1
|
Gu J, Zhou X, Sutherland L, Liszczak G, McKnight SL. A simple method for mapping the location of cross-β-forming regions within protein domains of low sequence complexity. Proc Natl Acad Sci U S A 2025; 122:e2503382122. [PMID: 40267128 PMCID: PMC12054801 DOI: 10.1073/pnas.2503382122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/21/2025] [Indexed: 04/25/2025] Open
Abstract
Protein domains of low sequence complexity are unable to fold into stable, three-dimensional structures. In test tube studies, these unusual polypeptide regions can self-associate in a manner causing phase separation from aqueous solution. This form of protein:protein interaction has been implicated in numerous examples of dynamic morphological organization within eukaryotic cells. In several cases, the basis for low complexity domain (LCD) self-association and phase separation has been traced to the formation of labile cross-β structures. The primary energetic force favoring formation of these transient and reversible structures is enabled by polypeptide backbone interactions. Short, contiguous networks of peptide backbone amino groups and carbonyl oxygens are zippered together intermolecularly by hydrogen bonding as described by Linus Pauling seven decades ago. Here, we describe a simple, molecular biological method useful for the identification of localized, self-associating regions within larger protein domains of low sequence complexity.
Collapse
Affiliation(s)
- Jinge Gu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Xiaoming Zhou
- Westlake University, Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang310024, PR China
| | - Lillian Sutherland
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Glen Liszczak
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Steven L. McKnight
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX75390
| |
Collapse
|
2
|
Uzgiris AJ, Ladic LA, Pfister SX. Advances in neurofilament light chain analysis. Adv Clin Chem 2025; 126:31-71. [PMID: 40185536 DOI: 10.1016/bs.acc.2025.01.006] [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: 04/07/2025]
Abstract
This chapter provides a comprehensive summary of clinical laboratory testing for neurofilament light chain (NfL) in neurologic disease. A primer on the NfL structure and function is presented with its potential use as a biomarker. The most widely utilized methods for NfL in biologic samples are highlighted and examined. Limitations of current knowledge are considered, as are outstanding questions related to dissemination and standardization of testing. Herein we focus on methods available today and those in development for clinical use. In the final section, a broad vision is presented of how NfL may be utilized in the future to improve diagnosis and treatment of neurologic diseases as well as for maintaining health.
Collapse
Affiliation(s)
- Arejas J Uzgiris
- Siemens Healthcare Diagnostics Inc., Tarrytown, NY, United States.
| | - Lance A Ladic
- Siemens Healthcare Diagnostics Inc., Tarrytown, NY, United States
| | - Sophia X Pfister
- Siemens Healthcare Diagnostics Inc., Tarrytown, NY, United States
| |
Collapse
|
3
|
Paumier JM, Zewe J, Panja C, Pergande MR, Venkatesan M, Israeli E, Prasad S, Snider N, Savas JN, Opal P. Neurofilament accumulation disrupts autophagy in giant axonal neuropathy. JCI Insight 2025; 10:e177999. [PMID: 40059823 PMCID: PMC11949051 DOI: 10.1172/jci.insight.177999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/22/2025] [Indexed: 03/29/2025] Open
Abstract
Neurofilament accumulation is associated with many neurodegenerative diseases, but it is the primary pathology in giant axonal neuropathy (GAN). This childhood-onset autosomal recessive disease is caused by loss-of-function mutations in gigaxonin, the E3 adaptor protein that enables neurofilament degradation. Using a combination of genetic and RNA interference approaches, we found that dorsal root ganglia from mice lacking gigaxonin have impaired autophagy and lysosomal degradation through 2 mechanisms. First, neurofilament accumulations interfere with the distribution of autophagic organelles, impairing their maturation and fusion with lysosomes. Second, the accumulations attract the chaperone 14-3-3, which is responsible for the proper localization of the key autophagy regulator transcription factor EB (TFEB). We propose that this dual disruption of autophagy contributes to the pathogenesis of other neurodegenerative diseases involving neurofilament accumulations.
Collapse
Affiliation(s)
- Jean-Michel Paumier
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - James Zewe
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Chiranjit Panja
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Melissa R. Pergande
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Meghana Venkatesan
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Eitan Israeli
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Shikha Prasad
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Natasha Snider
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Jeffrey N. Savas
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Puneet Opal
- Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Cell and Molecular Biology, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
4
|
Lin NH, Jian WS, Perng MD. Deletions in Glial Fibrillary Acidic Protein Leading to Alterations in Intermediate Filament Assembly and Network Formation. Int J Mol Sci 2025; 26:1913. [PMID: 40076540 PMCID: PMC11900225 DOI: 10.3390/ijms26051913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Glial fibrillary acidic protein (GFAP) is classified as a type III intermediate filament protein predominantly expressed in mature astrocytes. It has the ability to self-assemble into 10 nm filaments in vitro, making it particularly valuable for elucidating the sequences essential for filament assembly. In this study, we created a series of deletion mutants targeting sequences in the N-terminal, C-terminal, and central rod domains to explore the sequences critical for the assembly of GFAP into 10 nm filaments. The impact of these deletions on filament formation was evaluated through in vitro assembly studies and transduction assays conducted with primary astrocytes. Our data revealed that deletions at the carboxy end resulted in abnormalities in either filament diameter calibration or lateral association, whereas deletions at the amino-terminal end significantly disrupted the filament assembly process, particularly restricting filament elongation. Furthermore, we discovered that the filament-forming sequences within the rod domain varied in their contributions to filament assembly and network formation. These findings enhance our understanding of the GFAP assembly process in vitro and provide a detailed mapping of the essential regions required for GFAP assembly. These insights hold significant implications for Alexander disease arising from deletion mutations in GFAP.
Collapse
Affiliation(s)
- Ni-Hsuan Lin
- Institute of Molecular Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu 30043, Taiwan; (N.-H.L.); (W.-S.J.)
| | - Wan-Syuan Jian
- Institute of Molecular Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu 30043, Taiwan; (N.-H.L.); (W.-S.J.)
| | - Ming-Der Perng
- Institute of Molecular Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu 30043, Taiwan; (N.-H.L.); (W.-S.J.)
- School of Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu 30043, Taiwan
| |
Collapse
|
5
|
Xu P, Schumacher D, Liu C, Harms A, Dickmanns M, Beck F, Plitzko JM, Baumeister W, Søgaard-Andersen L. In situ architecture of a nucleoid-associated biomolecular co-condensate that regulates bacterial cell division. Proc Natl Acad Sci U S A 2025; 122:e2419610121. [PMID: 39739804 PMCID: PMC11725790 DOI: 10.1073/pnas.2419610121] [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: 09/24/2024] [Accepted: 11/22/2024] [Indexed: 01/02/2025] Open
Abstract
In most bacteria, cell division depends on the tubulin-homolog FtsZ that polymerizes in a GTP-dependent manner to form the cytokinetic Z-ring at the future division site. Subsequently, the Z-ring recruits, directly or indirectly, all other proteins of the divisome complex that executes cytokinesis. A critical step in this process is the precise positioning of the Z-ring at the future division site. While the divisome proteins are generally conserved, the regulatory systems that position the Z-ring are more diverse. However, these systems have in common that they modulate FtsZ polymerization. In Myxococcus, PomX, PomY, and PomZ form precisely one MDa-sized, nonstoichiometric, nucleoid-associated assembly that spatiotemporally guides Z-ring formation. Here, using cryo-correlative light and electron microscopy together with in situ cryoelectron tomography, we determine the PomXYZ assembly's architecture at close-to-live conditions. PomX forms a porous meshwork of randomly intertwined filaments. Templated by this meshwork, the phase-separating PomY protein forms a biomolecular condensate that compacts and bends the PomX filaments, resulting in the formation of a selective PomXYZ co-condensate that is associated to the nucleoid by PomZ. These studies reveal a hitherto undescribed supramolecular structure and provide a framework for understanding how a nonstoichiometric co-condensate forms, maintains number control, and nucleates GTP-dependent FtsZ polymerization to precisely regulate cell division.
Collapse
Affiliation(s)
- Peng Xu
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried82152, Germany
| | - Dominik Schumacher
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg35043, Germany
| | - Chuan Liu
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried82152, Germany
| | - Andrea Harms
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg35043, Germany
| | - Marcel Dickmanns
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried82152, Germany
| | - Florian Beck
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried82152, Germany
| | - Jürgen M. Plitzko
- Research Group CryoEM Technology, Max Planck Institute of Biochemistry, Martinsried82152, Germany
| | - Wolfgang Baumeister
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried82152, Germany
| | - Lotte Søgaard-Andersen
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg35043, Germany
| |
Collapse
|
6
|
Martínez-Cenalmor P, Martínez AE, Moneo-Corcuera D, González-Jiménez P, Pérez-Sala D. Oxidative stress elicits the remodeling of vimentin filaments into biomolecular condensates. Redox Biol 2024; 75:103282. [PMID: 39079387 PMCID: PMC11338992 DOI: 10.1016/j.redox.2024.103282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/21/2024] [Accepted: 07/21/2024] [Indexed: 08/23/2024] Open
Abstract
The intermediate filament protein vimentin performs an essential role in cytoskeletal interplay and dynamics, mechanosensing and cellular stress responses. In pathology, vimentin is a key player in tumorigenesis, fibrosis and infection. Vimentin filaments undergo distinct and versatile reorganizations, and behave as redox sensors. The vimentin monomer possesses a central α-helical rod domain flanked by N- and C-terminal low complexity domains. Interactions between this type of domains play an important function in the formation of phase-separated biomolecular condensates, which in turn are critical for the organization of cellular components. Here we show that several oxidants, including hydrogen peroxide and diamide, elicit the remodeling of vimentin filaments into small particles. Oxidative stress elicited by diamide induces a fast dissociation of filaments into circular, motile dots, which requires the presence of the single vimentin cysteine residue, C328. This effect is reversible, and filament reassembly can occur within minutes of oxidant removal. Diamide-elicited vimentin droplets recover fluorescence after photobleaching. Moreover, fusion of cells expressing differentially tagged vimentin allows the detection of dots positive for both tags, indicating that vimentin dots merge upon cell fusion. The aliphatic alcohol 1,6-hexanediol, known to alter interactions between low complexity domains, readily dissolves diamide-elicited vimentin dots at low concentrations, in a C328 dependent manner, and hampers reassembly. Taken together, these results indicate that vimentin oxidation promotes a fast and reversible filament remodeling into biomolecular condensate-like structures, and provide primary evidence of its regulated phase separation. Moreover, we hypothesize that filament to droplet transition could play a protective role against irreversible damage of the vimentin network by oxidative stress.
Collapse
Affiliation(s)
- Paula Martínez-Cenalmor
- Department of Cellular and Molecular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040, Madrid, Spain
| | - Alma E Martínez
- Department of Cellular and Molecular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040, Madrid, Spain
| | - Diego Moneo-Corcuera
- Department of Cellular and Molecular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040, Madrid, Spain
| | - Patricia González-Jiménez
- Department of Cellular and Molecular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040, Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Cellular and Molecular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040, Madrid, Spain.
| |
Collapse
|
7
|
Coelho-Rato LS, Parvanian S, Andrs Salajkova S, Medalia O, Eriksson JE. Intermediate filaments at a glance. J Cell Sci 2024; 137:jcs261386. [PMID: 39206824 DOI: 10.1242/jcs.261386] [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] [Indexed: 09/04/2024] Open
Abstract
Intermediate filaments (IFs) comprise a large family of versatile cytoskeletal proteins, divided into six subtypes with tissue-specific expression patterns. IFs have a wide repertoire of cellular functions, including providing structural support to cells, as well as active roles in mechanical support and signaling pathways. Consequently, defects in IFs are associated with more than 100 diseases. In this Cell Science at a Glance article, we discuss the established classes of IFs and their general features, their functions beyond structural support, and recent advances in the field. We also highlight their involvement in disease and potential use as clinical markers of pathological conditions. Finally, we provide our view on current knowledge gaps and the future directions of the IF field.
Collapse
Affiliation(s)
- Leila S Coelho-Rato
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
| | - Sepideh Parvanian
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA 02114, USA
| | - Sarka Andrs Salajkova
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - John E Eriksson
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
- Euro-Bioimaging ERIC, 20520 Turku, Finland
| |
Collapse
|
8
|
Odell J, Lammerding J. N-terminal tags impair the ability of lamin A to provide structural support to the nucleus. J Cell Sci 2024; 137:jcs262207. [PMID: 39092499 PMCID: PMC11361635 DOI: 10.1242/jcs.262207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024] Open
Abstract
Lamins are intermediate filament proteins that contribute to numerous cellular functions, including nuclear morphology and mechanical stability. The N-terminal head domain of lamin is crucial for higher order filament assembly and function, yet the effects of commonly used N-terminal tags on lamin function remain largely unexplored. Here, we systematically studied the effect of two differently sized tags on lamin A (LaA) function in a mammalian cell model engineered to allow for precise control of expression of tagged lamin proteins. Untagged, FLAG-tagged and GFP-tagged LaA completely rescued nuclear shape defects when expressed at similar levels in lamin A/C-deficient (Lmna-/-) MEFs, and all LaA constructs prevented increased nuclear envelope ruptures in these cells. N-terminal tags, however, altered the nuclear localization of LaA and impaired the ability of LaA to restore nuclear deformability and to recruit emerin to the nuclear membrane in Lmna-/- MEFs. Our finding that tags impede some LaA functions but not others might explain the partial loss of function phenotypes when tagged lamins are expressed in model organisms and should caution researchers using tagged lamins to study the nucleus.
Collapse
Affiliation(s)
- Jacob Odell
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Graduate Field of Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
9
|
Odell J, Lammerding J. N-terminal tags impair the ability of Lamin A to provide structural support to the nucleus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590311. [PMID: 39211210 PMCID: PMC11361184 DOI: 10.1101/2024.04.19.590311] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Lamins are intermediate filament proteins that contribute to numerous cellular functions, including nuclear morphology and mechanical stability. The N-terminal head domain of lamin is critical for higher order filament assembly and function, yet the effects of commonly used N-terminal tags on lamin function remain largely unexplored. Here, we systematically studied the effect of two differently sized tags on Lamin A (LaA) function in a mammalian cell model engineered to allow for precise control of expression of tagged lamin proteins. Untagged, FLAG-tagged, and GFP-tagged LaA completely rescued nuclear shape defects when expressed at similar levels in lamin A/C-deficient ( Lmna -/- ) MEFs, and all LaA constructs prevented increased nuclear envelope (NE) ruptures in these cells. N-terminal tags, however, altered the nuclear localization of LaA and impaired the ability of LaA to restore nuclear deformability and to recruit Emerin to the nuclear membrane in Lmna -/- MEFs. Our finding that tags impede some LaA functions but not others may explain the partial loss of function phenotypes when tagged lamins are expressed in model organisms and should caution researchers using tagged lamins to study the nucleus.
Collapse
|
10
|
Recine F, Vanni S, Bongiovanni A, Fausti V, Mercatali L, Miserocchi G, Liverani C, Pieri F, Casadei R, Cavaliere D, Falbo PT, Diano D, Ibrahim T, De Vita A. Clinical and translational implications of immunotherapy in sarcomas. Front Immunol 2024; 15:1378398. [PMID: 38983859 PMCID: PMC11231074 DOI: 10.3389/fimmu.2024.1378398] [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: 01/29/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024] Open
Abstract
Immunotherapy has emerged as promising treatment in sarcomas, but the high variability in terms of histology, clinical behavior and response to treatments determines a particular challenge for its role in these neoplasms. Tumor immune microenvironment (TiME) of sarcomas reflects the heterogeneity of these tumors originating from mesenchymal cells and encompassing more than 100 histologies. Advances in the understanding of the complexity of TiME have led to an improvement of the immunotherapeutic responsiveness in sarcomas, that at first showed disappointing results. The proposed immune-classification of sarcomas based on the interaction between immune cell populations and tumor cells showed to have a prognostic and potential predictive role for immunotherapies. Several studies have explored the clinical impact of immune therapies in the management of these histotypes leading to controversial results. The presence of Tumor Infiltrating Lymphocytes (TIL) seems to correlate with an improvement in the survival of patients and with a higher responsiveness to immunotherapy. In this context, it is important to consider that also immune-related genes (IRGs) have been demonstrated to have a key role in tumorigenesis and in the building of tumor immune microenvironment. The IRGs landscape in soft tissue and bone sarcomas is characterized by the connection between several tumor-related genes that can assume a potential prognostic and predictive therapeutic role. In this paper, we reviewed the state of art of the principal immune strategies in the management of sarcomas including their clinical and translational relevance.
Collapse
Affiliation(s)
- Federica Recine
- Medical Oncology Unit, Azienda Ospedaliera “San Giovanni Addolorata”, Roma, Italy
| | - Silvia Vanni
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Alberto Bongiovanni
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Valentina Fausti
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Laura Mercatali
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Giacomo Miserocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Chiara Liverani
- Medical Oncology Unit, Azienda Ospedaliera “San Giovanni Addolorata”, Roma, Italy
| | - Federica Pieri
- Pathology Unit, “Morgagni-Pierantoni” Hospital, Forlì, Italy
| | - Roberto Casadei
- Orthopedic Unit, “Morgagni-Pierantoni” Hospital, Forlì, Italy
| | - Davide Cavaliere
- General and Oncologic Surgery, “Morgagni-Pierantoni” Hospital, Forlì, Italy
| | - Pina Tiziana Falbo
- Medical Oncology Unit, Azienda Ospedaliera “San Giovanni Addolorata”, Roma, Italy
| | - Danila Diano
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Alessandro De Vita
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
11
|
Eibauer M, Weber MS, Kronenberg-Tenga R, Beales CT, Boujemaa-Paterski R, Turgay Y, Sivagurunathan S, Kraxner J, Köster S, Goldman RD, Medalia O. Vimentin filaments integrate low-complexity domains in a complex helical structure. Nat Struct Mol Biol 2024; 31:939-949. [PMID: 38632361 PMCID: PMC11189308 DOI: 10.1038/s41594-024-01261-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 03/01/2024] [Indexed: 04/19/2024]
Abstract
Intermediate filaments (IFs) are integral components of the cytoskeleton. They provide cells with tissue-specific mechanical properties and are involved in numerous cellular processes. Due to their intricate architecture, a 3D structure of IFs has remained elusive. Here we use cryo-focused ion-beam milling, cryo-electron microscopy and tomography to obtain a 3D structure of vimentin IFs (VIFs). VIFs assemble into a modular, intertwined and flexible helical structure of 40 α-helices in cross-section, organized into five protofibrils. Surprisingly, the intrinsically disordered head domains form a fiber in the lumen of VIFs, while the intrinsically disordered tails form lateral connections between the protofibrils. Our findings demonstrate how protein domains of low sequence complexity can complement well-folded protein domains to construct a biopolymer with striking mechanical strength and stretchability.
Collapse
Affiliation(s)
- Matthias Eibauer
- Department of Biochemistry, University of Zurich, Zurich, Switzerland.
| | - Miriam S Weber
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | | | - Charlie T Beales
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | | | - Yagmur Turgay
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Suganya Sivagurunathan
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Julia Kraxner
- Institute for X-Ray Physics, University of Göttingen, Göttingen, Germany
- MDC Berlin-Buch, Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - Sarah Köster
- Institute for X-Ray Physics, University of Göttingen, Göttingen, Germany
| | - Robert D Goldman
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Lomakin IB, Ho M, Bunick CG. Unveiling the secrets of vimentin filament architecture relevant to human disease. Nat Struct Mol Biol 2024; 31:849-851. [PMID: 38684931 PMCID: PMC11955283 DOI: 10.1038/s41594-024-01301-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Affiliation(s)
- Ivan B Lomakin
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Minh Ho
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Christopher G Bunick
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.
- Program in Translational Biomedicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
13
|
Tsilafakis K, Mavroidis M. Are the Head and Tail Domains of Intermediate Filaments Really Unstructured Regions? Genes (Basel) 2024; 15:633. [PMID: 38790262 PMCID: PMC11121635 DOI: 10.3390/genes15050633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/01/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Intermediate filaments (IFs) are integral components of the cytoskeleton which provide cells with tissue-specific mechanical properties and are involved in a plethora of cellular processes. Unfortunately, due to their intricate architecture, the 3D structure of the complete molecule of IFs has remained unresolved. Even though most of the rod domain structure has been revealed by means of crystallographic analyses, the flanked head and tail domains are still mostly unknown. Only recently have studies shed light on head or tail domains of IFs, revealing certainsecondary structures and conformational changes during IF assembly. Thus, a deeper understanding of their structure could provide insights into their function.
Collapse
Affiliation(s)
- Konstantinos Tsilafakis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece;
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Manolis Mavroidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece;
| |
Collapse
|
14
|
Ding EA, Kumar S. Neurofilament Biophysics: From Structure to Biomechanics. Mol Biol Cell 2024; 35:re1. [PMID: 38598299 PMCID: PMC11151108 DOI: 10.1091/mbc.e23-11-0438] [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: 01/29/2024] [Revised: 03/25/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
Neurofilaments (NFs) are multisubunit, neuron-specific intermediate filaments consisting of a 10-nm diameter filament "core" surrounded by a layer of long intrinsically disordered protein (IDP) "tails." NFs are thought to regulate axonal caliber during development and then stabilize the mature axon, with NF subunit misregulation, mutation, and aggregation featuring prominently in multiple neurological diseases. The field's understanding of NF structure, mechanics, and function has been deeply informed by a rich variety of biochemical, cell biological, and mouse genetic studies spanning more than four decades. These studies have contributed much to our collective understanding of NF function in axonal physiology and disease. In recent years, however, there has been a resurgence of interest in NF subunit proteins in two new contexts: as potential blood- and cerebrospinal fluid-based biomarkers of neuronal damage, and as model IDPs with intriguing properties. Here, we review established principles and more recent discoveries in NF structure and function. Where possible, we place these findings in the context of biophysics of NF assembly, interaction, and contributions to axonal mechanics.
Collapse
Affiliation(s)
- Erika A. Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720
| | - Sanjay Kumar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
15
|
Pajares MA, Pérez-Sala D. Type III intermediate filaments in redox interplay: key role of the conserved cysteine residue. Biochem Soc Trans 2024; 52:849-860. [PMID: 38451193 PMCID: PMC11088922 DOI: 10.1042/bst20231059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Intermediate filaments (IFs) are cytoskeletal elements involved in mechanotransduction and in the integration of cellular responses. They are versatile structures and their assembly and organization are finely tuned by posttranslational modifications. Among them, type III IFs, mainly vimentin, have been identified as targets of multiple oxidative and electrophilic modifications. A characteristic of most type III IF proteins is the presence in their sequence of a single, conserved cysteine residue (C328 in vimentin), that is a hot spot for these modifications and appears to play a key role in the ability of the filament network to respond to oxidative stress. Current structural models and experimental evidence indicate that this cysteine residue may occupy a strategic position in the filaments in such a way that perturbations at this site, due to chemical modification or mutation, impact filament assembly or organization in a structure-dependent manner. Cysteine-dependent regulation of vimentin can be modulated by interaction with divalent cations, such as zinc, and by pH. Importantly, vimentin remodeling induced by C328 modification may affect its interaction with cellular organelles, as well as the cross-talk between cytoskeletal networks, as seems to be the case for the reorganization of actin filaments in response to oxidants and electrophiles. In summary, the evidence herein reviewed delineates a complex interplay in which type III IFs emerge both as targets and modulators of redox signaling.
Collapse
Affiliation(s)
- María A. Pajares
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, C.S.I.C., Ramiro de Maeztu, 9, 28040 Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, C.S.I.C., Ramiro de Maeztu, 9, 28040 Madrid, Spain
| |
Collapse
|
16
|
Nanes BA, Bhatt K, Boujemaa-Paterski R, Azarova E, Munawar S, Rajendran D, Isogai T, Dean KM, Medalia O, Danuser G. Keratin isoform shifts modulate motility signals during wound healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.04.538989. [PMID: 37205459 PMCID: PMC10187270 DOI: 10.1101/2023.05.04.538989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Keratin intermediate filaments form strong mechanical scaffolds that confer structural stability to epithelial tissues, but the reason this function requires a protein family with 54 isoforms is not understood. During skin wound healing, a shift in keratin isoform expression alters the composition of keratin filaments. How this change modulates cellular function to support epidermal remodeling remains unclear. We report an unexpected effect of keratin isoform variation on kinase signal transduction. Increased expression of wound-associated keratin 6A, but not of steady-state keratin 5, potentiated keratinocyte migration and wound closure without compromising epidermal stability by activating myosin motors. This pathway depended on isoform-specific interaction between intrinsically disordered keratin head domains and non-filamentous vimentin shuttling myosin-activating kinases. These results substantially expand the functional repertoire of intermediate filaments from their canonical role as mechanical scaffolds to include roles as isoform-tuned signaling scaffolds that organize signal transduction cascades in space and time to influence epithelial cell state.
Collapse
Affiliation(s)
- Benjamin A Nanes
- Department of Dermatology, UT Southwestern Medical Center; Dallas, TX 75390, USA
- Lyda Hill Department of Bioinformatics and Cecil H and Ida Green Center for Systems Biology, UT Southwestern Medical Center; Dallas, TX 75390, USA
| | - Kushal Bhatt
- Lyda Hill Department of Bioinformatics and Cecil H and Ida Green Center for Systems Biology, UT Southwestern Medical Center; Dallas, TX 75390, USA
| | | | - Evgenia Azarova
- Lyda Hill Department of Bioinformatics and Cecil H and Ida Green Center for Systems Biology, UT Southwestern Medical Center; Dallas, TX 75390, USA
- Present address: Department of Materials Science and Engineering, Johns Hopkins University; Baltimore, MD 21218, USA
| | - Sabahat Munawar
- Lyda Hill Department of Bioinformatics and Cecil H and Ida Green Center for Systems Biology, UT Southwestern Medical Center; Dallas, TX 75390, USA
| | - Divya Rajendran
- Lyda Hill Department of Bioinformatics and Cecil H and Ida Green Center for Systems Biology, UT Southwestern Medical Center; Dallas, TX 75390, USA
| | - Tadamoto Isogai
- Lyda Hill Department of Bioinformatics and Cecil H and Ida Green Center for Systems Biology, UT Southwestern Medical Center; Dallas, TX 75390, USA
| | - Kevin M Dean
- Lyda Hill Department of Bioinformatics and Cecil H and Ida Green Center for Systems Biology, UT Southwestern Medical Center; Dallas, TX 75390, USA
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich; Zurich CH-8057, Switzerland
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics and Cecil H and Ida Green Center for Systems Biology, UT Southwestern Medical Center; Dallas, TX 75390, USA
| |
Collapse
|
17
|
McKnight SL. Protein domains of low sequence complexity-dark matter of the proteome. Genes Dev 2024; 38:205-212. [PMID: 38503517 PMCID: PMC11065162 DOI: 10.1101/gad.351465.123] [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] [Indexed: 03/21/2024]
Abstract
This perspective begins with a speculative consideration of the properties of the earliest proteins to appear during evolution. What did these primitive proteins look like, and how were they of benefit to early forms of life? I proceed to hypothesize that primitive proteins have been preserved through evolution and now serve diverse functions important to the dynamics of cell morphology and biological regulation. The primitive nature of these modern proteins is easy to spot. They are composed of a limited subset of the 20 amino acids used by traditionally evolved proteins and thus are of low sequence complexity. This chemical simplicity limits protein domains of low sequence complexity to forming only a crude and labile type of protein structure currently hidden from the computational powers of machine learning. I conclude by hypothesizing that this structural weakness represents the underlying virtue of proteins that, at least for the moment, constitute the dark matter of the proteome.
Collapse
Affiliation(s)
- Steven L McKnight
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75390-9152, USA
| |
Collapse
|
18
|
Sanchez‐Martinez S, Nguyen K, Biswas S, Nicholson V, Romanyuk AV, Ramirez J, Kc S, Akter A, Childs C, Meese EK, Usher ET, Ginell GM, Yu F, Gollub E, Malferrari M, Francia F, Venturoli G, Martin EW, Caporaletti F, Giubertoni G, Woutersen S, Sukenik S, Woolfson DN, Holehouse AS, Boothby TC. Labile assembly of a tardigrade protein induces biostasis. Protein Sci 2024; 33:e4941. [PMID: 38501490 PMCID: PMC10949331 DOI: 10.1002/pro.4941] [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: 11/01/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 03/20/2024]
Abstract
Tardigrades are microscopic animals that survive desiccation by inducing biostasis. To survive drying tardigrades rely on intrinsically disordered CAHS proteins, which also function to prevent perturbations induced by drying in vitro and in heterologous systems. CAHS proteins have been shown to form gels both in vitro and in vivo, which has been speculated to be linked to their protective capacity. However, the sequence features and mechanisms underlying gel formation and the necessity of gelation for protection have not been demonstrated. Here we report a mechanism of fibrillization and gelation for CAHS D similar to that of intermediate filament assembly. We show that in vitro, gelation restricts molecular motion, immobilizing and protecting labile material from the harmful effects of drying. In vivo, we observe that CAHS D forms fibrillar networks during osmotic stress. Fibrillar networking of CAHS D improves survival of osmotically shocked cells. We observe two emergent properties associated with fibrillization; (i) prevention of cell volume change and (ii) reduction of metabolic activity during osmotic shock. We find that there is no significant correlation between maintenance of cell volume and survival, while there is a significant correlation between reduced metabolism and survival. Importantly, CAHS D's fibrillar network formation is reversible and metabolic rates return to control levels after CAHS fibers are resolved. This work provides insights into how tardigrades induce reversible biostasis through the self-assembly of labile CAHS gels.
Collapse
Affiliation(s)
| | - K. Nguyen
- Department of Molecular BiologyUniversity of WyomingLaramieWyomingUSA
| | - S. Biswas
- Department of Molecular BiologyUniversity of WyomingLaramieWyomingUSA
| | - V. Nicholson
- Department of Molecular BiologyUniversity of WyomingLaramieWyomingUSA
| | - A. V. Romanyuk
- School of ChemistryUniversity of BristolBristolUK
- Max Planck‐Bristol Centre for Minimal BiologyUniversity of BristolBristolUK
| | - J. Ramirez
- Department of Molecular BiologyUniversity of WyomingLaramieWyomingUSA
| | - S. Kc
- Department of Molecular BiologyUniversity of WyomingLaramieWyomingUSA
| | - A. Akter
- Department of Molecular BiologyUniversity of WyomingLaramieWyomingUSA
| | - C. Childs
- Department of Molecular BiologyUniversity of WyomingLaramieWyomingUSA
| | - E. K. Meese
- Department of Molecular BiologyUniversity of WyomingLaramieWyomingUSA
| | - E. T. Usher
- Department of Biochemistry and Molecular BiophysicsWashington University School of MedicineSt. LouisMissouriUSA
- Center for Biomolecular CondensatesWashington University in St. LouisSt. LouisMissouriUSA
| | - G. M. Ginell
- Department of Biochemistry and Molecular BiophysicsWashington University School of MedicineSt. LouisMissouriUSA
- Center for Biomolecular CondensatesWashington University in St. LouisSt. LouisMissouriUSA
| | - F. Yu
- Quantitative Systems Biology ProgramUniversity of California MercedMercedCaliforniaUSA
| | - E. Gollub
- Department of Chemistry and BiochemistryUniversity of California MercedMercedCaliforniaUSA
| | - M. Malferrari
- Dipartimento di Chimica “Giacomo Ciamician”Università di BolognaBolognaItaly
| | - F. Francia
- Laboratorio di Biochimica e Biofisica Molecolare, Dipartimento di Farmacia e Biotecnologie, FaBiTUniversità di BolognaBolognaItaly
| | - G. Venturoli
- Laboratorio di Biochimica e Biofisica Molecolare, Dipartimento di Farmacia e Biotecnologie, FaBiTUniversità di BolognaBolognaItaly
- Consorzio Nazionale Interuniversitario per le Scienze Fisiche della Materia (CNISM), c/o Dipartimento di Fisica e Astronomia (DIFA)Università di BolognaBolognaItaly
| | - E. W. Martin
- Department of Structural BiologySt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - F. Caporaletti
- Van't Hoff Institute for Molecular SciencesUniversity of AmsterdamAmsterdamThe Netherlands
| | - G. Giubertoni
- Van't Hoff Institute for Molecular SciencesUniversity of AmsterdamAmsterdamThe Netherlands
| | - S. Woutersen
- Van't Hoff Institute for Molecular SciencesUniversity of AmsterdamAmsterdamThe Netherlands
| | - S. Sukenik
- Quantitative Systems Biology ProgramUniversity of California MercedMercedCaliforniaUSA
- Department of Chemistry and BiochemistryUniversity of California MercedMercedCaliforniaUSA
| | - D. N. Woolfson
- School of ChemistryUniversity of BristolBristolUK
- Max Planck‐Bristol Centre for Minimal BiologyUniversity of BristolBristolUK
- School of BiochemistryUniversity of Bristol, Biomedical Sciences BuildingBristolUK
| | - A. S. Holehouse
- Department of Biochemistry and Molecular BiophysicsWashington University School of MedicineSt. LouisMissouriUSA
- Center for Biomolecular CondensatesWashington University in St. LouisSt. LouisMissouriUSA
| | - T. C. Boothby
- Department of Molecular BiologyUniversity of WyomingLaramieWyomingUSA
| |
Collapse
|
19
|
Vlachakis D, Tsilafakis K, Kostavasili I, Kossida S, Mavroidis M. Unraveling Desmin's Head Domain Structure and Function. Cells 2024; 13:603. [PMID: 38607042 PMCID: PMC11012097 DOI: 10.3390/cells13070603] [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: 01/09/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
Understanding the structure and function of intermediate filaments (IFs) is necessary in order to explain why more than 70 related IF genes have evolved in vertebrates while maintaining such dramatically tissue-specific expression. Desmin is a member of the large multigene family of IF proteins and is specifically expressed in myocytes. In an effort to elucidate its muscle-specific behavior, we have used a yeast two-hybrid system in order to identify desmin's head binding partners. We described a mitochondrial and a lysosomal protein, NADH ubiquinone oxidoreductase core subunit S2 (NDUFS2), and saposin D, respectively, as direct desmin binding partners. In silico analysis indicated that both interactions at the atomic level occur in a very similar way, by the formation of a three-helix bundle with hydrophobic interactions in the interdomain space and hydrogen bonds at R16 and S32 of the desmin head domain. The interactions, confirmed also by GST pull-down assays, indicating the necessity of the desmin head domain and, furthermore, point out its role in function of mitochondria and lysosomes, organelles which are disrupted in myopathies due to desmin head domain mutations.
Collapse
Affiliation(s)
- Dimitrios Vlachakis
- Biotechnology Department, Agricultural University of Athens, 11855 Athens, Greece;
| | - Konstantinos Tsilafakis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece; (K.T.); (I.K.)
- Biochemistry & Biotechnology Department, University of Thessaly, 41500 Larisa, Greece
| | - Ioanna Kostavasili
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece; (K.T.); (I.K.)
| | - Sophia Kossida
- IMGT, The International ImMunoGeneTics Information System, National Center for Scientific Research (CNRS), Institute of Human Genetics (IGH), University of Montpellier (UM), 34090 Montpellier, France;
| | - Manolis Mavroidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece; (K.T.); (I.K.)
| |
Collapse
|
20
|
Pérez-Sala D, Quinlan RA. The redox-responsive roles of intermediate filaments in cellular stress detection, integration and mitigation. Curr Opin Cell Biol 2024; 86:102283. [PMID: 37989035 DOI: 10.1016/j.ceb.2023.102283] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 11/23/2023]
Abstract
Intermediate filaments are critical for cell and tissue homeostasis and for stress responses. Cytoplasmic intermediate filaments form versatile and dynamic assemblies that interconnect cellular organelles, participate in signaling and protect cells and tissues against stress. Here we have focused on their involvement in redox signaling and oxidative stress, which arises in numerous pathophysiological situations. We pay special attention to type III intermediate filaments, mainly vimentin, because it provides a physical interface for redox signaling, stress responses and mechanosensing. Vimentin possesses a single cysteine residue that is a target for multiple oxidants and electrophiles. This conserved residue fine tunes vimentin assembly, response to oxidative stress and crosstalk with other cellular structures. Here we integrate evidence from the intermediate filament and redox biology fields to propose intermediate filaments as redox sentinel networks of the cell. To support this, we appraise how vimentin detects and orchestrates cellular responses to oxidative and electrophilic stress.
Collapse
Affiliation(s)
- Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, C.S.I.C., 28040 Madrid, Spain.
| | - Roy A Quinlan
- Department of Biosciences, University of Durham, Upper Mountjoy Science Site, Durham, United Kingdom; Biophysical Sciences Institute, University of Durham, Durham, United Kingdom; Department of Biological Structure, University of Washington, Seattle, WA, United States.
| |
Collapse
|
21
|
Al-Ansari M, Fitzsimons T, Wei W, Goldberg MW, Kunieda T, Quinlan RA. The major inducible small heat shock protein HSP20-3 in the tardigrade Ramazzottius varieornatus forms filament-like structures and is an active chaperone. Cell Stress Chaperones 2024; 29:51-65. [PMID: 38330543 PMCID: PMC10939073 DOI: 10.1016/j.cstres.2023.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/03/2023] [Indexed: 02/10/2024] Open
Abstract
The tardigrade Ramazzottius varieornatus has remarkable resilience to a range of environmental stresses. In this study, we have characterised two members of the small heat shock protein (sHSP) family in R. varieornatus, HSP20-3 and HSP20-6. These are the most highly upregulated sHSPs in response to a 24 h heat shock at 35 0C of adult tardigrades with HSP20-3 being one of the most highly upregulated gene in the whole transcriptome. Both R. varieornatus sHSPs and the human sHSP, CRYAB (HSPB5), were produced recombinantly for comparative structure-function studies. HSP20-3 exhibited a superior chaperone activity than human CRYAB in a heat-induced protein aggregation assay. Both tardigrade sHSPs also formed larger oligomers than CRYAB as assessed by size exclusion chromatography and transmission electron microscopy of negatively stained samples. Whilst both HSP20-3 and HSP20-6 formed particles that were variable in size and larger than the particles formed by CRYAB, only HSP20-3 formed filament-like structures. The particles and filament-like structures formed by HSP20-3 appear inter-related as the filament-like structures often had particles located at their ends. Sequence analyses identified two unique features; an insertion in the middle region of the N-terminal domain (NTD) and preceding the critical-sequence identified in CRYAB, as well as a repeated QNTN-motif located in the C-terminal domain of HSP20-3. The NTD insertion is expected to affect protein-protein interactions and subunit oligomerisation. Removal of the repeated QNTN-motif abolished HSP20-3 chaperone activity and also affected the assembly of the filament-like structures. We discuss the potential contribution of HSP20-3 to protein condensate formation.
Collapse
Affiliation(s)
- Mohammad Al-Ansari
- Department of Biosciences, Upper Mountjoy Science Site, University of Durham, Durham DH1 3LE, UK; Department of Biochemistry, Health Sciences Centre, Kuwait University, Kuwait
| | - Taylor Fitzsimons
- Department of Biosciences, Upper Mountjoy Science Site, University of Durham, Durham DH1 3LE, UK
| | - Wenbin Wei
- Department of Biosciences, Upper Mountjoy Science Site, University of Durham, Durham DH1 3LE, UK.
| | - Martin W Goldberg
- Department of Biosciences, Upper Mountjoy Science Site, University of Durham, Durham DH1 3LE, UK
| | - Takekazu Kunieda
- Department of Biological Sciences, The University of Tokyo, Japan
| | - Roy A Quinlan
- Department of Biosciences, Upper Mountjoy Science Site, University of Durham, Durham DH1 3LE, UK; Department of Biological Structure, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
22
|
Ding L, Wei LW, Li TS, Chen J. Mental retardation, seizures and language delay caused by new SETD1B mutations: Three case reports. World J Clin Cases 2024; 12:383-391. [PMID: 38313655 PMCID: PMC10835677 DOI: 10.12998/wjcc.v12.i2.383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/01/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND The SETD1B gene is instrumental in human intelligence and nerve development. Mutations in the SETD1B gene have been linked in recent studies to neurodevelopmental disorders, seizures, and language delay. CASE SUMMARY This study aimed to analyze the clinical manifestations and treatment of three patients suffering from mental retardation, epilepsy, and language delay resulting from a new mutation in the SETD1B gene. Three individuals with these symptoms were selected, and their clinical symptoms, gene test results, and treatment were analyzed. This article discusses the impact of the SETD1B gene mutation on patients and outlines the treatment approach. Among the three patients (two females and one male, aged 8, 4, and 1, respectively), all exhibited psychomotor retardation, attention deficit, and hyperactivity disorder, and two had epilepsy. Antiepileptic treatment with sodium tripolyvalproate halted the seizures in the affected child, although mental development remained somewhat delayed. Whole exome sequencing revealed new mutations in the SETD1B gene for all patients, specifically with c.5473C>T (p.Arg1825trp), c.4120C>T (p.Gln1374*, 593), c.14_15insC (p.His5Hisfs*33). CONCLUSION Possessing the SETD1B gene mutation may cause mental retardation accompanied by seizures and language delay. Although the exact mechanism is not fully understood, interventions such as drug therapy, rehabilitation training, and family support can assist patients in managing their symptoms and enhancing their quality of life. Furthermore, genetic testing supplies healthcare providers with more precise diagnostic and therapeutic guidance, informs families about genetic disease risks, and contributes to understanding disease pathogenesis and drug research and development.
Collapse
Affiliation(s)
- Le Ding
- Department of Neurology, Children’s Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Li-Wan Wei
- Chigene (Beijing) Translational Medical Research Center, Co. Ltd., Beijing 101111, China
| | - Tai-Song Li
- Chigene (Beijing) Translational Medical Research Center, Co. Ltd., Beijing 101111, China
| | - Jing Chen
- Department of Neurology, Children’s Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| |
Collapse
|
23
|
Chen W, Liao Y, Sun P, Tu J, Zou Y, Fang J, Chen Z, Li H, Chen J, Peng Y, Wen L, Xie X. Construction of an ER stress-related prognostic signature for predicting prognosis and screening the effective anti-tumor drug in osteosarcoma. J Transl Med 2024; 22:66. [PMID: 38229155 PMCID: PMC10792867 DOI: 10.1186/s12967-023-04794-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/09/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Osteosarcoma is the most common malignant primary bone tumor in infants and adolescents. The lack of understanding of the molecular mechanisms underlying osteosarcoma progression and metastasis has contributed to a plateau in the development of current therapies. Endoplasmic reticulum (ER) stress has emerged as a significant contributor to the malignant progression of tumors, but its potential regulatory mechanisms in osteosarcoma progression remain unknown. METHODS In this study, we collected RNA sequencing and clinical data of osteosarcoma from The TCGA, GSE21257, and GSE33382 cohorts. Differentially expressed analysis and the least absolute shrinkage and selection operator regression analysis were conducted to identify prognostic genes and construct an ER stress-related prognostic signature (ERSRPS). Survival analysis and time dependent ROC analysis were performed to evaluate the predictive performance of the constructed prognostic signature. The "ESTIMATE" package and ssGSEA algorithm were utilized to evaluate the differences in immune cells infiltration between the groups. Cell-based assays, including CCK-8, colony formation, and transwell assays and co-culture system were performed to assess the effects of the target gene and small molecular drug in osteosarcoma. Animal models were employed to assess the anti-osteosarcoma effects of small molecular drug. RESULTS Five genes (BLC2, MAGEA3, MAP3K5, STC2, TXNDC12) were identified to construct an ERSRPS. The ER stress-related gene Stanniocalcin 2 (STC2) was identified as a risk gene in this signature. Additionally, STC2 knockdown significantly inhibited osteosarcoma cell proliferation, migration, and invasion. Furthermore, the ER stress-related gene STC2 was found to downregulate the expression of MHC-I molecules in osteosarcoma cells, and mediate immune responses through influencing the infiltration and modulating the function of CD8+ T cells. Patients categorized by risk scores showed distinct immune status, and immunotherapy response. ISOX was subsequently identified and validated as an effective anti-osteosarcoma drug through a combination of CMap database screening and in vitro and in vivo experiments. CONCLUSION The ERSRPS may guide personalized treatment decisions for osteosarcoma, and ISOX holds promise for repurposing in osteosarcoma treatment.
Collapse
Affiliation(s)
- Weidong Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yan Liao
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Pengxiao Sun
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jian Tu
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yutong Zou
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Ji Fang
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Ziyun Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Hongbo Li
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Junkai Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yuzhong Peng
- Macau University of Science and Technology, Macau, 999078, China
| | - Lili Wen
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Xianbiao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
24
|
Phillips CL, Faridounnia M, Armao D, Snider NT. Stability dynamics of neurofilament and GFAP networks and protein fragments. Curr Opin Cell Biol 2023; 85:102266. [PMID: 37866019 PMCID: PMC11402464 DOI: 10.1016/j.ceb.2023.102266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/21/2023] [Accepted: 09/24/2023] [Indexed: 10/24/2023]
Abstract
Neurofilaments (NFs) and GFAP are cytoskeletal intermediate filaments (IFs) that support cellular processes unfolding within the uniquely complex environments of neurons and astrocytes, respectively. This review highlights emerging concepts on the transitions between stable and destabilized IF networks in the nervous system. While self-association between transiently structured low-complexity IF domains promotes filament assembly, the opposing destabilizing actions of phosphorylation-mediated filament severing facilitate faster intracellular transport. Cellular proteases, including caspases and calpains, produce a variety of IF fragments, which may interact with N-degron and C-degron pathways of the protein degradation machinery. The rapid adoption of NF and GFAP-based clinical biomarker tests is contrasted with the lagging understanding of the dynamics between the native IF proteins and their fragments.
Collapse
Affiliation(s)
- Cassandra L Phillips
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, USA
| | - Maryam Faridounnia
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, USA
| | - Diane Armao
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, USA; Department of Radiology, University of North Carolina at Chapel Hill, USA
| | - Natasha T Snider
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, USA.
| |
Collapse
|
25
|
Buxboim A, Kronenberg-Tenga R, Salajkova S, Avidan N, Shahak H, Thurston A, Medalia O. Scaffold, mechanics and functions of nuclear lamins. FEBS Lett 2023; 597:2791-2805. [PMID: 37813648 DOI: 10.1002/1873-3468.14750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/05/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023]
Abstract
Nuclear lamins are type-V intermediate filaments that are involved in many nuclear processes. In mammals, A- and B-type lamins assemble into separate physical meshwork underneath the inner nuclear membrane, the nuclear lamina, with some residual fraction localized within the nucleoplasm. Lamins are the major part of the nucleoskeleton, providing mechanical strength and flexibility to protect the genome and allow nuclear deformability, while also contributing to gene regulation via interactions with chromatin. While lamins are the evolutionary ancestors of all intermediate filament family proteins, their ultimate filamentous assembly is markedly different from their cytoplasmic counterparts. Interestingly, hundreds of genetic mutations in the lamina proteins have been causally linked with a broad range of human pathologies, termed laminopathies. These include muscular, neurological and metabolic disorders, as well as premature aging diseases. Recent technological advances have contributed to resolving the filamentous structure of lamins and the corresponding lamina organization. In this review, we revisit the multiscale lamin organization and discuss its implications on nuclear mechanics and chromatin organization within lamina-associated domains.
Collapse
Affiliation(s)
- Amnon Buxboim
- The Rachel and Selim Benin School of Computer Science and Engineering and The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Israel
| | | | - Sarka Salajkova
- Department of Biochemistry, University of Zurich, Switzerland
| | - Nili Avidan
- The Rachel and Selim Benin School of Computer Science and Engineering and The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Israel
| | - Hen Shahak
- The Rachel and Selim Benin School of Computer Science and Engineering and The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Israel
| | - Alice Thurston
- Department of Biochemistry, University of Zurich, Switzerland
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Switzerland
| |
Collapse
|
26
|
Bucki R, Iwamoto DV, Shi X, Kerr KE, Byfield FJ, Suprewicz Ł, Skłodowski K, Sutaria J, Misiak P, Wilczewska AZ, Ramachandran S, Wolfe A, Thanh MTH, Whalen E, Patteson AE, Janmey PA. Extracellular vimentin is sufficient to promote cell attachment, spreading, and motility by a mechanism involving N-acetyl glucosamine-containing structures. J Biol Chem 2023; 299:104963. [PMID: 37356720 PMCID: PMC10392088 DOI: 10.1016/j.jbc.2023.104963] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/29/2023] [Accepted: 06/06/2023] [Indexed: 06/27/2023] Open
Abstract
Vimentin intermediate filaments form part of the cytoskeleton of mesenchymal cells, but under pathological conditions often associated with inflammation, vimentin filaments depolymerize as the result of phosphorylation or citrullination, and vimentin oligomers are secreted or released into the extracellular environment. In the extracellular space, vimentin can bind surfaces of cells and the extracellular matrix, and the interaction between extracellular vimentin and cells can trigger changes in cellular functions, such as activation of fibroblasts to a fibrotic phenotype. The mechanism by which extracellular vimentin binds external cell membranes and whether vimentin alone can act as an adhesive anchor for cells is largely uncharacterized. Here, we show that various cell types (normal and vimentin null fibroblasts, mesenchymal stem cells, and A549 lung carcinoma cells) attach to and spread on polyacrylamide hydrogel substrates covalently linked to vimentin. Using traction force microscopy and spheroid expansion assays, we characterize how different cell types respond to extracellular vimentin. Cell attachment to and spreading on vimentin-coated surfaces is inhibited by hyaluronic acid degrading enzymes, hyaluronic acid synthase inhibitors, soluble heparin or N-acetyl glucosamine, all of which are treatments that have little or no effect on the same cell types binding to collagen-coated hydrogels. These studies highlight the effectiveness of substrate-bound vimentin as a ligand for cells and suggest that carbohydrate structures, including the glycocalyx and glycosylated cell surface proteins that contain N-acetyl glucosamine, form a novel class of adhesion receptors for extracellular vimentin that can either directly support cell adhesion to a substrate or fine-tune the glycocalyx adhesive properties.
Collapse
Affiliation(s)
- Robert Bucki
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland.
| | - Daniel V Iwamoto
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xuechen Shi
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katherine E Kerr
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fitzroy J Byfield
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Łukasz Suprewicz
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Karol Skłodowski
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Julian Sutaria
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paweł Misiak
- Faculty of Chemistry, University of Białystok, Białystok, Poland
| | | | | | - Aaron Wolfe
- Ichor Life Sciences, Inc, LaFayette, New York, USA; Lewis School of Health Sciences, Clarkson University, Potsdam, New York, USA
| | - Minh-Tri Ho Thanh
- Physics Department, BioInspired Institute, Syracuse University, Syracuse, New York, USA
| | - Eli Whalen
- Physics Department, BioInspired Institute, Syracuse University, Syracuse, New York, USA
| | - Alison E Patteson
- Physics Department, BioInspired Institute, Syracuse University, Syracuse, New York, USA.
| | - Paul A Janmey
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
27
|
Leube RE, Quinlan RA. Editorial: The wetware credentials of intermediate filaments involves coordinating, organising and networking in cells and tissues. Front Cell Dev Biol 2023; 11:1146618. [PMID: 36861037 PMCID: PMC9969193 DOI: 10.3389/fcell.2023.1146618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Affiliation(s)
- Rudolf E. Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Aachen, Germany,*Correspondence: Rudolf E. Leube, ; Roy A. Quinlan,
| | - Roy A. Quinlan
- Department of Biosciences, University of Durham, Upper Mountjoy Science Site, Durham, United Kingdom,Biophysical Sciences Institute, University of Durham, Durham, United Kingdom,Department of Biological Structure, University of Washington, Seattle, WA, United States,*Correspondence: Rudolf E. Leube, ; Roy A. Quinlan,
| |
Collapse
|
28
|
Sun B, Kekenes-Huskey PM. Myofilament-associated proteins with intrinsic disorder (MAPIDs) and their resolution by computational modeling. Q Rev Biophys 2023; 56:e2. [PMID: 36628457 PMCID: PMC11070111 DOI: 10.1017/s003358352300001x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The cardiac sarcomere is a cellular structure in the heart that enables muscle cells to contract. Dozens of proteins belong to the cardiac sarcomere, which work in tandem to generate force and adapt to demands on cardiac output. Intriguingly, the majority of these proteins have significant intrinsic disorder that contributes to their functions, yet the biophysics of these intrinsically disordered regions (IDRs) have been characterized in limited detail. In this review, we first enumerate these myofilament-associated proteins with intrinsic disorder (MAPIDs) and recent biophysical studies to characterize their IDRs. We secondly summarize the biophysics governing IDR properties and the state-of-the-art in computational tools toward MAPID identification and characterization of their conformation ensembles. We conclude with an overview of future computational approaches toward broadening the understanding of intrinsic disorder in the cardiac sarcomere.
Collapse
Affiliation(s)
- Bin Sun
- Research Center for Pharmacoinformatics (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | | |
Collapse
|
29
|
Lee J, Cho H, Kwon I. Phase separation of low-complexity domains in cellular function and disease. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1412-1422. [PMID: 36175485 PMCID: PMC9534829 DOI: 10.1038/s12276-022-00857-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 11/09/2022]
Abstract
In this review, we discuss the ways in which recent studies of low-complexity (LC) domains have challenged our understanding of the mechanisms underlying cellular organization. LC sequences, long believed to function in the absence of a molecular structure, are abundant in the proteomes of all eukaryotic organisms. Over the past decade, the phase separation of LC domains has emerged as a fundamental mechanism driving dynamic multivalent interactions of many cellular processes. We review the key evidence showing the role of phase separation of individual proteins in organizing cellular assemblies and facilitating biological function while implicating the dynamics of phase separation as a key to biological validity and functional utility. We also highlight the evidence showing that pathogenic LC proteins alter various phase separation-dependent interactions to elicit debilitating human diseases, including cancer and neurodegenerative diseases. Progress in understanding the biology of phase separation may offer useful hints toward possible therapeutic interventions to combat the toxicity of pathogenic proteins.
Collapse
Affiliation(s)
- Jiwon Lee
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea.
| | - Ilmin Kwon
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea.
| |
Collapse
|
30
|
Liu P, Zhang S, Ma J, Jin D, Qin Y, Chen M. Vimentin inhibits α-tubulin acetylation via enhancing α-TAT1 degradation to suppress the replication of human parainfluenza virus type 3. PLoS Pathog 2022; 18:e1010856. [PMID: 36108090 PMCID: PMC9524669 DOI: 10.1371/journal.ppat.1010856] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/30/2022] [Accepted: 09/03/2022] [Indexed: 11/24/2022] Open
Abstract
We previously found that, among human parainfluenza virus type 3 (HPIV3) proteins, the interaction of nucleoprotein (N) and phosphoprotein (P) provides the minimal requirement for the formation of cytoplasmic inclusion bodies (IBs), which are sites of RNA synthesis, and that acetylated α-tubulin enhances IB fusion and viral replication. In this study, using immunoprecipitation and mass spectrometry assays, we determined that vimentin (VIM) specifically interacted with the N-P complex of HPIV3, and that the head domain of VIM was responsible for this interaction, contributing to the inhibition of IB fusion and viral replication. Furthermore, we found that VIM promoted the degradation of α-tubulin acetyltransferase 1 (α-TAT1), through its head region, thereby inhibiting the acetylation of α-tubulin, IB fusion, and viral replication. In addition, we identified a 20-amino-acid peptide derived from the head region of VIM that participated in the interaction with the N-P complex and inhibited viral replication. Our findings suggest that VIM inhibits the formation of HPIV3 IBs by downregulating α-tubulin acetylation via enhancing the degradation of α-TAT1. Our work sheds light on a new mechanism by which VIM suppresses HPIV3 replication.
Collapse
Affiliation(s)
- Pengfei Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Luo Jia Hill, Wuhan, China
| | - Shengwei Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Jingyi Ma
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Luo Jia Hill, Wuhan, China
| | - Dongning Jin
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Luo Jia Hill, Wuhan, China
| | - Yali Qin
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Luo Jia Hill, Wuhan, China
| | - Mingzhou Chen
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Luo Jia Hill, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
31
|
Zhou X, Sumrow L, Tashiro K, Sutherland L, Liu D, Qin T, Kato M, Liszczak G, McKnight SL. Mutations linked to neurological disease enhance self-association of low-complexity protein sequences. Science 2022; 377:eabn5582. [PMID: 35771920 PMCID: PMC9610444 DOI: 10.1126/science.abn5582] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Protein domains of low sequence complexity do not fold into stable, three-dimensional structures. Nevertheless, proteins with these sequences assist in many aspects of cell organization, including assembly of nuclear and cytoplasmic structures not surrounded by membranes. The dynamic nature of these cellular assemblies is caused by the ability of low-complexity domains (LCDs) to transiently self-associate through labile, cross-β structures. Mechanistic studies useful for the study of LCD self-association have evolved over the past decade in the form of simple assays of phase separation. Here, we have used such assays to demonstrate that the interactions responsible for LCD self-association can be dictated by labile protein structures poised close to equilibrium between the folded and unfolded states. Furthermore, missense mutations causing Charcot-Marie-Tooth disease, frontotemporal dementia, and Alzheimer's disease manifest their pathophysiology in vitro and in cultured cell systems by enhancing the stability of otherwise labile molecular structures formed upon LCD self-association.
Collapse
|
32
|
Aledo JC. A Census of Human Methionine-Rich Prion-like Domain-Containing Proteins. Antioxidants (Basel) 2022; 11:antiox11071289. [PMID: 35883780 PMCID: PMC9312190 DOI: 10.3390/antiox11071289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Methionine-rich prion-like proteins can regulate liquid–liquid phase separation processes in response to stresses. To date, however, very few proteins have been identified as methionine-rich prion-like. Herein, we have performed a computational survey of the human proteome to search for methionine-rich prion-like domains. We present a census of 51 manually curated methionine-rich prion-like proteins. Our results show that these proteins tend to be modular in nature, with molecular sizes significantly greater than those we would expect due to random sampling effects. These proteins also exhibit a remarkably high degree of spatial compaction when compared to average human proteins, even when protein size is accounted for. Computational evidence suggests that such a high degree of compactness might be due to the aggregation of methionine residues, pointing to a potential redox regulation of compactness. Gene ontology and network analyses, performed to shed light on the biological processes in which these proteins might participate, indicate that methionine-rich and non-methionine-rich prion-like proteins share gene ontology terms related to the regulation of transcription and translation but, more interestingly, these analyses also reveal that proteins from the methionine-rich group tend to share more gene ontology terms among them than they do with their non-methionine-rich prion-like counterparts.
Collapse
Affiliation(s)
- Juan Carlos Aledo
- Department of Molecular Biology and Biochemistry, University of Malaga, 29071 Malaga, Spain
| |
Collapse
|
33
|
Rosenberg GM, Murray KA, Salwinski L, Hughes MP, Abskharon R, Eisenberg DS. Bioinformatic identification of previously unrecognized amyloidogenic proteins. J Biol Chem 2022; 298:101920. [PMID: 35405097 PMCID: PMC9108986 DOI: 10.1016/j.jbc.2022.101920] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/04/2022] Open
Abstract
Low-complexity domains (LCDs) of proteins have been shown to self-associate, and pathogenic mutations within these domains often drive the proteins into amyloid aggregation associated with disease. These domains may be especially susceptible to amyloidogenic mutations because they are commonly intrinsically disordered and function in self-association. The question therefore arises whether a search for pathogenic mutations in LCDs of the human proteome can lead to identification of other proteins associated with amyloid disease. Here, we take a computational approach to identify documented pathogenic mutations within LCDs that may favor amyloid formation. Using this approach, we identify numerous known amyloidogenic mutations, including several such mutations within proteins previously unidentified as amyloidogenic. Among the latter group, we focus on two mutations within the TRK-fused gene protein (TFG), known to play roles in protein secretion and innate immunity, which are associated with two different peripheral neuropathies. We show that both mutations increase the propensity of TFG to form amyloid fibrils. We therefore conclude that TFG is a novel amyloid protein and propose that the diseases associated with its mutant forms may be amyloidoses.
Collapse
Affiliation(s)
- Gregory M Rosenberg
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, California, USA
| | - Kevin A Murray
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, California, USA
| | - Lukasz Salwinski
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, California, USA
| | - Michael P Hughes
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, California, USA; Department of Cell and Molecular Biology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Romany Abskharon
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, California, USA
| | - David S Eisenberg
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, California, USA.
| |
Collapse
|
34
|
Flynn LL, Li R, Pitout IL, Aung-Htut MT, Larcher LM, Cooper JAL, Greer KL, Hubbard A, Griffiths L, Bond CS, Wilton SD, Fox AH, Fletcher S. Single Stranded Fully Modified-Phosphorothioate Oligonucleotides can Induce Structured Nuclear Inclusions, Alter Nuclear Protein Localization and Disturb the Transcriptome In Vitro. Front Genet 2022; 13:791416. [PMID: 35464859 PMCID: PMC9019733 DOI: 10.3389/fgene.2022.791416] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/02/2022] [Indexed: 01/12/2023] Open
Abstract
Oligonucleotides and nucleic acid analogues that alter gene expression are now showing therapeutic promise in human disease. Whilst the modification of synthetic nucleic acids to protect against nuclease degradation and to influence drug function is common practice, such modifications may also confer unexpected physicochemical and biological properties. Gapmer mixed-modified and DNA oligonucleotides on a phosphorothioate backbone can bind non-specifically to intracellular proteins to form a variety of toxic inclusions, driven by the phosphorothioate linkages, but also influenced by the oligonucleotide sequence. Recently, the non-antisense or other off-target effects of 2′ O- fully modified phosphorothioate linkage oligonucleotides are becoming better understood. Here, we report chemistry-specific effects of oligonucleotides composed of modified or unmodified bases, with phosphorothioate linkages, on subnuclear organelles and show altered distribution of nuclear proteins, the appearance of highly stable and strikingly structured nuclear inclusions, and disturbed RNA processing in primary human fibroblasts and other cultured cells. Phosphodiester, phosphorodiamidate morpholino oligomers, and annealed complimentary phosphorothioate oligomer duplexes elicited no such consequences. Disruption of subnuclear structures and proteins elicit severe phenotypic disturbances, revealed by transcriptomic analysis of transfected fibroblasts exhibiting such disruption. Our data add to the growing body of evidence of off-target effects of some phosphorothioate nucleic acid drugs in primary cells and suggest alternative approaches to mitigate these effects.
Collapse
Affiliation(s)
- Loren L Flynn
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia.,Black Swan Pharmaceuticals, Wake Forest, NC, United States
| | - Ruohan Li
- Cell and Tissue Therapies WA, Royal Perth Hospital, Perth, WA, Australia.,School of Human Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Ianthe L Pitout
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,PYC Therapeutics, Nedlands, WA, Australia
| | - May T Aung-Htut
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Leon M Larcher
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Jack A L Cooper
- School of Human Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Kane L Greer
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Alysia Hubbard
- Centre for Microscopy, Characterization and Analysis, The University of Western Australia, Nedlands, WA, Australia
| | - Lisa Griffiths
- Anatomical Pathology, Department of Health, Nedlands, WA, Australia
| | - Charles S Bond
- School of Molecular Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Steve D Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Archa H Fox
- School of Human Sciences, The University of Western Australia, Nedlands, WA, Australia.,School of Molecular Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Sue Fletcher
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia.,PYC Therapeutics, Nedlands, WA, Australia
| |
Collapse
|
35
|
Shiota T, Nagata R, Kikuchi S, Nanaura H, Matsubayashi M, Nakanishi M, Kobashigawa S, Isozumi N, Kiriyama T, Nagayama K, Sugie K, Yamashiro Y, Mori E. C9orf72-Derived Proline:Arginine Poly-Dipeptides Modulate Cytoskeleton and Mechanical Stress Response. Front Cell Dev Biol 2022; 10:750829. [PMID: 35399536 PMCID: PMC8983821 DOI: 10.3389/fcell.2022.750829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 03/07/2022] [Indexed: 11/28/2022] Open
Abstract
Proline:arginine (PR) poly-dipeptides from the GGGGCC repeat expansion in C9orf72 have cytotoxicity and bind intermediate filaments (IFs). However, it remains unknown how PR poly-dipeptides affect cytoskeletal organization and focal adhesion (FA) formation. Here, we show that changes to the cytoskeleton and FA by PR poly-dipeptides result in the alteration of cell stiffness and mechanical stress response. PR poly-dipeptides increased the junctions and branches of the IF network and increased cell stiffness. They also changed the distribution of actin filaments and increased the size of FA and intracellular calcium concentration. PR poly-dipeptides or an inhibitor of IF organization prevented cell detachment. Furthermore, PR poly-dipeptides induced upregulation of mechanical stress response factors and led to a maladaptive response to cyclic stretch. These results suggest that the effects of PR poly-dipeptides on mechanical properties and mechanical stress response may serve as a pathogenesis of C9orf72-related neurodegeneration.
Collapse
Affiliation(s)
- Tomo Shiota
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Riko Nagata
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Sotaro Kikuchi
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Hitoki Nanaura
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Masaya Matsubayashi
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Mari Nakanishi
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Shinko Kobashigawa
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Noriyoshi Isozumi
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
| | - Takao Kiriyama
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Kazuaki Nagayama
- Micro-Nano Biomechanics Laboratory, Department of Mechanical Systems Engineering, Ibaraki University, Hitachi, Japan
| | - Kazuma Sugie
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Yoshito Yamashiro
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, Tsukuba, Japan
- *Correspondence: Yoshito Yamashiro, ; Eiichiro Mori,
| | - Eiichiro Mori
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Japan
- V-iCliniX Laboratory, Nara Medical University, Kashihara, Japan
- *Correspondence: Yoshito Yamashiro, ; Eiichiro Mori,
| |
Collapse
|
36
|
Murray KA, Hughes MP, Hu CJ, Sawaya MR, Salwinski L, Pan H, French SW, Seidler PM, Eisenberg DS. Identifying amyloid-related diseases by mapping mutations in low-complexity protein domains to pathologies. Nat Struct Mol Biol 2022; 29:529-536. [PMID: 35637421 PMCID: PMC9205782 DOI: 10.1038/s41594-022-00774-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 04/08/2022] [Indexed: 01/19/2023]
Abstract
Proteins including FUS, hnRNPA2, and TDP-43 reversibly aggregate into amyloid-like fibrils through interactions of their low-complexity domains (LCDs). Mutations in LCDs can promote irreversible amyloid aggregation and disease. We introduce a computational approach to identify mutations in LCDs of disease-associated proteins predicted to increase propensity for amyloid aggregation. We identify several disease-related mutations in the intermediate filament protein keratin-8 (KRT8). Atomic structures of wild-type and mutant KRT8 segments confirm the transition to a pleated strand capable of amyloid formation. Biochemical analysis reveals KRT8 forms amyloid aggregates, and the identified mutations promote aggregation. Aggregated KRT8 is found in Mallory-Denk bodies, observed in hepatocytes of livers with alcoholic steatohepatitis (ASH). We demonstrate that ethanol promotes KRT8 aggregation, and KRT8 amyloids co-crystallize with alcohol. Lastly, KRT8 aggregation can be seeded by liver extract from people with ASH, consistent with the amyloid nature of KRT8 aggregates and the classification of ASH as an amyloid-related condition.
Collapse
Affiliation(s)
- Kevin A. Murray
- grid.19006.3e0000 0000 9632 6718Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, CA USA
| | - Michael P. Hughes
- grid.19006.3e0000 0000 9632 6718Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, CA USA
| | - Carolyn J. Hu
- grid.19006.3e0000 0000 9632 6718Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, CA USA
| | - Michael R. Sawaya
- grid.19006.3e0000 0000 9632 6718Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, CA USA
| | - Lukasz Salwinski
- grid.19006.3e0000 0000 9632 6718Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, CA USA
| | - Hope Pan
- grid.19006.3e0000 0000 9632 6718Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, CA USA
| | - Samuel W. French
- grid.19006.3e0000 0000 9632 6718Department of Pathology & Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| | - Paul M. Seidler
- grid.42505.360000 0001 2156 6853Department of Pharmacology and Pharmaceutical Science, University of Southern California, Los Angeles, CA USA
| | - David S. Eisenberg
- grid.19006.3e0000 0000 9632 6718Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, UCLA, Los Angeles, CA USA
| |
Collapse
|
37
|
Abstract
This review covers research findings reported over the past decade concerning the ability of low complexity (LC) domains to self-associate in a manner leading to their phase separation from aqueous solution. We focus our message upon the reductionist use of two forms of phase separation as biochemical assays to study how LC domains might function in living cells. Cells and their varied compartments represent extreme examples of material condensates. Over the past half century, biochemists, structural biologists, and molecular biologists have resolved the mechanisms driving innumerable forms of macromolecular condensation. In contrast, we remain largely ignorant as to how 10%-20% of our proteins actually work to assist in cell organization. This enigmatic 10%-20% of the proteome corresponds to gibberish-like LC sequences. We contend that many of these LC sequences move in and out of a structurally ordered, self-associated state as a means of offering a combination of organizational specificity and dynamic pliability to living cells. Finally, we speculate that ancient proteins may have behaved similarly, helping to condense, organize, and protect RNA early during evolution.
Collapse
Affiliation(s)
- Masato Kato
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75390, USA
- Institute for Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Xiaoming Zhou
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Steven L McKnight
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
38
|
Sundaram MV. Intermediate filaments: New insights are bublin up. Curr Biol 2021; 31:R719-R721. [PMID: 34102119 DOI: 10.1016/j.cub.2021.04.031] [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] [Indexed: 11/16/2022]
Abstract
Cytoplasmic intermediate filaments affect cell shape and tissue integrity, and mutations in the proteins that make up these filaments contribute to many human diseases. A new study has identified a conserved protein, BBLN-1/bublin, that is important for intermediate filament organization.
Collapse
Affiliation(s)
- Meera V Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Room 446a, Clinical Research Building, 415 Curie Boulevard, Philadelphia, PA 19104-6145, USA.
| |
Collapse
|
39
|
Skeletal and Cardiac Muscle Disorders Caused by Mutations in Genes Encoding Intermediate Filament Proteins. Int J Mol Sci 2021; 22:ijms22084256. [PMID: 33923914 PMCID: PMC8073371 DOI: 10.3390/ijms22084256] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/12/2021] [Accepted: 04/15/2021] [Indexed: 02/08/2023] Open
Abstract
Intermediate filaments are major components of the cytoskeleton. Desmin and synemin, cytoplasmic intermediate filament proteins and A-type lamins, nuclear intermediate filament proteins, play key roles in skeletal and cardiac muscle. Desmin, encoded by the DES gene (OMIM *125660) and A-type lamins by the LMNA gene (OMIM *150330), have been involved in striated muscle disorders. Diseases include desmin-related myopathy and cardiomyopathy (desminopathy), which can be manifested with dilated, restrictive, hypertrophic, arrhythmogenic, or even left ventricular non-compaction cardiomyopathy, Emery–Dreifuss Muscular Dystrophy (EDMD2 and EDMD3, due to LMNA mutations), LMNA-related congenital Muscular Dystrophy (L-CMD) and LMNA-linked dilated cardiomyopathy with conduction system defects (CMD1A). Recently, mutations in synemin (SYNM gene, OMIM *606087) have been linked to cardiomyopathy. This review will summarize clinical and molecular aspects of desmin-, lamin- and synemin-related striated muscle disorders with focus on LMNA and DES-associated clinical entities and will suggest pathogenetic hypotheses based on the interplay of desmin and lamin A/C. In healthy muscle, such interplay is responsible for the involvement of this network in mechanosignaling, nuclear positioning and mitochondrial homeostasis, while in disease it is disturbed, leading to myocyte death and activation of inflammation and the associated secretome alterations.
Collapse
|
40
|
Assembly of NFL and desmin intermediate filaments: Headed in the right direction. Proc Natl Acad Sci U S A 2021; 118:2102176118. [PMID: 33707309 PMCID: PMC8020673 DOI: 10.1073/pnas.2102176118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|