at higher HMM motor density. This finding is consistent with cooperativity between actin filament severing by myosin-induced forces and by gelsolin. We also observed reduced sliding velocity of the HMM-propelled filaments in the presence of gelsolin, providing further support of myosin-gelsolin cooperativity. Total internal reflection fluorescence microscopy–based single molecule studies corroborated that the velocity reduction was a direct effect of gelsolin binding to the filament and revealed different filament severing pattern of stationary and HMM propelled filaments. Overall, the results corroborate cooperative effects between gelsolin-induced alterations in the actin filaments and changes due to myosin motor activity leading to enhanced F-actin severing of possible physiological relevance.
Collapse
Affiliation(s)
- Venukumar Vemula
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Tamás Huber
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary
| | - Marko Ušaj
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Beáta Bugyi
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary.
| | - Alf Månsson
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden.
| |
Collapse
|
18
|
Pintér R, Huber T, Bukovics P, Gaszler P, Vig AT, Tóth MÁ, Gazsó-Gerhát G, Farkas D, Migh E, Mihály J, Bugyi B. The Activities of the Gelsolin Homology Domains of Flightless-I in Actin Dynamics. Front Mol Biosci 2020; 7:575077. [PMID: 33033719 PMCID: PMC7509490 DOI: 10.3389/fmolb.2020.575077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Flightless-I is a unique member of the gelsolin superfamily alloying six gelsolin homology domains and leucine-rich repeats. Flightless-I is an established regulator of the actin cytoskeleton, however, its biochemical activities in actin dynamics are still largely elusive. To better understand the biological functioning of Flightless-I we studied the actin activities of Drosophila Flightless-I by in vitro bulk fluorescence spectroscopy and single filament fluorescence microscopy, as well as in vivo genetic approaches. Flightless-I was found to interact with actin and affects actin dynamics in a calcium-independent fashion in vitro. Our work identifies the first three gelsolin homology domains (1–3) of Flightless-I as the main actin-binding site; neither the other three gelsolin homology domains (4–6) nor the leucine-rich repeats bind actin. Flightless-I inhibits polymerization by high-affinity (∼nM) filament barbed end capping, moderately facilitates nucleation by low-affinity (∼μM) monomer binding, and does not sever actin filaments. Our work reveals that in the presence of profilin Flightless-I is only able to cap actin filament barbed ends but fails to promote actin assembly. In line with the in vitro data, while gelsolin homology domains 4–6 have no effect on in vivo actin polymerization, overexpression of gelsolin homology domains 1–3 prevents the formation of various types of actin cables in the developing Drosophila egg chambers. We also show that the gelsolin homology domains 4–6 of Flightless-I interact with the C-terminus of Drosophila Disheveled-associated activator of morphogenesis formin and negatively regulates its actin assembly activity.
Collapse
Affiliation(s)
- Réka Pintér
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary
| | - Tamás Huber
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Bukovics
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Gaszler
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary
| | - Andrea Teréz Vig
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary
| | - Mónika Ágnes Tóth
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary
| | - Gabriella Gazsó-Gerhát
- Biological Research Centre Szeged, Institute of Genetics, Szeged, Hungary.,Faculty of Science and Informatics, Doctoral School in Biology, University of Szeged, Szeged, Hungary
| | - Dávid Farkas
- Biological Research Centre Szeged, Institute of Genetics, Szeged, Hungary
| | - Ede Migh
- Biological Research Centre Szeged, Institute of Genetics, Szeged, Hungary
| | - József Mihály
- Biological Research Centre Szeged, Institute of Genetics, Szeged, Hungary
| | - Beáta Bugyi
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary.,Szentágothai Research Center, Pécs, Hungary
| |
Collapse
|
19
|
Novel inter-domain Ca2+-binding site in the gelsolin superfamily protein fragmin. J Muscle Res Cell Motil 2019; 41:153-162. [DOI: 10.1007/s10974-019-09571-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 12/14/2019] [Indexed: 12/30/2022]
|
20
|
Abstract
In the disease familial amyloidosis, Finnish type (FAF) the mechanism by which point mutations in gelsolin domain 2 (G2) lead to furin cleavage is not understood for the intact protein. Here, we determine that FAF mutants adopt similar conformations to the wild-type protein. However, the mutations appear to affect the dynamics of domain:domain interactions. Thus, proper domain:domain interactions are needed to protect G2 from protease cleavage. We make mutations in the following domain (G3) that functionally mimic the FAF mutations in G2. We conclude that G2 is on the limits of stability, and perturbations that affect domain:domain stabilizing interactions tip the balance toward cleavage. These data explain how multiple FAF mutations give rise to amyloid formation. In the disease familial amyloidosis, Finnish type (FAF), also known as AGel amyloidosis (AGel), the mechanism by which point mutations in the calcium-regulated actin-severing protein gelsolin lead to furin cleavage is not understood in the intact protein. Here, we provide a structural and biochemical characterization of the FAF variants. X-ray crystallography structures of the FAF mutant gelsolins demonstrate that the mutations do not significantly disrupt the calcium-free conformations of gelsolin. Small-angle X-ray–scattering (SAXS) studies indicate that the FAF calcium-binding site mutants are slower to activate, whereas G167R is as efficient as the wild type. Actin-regulating studies of the gelsolins at the furin cleavage pH (6.5) show that the mutant gelsolins are functional, suggesting that they also adopt relatively normal active conformations. Deletion of gelsolin domains leads to sensitization to furin cleavage, and nanobody-binding protects against furin cleavage. These data indicate instability in the second domain of gelsolin (G2), since loss or gain of G2-stabilizing interactions impacts the efficiency of cleavage by furin. To demonstrate this principle, we engineered non-FAF mutations in G3 that disrupt the G2-G3 interface in the calcium-activated structure. These mutants led to increased furin cleavage. We carried out molecular dynamics (MD) simulations on the FAF and non-FAF mutant G2-G3 fragments of gelsolin. All mutants showed an increase in the distance between the center of masses of the 2 domains (G2 and G3). Since G3 covers the furin cleavage site on G2 in calcium-activated gelsolin, this suggests that destabilization of this interface is a critical step in cleavage.
Collapse
|
21
|
Szatmári D, Xue B, Kannan B, Burtnick LD, Bugyi B, Nyitrai M, Robinson RC. ATP competes with PIP2 for binding to gelsolin. PLoS One 2018; 13:e0201826. [PMID: 30086165 PMCID: PMC6080781 DOI: 10.1371/journal.pone.0201826] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/23/2018] [Indexed: 01/08/2023] Open
Abstract
Gelsolin is a severing and capping protein that targets filamentous actin and regulates filament lengths near plasma membranes, contributing to cell movement and plasma membrane morphology. Gelsolin binds to the plasma membrane via phosphatidylinositol 4,5-bisphosphate (PIP2) in a state that cannot cap F-actin, and gelsolin-capped actin filaments are uncapped by PIP2 leading to filament elongation. The process by which gelsolin is removed from PIP2 at the plasma membrane is currently unknown. Gelsolin also binds ATP with unknown function. Here we characterize the role of ATP on PIP2-gelsolin complex dynamics. Fluorophore-labeled PIP2 and ATP were used to study their interactions with gelsolin using steady-state fluorescence anisotropy, and Alexa488-labeled gelsolin was utilized to reconstitute the regulation of gelsolin binding to PIP2-containing phospholipid vesicles by ATP. Under physiological salt conditions ATP competes with PIP2 for binding to gelsolin, while calcium causes the release of ATP from gelsolin. These data suggest a cycle for gelsolin activity. Firstly, calcium activates ATP-bound gelsolin allowing it to sever and cap F-actin. Secondly, PIP2-binding removes the gelsolin cap from F-actin at low calcium levels, leading to filament elongation. Finally, ATP competes with PIP2 to release the calcium-free ATP-bound gelsolin, allowing it to undergo a further round of severing.
Collapse
Affiliation(s)
- Dávid Szatmári
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
- University of Pécs, Medical School, Department of Biophysics, Pécs, Hungary
| | - Bo Xue
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
- Department of Biochemistry, National University of Singapore, Singapore, Singapore
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Balakrishnan Kannan
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Leslie D. Burtnick
- Department of Chemistry and Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Beáta Bugyi
- University of Pécs, Medical School, Department of Biophysics, Pécs, Hungary
- Szentágothai Research Center, Pécs, Hungary
| | - Miklós Nyitrai
- University of Pécs, Medical School, Department of Biophysics, Pécs, Hungary
- Szentágothai Research Center, Pécs, Hungary
| | - Robert C. Robinson
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
- Department of Biochemistry, National University of Singapore, Singapore, Singapore
- Research Institute for Interdisciplinary Science, Okayama University, Okayama, Japan
| |
Collapse
|
22
|
Rao J, Ashraf S, Tan W, van der Ven AT, Gee HY, Braun DA, Fehér K, George SP, Esmaeilniakooshkghazi A, Choi WI, Jobst-Schwan T, Schneider R, Schmidt JM, Widmeier E, Warejko JK, Hermle T, Schapiro D, Lovric S, Shril S, Daga A, Nayir A, Shenoy M, Tse Y, Bald M, Helmchen U, Mir S, Berdeli A, Kari JA, El Desoky S, Soliman NA, Bagga A, Mane S, Jairajpuri MA, Lifton RP, Khurana S, Martins JC, Hildebrandt F. Advillin acts upstream of phospholipase C ϵ1 in steroid-resistant nephrotic syndrome. J Clin Invest 2017; 127:4257-4269. [PMID: 29058690 DOI: 10.1172/jci94138] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 09/19/2017] [Indexed: 11/17/2022] Open
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is a frequent cause of chronic kidney disease. Here, we identified recessive mutations in the gene encoding the actin-binding protein advillin (AVIL) in 3 unrelated families with SRNS. While all AVIL mutations resulted in a marked loss of its actin-bundling ability, truncation of AVIL also disrupted colocalization with F-actin, thereby leading to impaired actin binding and severing. Additionally, AVIL colocalized and interacted with the phospholipase enzyme PLCE1 and with the ARP2/3 actin-modulating complex. Knockdown of AVIL in human podocytes reduced actin stress fibers at the cell periphery, prevented recruitment of PLCE1 to the ARP3-rich lamellipodia, blocked EGF-induced generation of diacylglycerol (DAG) by PLCE1, and attenuated the podocyte migration rate (PMR). These effects were reversed by overexpression of WT AVIL but not by overexpression of any of the 3 patient-derived AVIL mutants. The PMR was increased by overexpression of WT Avil or PLCE1, or by EGF stimulation; however, this increased PMR was ameliorated by inhibition of the ARP2/3 complex, indicating that ARP-dependent lamellipodia formation occurs downstream of AVIL and PLCE1 function. Together, these results delineate a comprehensive pathogenic axis of SRNS that integrates loss of AVIL function with alterations in the action of PLCE1, an established SRNS protein.
Collapse
Affiliation(s)
- Jia Rao
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Medicine, Nephrology, Children's Hospital of Fudan University, Shanghai, China
| | - Shazia Ashraf
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Weizhen Tan
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Amelie T van der Ven
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Heon Yung Gee
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Daniela A Braun
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Krisztina Fehér
- NMR and Structure Analysis Group, Department of Organic and Macromolecular Chemistry, University of Gent, Gent, Belgium
| | - Sudeep P George
- Department of Biology and Biochemistry, University of Houston, Houston,Texas, USA
| | | | - Won-Il Choi
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tilman Jobst-Schwan
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ronen Schneider
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Johanna Magdalena Schmidt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Eugen Widmeier
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jillian K Warejko
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tobias Hermle
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David Schapiro
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Svjetlana Lovric
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shirlee Shril
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ankana Daga
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ahmet Nayir
- Department of Pediatric Nephrology, Faculty of Medicine, University of Istanbul, Istanbul, Turkey
| | - Mohan Shenoy
- Department of Pediatric Nephrology, Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Yincent Tse
- Department of Pediatric Nephrology, Great North Children's Hospital, Newcastle Upon Tyne, United Kingdom
| | - Martin Bald
- Olga Children's Hospital, Clinic Stuttgart, Stuttgart, Germany
| | - Udo Helmchen
- Institute of Pathology, Kidney Registry, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Sevgi Mir
- Department of Pediatrics, Molecular Medicine Laboratory, Ege University, Izmir, Turkey
| | - Afig Berdeli
- Department of Pediatrics, Molecular Medicine Laboratory, Ege University, Izmir, Turkey
| | - Jameela A Kari
- Pediatric Nephrology Center of Excellence and Pediatric Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sherif El Desoky
- Pediatric Nephrology Center of Excellence and Pediatric Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Neveen A Soliman
- Department of Pediatrics, Center of Pediatric Nephrology and Transplantation, Kasr Al Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Arvind Bagga
- Division of Pediatric Nephrology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Shrikant Mane
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Seema Khurana
- Department of Biology and Biochemistry, University of Houston, Houston,Texas, USA.,Baylor College of Medicine, Houston, Texas, USA
| | - Jose C Martins
- NMR and Structure Analysis Group, Department of Organic and Macromolecular Chemistry, University of Gent, Gent, Belgium
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Gelsolin dysfunction causes photoreceptor loss in induced pluripotent cell and animal retinitis pigmentosa models. Nat Commun 2017; 8:271. [PMID: 28814713 PMCID: PMC5559447 DOI: 10.1038/s41467-017-00111-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/02/2017] [Indexed: 12/27/2022] Open
Abstract
Mutations in the Retinitis Pigmentosa GTPase Regulator (RPGR) cause X-linked RP (XLRP), an untreatable, inherited retinal dystrophy that leads to premature blindness. RPGR localises to the photoreceptor connecting cilium where its function remains unknown. Here we show, using murine and human induced pluripotent stem cell models, that RPGR interacts with and activates the actin-severing protein gelsolin, and that gelsolin regulates actin disassembly in the connecting cilium, thus facilitating rhodopsin transport to photoreceptor outer segments. Disease-causing RPGR mutations perturb this RPGR-gelsolin interaction, compromising gelsolin activation. Both RPGR and Gelsolin knockout mice show abnormalities of actin polymerisation and mislocalisation of rhodopsin in photoreceptors. These findings reveal a clinically-significant role for RPGR in the activation of gelsolin, without which abnormalities in actin polymerisation in the photoreceptor connecting cilia cause rhodopsin mislocalisation and eventual retinal degeneration in XLRP. Mutations in the Retinitis Pigmentosa GTPase Regulator (RPGR) cause retinal dystrophy, but how this arises at a molecular level is unclear. Here, the authors show in induced pluripotent stem cells and mouse knockouts that RPGR mediates actin dynamics in photoreceptors via the actin-severing protein, gelsolin.
Collapse
|
24
|
Visualizing Temperature Mediated Activation of Gelsolin and Its Deactivation By Pip 2: A Saxs Based Study. Sci Rep 2017; 7:4670. [PMID: 28680082 PMCID: PMC5498565 DOI: 10.1038/s41598-017-04975-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 05/23/2017] [Indexed: 11/21/2022] Open
Abstract
This is the first report describing temperature based initiation of gelsolin’s F-actin depolymerization activity, even in absence of free Ca2+ or low pH. Small angle X-ray scattering (SAXS) and circular dichroism (CD) studies revealed that temperature in the range of 30–40 °C is capable of opening the G1 domain alone, as remaining domains are held together by the Ca2+-sensitive C-tail latch without any loss in the secondary structural content. Full opening of all domains of tail-less gelsolin, and retention of closed shape for G2–G6 gelsolin merely by heating, further substantiated our findings. The Ca2+/pH independent activity of gelsolin near physiological temperature brought out a query: whether gelsolin is always active, and if not, what might deactivate it? Earlier, PIP2 has been reported to render gelsolin inactive with no structural insight. Reduction in shape parameters and modeling revealed that PIP2 reverses the temperature induced extension of g1-g2 linker leading to a compact shape seen for Ca2+-free gelsolin. Similar results for partially activated gelsolin (by low pH or Ca2+ ions below 0.1 μM) imply that inside cells, depolymerization, capping, and nucleation of F-actin by gelsolin is regulated by the culmination of local Ca2+ ion concentration, pH, temperature and PIP2 levels.
Collapse
|
25
|
Verhelle A, Nair N, Everaert I, Van Overbeke W, Supply L, Zwaenepoel O, Peleman C, Van Dorpe J, Lahoutte T, Devoogdt N, Derave W, Chuah MK, VandenDriessche T, Gettemans J. AAV9 delivered bispecific nanobody attenuates amyloid burden in the gelsolin amyloidosis mouse model. Hum Mol Genet 2017; 26:1353-1364. [PMID: 28334940 DOI: 10.1093/hmg/ddx056] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/07/2017] [Indexed: 12/23/2022] Open
Abstract
Gelsolin amyloidosis is a dominantly inherited, incurable type of amyloidosis. A single point mutation in the gelsolin gene (G654A is most common) results in the loss of a Ca2+ binding site in the second gelsolin domain. Consequently, this domain partly unfolds and exposes an otherwise buried furin cleavage site at the surface. During secretion of mutant plasma gelsolin consecutive cleavage by furin and MT1-MMP results in the production of 8 and 5 kDa amyloidogenic peptides. Nanobodies that are able to (partly) inhibit furin or MT1-MMP proteolysis have previously been reported. In this study, the nanobodies have been combined into a single bispecific format able to simultaneously shield mutant plasma gelsolin from intracellular furin and extracellular MT1-MMP activity. We report the successful in vivo expression of this bispecific nanobody following adeno-associated virus serotype 9 gene therapy in gelsolin amyloidosis mice. Using SPECT/CT and immunohistochemistry, a reduction in gelsolin amyloid burden was detected which translated into improved muscle contractile properties. We conclude that a nanobody-based gene therapy using adeno-associated viruses shows great potential as a novel strategy in gelsolin amyloidosis and potentially other amyloid diseases.
Collapse
Affiliation(s)
- Adriaan Verhelle
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Nisha Nair
- Department of Gene Therapy & Regenerative Medicine, Free University of Brussels (VUB), Brussels, Belgium
| | - Inge Everaert
- Department of Movement and Sport Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Wouter Van Overbeke
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Lynn Supply
- Department of Medical and Forensic Pathology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Cindy Peleman
- In Vivo Cellular and Molecular Imaging Laboratory, Free University of Brussels (VUB), Brussels, Belgium
| | - Jo Van Dorpe
- Department of Medical and Forensic Pathology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Tony Lahoutte
- In Vivo Cellular and Molecular Imaging Laboratory, Free University of Brussels (VUB), Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Free University of Brussels (VUB), Brussels, Belgium
| | - Wim Derave
- Department of Movement and Sport Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Marinee K Chuah
- Department of Gene Therapy & Regenerative Medicine, Free University of Brussels (VUB), Brussels, Belgium.,Department of Cardiovascular Sciences, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - Thierry VandenDriessche
- Department of Gene Therapy & Regenerative Medicine, Free University of Brussels (VUB), Brussels, Belgium.,Department of Cardiovascular Sciences, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - Jan Gettemans
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
26
|
Thiruketheeswaran P, Thomalla P, Krüger E, Hinssen H, D'Haese J. Four paralog gelsolin genes are differentially expressed in the earthworm Lumbricus terrestris. Comp Biochem Physiol B Biochem Mol Biol 2017; 208-209:58-67. [PMID: 28400331 DOI: 10.1016/j.cbpb.2017.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/04/2017] [Accepted: 04/06/2017] [Indexed: 11/25/2022]
Abstract
We have identified and characterized four distinct variants of the gelsolin-related protein (EWAM P1-P4) in the earthworm L. terrestris. All of these proteins biochemically qualify as gelsolins since they sever actin filaments in a calcium dependent manner. P1, P2 and P3 are present in the Lumbricus body wall muscle whereas in the gizzard muscle P3 and P4 were found. P1-P4 are encoded by four paralog genes and are differentially expressed in various muscle cell tissues. While the genes for P1 and P2 contain one intron, there was no intron in both P3 and P4 genes. The coding sequences consist of 1104bp (368 amino acids) for P1/P4 and 1101bp (367 amino acids) for P2/P3. Corresponding genes were confirmed by northern blot analysis which revealed three (calculated lengths: 3100, 2300 and 2100 nucleotides) and two (calculated lengths: 2300 and 1700 nucleotides) mRNA transcripts in the body wall and the gizzard, respectively. EWAM mRNA was localized by fluorescence in situ hybridization in the body wall and the gizzard muscle. P1 mRNA was detected in the inner proximal layers of both the circular and longitudinal muscle of the body wall whereas in the gizzard no significant staining was observed for P1. P2-P4 mRNAs were abundant in the outer distal layers of both the circular and the longitudinal muscles of both body wall and gizzard. The differential expression of four paralog gelsolin genes suggests a functional adaptation of different muscle cells with respect to actin filament turnover and modulation of its polymer state.
Collapse
Affiliation(s)
- Prasath Thiruketheeswaran
- Institute for Cell Biology, Department Biology, Heinrich-Heine-University Düsseldorf, Universitätsstrasse 1, D-40225 Düsseldorf, Germany
| | - Paul Thomalla
- Institute for Cell Biology, Department Biology, Heinrich-Heine-University Düsseldorf, Universitätsstrasse 1, D-40225 Düsseldorf, Germany
| | - Evelyn Krüger
- Institute for Cell Biology, Department Biology, Heinrich-Heine-University Düsseldorf, Universitätsstrasse 1, D-40225 Düsseldorf, Germany
| | - Horst Hinssen
- Biochemical Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, D-33615 Bielefeld, Germany
| | - Jochen D'Haese
- Institute for Cell Biology, Department Biology, Heinrich-Heine-University Düsseldorf, Universitätsstrasse 1, D-40225 Düsseldorf, Germany.
| |
Collapse
|
27
|
Thiruketheeswaran P, Greven H, D'Haese J. Gelsolin in Onychophora and Tardigrada with notes on its variability in the Ecdysozoa. Comp Biochem Physiol B Biochem Mol Biol 2016; 203:47-52. [PMID: 27627778 DOI: 10.1016/j.cbpb.2016.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/31/2016] [Accepted: 09/08/2016] [Indexed: 01/25/2023]
Abstract
Rearrangements of the filamentous actin network involve a broad range of actin binding proteins. Among these, the gelsolin proteins sever actin filaments, cap their fast growing end and nucleate actin assembly in a calcium-dependent manner. Here, we focus on the gelsolin of the onychophoran Peripatoides novaezealandiae and the eutardigrade Hypsibius dujardini. From the cDNA of P. novaezealandiae we obtained the complete coding sequence with an open reading frame of 2178bp. It encodes a protein of 726 amino acids with a calculated molecular mass of 82,610.9Da and a pI of 5.57. This sequence is comprised of six segments (S1-S6). However, analysis of data from TardiBase reveals that the gelsolin of the eutardigrade Hypsibius dujardini has only three segments (S1-S3). The coding sequence consist of 1119bp for 373 amino acids with a calculated molecular mass of 42,440.95Da and a pI of 6.17. The Peripatoides and Hypsibius gelsolin revealed both conserved binding motifs for G-actin, F-actin and phosphatidylinositol 4,5-bisphosphate (PIP2), along with a full set of type-1 and type-2 Ca2+-binding sites which could result in the binding of eight and four calcium ions, respectively. Both gelsolin proteins lack a C-terminal latch-helix indicating a more rapid activation in the submicromolar Ca2+ range. We suggest that a gelsolin with three segments was present in the last common ancestor of the ecdysozoan clade Panarthropoda (Onychophora, Tardigrada, Arthropoda), primarily because the gelsolin of all non-Ecdysozoa studied so far (except Chordata) reveals this number of segments. Mapping of our molecular data onto a well-established phylogeny revealed that the number of gelsolin segments does not correlate with the phylogenetic lineage but rather with particular functional demands to alter the kinetics of actin polymerization.
Collapse
Affiliation(s)
- Prasath Thiruketheeswaran
- Institute for Cell Biology, Department Biology, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, D-40225 Düsseldorf, Germany
| | - Hartmut Greven
- Institute for Cell Biology, Department Biology, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, D-40225 Düsseldorf, Germany
| | - Jochen D'Haese
- Institute for Cell Biology, Department Biology, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, D-40225 Düsseldorf, Germany.
| |
Collapse
|
28
|
Pothana L, Devi L, Venna NK, Pentakota N, Varma VP, Jose J, Goel S. Replacement of serum with ocular fluid for cryopreservation of immature testes. Cryobiology 2016; 73:356-366. [PMID: 27693391 DOI: 10.1016/j.cryobiol.2016.09.169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/26/2016] [Accepted: 09/26/2016] [Indexed: 11/25/2022]
Abstract
Cryopreservation of immature testis is a feasible approach for germplasm preservation of male animals. Combinations of dimethyl sulfoxide (DMSO) and foetal bovine serum (FBS) are used for testis cryopreservation. However, an alternative to FBS is needed, because FBS is expensive. Buffalo ocular fluid (BuOF), a slaughter house by-product, could be an economical option. The objective of the present study was to assess whether BuOF can replace FBS for cryopreservation of immature mouse (Mus musculus), rat (Rattus norvegicus), and buffalo (Bubalus bubalis) testes. Results showed that rodent and buffalo testes frozen in DMSO (10% for rodents and 20% for buffalo) with 20% FBS or BuOF had similar numbers of viable and DNA-damaged cells (P > 0.05). The expression of cell proliferation- (PCNA) and apoptosis-specific proteins (Annexin V and BAX/BCL2 ratio) were also comparable in mouse and buffalo testes frozen in DMSO with FBS or BuOF (P > 0.05). Interestingly, rat testis frozen in DMSO with BuOF had lower expression of Annexin V protein than testis frozen in DMSO with FBS (P < 0.05). The percentage of meiotic germ cells (pachytene-stage spermatocytes) in xenografts from testis frozen either in DMSO with BuOF or FBS did not significantly differ in rats or buffalo (P > 0.05). These findings provide evidence that BuOF has potential to replace FBS for cryopreservation of immature rodent and buffalo testis. Further investigation is needed to explore whether BuOF can replace FBS for testis cryopreservation of other species.
Collapse
Affiliation(s)
- Lavanya Pothana
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Lalitha Devi
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Naresh Kumar Venna
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Niharika Pentakota
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Vivek Phani Varma
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Jedy Jose
- Animal House, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India
| | - Sandeep Goel
- Laboratory for the Conservation of Endangered Species, Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Uppal Road, Hyderabad, 500 007, India.
| |
Collapse
|
29
|
Zhang S, Liu C, Wang J, Ren Z, Staiger CJ, Ren H. A Processive Arabidopsis Formin Modulates Actin Filament Dynamics in Association with Profilin. MOLECULAR PLANT 2016; 9:900-10. [PMID: 26996265 DOI: 10.1016/j.molp.2016.03.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 02/28/2016] [Accepted: 03/04/2016] [Indexed: 05/08/2023]
Abstract
Formins are conserved regulators of actin cytoskeletal organization and dynamics that have been implicated to be important for cell division and cell polarity. The mechanism by which diverse formins regulate actin dynamics in plants is still not well understood. Using in vitro single-molecule imaging technology, we directly observed that the FH1-FH2 domain of an Arabidopsis thaliana formin, AtFH14, processively attaches to the barbed end of actin filaments as a dimer and slows their elongation rate by 90%. The attachment persistence of FH1-FH2 is concentration dependent. Furthermore, by use of the triple-color total internal reflection fluorescence microscopy, we found that ABP29, a barbed-end capping protein, competes with FH1-FH2 at the filament barbed end, where its binding is mutually exclusive with AtFH14. In the presence of different plant profilin isoforms, FH1-FH2 enhances filament elongation rates from about 10 to 42 times. Filaments buckle when FH1-FH2 is anchored specifically to cover slides, further indicating that AtFH14 moves processively on the elongating barbed end. At high concentration, AtFH14 bundles actin filaments randomly into antiparallel or parallel spindle-like structures; however, the FH1-FH2-mediated bundles become thinner and longer in the presence of plant profilins. This is the direct demonstration of a processive formin from plants. Our results also illuminate the molecular mechanism of AtFH14 in regulating actin dynamics via association with profilin.
Collapse
Affiliation(s)
- Sha Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Chang Liu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Jiaojiao Wang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Zhanhong Ren
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Christopher J Staiger
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Research Building, West Lafayette, IN 47907-2064, USA
| | - Haiyun Ren
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, People's Republic of China.
| |
Collapse
|
30
|
Regulation of the Postsynaptic Compartment of Excitatory Synapses by the Actin Cytoskeleton in Health and Its Disruption in Disease. Neural Plast 2016; 2016:2371970. [PMID: 27127658 PMCID: PMC4835652 DOI: 10.1155/2016/2371970] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 03/09/2016] [Indexed: 02/07/2023] Open
Abstract
Disruption of synaptic function at excitatory synapses is one of the earliest pathological changes seen in wide range of neurological diseases. The proper control of the segregation of neurotransmitter receptors at these synapses is directly correlated with the intact regulation of the postsynaptic cytoskeleton. In this review, we are discussing key factors that regulate the structure and dynamics of the actin cytoskeleton, the major cytoskeletal building block that supports the postsynaptic compartment. Special attention is given to the complex interplay of actin-associated proteins that are found in the synaptic specialization. We then discuss our current understanding of how disruption of these cytoskeletal elements may contribute to the pathological events observed in the nervous system under disease conditions with a particular focus on Alzheimer's disease pathology.
Collapse
|
31
|
Svanström A, Grantham J. The molecular chaperone CCT modulates the activity of the actin filament severing and capping protein gelsolin in vitro. Cell Stress Chaperones 2016; 21:55-62. [PMID: 26364302 PMCID: PMC4679748 DOI: 10.1007/s12192-015-0637-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/26/2015] [Accepted: 08/31/2015] [Indexed: 10/23/2022] Open
Abstract
The oligomeric molecular chaperone CCT is essential for the folding of the highly abundant protein actin, which in its native state forms actin filaments that generate the traction forces required for cell motility. In addition to folding proteins, CCT can provide a platform for protein complex assembly and binds actin filaments assembled in vitro. Some individual subunits of CCT, when monomeric, have been shown to be functionally active, and in particular, the CCTepsilon subunit is involved in the serum response factor pathway that controls actin transcription. Thus, there is a complex interplay between CCT and actin that extends beyond actin folding. CCT has recently been shown to bind gelsolin, an actin filament severing protein that increases actin dynamics by generating filament ends for further actin polymerization. However, the biological significance of the CCT:gelsolin interaction is unknown. Here, using a co-immunoprecipitation assay, we show that CCT binds directly to gelsolin in its calcium-activated, actin-severing conformation. Furthermore, using actin filaments retained from fixed and permeabilized cells, we demonstrate that CCT can inhibit the actin filament severing activity of gelsolin. As our work and that of others shows gelsolin is not folded by CCT, the CCT:gelsolin interaction represents a novel mode of binding where CCT may modulate protein activity. The data presented here reveal an additional level of interplay between CCT and actin mediated via gelsolin, suggesting that CCT may influence processes depending on gelsolin activity, such as cell motility.
Collapse
Affiliation(s)
- Andreas Svanström
- Department of Chemistry and Molecular Biology, University of Gothenburg, Medicinaregatan 9C, 40530, Gothenburg, Sweden
| | - Julie Grantham
- Department of Chemistry and Molecular Biology, University of Gothenburg, Medicinaregatan 9C, 40530, Gothenburg, Sweden.
| |
Collapse
|
32
|
Qian D, Nan Q, Yang Y, Li H, Zhou Y, Zhu J, Bai Q, Zhang P, An L, Xiang Y. Gelsolin-Like Domain 3 Plays Vital Roles in Regulating the Activities of the Lily Villin/Gelsolin/Fragmin Superfamily. PLoS One 2015; 10:e0143174. [PMID: 26587673 PMCID: PMC4654503 DOI: 10.1371/journal.pone.0143174] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 11/02/2015] [Indexed: 02/08/2023] Open
Abstract
The villin/gelsolin/fragmin superfamily is a major group of Ca2+-dependent actin-binding proteins (ABPs) involved in various cellular processes. Members of this superfamily typically possess three or six tandem gelsolin-like (G) domains, and each domain plays a distinct role in actin filament dynamics. Although the activities of most G domains have been characterized, the biochemical function of the G3 domain remains poorly understood. In this study, we carefully compared the detailed biochemical activities of ABP29 (a new member of this family that contains the G1-G2 domains of lily ABP135) and ABP135G1-G3 (which contains the G1-G3 domains of lily ABP135). In the presence of high Ca2+ levels in vitro (200 and 10 μM), ABP135G1-G3 exhibited greater actin severing and/or depolymerization and nucleating activities than ABP29, and these proteins had similar actin capping activities. However, in the presence of low levels of Ca2+ (41 nM), ABP135G1-G3 had a weaker capping activity than ABP29. In addition, ABP29 inhibited F-actin depolymerization, as shown by dilution-mediated depolymerization assay, differing from the typical superfamily proteins. In contrast, ABP135G1-G3 accelerated F-actin depolymerization. All of these results demonstrate that the G3 domain plays specific roles in regulating the activities of the lily villin/gelsolin/fragmin superfamily proteins.
Collapse
Affiliation(s)
- Dong Qian
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Qiong Nan
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yueming Yang
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Hui Li
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yuelong Zhou
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jingen Zhu
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Qifeng Bai
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Pan Zhang
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Lizhe An
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yun Xiang
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
33
|
Calcium-controlled conformational choreography in the N-terminal half of adseverin. Nat Commun 2015; 6:8254. [PMID: 26365202 PMCID: PMC4647846 DOI: 10.1038/ncomms9254] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 08/03/2015] [Indexed: 01/23/2023] Open
Abstract
Adseverin is a member of the calcium-regulated gelsolin superfamily of actin-binding proteins. Here we report the crystal structure of the calcium-free N-terminal half of adseverin (iA1-A3) and the Ca(2+)-bound structure of A3, which reveal structural similarities and differences with gelsolin. Solution small-angle X-ray scattering combined with ensemble optimization revealed a dynamic Ca(2+)-dependent equilibrium between inactive, intermediate and active conformations. Increasing calcium concentrations progressively shift this equilibrium from a main population of inactive conformation to the active form. Molecular dynamics simulations of iA1-A3 provided insights into Ca(2+)-induced destabilization, implicating a critical role for the A2 type II calcium-binding site and the A2A3 linker in the activation process. Finally, mutations that disrupt the A1/A3 interface increase Ca(2+)-independent F-actin severing by A1-A3, albeit at a lower efficiency than observed for gelsolin domains G1-G3. Together, these data address the calcium dependency of A1-A3 activity in relation to the calcium-independent activity of G1-G3.
Collapse
|
34
|
Varma VP, Devi L, Venna NK, Murthy CLN, Idris MM, Goel S. Ocular Fluid As a Replacement for Serum in Cell Cryopreservation Media. PLoS One 2015; 10:e0131291. [PMID: 26135924 PMCID: PMC4489643 DOI: 10.1371/journal.pone.0131291] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 06/01/2015] [Indexed: 11/19/2022] Open
Abstract
Cryostorage is of immense interest in biomedical research, especially for stem cell-based therapies and fertility preservation. Several protocols have been developed for efficient cryopreservation of cells and tissues, and a combination of dimethyl sulfoxide (DMSO) and fetal bovine serum (FBS) is commonly used. However, there is a need for an alternative to FBS because of ethical reasons, high cost, and risk of contamination with blood-borne diseases. The objective of the present study was to examine the possibility of using buffalo (Bubalus bubalis) ocular fluid (BuOF) to replace FBS in cryomedia. Frozen-thawed cells, which were cryopreserved in a cryomedia with BuOF, were assessed for viability, early and late apoptosis, and proliferation. Three cell lines (CHO, HEK, and C18-4), mouse embryonic stem (mES) cells, and primary cells, such as mouse embryonic fibroblast (MEF) cells, human peripheral blood mononuclear cells (hPBMCs), and mouse bone marrow cells (mBMCs), were cryopreserved in cryomedia containing 10% DMSO (D10) with 20% FBS (D10S20) or D10 with 20% BuOF (D10O20). For all three cell lines and mES cells cryopreserved in either D10S20 or D10O20, thawed cells showed no difference in cell viability or cell recovery. Western blot analysis of frozen-thawed-cultured cells revealed that the expression of Annexin V and proliferating cell nuclear antigen (PCNA) proteins, and the ratio of BAX/BCL2 proteins were similar in all three cell lines, mES cells, and hPBMCs cryopreserved in D10S20 and D10O20. However, initial cell viability, cell recovery after culture, and PCNA expression were significantly lower in MEF cells, and the BAX/BCL2 protein ratio was elevated in mBMCs cryopreserved in D10O20. Biochemical and proteomic analysis of BuOF showed the presence of several components that may have roles in imparting the cryoprotective property of BuOF. These results encourage further research to develop an efficient serum-free cryomedia for several cell types using BuOF.
Collapse
Affiliation(s)
- Vivek Phani Varma
- Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Hyderabad, India
| | - Lalitha Devi
- Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Hyderabad, India
| | - Naresh Kumar Venna
- Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Hyderabad, India
| | - Ch Lakshmi N. Murthy
- Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Hyderabad, India
| | - Mohammed M. Idris
- Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Hyderabad, India
| | - Sandeep Goel
- Centre for Cellular and Molecular Biology, Council for Scientific and Industrial Research, Hyderabad, India
- * E-mail:
| |
Collapse
|
35
|
Lee WL, Grimes JM, Robinson RC. Yersinia effector YopO uses actin as bait to phosphorylate proteins that regulate actin polymerization. Nat Struct Mol Biol 2015; 22:248-55. [PMID: 25664724 DOI: 10.1038/nsmb.2964] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/30/2014] [Indexed: 11/09/2022]
Abstract
Pathogenic Yersinia species evade host immune systems through the injection of Yersinia outer proteins (Yops) into phagocytic cells. One Yop, YopO, also known as YpkA, induces actin-filament disruption, impairing phagocytosis. Here we describe the X-ray structure of Yersinia enterocolitica YopO in complex with actin, which reveals that YopO binds to an actin monomer in a manner that blocks polymerization yet allows the bound actin to interact with host actin-regulating proteins. SILAC-MS and biochemical analyses confirm that actin-polymerization regulators such as VASP, EVL, WASP, gelsolin and the formin diaphanous 1 are directly sequestered and phosphorylated by YopO through formation of ternary complexes with actin. This leads to a model in which YopO at the membrane sequesters actin from polymerization while using the bound actin as bait to recruit, phosphorylate and misregulate host actin-regulating proteins to disrupt phagocytosis.
Collapse
Affiliation(s)
- Wei Lin Lee
- 1] Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore. [2] Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Jonathan M Grimes
- 1] Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK. [2] Diamond Light Source, Oxfordshire, UK
| | - Robert C Robinson
- 1] Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore. [2] Department of Biochemistry, National University of Singapore, Singapore
| |
Collapse
|
36
|
Van Overbeke W, Wongsantichon J, Everaert I, Verhelle A, Zwaenepoel O, Loonchanta A, Burtnick LD, De Ganck A, Hochepied T, Haigh J, Cuvelier C, Derave W, Robinson RC, Gettemans J. An ER-directed gelsolin nanobody targets the first step in amyloid formation in a gelsolin amyloidosis mouse model. Hum Mol Genet 2015; 24:2492-507. [PMID: 25601851 DOI: 10.1093/hmg/ddv010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/14/2015] [Indexed: 01/01/2023] Open
Abstract
Hereditary gelsolin amyloidosis is an autosomal dominantly inherited amyloid disorder. A point mutation in the GSN gene (G654A being the most common one) results in disturbed calcium binding by the second gelsolin domain (G2). As a result, the folding of G2 is hampered, rendering the mutant plasma gelsolin susceptible to a proteolytic cascade. Consecutive cleavage by furin and MT1-MMP-like proteases generates 8 and 5 kDa amyloidogenic peptides that cause neurological, ophthalmological and dermatological findings. To this day, no specific treatment is available to counter the pathogenesis. Using GSN nanobody 11 as a molecular chaperone, we aimed to protect mutant plasma gelsolin from furin proteolysis in the trans-Golgi network. We report a transgenic, GSN nanobody 11 secreting mouse that was used for crossbreeding with gelsolin amyloidosis mice. Insertion of the therapeutic nanobody gene into the gelsolin amyloidosis mouse genome resulted in improved muscle contractility. X-ray crystal structure determination of the gelsolin G2:Nb11 complex revealed that Nb11 does not directly block the furin cleavage site. We conclude that nanobodies can be used to shield substrates from aberrant proteolysis and this approach might establish a novel therapeutic strategy in amyloid diseases.
Collapse
Affiliation(s)
| | - Jantana Wongsantichon
- Institute of Molecular and Cellular Biology, A*STAR, Biopolis, Singapore 138673, Singapore
| | - Inge Everaert
- Department of Movement and Sport Sciences, Faculty of Medicine and Health Sciences
| | - Adriaan Verhelle
- Department of Biochemistry, Faculty of Medicine and Health Sciences
| | | | - Anantasak Loonchanta
- Department of Chemistry and Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leslie D Burtnick
- Department of Chemistry and Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ariane De Ganck
- Department of Biochemistry, Faculty of Medicine and Health Sciences
| | - Tino Hochepied
- Department for Molecular Biomedical Research, VIB, Ghent, Belgium, Department of Biomedical Molecular Biology and
| | - Jody Haigh
- Department of Biomedical Molecular Biology and Vascular Cell Biology Unit, VIB Inflammation Research Centre, Ghent, Belgium and Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Department of Clinical Haematology, Monash University and Alfred Health Centre, Melbourne, Australia
| | - Claude Cuvelier
- Department of Pathology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Wim Derave
- Department of Movement and Sport Sciences, Faculty of Medicine and Health Sciences
| | - Robert C Robinson
- Institute of Molecular and Cellular Biology, A*STAR, Biopolis, Singapore 138673, Singapore, Department of Biochemistry, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore,
| | - Jan Gettemans
- Department of Biochemistry, Faculty of Medicine and Health Sciences,
| |
Collapse
|
37
|
Povarova OI, Uversky VN, Kuznetsova IM, Turoverov KK. Actinous enigma or enigmatic actin: Folding, structure, and functions of the most abundant eukaryotic protein. INTRINSICALLY DISORDERED PROTEINS 2014; 2:e34500. [PMID: 28232879 PMCID: PMC5314930 DOI: 10.4161/idp.34500] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 08/12/2014] [Accepted: 08/13/2014] [Indexed: 02/06/2023]
Abstract
Being the most abundant protein of the eukaryotic cell, actin continues to keep its secrets for more than 60 years. Everything about this protein, its structure, functions, and folding, is mysteriously counterintuitive, and this review represents an attempt to solve some of the riddles and conundrums commonly found in the field of actin research. In fact, actin is a promiscuous binder with a wide spectrum of biological activities. It can exist in at least three structural forms, globular, fibrillar, and inactive (G-, F-, and I-actin, respectively). G-actin represents a thermodynamically instable, quasi-stationary state, which is formed in vivo as a result of the energy-intensive, complex posttranslational folding events controlled and driven by cellular folding machinery. The G-actin structure is dependent on the ATP and Mg2+ binding (which in vitro is typically substituted by Ca2+) and protein is easily converted to the I-actin by the removal of metal ions and by action of various denaturing agents (pH, temperature, and chemical denaturants). I-actin cannot be converted back to the G-form. Foldable and “natively folded” forms of actin are always involved in interactions either with the specific protein partners, such as Hsp70 chaperone, prefoldin, and the CCT chaperonin during the actin folding in vivo or with Mg2+ and ATP as it takes place in the G-form. We emphasize that the solutions for the mysteries of actin multifunctionality, multistructurality, and trapped unfolding can be found in the quasi-stationary nature of this enigmatic protein, which clearly possesses many features attributed to both globular and intrinsically disordered proteins.
Collapse
Affiliation(s)
- Olga I Povarova
- Laboratory of structural dynamics, stability, and folding of proteins; Institute of Cytology; Russian Academy of Sciences; St. Petersburg, Russia
| | - Vladimir N Uversky
- Laboratory of structural dynamics, stability, and folding of proteins; Institute of Cytology; Russian Academy of Sciences; St. Petersburg, Russia; Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute; Morsani College of Medicine; University of South Florida; Tampa, FL USA; Institute for Biological Instrumentation; Russian Academy of Sciences; Pushchino, Russia; Biology Department; Faculty of Science; King Abdulaziz University; Jeddah, Kingdom of Saudi Arabia
| | - Irina M Kuznetsova
- Laboratory of structural dynamics, stability, and folding of proteins; Institute of Cytology; Russian Academy of Sciences; St. Petersburg, Russia; St. Petersburg State Polytechnical University; St. Petersburg, Russia
| | - Konstantin K Turoverov
- Laboratory of structural dynamics, stability, and folding of proteins; Institute of Cytology; Russian Academy of Sciences; St. Petersburg, Russia; St. Petersburg State Polytechnical University; St. Petersburg, Russia
| |
Collapse
|
38
|
Single-molecule force spectroscopy reveals force-enhanced binding of calcium ions by gelsolin. Nat Commun 2014; 5:4623. [PMID: 25100107 PMCID: PMC4143929 DOI: 10.1038/ncomms5623] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 07/08/2014] [Indexed: 01/30/2023] Open
Abstract
Force is increasingly recognized as an important element in controlling biological processes. Forces can deform native protein conformations leading to protein-specific effects. Protein–protein binding affinities may be decreased, or novel protein–protein interaction sites may be revealed, on mechanically stressing one or more components. Here we demonstrate that the calcium-binding affinity of the sixth domain of the actin-binding protein gelsolin (G6) can be enhanced by mechanical force. Our kinetic model suggests that the calcium-binding affinity of G6 increases exponentially with force, up to the point of G6 unfolding. This implies that gelsolin may be activated at lower calcium ion levels when subjected to tensile forces. The demonstration that cation–protein binding affinities can be force-dependent provides a new understanding of the complex behaviour of cation-regulated proteins in stressful cellular environments, such as those found in the cytoskeleton-rich leading edge and at cell adhesions. The application of force can influence biological processes such as ligand and protein–protein binding, with mechanical stress typically hindering such interactions. Here, the authors use atomic force microscopy to show that the binding of calcium to gelsolin can be improved under stress.
Collapse
|
39
|
Chaperone nanobodies protect gelsolin against MT1-MMP degradation and alleviate amyloid burden in the gelsolin amyloidosis mouse model. Mol Ther 2014; 22:1768-78. [PMID: 25023329 DOI: 10.1038/mt.2014.132] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 07/07/2014] [Indexed: 01/11/2023] Open
Abstract
Gelsolin amyloidosis is an autosomal dominant incurable disease caused by a point mutation in the GSN gene (G654A/T), specifically affecting secreted plasma gelsolin. Incorrect folding of the mutant (D187N/Y) second gelsolin domain leads to a pathological proteolytic cascade. D187N/Y gelsolin is first cleaved by furin in the trans-Golgi network, generating a 68 kDa fragment (C68). Upon secretion, C68 is cleaved by MT1-MMP-like proteases in the extracellular matrix, releasing 8 kDa and 5 kDa amyloidogenic peptides which aggregate in multiple tissues and cause disease-associated symptoms. We developed nanobodies that recognize the C68 fragment, but not native wild type gelsolin, and used these as molecular chaperones to mitigate gelsolin amyloid buildup in a mouse model that recapitulates the proteolytic cascade. We identified gelsolin nanobodies that potently reduce C68 proteolysis by MT1-MMP in vitro. Converting these nanobodies into an albumin-binding format drastically increased their serum half-life in mice, rendering them suitable for intraperitoneal injection. A 12-week treatment schedule of heterozygote D187N gelsolin transgenic mice with recombinant bispecific gelsolin-albumin nanobody significantly decreased gelsolin buildup in the endomysium and concomitantly improved muscle contractile properties. These findings demonstrate that nanobodies may be of considerable value in the treatment of gelsolin amyloidosis and related diseases.
Collapse
|
40
|
The proteome of the differentiating mesencephalic progenitor cell line CSM14.1 in vitro. BIOMED RESEARCH INTERNATIONAL 2014; 2014:351821. [PMID: 24592386 PMCID: PMC3925624 DOI: 10.1155/2014/351821] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 12/16/2013] [Indexed: 11/23/2022]
Abstract
The treatment of Parkinson's disease by transplantation of dopaminergic (DA) neurons from human embryonic mesencephalic tissue is a promising approach. However, the origin of these cells causes major problems: availability and standardization of the graft. Therefore, the generation of unlimited numbers of DA neurons from various types of stem or progenitor cells has been brought into focus. A source for DA neurons might be conditionally immortalized progenitor cells. The temperature-sensitive immortalized cell line CSM14.1 derived from the mesencephalon of an embryonic rat has been used successfully for transplantation experiments. This cell line was analyzed by unbiased stereology of cell type specific marker proteins and 2D-gel electrophoresis followed by mass spectrometry to characterize the differentially expressed proteome. Undifferentiated CSM14.1 cells only expressed the stem cell marker nestin, whereas differentiated cells expressed GFAP or NeuN and tyrosine hydroxylase. An increase of the latter cells during differentiation could be shown. By using proteomics an explanation on the protein level was found for the observed changes in cell morphology during differentiation, when CSM14.1 cells possessed the morphology of multipolar neurons. The results obtained in this study confirm the suitability of CSM14.1 cells as an in vitro model for the study of neuronal and dopaminergic differentiation in rats.
Collapse
|
41
|
Ghoshdastider U, Popp D, Burtnick LD, Robinson RC. The expanding superfamily of gelsolin homology domain proteins. Cytoskeleton (Hoboken) 2013; 70:775-95. [DOI: 10.1002/cm.21149] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 10/11/2013] [Accepted: 10/02/2013] [Indexed: 12/29/2022]
Affiliation(s)
- Umesh Ghoshdastider
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science; Technology and Research); Biopolis 138673 Singapore
| | - David Popp
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science; Technology and Research); Biopolis 138673 Singapore
| | - Leslie D. Burtnick
- Department of Chemistry and Centre for Blood Research; Life Sciences Institute; University of British Columbia; Vancouver British Columbia V6T 1Z1 Canada
| | - Robert C. Robinson
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science; Technology and Research); Biopolis 138673 Singapore
- Department of Biochemistry; National University of Singapore; Singapore 117597 Singapore
- School of Biological Sciences; Nanyang Technological University; Singapore 637551 Singapore
| |
Collapse
|
42
|
Guardians of the actin monomer. Eur J Cell Biol 2013; 92:316-32. [DOI: 10.1016/j.ejcb.2013.10.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 10/03/2013] [Accepted: 10/23/2013] [Indexed: 11/22/2022] Open
|
43
|
Peddada N, Sagar A, Rathore YS, Choudhary V, Pattnaik UBK, Khatri N, Garg R, Ashish. Global shapes of F-actin depolymerization-competent minimal gelsolins: insight into the role of g2-g3 linker in pH/Ca2+ insensitivity of the first half. J Biol Chem 2013; 288:28266-82. [PMID: 23940055 DOI: 10.1074/jbc.m113.463224] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Because of its ability to rapidly depolymerize F-actin, plasma gelsolin has emerged as a therapeutic molecule in different disease conditions. High amounts of exogenous gelsolin are, however, required to treat animal models of different diseases. Knowing that the F-actin depolymerizing property of gelsolin resides in its N terminus, we made several truncated versions of plasma gelsolin. The smaller versions, particularly the one composed of the first 28-161 residues, depolymerized the F-actin much faster than the native gelsolin and other truncates at the same molar ratios. Although G1-G3 loses its dependence on Ca(2+) or low pH for the actin depolymerization function, interestingly, G1-G2 and its smaller versions were found to regain this requirement. Small angle x-ray scattering-based shape reconstructions revealed that G1-G3 adopts an open shape in both the presence and the absence of Ca(2+) as well as low pH, whereas G1-G2 and residues 28-161 prefer collapsed states in Ca(2+)-free conditions at pH 8. The mutations in the g2-g3 linker resulted in the calcium sensitivity of the mutant G1-G3 for F-actin depolymerization activity, although the F-actin-binding sites remained exposed in the mutant G1-G3 as well as in the smaller truncates even in the Ca(2+)-free conditions at pH 8. Furthermore, unlike wild type G1-G3, calcium-sensitive mutants of G1-G3 acquired closed shapes in the absence of free calcium, implying a role of g2-g3 linker in determining the open F-actin depolymerizing-competent shape of G1-G3 in this condition. We demonstrate that the mobility of the G1 domain, essential for F-actin depolymerization, is indirectly regulated by the gelsolin-like sequence of g2-g3 linker.
Collapse
Affiliation(s)
- Nagesh Peddada
- From the Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh 160036, India
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Isoforms of gelsolin from lobster striated muscles differ in Calcium-dependence. Arch Biochem Biophys 2013; 536:38-45. [DOI: 10.1016/j.abb.2013.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Revised: 05/07/2013] [Accepted: 05/10/2013] [Indexed: 11/30/2022]
|
45
|
Khaitlina S, Fitz H, Hinssen H. The interaction of gelsolin with tropomyosin modulates actin dynamics. FEBS J 2013; 280:4600-11. [PMID: 23844991 DOI: 10.1111/febs.12431] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 06/24/2013] [Accepted: 07/05/2013] [Indexed: 12/19/2022]
Abstract
We have investigated the interactions between the actin-binding proteins gelsolin and tropomyosin, with special respect to any effects on the functional properties of gelsolin. Limited proteolysis indicated that the loop connecting the gelsolin domains G3 and G4 is involved in tropomyosin binding. Under nonpolymerizing conditions, binding of tropomyosin neither prevented the formation of a 2: 1actin-gelsolin complex, nor did it affect the nucleating activity of gelsolin in actin polymerization, likely as a result of competitive displacement of tropomyosin from gelsolin. To evaluate the effect of tropomyosin on the actin filament severing activity of gelsolin, we measured both filamentous actin (F-actin) viscosity and the relative number concentrations of filaments after fragmentation, either by gelsolin alone or by gelsolin-tropomyosin complexes. The interaction of gelsolin with tropomyosin caused a reduction in F-actin severing activity of up to 80% compared to gelsolin alone. Thus, being bound to gelsolin, tropomyosin prevented gelsolin from severing actin filaments. By contrast, the severing activity of gelsolin for F-actin/tropomyosin was similar to that for F-actin alone even at a tropomyosin : actin saturation ratio of 1: 7. Thus, when bound to actin filaments, tropomyosin did not significantly inhibit the severing of filaments by gelsolin. The interaction between gelsolin and tropomyosin was largely independent of the muscle actin and tropomyosin isoforms investigated. The results obtained in the present study suggest that tropomyosin is involved in the modulation of actin dynamics not via the protection of filaments against severing, but rather by binding gelsolin in solution to prevent it from severing and to promote the formation of new actin filaments.
Collapse
Affiliation(s)
- Sofia Khaitlina
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia
| | | | | |
Collapse
|
46
|
Joseph N, Reicher B, Barda-Saad M. The calcium feedback loop and T cell activation: how cytoskeleton networks control intracellular calcium flux. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:557-68. [PMID: 23860253 DOI: 10.1016/j.bbamem.2013.07.009] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 06/30/2013] [Accepted: 07/08/2013] [Indexed: 12/31/2022]
Abstract
During T cell activation, the engagement of a T cell with an antigen-presenting cell (APC) results in rapid cytoskeletal rearrangements and a dramatic increase of intracellular calcium (Ca(2+)) concentration, downstream to T cell antigen receptor (TCR) ligation. These events facilitate the organization of an immunological synapse (IS), which supports the redistribution of receptors, signaling molecules and organelles towards the T cell-APC interface to induce downstream signaling events, ultimately supporting T cell effector functions. Thus, Ca(2+) signaling and cytoskeleton rearrangements are essential for T cell activation and T cell-dependent immune response. Rapid release of Ca(2+) from intracellular stores, e.g. the endoplasmic reticulum (ER), triggers the opening of Ca(2+) release-activated Ca(2+) (CRAC) channels, residing in the plasma membrane. These channels facilitate a sustained influx of extracellular Ca(2+) across the plasma membrane in a process termed store-operated Ca(2+) entry (SOCE). Because CRAC channels are themselves inhibited by Ca(2+) ions, additional factors are suggested to enable the sustained Ca(2+) influx required for T cell function. Among these factors, we focus here on the contribution of the actin and microtubule cytoskeleton. The TCR-mediated increase in intracellular Ca(2+) evokes a rapid cytoskeleton-dependent polarization, which involves actin cytoskeleton rearrangements and microtubule-organizing center (MTOC) reorientation. Here, we review the molecular mechanisms of Ca(2+) flux and cytoskeletal rearrangements, and further describe the way by which the cytoskeletal networks feedback to Ca(2+) signaling by controlling the spatial and temporal distribution of Ca(2+) sources and sinks, modulating TCR-dependent Ca(2+) signals, which are required for an appropriate T cell response. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.
Collapse
Affiliation(s)
- Noah Joseph
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Barak Reicher
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Mira Barda-Saad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| |
Collapse
|
47
|
Nag S, Larsson M, Robinson RC, Burtnick LD. Gelsolin: The tail of a molecular gymnast. Cytoskeleton (Hoboken) 2013; 70:360-84. [DOI: 10.1002/cm.21117] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 05/24/2013] [Indexed: 12/14/2022]
Affiliation(s)
| | - Mårten Larsson
- Institute of Molecular and Cell Biology, A*STAR; Singapore
| | | | - Leslie D. Burtnick
- Department of Chemistry and Centre for Blood Research; Life Sciences Institute, University of British Columbia; Vancouver; British Columbia; Canada
| |
Collapse
|
48
|
Liu Z, Ono S. Regulatory role of the second gelsolin-like domain of Caenorhabditis elegans gelsolin-like protein 1 (GSNL-1) in its calcium-dependent conformation and actin-regulatory activities. Cytoskeleton (Hoboken) 2013; 70:228-39. [PMID: 23475707 DOI: 10.1002/cm.21103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 01/23/2013] [Accepted: 02/25/2013] [Indexed: 12/22/2022]
Abstract
Caenorhabditis elegans gelsolin-like protein-1 (GSNL-1) is an unconventional member of the gelsolin family of actin-regulatory proteins. Unlike typical gelsolin-related proteins with three or six G domains, GSNL-1 has four gelsolin-like (G) domains (G1-G4) and exhibits calcium-dependent actin filament severing and capping activities. The first G domain (G1) of GSNL-1 is necessary for its actin-regulatory activities. However, how other domains in GSNL-1 participate in regulation of its functions is not understood. Here, we report biochemical evidence that the second G domain (G2) of GSNL-1 has a regulatory role in its calcium-dependent conformation and actin-regulatory activities. Comparison of the sequences of gelsolin-related proteins from various species indicates that sequences of G2 are highly conserved. Among the conserved residues in G2, we focused on D162 of GSNL-1, since equivalent residues in gelsolin and severin are part of the calcium-binding sites and is a pathogenic mutation site in human gelsolin causing familial amyloidosis, Finish-type. The D162N mutation does not alter the inactive and fully calcium-activated states of GSNL-1 for actin filament severing (at 20 nM GSNL-1) and capping activities (at 50 nM GSNL-1). However, under these conditions, the mutant shows reduced calcium sensitivity for activation. By contrast, the D162N mutation strongly enhances susceptibility of GSNL-1 to chymotrypsin digestion only at high calcium concentrations but not at low calcium concentrations. The mutation also reduces affinity of GSNL-1 with actin monomers. These results suggest that G2 of GSNL-1 functions as a regulatory domain for its calcium-dependent actin-regulatory activities by mediating conformational changes of the GSNL-1 molecule.
Collapse
Affiliation(s)
- Zhongmei Liu
- Department of Pathology, Emory University, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
49
|
Arora PD, Wang Y, Bresnick A, Dawson J, Janmey PA, McCulloch CA. Collagen remodeling by phagocytosis is determined by collagen substrate topology and calcium-dependent interactions of gelsolin with nonmuscle myosin IIA in cell adhesions. Mol Biol Cell 2013; 24:734-47. [PMID: 23325791 PMCID: PMC3596245 DOI: 10.1091/mbc.e12-10-0754] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cell adhesion to collagen presented on beads activates Ca2+ entry and promotes the formation of phagosomes enriched with NMMIIA and gelsolin. The Ca2+-dependent interaction of gelsolin and NMMIIA in turn enables actin remodeling and enhances collagen degradation by phagocytosis. We examine how collagen substrate topography, free intracellular calcium ion concentration ([Ca2+]i, and the association of gelsolin with nonmuscle myosin IIA (NMMIIA) at collagen adhesions are regulated to enable collagen phagocytosis. Fibroblasts plated on planar, collagen-coated substrates show minimal increase of [Ca2+]i, minimal colocalization of gelsolin and NMMIIA in focal adhesions, and minimal intracellular collagen degradation. In fibroblasts plated on collagen-coated latex beads there are large increases of [Ca2+]i, time- and Ca2+-dependent enrichment of NMMIIA and gelsolin at collagen adhesions, and abundant intracellular collagen degradation. NMMIIA knockdown retards gelsolin recruitment to adhesions and blocks collagen phagocytosis. Gelsolin exhibits tight, Ca2+-dependent binding to full-length NMMIIA. Gelsolin domains G4–G6 selectively require Ca2+ to interact with NMMIIA, which is restricted to residues 1339–1899 of NMMIIA. We conclude that cell adhesion to collagen presented on beads activates Ca2+ entry and promotes the formation of phagosomes enriched with NMMIIA and gelsolin. The Ca2+ -dependent interaction of gelsolin and NMMIIA in turn enables actin remodeling and enhances collagen degradation by phagocytosis.
Collapse
Affiliation(s)
- P D Arora
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | |
Collapse
|
50
|
Interactions between the nucleosome histone core and Arp8 in the INO80 chromatin remodeling complex. Proc Natl Acad Sci U S A 2012; 109:20883-8. [PMID: 23213201 DOI: 10.1073/pnas.1214735109] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Actin-related protein Arp8 is a component of the INO80 chromatin remodeling complex. Yeast Arp8 (yArp8) comprises two domains: a 25-KDa N-terminal domain, found only in yeast, and a 75-KDa C-terminal domain (yArp8CTD) that contains the actin fold and is conserved across other species. The crystal structure shows that yArp8CTD contains three insertions within the actin core. Using a combination of biochemistry and EM, we show that Arp8 forms a complex with nucleosomes, and that the principal interactions are via the H3 and H4 histones, mediated through one of the yArp8 insertions. We show that recombinant yArp8 exists in monomeric and dimeric states, but the dimer is the biologically relevant form required for stable interactions with histones that exploits the twofold symmetry of the nucleosome core. Taken together, these data provide unique insight into the stoichiometry, architecture, and molecular interactions between components of the INO80 remodeling complex and nucleosomes, providing a first step toward building up the structure of the complex.
Collapse
|