51
|
Chase AR, Laudermilch E, Schlieker C. Torsin ATPases: Harnessing Dynamic Instability for Function. Front Mol Biosci 2017; 4:29. [PMID: 28553638 PMCID: PMC5425593 DOI: 10.3389/fmolb.2017.00029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 04/25/2017] [Indexed: 12/11/2022] Open
Abstract
Torsins are essential, disease-relevant AAA+ (ATPases associated with various cellular activities) proteins residing in the endoplasmic reticulum and perinuclear space, where they are implicated in a variety of cellular functions. Recently, new structural and functional details about Torsins have emerged that will have a profound influence on unraveling the precise mechanistic details of their yet-unknown mode of action in the cell. While Torsins are phylogenetically related to Clp/HSP100 proteins, they exhibit comparatively weak ATPase activities, which are tightly controlled by virtue of an active site complementation through accessory cofactors. This control mechanism is offset by a TorsinA mutation implicated in the severe movement disorder DYT1 dystonia, suggesting a critical role for the functional Torsin-cofactor interplay in vivo. Notably, TorsinA lacks aromatic pore loops that are both conserved and critical for the processive unfolding activity of Clp/HSP100 proteins. Based on these distinctive yet defining features, we discuss how the apparent dynamic nature of the Torsin-cofactor system can inform emerging models and hypotheses for Torsin complex formation and function. Specifically, we propose that the dynamic assembly and disassembly of the Torsin/cofactor system is a critical property that is required for Torsins' functional roles in nuclear trafficking and nuclear pore complex assembly or homeostasis that merit further exploration. Insights obtained from these future studies will be a valuable addition to our understanding of disease etiology of DYT1 dystonia.
Collapse
Affiliation(s)
- Anna R Chase
- Department of Molecular Biophysics and Biochemistry, Yale UniversityNew Haven, CT, USA
| | - Ethan Laudermilch
- Department of Molecular Biophysics and Biochemistry, Yale UniversityNew Haven, CT, USA
| | - Christian Schlieker
- Department of Molecular Biophysics and Biochemistry, Yale UniversityNew Haven, CT, USA.,Department of Cell Biology, Yale School of MedicineNew Haven, CT, USA
| |
Collapse
|
52
|
Saunders CA, Harris NJ, Willey PT, Woolums BM, Wang Y, McQuown AJ, Schoenhofen A, Worman HJ, Dauer WT, Gundersen GG, Luxton GWG. TorsinA controls TAN line assembly and the retrograde flow of dorsal perinuclear actin cables during rearward nuclear movement. J Cell Biol 2017; 216:657-674. [PMID: 28242745 PMCID: PMC5350507 DOI: 10.1083/jcb.201507113] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 11/04/2016] [Accepted: 12/21/2016] [Indexed: 12/29/2022] Open
Abstract
The nucleus is positioned toward the rear of most migratory cells. In fibroblasts and myoblasts polarizing for migration, retrograde actin flow moves the nucleus rearward, resulting in the orientation of the centrosome in the direction of migration. In this study, we report that the nuclear envelope-localized AAA+ (ATPase associated with various cellular activities) torsinA (TA) and its activator, the inner nuclear membrane protein lamina-associated polypeptide 1 (LAP1), are required for rearward nuclear movement during centrosome orientation in migrating fibroblasts. Both TA and LAP1 contributed to the assembly of transmembrane actin-associated nuclear (TAN) lines, which couple the nucleus to dorsal perinuclear actin cables undergoing retrograde flow. In addition, TA localized to TAN lines and was necessary for the proper mobility of EGFP-mini-nesprin-2G, a functional TAN line reporter construct, within the nuclear envelope. Furthermore, TA and LAP1 were indispensable for the retrograde flow of dorsal perinuclear actin cables, supporting the recently proposed function for the nucleus in spatially organizing actin flow and cytoplasmic polarity. Collectively, these results identify TA as a key regulator of actin-dependent rearward nuclear movement during centrosome orientation.
Collapse
Affiliation(s)
- Cosmo A Saunders
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455
| | - Nathan J Harris
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455
| | - Patrick T Willey
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455
| | - Brian M Woolums
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455
| | - Yuexia Wang
- Department of Medicine, Columbia University, New York, NY 10032.,Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - Alex J McQuown
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455
| | - Amy Schoenhofen
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455
| | - Howard J Worman
- Department of Medicine, Columbia University, New York, NY 10032.,Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - William T Dauer
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109.,Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - Gregg G Gundersen
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - G W Gant Luxton
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
53
|
Abstract
As a compartment border, the nuclear envelope (NE) needs to serve as both a protective membrane shell for the genome and a versatile communication interface between the nucleus and the cytoplasm. Despite its important structural role in sheltering the genome, the NE is a dynamic and highly adaptable boundary that changes composition during differentiation, deforms in response to mechanical challenges, can be repaired upon rupture and even rapidly disassembles and reforms during open mitosis. NE remodelling is fundamentally involved in cell growth, division and differentiation, and if perturbed can lead to devastating diseases such as muscular dystrophies or premature ageing.
Collapse
|
54
|
Cascalho A, Jacquemyn J, Goodchild RE. Membrane defects and genetic redundancy: Are we at a turning point for DYT1 dystonia? Mov Disord 2016; 32:371-381. [PMID: 27911022 DOI: 10.1002/mds.26880] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/24/2016] [Accepted: 10/29/2016] [Indexed: 12/11/2022] Open
Abstract
Heterozygosity for a 3-base pair deletion (ΔGAG) in TOR1A/torsinA is one of the most common causes of hereditary dystonia. In this review, we highlight current understanding of how this mutation causes disease from research spanning structural biochemistry, cell science, neurobiology, and several model organisms. We now know that homozygosity for ΔGAG has the same effects as Tor1aKO , implicating a partial loss of function mechanism in the ΔGAG/+ disease state. In addition, torsinA loss specifically affects neurons in mice, even though the gene is broadly expressed, apparently because of differential expression of homologous torsinB. Furthermore, certain neuronal subtypes are more severely affected by torsinA loss. Interestingly, these include striatal cholinergic interneurons that display abnormal responses to dopamine in several Tor1a animal models. There is also progress on understanding torsinA molecular cell biology. The structural basis of how ΔGAG inhibits torsinA ATPase activity is defined, although mutant torsinAΔGAG protein also displays some characteristics suggesting it contributes to dystonia by a gain-of-function mechanism. Furthermore, a consistent relationship is emerging between torsin dysfunction and membrane biology, including an evolutionarily conserved regulation of lipid metabolism. Considered together, these findings provide major advances toward understanding the molecular, cellular, and neurobiological pathologies of DYT1/TOR1A dystonia that can hopefully be exploited for new approaches to treat this disease. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ana Cascalho
- Vlaams Instituut voor Biotechnologie Centre for the Biology of Disease, Leuven, Belgium.,KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Julie Jacquemyn
- Vlaams Instituut voor Biotechnologie Centre for the Biology of Disease, Leuven, Belgium.,KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Rose E Goodchild
- Vlaams Instituut voor Biotechnologie Centre for the Biology of Disease, Leuven, Belgium.,KU Leuven, Department of Human Genetics, Leuven, Belgium
| |
Collapse
|
55
|
Laudermilch E, Tsai PL, Graham M, Turner E, Zhao C, Schlieker C. Dissecting Torsin/cofactor function at the nuclear envelope: a genetic study. Mol Biol Cell 2016; 27:3964-3971. [PMID: 27798237 PMCID: PMC5156537 DOI: 10.1091/mbc.e16-07-0511] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/18/2016] [Accepted: 10/20/2016] [Indexed: 01/12/2023] Open
Abstract
Torsins are essential, disease-relevant ATPases, but their function is unknown. Monitoring of nuclear envelope morphology after deletion of multiple Torsins or their cofactors reveals a robust inner nuclear membrane–blebbing phenotype in HeLa cells. Nucleoporins and ubiquitin are defining molecular components of these omega-shaped blebs. The human genome encodes four Torsin ATPases, the functions of which are poorly understood. In this study, we use CRISPR/Cas9 engineering to delete all four Torsin ATPases individually and in combination. Using nuclear envelope (NE) blebbing as a phenotypic measure, we establish a direct correlation between the number of inactivated Torsin alleles and the occurrence of omega-shaped herniations within the lumen of the NE. A similar, although not identical, redundancy is observed for LAP1 and LULL1, which serve as regulatory cofactors for a subset of Torsin ATPases. Unexpectedly, deletion of Tor2A in a TorA/B/3A-deficient background results in a stark increase of bleb formation, even though Tor2A does not respond to LAP1/LULL1 stimulation. The robustness of the observed phenotype in Torsin-deficient cells enables a structural analysis via electron microscopy tomography and a compositional analysis via immunogold labeling. Ubiquitin and nucleoporins were identified as distinctively localizing components of the omega-shaped bleb structure. These findings suggest a functional link between the Torsin/cofactor system and NE/nuclear pore complex biogenesis or homeostasis and establish a Torsin-deficient cell line as a valuable experimental platform with which to decipher Torsin function.
Collapse
Affiliation(s)
- Ethan Laudermilch
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520
| | - Pei-Ling Tsai
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520
| | - Morven Graham
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520
| | - Elizabeth Turner
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520
| | - Chenguang Zhao
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520
| | - Christian Schlieker
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520 .,Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520
| |
Collapse
|
56
|
Tanabe LM, Liang CC, Dauer WT. Neuronal Nuclear Membrane Budding Occurs during a Developmental Window Modulated by Torsin Paralogs. Cell Rep 2016; 16:3322-3333. [PMID: 27653693 PMCID: PMC5061049 DOI: 10.1016/j.celrep.2016.08.044] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/19/2016] [Accepted: 08/14/2016] [Indexed: 01/26/2023] Open
Abstract
DYT1 dystonia is a neurodevelopmental disease that manifests during a discrete period of childhood. The disease is caused by impaired function of torsinA, a protein linked to nuclear membrane budding. The relationship of NE budding to neural development and CNS function is unclear, however, obscuring its potential role in dystonia pathogenesis. We find NE budding begins and resolves during a discrete neurodevelopmental window in torsinA null neurons in vivo. The developmental resolution of NE budding corresponds to increased torsinB protein, while ablating torsinB from torsinA null neurons prevents budding resolution and causes lethal neural dysfunction. Developmental changes in torsinB also correlate with NE bud formation in differentiating DYT1 embryonic stem cells, and overexpression of torsinA or torsinB rescues NE bud formation in this system. These findings identify a torsinA neurodevelopmental window that is essential for normal CNS function and have important implications for dystonia pathogenesis and therapeutics.
Collapse
Affiliation(s)
- Lauren M Tanabe
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chun-Chi Liang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - William T Dauer
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
57
|
Molecular and cellular basis for the unique functioning of Nrf1, an indispensable transcription factor for maintaining cell homoeostasis and organ integrity. Biochem J 2016; 473:961-1000. [PMID: 27060105 DOI: 10.1042/bj20151182] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/26/2016] [Indexed: 12/30/2022]
Abstract
The consensuscis-regulatory AP-1 (activator protein-1)-like AREs (antioxidant-response elements) and/or EpREs (electrophile-response elements) allow for differential recruitment of Nrf1 [NF-E2 (nuclear factor-erythroid 2)-related factor 1], Nrf2 and Nrf3, together with each of their heterodimeric partners (e.g. sMaf, c-Jun, JunD or c-Fos), to regulate different sets of cognate genes. Among them, NF-E2 p45 and Nrf3 are subject to tissue-specific expression in haemopoietic and placental cell lineages respectively. By contrast, Nrf1 and Nrf2 are two important transcription factors expressed ubiquitously in various vertebrate tissues and hence may elicit putative combinational or competitive functions. Nevertheless, they have de facto distinct biological activities because knockout of their genes in mice leads to distinguishable phenotypes. Of note, Nrf2 is dispensable during development and growth, albeit it is accepted as a master regulator of antioxidant, detoxification and cytoprotective genes against cellular stress. Relative to the water-soluble Nrf2, less attention has hitherto been drawn to the membrane-bound Nrf1, even though it has been shown to be indispensable for embryonic development and organ integrity. The biological discrepancy between Nrf1 and Nrf2 is determined by differences in both their primary structures and topovectorial subcellular locations, in which they are subjected to distinct post-translational processing so as to mediate differential expression of ARE-driven cytoprotective genes. In the present review, we focus on the molecular and cellular basis for Nrf1 and its isoforms, which together exert its essential functions for maintaining cellular homoeostasis, normal organ development and growth during life processes. Conversely, dysfunction of Nrf1 results in spontaneous development of non-alcoholic steatohepatitis, hepatoma, diabetes and neurodegenerative diseases in animal models.
Collapse
|
58
|
Grillet M, Dominguez Gonzalez B, Sicart A, Pöttler M, Cascalho A, Billion K, Hernandez Diaz S, Swerts J, Naismith TV, Gounko NV, Verstreken P, Hanson PI, Goodchild RE. Torsins Are Essential Regulators of Cellular Lipid Metabolism. Dev Cell 2016; 38:235-47. [PMID: 27453503 DOI: 10.1016/j.devcel.2016.06.017] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 05/18/2016] [Accepted: 06/12/2016] [Indexed: 01/10/2023]
Abstract
Torsins are developmentally essential AAA+ proteins, and mutation of human torsinA causes the neurological disease DYT1 dystonia. They localize in the ER membranes, but their cellular function remains unclear. We now show that dTorsin is required in Drosophila adipose tissue, where it suppresses triglyceride levels, promotes cell growth, and elevates membrane lipid content. We also see that human torsinA at the inner nuclear membrane is associated with membrane expansion and elevated cellular lipid content. Furthermore, the key lipid metabolizing enzyme, lipin, is mislocalized in dTorsin-KO cells, and dTorsin increases levels of the lipin substrate, phosphatidate, and reduces the product, diacylglycerol. Finally, genetic suppression of dLipin rescues dTorsin-KO defects, including adipose cell size, animal growth, and survival. These findings identify that torsins are essential regulators of cellular lipid metabolism and implicate disturbed lipid biology in childhood-onset DYT1 dystonia.
Collapse
Affiliation(s)
- Micheline Grillet
- VIB Centre for the Biology of Disease, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Beatriz Dominguez Gonzalez
- VIB Centre for the Biology of Disease, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Adria Sicart
- VIB Centre for the Biology of Disease, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Maria Pöttler
- VIB Centre for the Biology of Disease, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Ana Cascalho
- VIB Centre for the Biology of Disease, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Karolien Billion
- VIB Centre for the Biology of Disease, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Sergio Hernandez Diaz
- VIB Centre for the Biology of Disease, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Jef Swerts
- VIB Centre for the Biology of Disease, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Teresa V Naismith
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Natalia V Gounko
- VIB Centre for the Biology of Disease, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium; Electron Microscopy Platform, VIB Bio-Imaging Core, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Patrik Verstreken
- VIB Centre for the Biology of Disease, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Phyllis I Hanson
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Rose E Goodchild
- VIB Centre for the Biology of Disease, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium.
| |
Collapse
|
59
|
Phenotypic lentivirus screens to identify functional single domain antibodies. Nat Microbiol 2016; 1:16080. [PMID: 27573105 PMCID: PMC5010022 DOI: 10.1038/nmicrobiol.2016.80] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/28/2016] [Indexed: 12/02/2022]
Abstract
Manipulation of proteins is key in assessing their in
vivo function. While genetic ablation is straightforward,
reversible and specific perturbation of protein function remains a challenge.
Single domain antibody fragments, such as camelid-derived VHHs, can serve as
inhibitors or activators of intracellular protein function, but functional
testing of identified VHHs is laborious. To address this challenge, we developed
a lentiviral screening approach to identify VHHs that elicit a phenotype when
expressed intracellularly. We identified 19 antiviral VHHs that protect human
A549 cells from lethal infection with influenza A virus (IAV) or vesicular
stomatitis virus (VSV), respectively. Both negative-sense RNA viruses are
vulnerable to VHHs uniquely specific for their respective nucleoproteins.
Antiviral VHHs prevented nuclear import of viral ribonucleoproteins or mRNA
transcription, respectively, and may provide clues for novel antiviral reagents.
In principle, the screening approach described here should be applicable to
identify inhibitors of any pathogen or biological pathway.
Collapse
|
60
|
Hagen C, Dent KC, Zeev-Ben-Mordehai T, Grange M, Bosse JB, Whittle C, Klupp BG, Siebert CA, Vasishtan D, Bäuerlein FJB, Cheleski J, Werner S, Guttmann P, Rehbein S, Henzler K, Demmerle J, Adler B, Koszinowski U, Schermelleh L, Schneider G, Enquist LW, Plitzko JM, Mettenleiter TC, Grünewald K. Structural Basis of Vesicle Formation at the Inner Nuclear Membrane. Cell 2016; 163:1692-701. [PMID: 26687357 PMCID: PMC4701712 DOI: 10.1016/j.cell.2015.11.029] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/11/2015] [Accepted: 11/06/2015] [Indexed: 12/22/2022]
Abstract
Vesicular nucleo-cytoplasmic transport is becoming recognized as a general cellular mechanism for translocation of large cargoes across the nuclear envelope. Cargo is recruited, enveloped at the inner nuclear membrane (INM), and delivered by membrane fusion at the outer nuclear membrane. To understand the structural underpinning for this trafficking, we investigated nuclear egress of progeny herpesvirus capsids where capsid envelopment is mediated by two viral proteins, forming the nuclear egress complex (NEC). Using a multi-modal imaging approach, we visualized the NEC in situ forming coated vesicles of defined size. Cellular electron cryo-tomography revealed a protein layer showing two distinct hexagonal lattices at its membrane-proximal and membrane-distant faces, respectively. NEC coat architecture was determined by combining this information with integrative modeling using small-angle X-ray scattering data. The molecular arrangement of the NEC establishes the basic mechanism for budding and scission of tailored vesicles at the INM. Multimodal imaging reveals mechanism of vesicle formation at inner nuclear membrane Nucleo-cytoplasmic cargo vesicle coat in situ comprises two distinct lattices Lattices are formed by hexameric building blocks made of the nuclear egress complex Induction of membrane curvature based solely on heterodimeric interactions
Collapse
Affiliation(s)
- Christoph Hagen
- Oxford Particle Imaging Centre, Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Kyle C Dent
- Oxford Particle Imaging Centre, Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
| | - Tzviya Zeev-Ben-Mordehai
- Oxford Particle Imaging Centre, Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Michael Grange
- Oxford Particle Imaging Centre, Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Jens B Bosse
- Department of Molecular Biology, Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Cathy Whittle
- Oxford Particle Imaging Centre, Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Barbara G Klupp
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - C Alistair Siebert
- Oxford Particle Imaging Centre, Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Daven Vasishtan
- Oxford Particle Imaging Centre, Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Felix J B Bäuerlein
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Juliana Cheleski
- Oxford Particle Imaging Centre, Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Stephan Werner
- Helmholtz Zentrum Berlin für Materialien und Energie GmbH, Wilhelm-Conrad-Röntgen Campus, 12489 Berlin, Germany
| | - Peter Guttmann
- Helmholtz Zentrum Berlin für Materialien und Energie GmbH, Wilhelm-Conrad-Röntgen Campus, 12489 Berlin, Germany
| | - Stefan Rehbein
- Helmholtz Zentrum Berlin für Materialien und Energie GmbH, Wilhelm-Conrad-Röntgen Campus, 12489 Berlin, Germany
| | - Katja Henzler
- Helmholtz Zentrum Berlin für Materialien und Energie GmbH, Wilhelm-Conrad-Röntgen Campus, 12489 Berlin, Germany
| | - Justin Demmerle
- Micron Oxford, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Barbara Adler
- Max von Pettenkofer-Institut, Ludwig-Maximilians-Universität München, Pettenkoferstr. 9a, 80336 Munich, Germany
| | - Ulrich Koszinowski
- Max von Pettenkofer-Institut, Ludwig-Maximilians-Universität München, Pettenkoferstr. 9a, 80336 Munich, Germany
| | - Lothar Schermelleh
- Micron Oxford, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Gerd Schneider
- Helmholtz Zentrum Berlin für Materialien und Energie GmbH, Wilhelm-Conrad-Röntgen Campus, 12489 Berlin, Germany
| | - Lynn W Enquist
- Department of Molecular Biology, Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Jürgen M Plitzko
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Thomas C Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany.
| | - Kay Grünewald
- Oxford Particle Imaging Centre, Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK.
| |
Collapse
|
61
|
Schmidt FI, Lu A, Chen JW, Ruan J, Tang C, Wu H, Ploegh HL. A single domain antibody fragment that recognizes the adaptor ASC defines the role of ASC domains in inflammasome assembly. J Exp Med 2016; 213:771-90. [PMID: 27069117 PMCID: PMC4854733 DOI: 10.1084/jem.20151790] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/01/2016] [Indexed: 01/09/2023] Open
Abstract
Ploegh et al. raised an alpaca single-domain antibody (VHH) against the inflammasome adaptor ASC. VHHASC blocks inflammasome activation in vitro and in living cells, and demonstrates a role of the ASC CARD domain in cross-linking ASC Pyrin domain filaments. Myeloid cells assemble inflammasomes in response to infection or cell damage; cytosolic sensors activate pro–caspase-1, indirectly for the most part, via the adaptors ASC and NLRC4. This leads to secretion of proinflammatory cytokines and pyroptosis. To explore complex formation under physiological conditions, we generated an alpaca single domain antibody, VHHASC, which specifically recognizes the CARD of human ASC via its type II interface. VHHASC not only impairs ASCCARD interactions in vitro, but also inhibits inflammasome activation in response to NLRP3, AIM2, and NAIP triggers when expressed in living cells, highlighting a role of ASC in all three types of inflammasomes. VHHASC leaves the Pyrin domain of ASC functional and stabilizes a filamentous intermediate of inflammasome activation. Incorporation of VHHASC-EGFP into these structures allowed the visualization of endogenous ASCPYD filaments for the first time. These data revealed that cross-linking of ASCPYD filaments via ASCCARD mediates the assembly of ASC foci.
Collapse
Affiliation(s)
| | - Alvin Lu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Jeff W Chen
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
| | - Jianbin Ruan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Catherine Tang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115 Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Hidde L Ploegh
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142 Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
62
|
A model for coordinating nuclear mechanics and membrane remodeling to support nuclear integrity. Curr Opin Cell Biol 2016; 41:9-17. [PMID: 27031045 DOI: 10.1016/j.ceb.2016.03.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 11/20/2022]
Abstract
A polymer network of intranuclear lamin filaments underlies the nuclear envelope and provides mechanical stability to the nucleus in metazoans. Recent work demonstrates that the expression of A-type lamins scales positively with the stiffness of the cellular environment, thereby coupling nuclear and extracellular mechanics. Using the spectrin-actin network at the erythrocyte plasma membrane as a model, we contemplate how the relative stiffness of the nuclear scaffold impinges on the growing number of interphase-specific nuclear envelope remodeling events, including recently discovered, nuclear envelope-specialized quality control mechanisms. We suggest that a stiffer lamina impedes these remodeling events, necessitating local lamina remodeling and/or concomitant scaling of the efficacy of membrane-remodeling machineries that act at the nuclear envelope.
Collapse
|
63
|
Zhao C, Brown RSH, Tang CHA, Hu CCA, Schlieker C. Site-specific Proteolysis Mobilizes TorsinA from the Membrane of the Endoplasmic Reticulum (ER) in Response to ER Stress and B Cell Stimulation. J Biol Chem 2016; 291:9469-81. [PMID: 26953341 DOI: 10.1074/jbc.m115.709337] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Indexed: 01/01/2023] Open
Abstract
Torsin ATPases are the only representatives of the AAA+ ATPase family that reside in the lumen of the endoplasmic reticulum (ER) and nuclear envelope. Two of these, TorsinA and TorsinB, are anchored to the ER membrane by virtue of an N-terminal hydrophobic domain. Here we demonstrate that the imposition of ER stress leads to a proteolytic cleavage event that selectively removes the hydrophobic domain from the AAA+ domain of TorsinA, which retains catalytic activity. Both the pharmacological inhibition profile and the identified cleavage site between two juxtaposed cysteine residues are distinct from those of presently known proteases, suggesting that a hitherto uncharacterized, membrane-associated protease accounts for TorsinA processing. This processing occurs not only in stress-exposed cell lines but also in primary cells from distinct organisms including stimulated B cells, indicating that Torsin conversion in response to physiologically relevant stimuli is an evolutionarily conserved process. By establishing 5-nitroisatin as a cell-permeable inhibitor for Torsin processing, we provide the methodological framework for interfering with Torsin processing in a wide range of primary cells without the need for genetic manipulation.
Collapse
Affiliation(s)
- Chenguang Zhao
- From the Departments of Molecular Biophysics and Biochemistry and
| | | | - Chih-Hang Anthony Tang
- the Department of Translational Tumor Immunology, The Wistar Institute, Philadelphia, Pennsylvania 19104
| | - Chih-Chi Andrew Hu
- the Department of Translational Tumor Immunology, The Wistar Institute, Philadelphia, Pennsylvania 19104
| | - Christian Schlieker
- From the Departments of Molecular Biophysics and Biochemistry and Cell Biology, Yale University, New Haven, Connecticut 06520 and
| |
Collapse
|
64
|
Saunders CA, Luxton GWG. LINCing defective nuclear-cytoskeletal coupling and DYT1 dystonia. Cell Mol Bioeng 2016; 9:207-216. [PMID: 27499815 DOI: 10.1007/s12195-016-0432-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Mechanical forces generated by nuclear-cytoskeletal coupling through the LINC (linker of nucleoskeleton and cytoskeleton) complex, an evolutionarily conserved molecular bridge in the nuclear envelope (NE), are critical for the execution of wholesale nuclear positioning events in migrating and dividing cells, chromosome dynamics during meiosis, and mechanotransduction. LINC complexes consist of outer (KASH (Klarsicht, ANC-1, and Syne homology)) and inner (SUN (Sad1, UNC-84)) nuclear membrane proteins. KASH proteins interact with the cytoskeleton in the cytoplasm and SUN proteins in the perinuclear space of the NE. In the nucleoplasm, SUN proteins interact with A-type nuclear lamins and chromatin-binding proteins. Recent structural insights into the KASH-SUN interaction have generated several questions regarding how LINC complex assembly and function might be regulated within the perinuclear space. Here we discuss potential LINC regulatory mechanisms and focus on the potential role of AAA+ (ATPases associated with various cellular activities) protein, torsinA, as a LINC complex regulator within the NE. We also examine how defects in LINC complex regulation by torsinA may contribute to the pathogenesis of the human neurological movement disorder, DYT1 dystonia.
Collapse
Affiliation(s)
- Cosmo A Saunders
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455
| | - G W Gant Luxton
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
65
|
Abstract
Mechanoresponses in mesenchymal stem cells (MSCs) guide both differentiation and function. In this review, we focus on advances in0 our understanding of how the cytoplasmic cytoskeleton, nuclear envelope and nucleoskeleton, which are connected via LINC (Linker of Nucleoskeleton and Cytoskeleton) complexes, are emerging as an integrated dynamic signaling platform to regulate MSC mechanobiology. This dynamic interconnectivity affects mechanical signaling and transfer of signals into the nucleus. In this way, nuclear and LINC-mediated cytoskeletal connectivity play a critical role in maintaining mechanical signaling that affects MSC fate by serving as both mechanosensory and mechanoresponsive structures. We review disease and age related compromises of LINC complexes and nucleoskeleton that contribute to the etiology of musculoskeletal diseases. Finally we invite the idea that acquired dysfunctions of LINC might be a contributing factor to conditions such as aging, microgravity and osteoporosis and discuss potential mechanical strategies to modulate LINC connectivity to combat these conditions.
Collapse
|
66
|
Hellberg T, Paßvogel L, Schulz KS, Klupp BG, Mettenleiter TC. Nuclear Egress of Herpesviruses: The Prototypic Vesicular Nucleocytoplasmic Transport. Adv Virus Res 2016; 94:81-140. [PMID: 26997591 DOI: 10.1016/bs.aivir.2015.10.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Herpesvirus particles mature in two different cellular compartments. While capsid assembly and packaging of the genomic linear double-stranded DNA occur in the nucleus, virion formation takes place in the cytoplasm by the addition of numerous tegument proteins as well as acquisition of the viral envelope by budding into cellular vesicles derived from the trans-Golgi network containing virally encoded glycoproteins. To gain access to the final maturation compartment, herpesvirus nucleocapsids have to cross a formidable barrier, the nuclear envelope (NE). Since the ca. 120 nm diameter capsids are unable to traverse via nuclear pores, herpesviruses employ a vesicular transport through both leaflets of the NE. This process involves proteins which support local dissolution of the nuclear lamina to allow access of capsids to the inner nuclear membrane (INM), drive vesicle formation from the INM and mediate inclusion of the capsid as well as scission of the capsid-containing vesicle (also designated as "primary virion"). Fusion of the vesicle membrane (i.e., the "primary envelope") with the outer nuclear membrane subsequently results in release of the nucleocapsid into the cytoplasm for continuing virion morphogenesis. While this process has long been thought to be unique for herpesviruses, a similar pathway for nuclear egress of macromolecular complexes has recently been observed in Drosophila. Thus, herpesviruses may have coopted a hitherto unrecognized cellular mechanism of vesicle-mediated nucleocytoplasmic transport. This could have far reaching consequences for our understanding of cellular functions as again unraveled by the study of viruses.
Collapse
Affiliation(s)
- Teresa Hellberg
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Lars Paßvogel
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Katharina S Schulz
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Barbara G Klupp
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Thomas C Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany.
| |
Collapse
|
67
|
Torsin ATPases: structural insights and functional perspectives. Curr Opin Cell Biol 2016; 40:1-7. [PMID: 26803745 DOI: 10.1016/j.ceb.2016.01.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 12/22/2015] [Accepted: 01/02/2016] [Indexed: 12/29/2022]
Abstract
Torsin ATPases are the only members of the AAA+ ATPase family that localize to the endoplasmic reticulum and contiguous perinuclear space. Accordingly, they are well positioned to perform essential work in these compartments, but their precise functions remain elusive. Recent studies have deciphered an unusual ATPase activation mechanism relying on Torsin-associated transmembrane cofactors, LAP1 or LULL1. These findings profoundly change our molecular view of the Torsin machinery and rationalize several human mutations in TorsinA or LAP1 leading to congenital disorders, symptoms of which have recently been recapitulated in mouse models. Here, we review these recent advances in the Torsin field and discuss the most pressing questions in relation to nuclear envelope dynamics.
Collapse
|
68
|
Demircioglu FE, Sosa BA, Ingram J, Ploegh HL, Schwartz TU. Structures of TorsinA and its disease-mutant complexed with an activator reveal the molecular basis for primary dystonia. eLife 2016; 5:e17983. [PMID: 27490483 PMCID: PMC4999309 DOI: 10.7554/elife.17983] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/03/2016] [Indexed: 01/07/2023] Open
Abstract
The most common cause of early onset primary dystonia, a neuromuscular disease, is a glutamate deletion (ΔE) at position 302/303 of TorsinA, a AAA+ ATPase that resides in the endoplasmic reticulum. While the function of TorsinA remains elusive, the ΔE mutation is known to diminish binding of two TorsinA ATPase activators: lamina-associated protein 1 (LAP1) and its paralog, luminal domain like LAP1 (LULL1). Using a nanobody as a crystallization chaperone, we obtained a 1.4 Å crystal structure of human TorsinA in complex with LULL1. This nanobody likewise stabilized the weakened TorsinAΔE-LULL1 interaction, which enabled us to solve its structure at 1.4 Å also. A comparison of these structures shows, in atomic detail, the subtle differences in activator interactions that separate the healthy from the diseased state. This information may provide a structural platform for drug development, as a small molecule that rescues TorsinAΔE could serve as a cure for primary dystonia.
Collapse
Affiliation(s)
- F Esra Demircioglu
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Brian A Sosa
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Jessica Ingram
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - Hidde L Ploegh
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - Thomas U Schwartz
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States,
| |
Collapse
|
69
|
Teng H, Sui X, Zhou C, Shen C, Yang Y, Zhang P, Guo X, Huo R. Fatty acid degradation plays an essential role in proliferation of mouse female primordial germ cells via the p53-dependent cell cycle regulation. Cell Cycle 2015; 15:425-31. [PMID: 26716399 DOI: 10.1080/15384101.2015.1127473] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Primordial germ cells (PGCs) are embryonic founders of germ cells that ultimately differentiate into oocytes and spermatogonia. Embryonic proliferation of PGCs starting from E11.5 ensures the presence of germ cells in adulthood, especially in female mammals whose total number of oocytes declines after this initial proliferation period. To better understand mechanisms underlying PGC proliferation in female mice, we constructed a proteome profile of female mouse gonads at E11.5. Subsequent KEGG pathway analysis of the 3,662 proteins profiled showed significant enrichment of pathways involved in fatty acid degradation. Further, the number of PGCs found in in vitro cultured fetal gonads significantly decreased with application of etomoxir, an inhibitor of the key rate-limiting enzyme of fatty acid degradation carnitine acyltransferase I (CPT1). Decrease in PGCs was further determined to be the result of reduced proliferation rather than apoptosis. The inhibition of fatty acid degradation by etomoxir has the potential to activate the Ca(2+)/CamKII/5'-adenosine monophosphate-activated protein kinase (AMPK) pathway; while as an upstream activator, activated AMPK can function as activator of p53 to induce cell cycle arrest. Thus, we detected the expressional level of AMPK, phosphorylated AMPK (P-AMPK), phosphorylated p53 (P-p53) and cyclin-dependent kinase inhibitor 1 (p21) by Western blots, the results showed increased expression of them after treatment with etomoxir, suggested the activation of p53 pathway was the reason for reduced proliferation of PGCs. Finally, the involvement of p53-dependent G1 cell cycle arrest in defective proliferation of PGCs was verified by rescue experiments. Our results demonstrate that fatty acid degradation plays an important role in proliferation of female PGCs via the p53-dependent cell cycle regulation.
Collapse
Affiliation(s)
- Hui Teng
- a State Key Laboratory of Reproductive Medicine , Department of Histology and Embryology, Nanjing Medical University , Nanjing , P.R. China
| | - Xuesong Sui
- a State Key Laboratory of Reproductive Medicine , Department of Histology and Embryology, Nanjing Medical University , Nanjing , P.R. China
| | - Cheng Zhou
- a State Key Laboratory of Reproductive Medicine , Department of Histology and Embryology, Nanjing Medical University , Nanjing , P.R. China
| | - Cong Shen
- a State Key Laboratory of Reproductive Medicine , Department of Histology and Embryology, Nanjing Medical University , Nanjing , P.R. China
| | - Ye Yang
- a State Key Laboratory of Reproductive Medicine , Department of Histology and Embryology, Nanjing Medical University , Nanjing , P.R. China
| | - Pang Zhang
- a State Key Laboratory of Reproductive Medicine , Department of Histology and Embryology, Nanjing Medical University , Nanjing , P.R. China
| | - Xuejiang Guo
- a State Key Laboratory of Reproductive Medicine , Department of Histology and Embryology, Nanjing Medical University , Nanjing , P.R. China
| | - Ran Huo
- a State Key Laboratory of Reproductive Medicine , Department of Histology and Embryology, Nanjing Medical University , Nanjing , P.R. China
| |
Collapse
|
70
|
Abstract
Torsin ATPases (Torsins) belong to the widespread AAA+ (ATPases associated with a variety of cellular activities) family of ATPases, which share structural similarity but have diverse cellular functions. Torsins are outliers in this family because they lack many characteristics of typical AAA+ proteins, and they are the only members of the AAA+ family located in the endoplasmic reticulum and contiguous perinuclear space. While it is clear that Torsins have essential roles in many, if not all metazoans, their precise cellular functions remain elusive. Studying Torsins has significant medical relevance since mutations in Torsins or Torsin-associated proteins result in a variety of congenital human disorders, the most frequent of which is early-onset torsion (DYT1) dystonia, a severe movement disorder. A better understanding of the Torsin system is needed to define the molecular etiology of these diseases, potentially enabling corrective therapy. Here, we provide a comprehensive overview of the Torsin system in metazoans, discuss functional clues obtained from various model systems and organisms and provide a phylogenetic and structural analysis of Torsins and their regulatory cofactors in relation to disease-causative mutations. Moreover, we review recent data that have led to a dramatically improved understanding of these machines at a molecular level, providing a foundation for investigating the molecular defects underlying the associated movement disorders. Lastly, we discuss our ideas on how recent progress may be utilized to inform future studies aimed at determining the cellular role(s) of these atypical molecular machines and their implications for dystonia treatment options.
Collapse
Affiliation(s)
- April E Rose
- a Department of Molecular Biophysics and Biochemistry , Yale University , New Haven , CT , USA and
| | - Rebecca S H Brown
- a Department of Molecular Biophysics and Biochemistry , Yale University , New Haven , CT , USA and
| | - Christian Schlieker
- a Department of Molecular Biophysics and Biochemistry , Yale University , New Haven , CT , USA and.,b Department of Cell Biology , Yale School of Medicine , New Haven , CT , USA
| |
Collapse
|
71
|
Genetic mutations strengthen functional association of LAP1 with DYT1 dystonia and muscular dystrophy. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2015; 766:42-7. [DOI: 10.1016/j.mrrev.2015.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/30/2015] [Accepted: 07/31/2015] [Indexed: 12/30/2022]
|
72
|
Weisheit CE, Dauer WT. A novel conditional knock-in approach defines molecular and circuit effects of the DYT1 dystonia mutation. Hum Mol Genet 2015; 24:6459-72. [PMID: 26370418 DOI: 10.1093/hmg/ddv355] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/01/2015] [Indexed: 01/15/2023] Open
Abstract
DYT1 dystonia, the most common inherited form of primary dystonia, is a neurodevelopmental disease caused by a dominant mutation in TOR1A. This mutation ('ΔE') removes a single glutamic acid from the encoded protein, torsinA. The effects of this mutation, at the molecular and circuit levels, and the reasons for its neurodevelopmental onset, remain incompletely understood. To uniquely address key questions of disease pathogenesis, we generated a conditional Tor1a knock-in allele that is converted from wild-type to DYT1 mutant ('induced' ΔE: Tor1a(i-ΔE)), following Cre recombination. We used this model to perform a gene dosage study exploring the effects of the ΔE mutation at the molecular, neuropathological and organismal levels. These analyses demonstrated that ΔE-torsinA is a hypomorphic allele and showed no evidence for any gain-of-function toxic properties. The unique capabilities of this model also enabled us to test a circuit-level hypothesis of DYT1 dystonia, which predicts that expression of the DYT1 genotype (Tor1a(ΔE/+)) selectively within hindbrain structures will produce an overtly dystonic animal. In contrast to this prediction, we find no effect of this anatomic-specific expression of the DYT1 genotype, a finding that has important implications for the interpretation of the human and mouse diffusion tensor-imaging studies upon which it is based. These studies advance understanding of the molecular effects of the ΔE mutation, challenge current concepts of the circuit dysfunction that characterize the disease and establish a powerful tool that will be valuable for future studies of disease pathophysiology.
Collapse
Affiliation(s)
| | - William T Dauer
- Department of Neurology and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
73
|
Goodchild RE, Buchwalter AL, Naismith TV, Holbrook K, Billion K, Dauer WT, Liang CC, Dear ML, Hanson PI. Access of torsinA to the inner nuclear membrane is activity dependent and regulated in the endoplasmic reticulum. J Cell Sci 2015; 128:2854-65. [PMID: 26092934 DOI: 10.1242/jcs.167452] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 06/12/2015] [Indexed: 11/20/2022] Open
Abstract
TorsinA (also known as torsin-1A) is a membrane-embedded AAA+ ATPase that has an important role in the nuclear envelope lumen. However, most torsinA is localized in the peripheral endoplasmic reticulum (ER) lumen where it has a slow mobility that is incompatible with free equilibration between ER subdomains. We now find that nuclear-envelope-localized torsinA is present on the inner nuclear membrane (INM) and ask how torsinA reaches this subdomain. The ER system contains two transmembrane proteins, LAP1 and LULL1 (also known as TOR1AIP1 and TOR1AIP2, respectively), that reversibly co-assemble with and activate torsinA. Whereas LAP1 localizes on the INM, we show that LULL1 is in the peripheral ER and does not enter the INM. Paradoxically, interaction between torsinA and LULL1 in the ER targets torsinA to the INM. Native gel electrophoresis reveals torsinA oligomeric complexes that are destabilized by LULL1. Mutations in torsinA or LULL1 that inhibit ATPase activity reduce the access of torsinA to the INM. Furthermore, although LULL1 binds torsinA in the ER lumen, its effect on torsinA localization requires cytosolic-domain-mediated oligomerization. These data suggest that LULL1 oligomerizes to engage and transiently disassemble torsinA oligomers, and is thereby positioned to transduce cytoplasmic signals to the INM through torsinA.
Collapse
Affiliation(s)
- Rose E Goodchild
- VIB Centre for the Biology of Disease and KU Leuven, Department of Human Genetics, Campus Gasthuisberg, Leuven 3000, Belgium
| | - Abigail L Buchwalter
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Teresa V Naismith
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Kristen Holbrook
- Department of Biochemistry, Cell and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Karolien Billion
- VIB Centre for the Biology of Disease and KU Leuven, Department of Human Genetics, Campus Gasthuisberg, Leuven 3000, Belgium
| | - William T Dauer
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Chun-Chi Liang
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mary Lynn Dear
- Department of Biochemistry, Cell and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Phyllis I Hanson
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63130, USA
| |
Collapse
|
74
|
The Torsin Activator LULL1 Is Required for Efficient Growth of Herpes Simplex Virus 1. J Virol 2015; 89:8444-52. [PMID: 26041288 DOI: 10.1128/jvi.01143-15] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 05/26/2015] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED TorsinA is a membrane-tethered AAA+ ATPase implicated in nuclear envelope dynamics as well as the nuclear egress of herpes simplex virus 1 (HSV-1). The activity of TorsinA and the related ATPase TorsinB strictly depends on LAP1 and LULL1, type II transmembrane proteins that are integral parts of the Torsin/cofactor AAA ring, forming a composite, membrane-spanning assembly. Here, we use CRISPR/Cas9-mediated genome engineering to create single- and double knockout (KO) cell lines of TorA and TorB as well as their activators, LAP1 and LULL1, to investigate the effect on HSV-1 production. Consistent with LULL1 being the more potent Torsin activator, a LULL1 KO reduces HSV-1 growth by one order of magnitude, while the deletion of other components of the Torsin system in combination causes subtle defects. Notably, LULL1 deficiency leads to a 10-fold decrease in the number of viral genomes per host cell without affecting viral protein production, allowing us to tentatively assign LULL1 to an unexpected role that precedes HSV-1 nuclear egress. IMPORTANCE In this study, we conduct the first comprehensive genetic and phenotypic analysis of the Torsin/cofactor system in the context of HSV-1 infection, establishing LULL1 as the most important component of the Torsin system with respect to viral production.
Collapse
|
75
|
Affiliation(s)
- John McCullough
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| | - Wesley I Sundquist
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
76
|
Truttmann MC, Wu Q, Stiegeler S, Duarte JN, Ingram J, Ploegh HL. HypE-specific nanobodies as tools to modulate HypE-mediated target AMPylation. J Biol Chem 2015; 290:9087-100. [PMID: 25678711 DOI: 10.1074/jbc.m114.634287] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Indexed: 11/06/2022] Open
Abstract
The covalent addition of mono-AMP to target proteins (AMPylation) by Fic domain-containing proteins is a poorly understood, yet highly conserved post-translational modification. Here, we describe the generation, evaluation, and application of four HypE-specific nanobodies: three that inhibit HypE-mediated target AMPylation in vitro and one that acts as an activator. All heavy chain-only antibody variable domains bind HypE when expressed as GFP fusions in intact cells. We observed localization of HypE at the nuclear envelope and further identified histones H2-H4, but not H1, as novel in vitro targets of the human Fic protein. Its role in histone modification provides a possible link between AMPylation and regulation of gene expression.
Collapse
Affiliation(s)
- Matthias C Truttmann
- From the Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142 and
| | - Qin Wu
- From the Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142 and
| | - Sarah Stiegeler
- From the Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142 and
| | - Joao N Duarte
- From the Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142 and
| | - Jessica Ingram
- From the Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142 and
| | - Hidde L Ploegh
- From the Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142 and the Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
77
|
Desmyter A, Spinelli S, Roussel A, Cambillau C. Camelid nanobodies: killing two birds with one stone. Curr Opin Struct Biol 2015; 32:1-8. [PMID: 25614146 DOI: 10.1016/j.sbi.2015.01.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 12/23/2014] [Accepted: 01/05/2015] [Indexed: 01/09/2023]
Abstract
In recent years, the use of single-domain camelid immunoglobulins, termed vHHs or nanobodies, has seen increasing growth in biotechnology, pharmaceutical applications and structure/function research. The usefulness of nanobodies in structural biology is now firmly established, as they provide access to new epitopes in concave and hinge regions - and stabilize them. These sites are often associated with enzyme inhibition or receptor neutralization, and, at the same time, provide favorable surfaces for crystal packing. Remarkable results have been achieved by using nanobodies with flexible multi-domain proteins, large complexes and, last but not least, membrane proteins. While generating nanobodies is still a rather long and expensive procedure, the advent of naive libraries might be expected to facilitate the whole process.
Collapse
Affiliation(s)
- Aline Desmyter
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, France; Centre National de la Recherche Scientifique, AFMB, UMR 7257, case 932, 13288 Marseille Cedex 09, France
| | - Silvia Spinelli
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, France; Centre National de la Recherche Scientifique, AFMB, UMR 7257, case 932, 13288 Marseille Cedex 09, France
| | - Alain Roussel
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, France; Centre National de la Recherche Scientifique, AFMB, UMR 7257, case 932, 13288 Marseille Cedex 09, France
| | - Christian Cambillau
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, France; Centre National de la Recherche Scientifique, AFMB, UMR 7257, case 932, 13288 Marseille Cedex 09, France.
| |
Collapse
|
78
|
Li H, Wu HC, Liu Z, Zacchi LF, Brodsky JL, Zolkiewski M. Intracellular complexes of the early-onset torsion dystonia-associated AAA+ ATPase TorsinA. SPRINGERPLUS 2014; 3:743. [PMID: 25674472 PMCID: PMC4320221 DOI: 10.1186/2193-1801-3-743] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 12/09/2014] [Indexed: 01/25/2023]
Abstract
A single GAG codon deletion in the gene encoding torsinA is linked to most cases of early-onset torsion dystonia. TorsinA is an ER-localized membrane-associated ATPase from the AAA+ superfamily with an unknown biological function. We investigated the formation of oligomeric complexes of torsinA in cultured mammalian cells and found that wild type torsinA associates into a complex with a molecular weight consistent with that of a homohexamer. Interestingly, the dystonia-linked variant torsinAΔE displayed a reduced propensity to form the oligomers compared to the wild type protein. We also discovered that the deletion of the N-terminal membrane-associating region of torsinA abolished oligomer formation. Our results demonstrate that the dystonia-linked mutation in the torsinA gene produces a protein variant that is deficient in maintaining its oligomeric state and suggest that ER membrane association is required to stabilize the torsinA complex.
Collapse
Affiliation(s)
- Hui Li
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506 USA
| | - Hui-Chuan Wu
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506 USA
| | - Zhonghua Liu
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506 USA ; Department of Embryology, Carnegie Institution, Baltimore, MD 21218 USA
| | - Lucia F Zacchi
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Michal Zolkiewski
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506 USA
| |
Collapse
|
79
|
Abstract
Torsins are membrane-associated ATPases whose activity is dependent on two activating cofactors, lamina-associated polypeptide 1 (LAP1) and luminal domain-like LAP1 (LULL1). The mechanism by which these cofactors regulate Torsin activity has so far remained elusive. In this study, we identify a conserved domain in these activators that is predicted to adopt a fold resembling an AAA+ (ATPase associated with a variety of cellular activities) domain. Within these domains, a strictly conserved Arg residue present in both activating cofactors, but notably missing in Torsins, aligns with a key catalytic Arg found in AAA+ proteins. We demonstrate that cofactors and Torsins associate to form heterooligomeric assemblies with a defined Torsin-activator interface. In this arrangement, the highly conserved Arg residue present in either cofactor comes into close proximity with the nucleotide bound in the neighboring Torsin subunit. Because this invariant Arg is strictly required to stimulate Torsin ATPase activity but is dispensable for Torsin binding, we propose that LAP1 and LULL1 regulate Torsin ATPase activity through an active site complementation mechanism.
Collapse
|