51
|
Andley UP, Tycksen E, McGlasson-Naumann BN, Hamilton PD. Probing the changes in gene expression due to α-crystallin mutations in mouse models of hereditary human cataract. PLoS One 2018; 13:e0190817. [PMID: 29338044 PMCID: PMC5770019 DOI: 10.1371/journal.pone.0190817] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 12/20/2017] [Indexed: 11/30/2022] Open
Abstract
The mammalian eye lens expresses a high concentration of crystallins (α, β and γ-crystallins) to maintain the refractive index essential for lens transparency. Crystallins are long-lived proteins that do not turnover throughout life. The structural destabilization of crystallins by UV exposure, glycation, oxidative stress and mutations in crystallin genes leads to protein aggregation and development of cataracts. Several destabilizing mutations in crystallin genes are linked with human autosomal dominant hereditary cataracts. To investigate the mechanism by which the α-crystallin mutations Cryaa-R49C and Cryab-R120G lead to cataract formation, we determined whether these mutations cause an altered expression of specific transcripts in the lens at an early postnatal age by RNA-seq analysis. Using knock-in mouse models previously generated in our laboratory, in the present work, we identified genes that exhibited altered abundance in the mutant lenses, including decreased transcripts for Clic5, an intracellular water channel in Cryaa-R49C heterozygous mutant lenses, and increased transcripts for Eno1b in Cryab-R120G heterozygous mutant lenses. In addition, RNA-seq analysis revealed increased histones H2B, H2A, and H4 gene expression in Cryaa-R49C mutant lenses, suggesting that the αA-crystallin mutation regulates histone expression via a transcriptional mechanism. Additionally, these studies confirmed the increased expression of histones H2B, H2A, and H4 by proteomic analysis of Cryaa-R49C knock-in and Cryaa;Cryab gene knockout lenses reported previously. Taken together, these findings offer additional insight into the early transcriptional changes caused by Cryaa and Cryab mutations associated with autosomal dominant human cataracts, and indicate that the transcript levels of certain genes are affected by the expression of mutant α-crystallin in vivo.
Collapse
Affiliation(s)
- Usha P. Andley
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| | - Eric Tycksen
- Genome Technology Access Center, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Brittney N. McGlasson-Naumann
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Paul D. Hamilton
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
52
|
Araco M, Merlo M, Carr-White G, Sinagra G. Genetic bases of dilated cardiomyopathy. J Cardiovasc Med (Hagerstown) 2017; 18:123-130. [PMID: 27661610 DOI: 10.2459/jcm.0000000000000432] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cardiomyopathies represent a wide and heterogeneous group of diseases wherein a genetic cause has been consistently identified.Dilated cardiomyopathy (DCM) is characterized by ventricular dilation and progressive systolic dysfunction, and it is the most common form of cardiomyopathy.Causative genetic mutations have been identified in more than 40 genes encoding proteins belonging to different cellular structures and pathways.A great diversity of pathways has been implied in the pathogenesis of DCM, depending on the affected genes and on the dislodged intracellular structures or mechanisms.This review describes the major genes and focus on the pathophysiologic mechanisms of DCM, with a special consideration of the most recent discoveries in the field.
Collapse
Affiliation(s)
- Marco Araco
- aDepartment of Cardiology, Guys and St Thomas NHS Trust, London, United Kingdom bDivision of Cardiology, Cardiovascular Department, Ospedali Riuniti and University of Trieste, Trieste, Italy
| | | | | | | |
Collapse
|
53
|
Arbach H, Butler C, McMenimen KA. Chaperone activity of human small heat shock protein-GST fusion proteins. Cell Stress Chaperones 2017; 22:503-515. [PMID: 28130664 PMCID: PMC5465028 DOI: 10.1007/s12192-017-0764-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/09/2017] [Accepted: 01/11/2017] [Indexed: 12/17/2022] Open
Abstract
Small heat shock proteins (sHsps) are a ubiquitous part of the machinery that maintains cellular protein homeostasis by acting as molecular chaperones. sHsps bind to and prevent the aggregation of partially folded substrate proteins in an ATP-independent manner. sHsps are dynamic, forming an ensemble of structures from dimers to large oligomers through concentration-dependent equilibrium dissociation. Based on structural studies and mutagenesis experiments, it is proposed that the dimer is the smallest active chaperone unit, while larger oligomers may act as storage depots for sHsps or play additional roles in chaperone function. The complexity and dynamic nature of their structural organization has made elucidation of their chaperone function challenging. HspB1 and HspB5 are two canonical human sHsps that vary in sequence and are expressed in a wide variety of tissues. In order to determine the role of the dimer in chaperone activity, glutathione-S-transferase (GST) was genetically linked as a fusion protein to the N-terminus regions of both HspB1 and HspB5 (also known as Hsp27 and αB-crystallin, respectively) proteins in order to constrain oligomer formation of HspB1 and HspB5, by using GST, since it readily forms a dimeric structure. We monitored the chaperone activity of these fusion proteins, which suggest they primarily form dimers and monomers and function as active molecular chaperones. Furthermore, the two different fusion proteins exhibit different chaperone activity for two model substrate proteins, citrate synthase (CS) and malate dehydrogenase (MDH). GST-HspB1 prevents more aggregation of MDH compared to GST-HspB5 and wild type HspB1. However, when CS is the substrate, both GST-HspB1 and GST-HspB5 are equally effective chaperones. Furthermore, wild type proteins do not display equal activity toward the substrates, suggesting that each sHsp exhibits different substrate specificity. Thus, substrate specificity, as described here for full-length GST fusion proteins with MDH and CS, is modulated by both sHsp oligomeric conformation and by variations of sHsp sequences.
Collapse
Affiliation(s)
- Hannah Arbach
- Department of Chemistry, Mount Holyoke College, 50 College Street, South Hadley, MA, 01075, USA
| | - Caley Butler
- Department of Chemistry, Mount Holyoke College, 50 College Street, South Hadley, MA, 01075, USA
| | - Kathryn A McMenimen
- Department of Chemistry, Mount Holyoke College, 50 College Street, South Hadley, MA, 01075, USA.
| |
Collapse
|
54
|
Dabbaghizadeh A, Finet S, Morrow G, Moutaoufik MT, Tanguay RM. Oligomeric structure and chaperone-like activity of Drosophila melanogaster mitochondrial small heat shock protein Hsp22 and arginine mutants in the alpha-crystallin domain. Cell Stress Chaperones 2017; 22:577-588. [PMID: 28389817 PMCID: PMC5465034 DOI: 10.1007/s12192-017-0784-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 02/24/2017] [Accepted: 02/28/2017] [Indexed: 01/17/2023] Open
Abstract
The structure and chaperone function of DmHsp22WT, a small Hsp of Drosophila melanogaster localized within mitochondria were examined. Mutations of conserved arginine mutants within the alpha-crystallin domain (ACD) domain (R105G, R109G, and R110G) were introduced, and their effects on oligomerization and chaperone function were assessed. Arginine to glycine mutations do not induce significant changes in tryptophan fluorescence, and the mutated proteins form oligomers that are of equal or smaller size than the wild-type protein. They all form oligomer with one single peak as determined by size exclusion chromatography. While all mutants demonstrate the same efficiency as the DmHsp22WT in a DTT-induced insulin aggregation assay, all are more efficient chaperones to prevent aggregation of malate dehydrogenase. Arginine mutants of DmHsp22 are efficient chaperones to retard aggregation of CS and Luc. In summary, this study shows that mutations of arginine to glycine in DmHsp22 ACD induce a number of structural changes, some of which differ from those described in mammalian sHsps. Interestingly, only the R110G-DmHsp22 mutant, and not the expected R109G equivalent to human R140-HspB1, R116-HspB4, and R120-HspB5, showed different structural properties compared with the DmHsp22WT.
Collapse
Affiliation(s)
- Afrooz Dabbaghizadeh
- Laboratoire de génétique cellulaire et développementale, Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de médecine, Institut de biologie intégrative et des systèmes (IBIS) and PROTEO, Université Laval, Québec, Québec, G1V 0A6, Canada
| | - Stéphanie Finet
- IMPMC UMR7590, CNRS, Sorbonne-Universités, MNHN, IRD, 4 place Jussieu, Paris, France
| | - Genevieve Morrow
- Laboratoire de génétique cellulaire et développementale, Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de médecine, Institut de biologie intégrative et des systèmes (IBIS) and PROTEO, Université Laval, Québec, Québec, G1V 0A6, Canada
| | - Mohamed Taha Moutaoufik
- Laboratoire de génétique cellulaire et développementale, Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de médecine, Institut de biologie intégrative et des systèmes (IBIS) and PROTEO, Université Laval, Québec, Québec, G1V 0A6, Canada
| | - Robert M Tanguay
- Laboratoire de génétique cellulaire et développementale, Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de médecine, Institut de biologie intégrative et des systèmes (IBIS) and PROTEO, Université Laval, Québec, Québec, G1V 0A6, Canada.
| |
Collapse
|
55
|
Moutaoufik MT, Morrow G, Maaroufi H, Férard C, Finet S, Tanguay RM. Oligomerization and chaperone-like activity of Drosophila melanogaster small heat shock protein DmHsp27 and three arginine mutants in the alpha-crystallin domain. Cell Stress Chaperones 2017; 22:455-466. [PMID: 27933579 PMCID: PMC5465024 DOI: 10.1007/s12192-016-0748-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/09/2016] [Accepted: 11/10/2016] [Indexed: 11/30/2022] Open
Abstract
The small Hsp DmHsp27 from Drosophila melanogaster is one of the few small heat shock proteins (sHsps) found within the nucleus. We report that its dimerization is independent of disulfide bond formation and seems to rely on salt bridges. Unlike metazoan sHsps, DmHsp27 forms two populations of oligomers not in equilibrium. Mutations at highly conserved arginine residues in mammalian sHsps have been reported to be associated with protein conformational defects and intracellular aggregation. Independent mutation of three highly conserved arginines (R122, R131, and R135) to glycine in DmHsp27 results in only one population of higher molecular weight form. In vitro, the chaperone-like activity of wild-type DmHsp27 was comparable with that of its two isolated populations and to the single population of the R122G, R131G, and R135G using luciferase as substrate. However, using insulin, the chaperone-like activity of wild-type DmHsp27 was lower than that of R122G and R131G mutants. Altogether, the results characterize wild-type DmHsp27 and its alpha-crystallin domain (ACD) arginine mutants and may give insight into protection mechanism of sHsps.
Collapse
Affiliation(s)
- Mohamed Taha Moutaoufik
- Laboratoire de génétique cellulaire et développementale, Département de biologie moléculaire, de biochimie médicale et de pathologie, Institut de biologie intégrative et des systèmes (IBIS) and PROTEO, Université Laval, Québec, G1V 0A6, Canada
| | - Geneviève Morrow
- Laboratoire de génétique cellulaire et développementale, Département de biologie moléculaire, de biochimie médicale et de pathologie, Institut de biologie intégrative et des systèmes (IBIS) and PROTEO, Université Laval, Québec, G1V 0A6, Canada
| | - Halim Maaroufi
- Plate-forme de bio-informatique, Institut de biologie intégrative et des systèmes (IBIS), Université Laval, Québec, G1V 0A6, Canada
| | - Céline Férard
- IMPMC UMR7590, CNRS, UPMC Paris 6, 4 place Jussieu, Paris, France
| | - Stéphanie Finet
- IMPMC UMR7590, CNRS, UPMC Paris 6, 4 place Jussieu, Paris, France
| | - Robert M Tanguay
- Laboratoire de génétique cellulaire et développementale, Département de biologie moléculaire, de biochimie médicale et de pathologie, Institut de biologie intégrative et des systèmes (IBIS) and PROTEO, Université Laval, Québec, G1V 0A6, Canada.
| |
Collapse
|
56
|
Carra S, Alberti S, Arrigo PA, Benesch JL, Benjamin IJ, Boelens W, Bartelt-Kirbach B, Brundel BJJM, Buchner J, Bukau B, Carver JA, Ecroyd H, Emanuelsson C, Finet S, Golenhofen N, Goloubinoff P, Gusev N, Haslbeck M, Hightower LE, Kampinga HH, Klevit RE, Liberek K, Mchaourab HS, McMenimen KA, Poletti A, Quinlan R, Strelkov SV, Toth ME, Vierling E, Tanguay RM. The growing world of small heat shock proteins: from structure to functions. Cell Stress Chaperones 2017; 22:601-611. [PMID: 28364346 PMCID: PMC5465036 DOI: 10.1007/s12192-017-0787-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2017] [Indexed: 12/21/2022] Open
Abstract
Small heat shock proteins (sHSPs) are present in all kingdoms of life and play fundamental roles in cell biology. sHSPs are key components of the cellular protein quality control system, acting as the first line of defense against conditions that affect protein homeostasis and proteome stability, from bacteria to plants to humans. sHSPs have the ability to bind to a large subset of substrates and to maintain them in a state competent for refolding or clearance with the assistance of the HSP70 machinery. sHSPs participate in a number of biological processes, from the cell cycle, to cell differentiation, from adaptation to stressful conditions, to apoptosis, and, even, to the transformation of a cell into a malignant state. As a consequence, sHSP malfunction has been implicated in abnormal placental development and preterm deliveries, in the prognosis of several types of cancer, and in the development of neurological diseases. Moreover, mutations in the genes encoding several mammalian sHSPs result in neurological, muscular, or cardiac age-related diseases in humans. Loss of protein homeostasis due to protein aggregation is typical of many age-related neurodegenerative and neuromuscular diseases. In light of the role of sHSPs in the clearance of un/misfolded aggregation-prone substrates, pharmacological modulation of sHSP expression or function and rescue of defective sHSPs represent possible routes to alleviate or cure protein conformation diseases. Here, we report the latest news and views on sHSPs discussed by many of the world's experts in the sHSP field during a dedicated workshop organized in Italy (Bertinoro, CEUB, October 12-15, 2016).
Collapse
Affiliation(s)
- Serena Carra
- Department of Biomedical, Metabolic and Neural Sciences, and Centre for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, via G. Campi 287, 41125 Modena, Italy
| | - Simon Alberti
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Patrick A. Arrigo
- Université de Lyon, 69622 Lyon, France
- CNRS, UMR 5310, INSERM U1217, Institut NeuroMyoGène, Université Lyon 1, 69100 Villeurbanne, France
| | | | - Ivor J. Benjamin
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112-5650 USA
| | - Wilbert Boelens
- Biomolecular Chemistry, 284, Radboud University, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | - Bianca J. J. M. Brundel
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Bernd Bukau
- Center for Molecular Biology of the University of Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - John A. Carver
- The Research School of Chemistry, The Australian National University, Acton, ACT 2601 Australia
| | - Heath Ecroyd
- Illawara Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Wollongong, NSW 2522 Australia
| | - Cecilia Emanuelsson
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, 221 00 Lund, Sweden
| | - Stephanie Finet
- IMPMC UMR7590, CNRS, UPMC Paris 6, 4 place Jussieu, Paris, France
| | - Nikola Golenhofen
- Institute of Anatomy and Cell Biology, University of Ulm, 89081 Ulm, Germany
| | - Pierre Goloubinoff
- Department of Plant Molecular Biology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Nikolai Gusev
- Department of Biochemistry, School of Biology, Moscow State University, Moscow, 119991 Russia
| | | | - Lawrence E. Hightower
- Department of Molecular & Cell Biology, University of Connecticut, 91 North Eagleville Road, Storrs, CT 06269-3125 USA
| | - Harm H. Kampinga
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Rachel E. Klevit
- Department of Biochemistry, University of Washington, Seattle, WA 98195 USA
| | - Krzysztof Liberek
- Department of Molecular and Cellular Biology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
| | - Hassane S. Mchaourab
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232 USA
| | - Kathryn A. McMenimen
- Departments of Pathology, Biological Chemistry, and Medicinal Chemistry and the Life Sciences Institute, University of Michigan, Ann Arbor, MI USA
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Milan, Italy
| | - Roy Quinlan
- Department of Biosciences and the Biophysical Sciences Institute, University of Durham, Durham, UK
| | - Sergei V. Strelkov
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Melinda E. Toth
- Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Elizabeth Vierling
- Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003 USA
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721 USA
| | - Robert M. Tanguay
- Laboratory of Cell & Developmental Genetics, IBIS, and Department of Molecular Biology, Medical Biochemistry and Pathology, Medical School, Université Laval, Québec (Qc), G1V 0A6 Canada
| |
Collapse
|
57
|
Hu X, Van Marion DMS, Wiersma M, Zhang D, Brundel BJJM. The protective role of small heat shock proteins in cardiac diseases: key role in atrial fibrillation. Cell Stress Chaperones 2017; 22:665-674. [PMID: 28484965 PMCID: PMC5465041 DOI: 10.1007/s12192-017-0799-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 02/06/2023] Open
Abstract
Atrial fibrillation (AF) is the most common tachyarrhythmia which is associated with increased morbidity and mortality. AF usually progresses from a self-terminating paroxysmal to persistent disease. It has been recognized that AF progression is driven by structural remodeling of cardiomyocytes, which results in electrical and contractile dysfunction of the atria. We recently showed that structural remodeling is rooted in derailment of proteostasis, i.e., homeostasis of protein production, function, and degradation. Since heat shock proteins (HSPs) play an important role in maintaining a healthy proteostasis, the role of HSPs was investigated in AF. It was found that especially small heat shock protein (HSPB) levels get exhausted in atrial tissue of patients with persistent AF and that genetic or pharmacological induction of HSPB protects against cardiomyocyte remodeling in experimental models for AF. In this review, we provide an overview of HSPBs as a potential therapeutic target for normalizing proteostasis and suppressing the substrates for AF progression in experimental and clinical AF and discuss HSP activators as a promising therapy to prevent AF onset and progression.
Collapse
Affiliation(s)
- Xu Hu
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Denise M S Van Marion
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Marit Wiersma
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Deli Zhang
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
58
|
Brodehl A, Gaertner-Rommel A, Klauke B, Grewe SA, Schirmer I, Peterschröder A, Faber L, Vorgerd M, Gummert J, Anselmetti D, Schulz U, Paluszkiewicz L, Milting H. The novel αB-crystallin (CRYAB) mutation p.D109G causes restrictive cardiomyopathy. Hum Mutat 2017; 38:947-952. [PMID: 28493373 DOI: 10.1002/humu.23248] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/03/2017] [Accepted: 05/03/2017] [Indexed: 11/09/2022]
Abstract
Restrictive cardiomyopathy (RCM) is a rare heart disease characterized by diastolic dysfunction and atrial enlargement. The genetic etiology of RCM is not completely known. We identified by a next-generation sequencing panel the novel CRYAB missense mutation c.326A>G, p.D109G in a small family with RCM in combination with skeletal myopathy with an early onset of the disease. CRYAB encodes αB-crystallin, a member of the small heat shock protein family, which is highly expressed in cardiac and skeletal muscle. In addition to in silico prediction analysis, our structural analysis of explanted myocardial tissue of a mutation carrier as well as in vitro cell transfection experiments revealed abnormal protein aggregation of mutant αB-crystallin and desmin, supporting the deleterious effect of this novel mutation. In conclusion, CRYAB appears to be a novel RCM gene, which might have relevance for the molecular diagnosis and the genetic counseling of further affected families in the future.
Collapse
Affiliation(s)
- Andreas Brodehl
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, Erich and Hanna Klessmann Institute for Cardiovascular Research & Development (EHKI), Bad Oeynhausen, Germany
| | - Anna Gaertner-Rommel
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, Erich and Hanna Klessmann Institute for Cardiovascular Research & Development (EHKI), Bad Oeynhausen, Germany
| | - Bärbel Klauke
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, Erich and Hanna Klessmann Institute for Cardiovascular Research & Development (EHKI), Bad Oeynhausen, Germany
| | - Simon Andre Grewe
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, Erich and Hanna Klessmann Institute for Cardiovascular Research & Development (EHKI), Bad Oeynhausen, Germany
| | - Ilona Schirmer
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, Erich and Hanna Klessmann Institute for Cardiovascular Research & Development (EHKI), Bad Oeynhausen, Germany
| | - Andreas Peterschröder
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, Institute of Radiology, Nuclear Medicine and Molecular Imaging, Bad Oeynhausen, Germany
| | - Lothar Faber
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, Clinic of Cardiology, Bad Oeynhausen, Germany
| | - Matthias Vorgerd
- Department of Neurology, BG-University Hospital Bergmannsheil, Bochum, Germany
| | - Jan Gummert
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, Erich and Hanna Klessmann Institute for Cardiovascular Research & Development (EHKI), Bad Oeynhausen, Germany
| | - Dario Anselmetti
- Bielefeld University and Bielefeld Institute for Nanoscience (BINAS), Faculty of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld, Germany
| | - Uwe Schulz
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, Erich and Hanna Klessmann Institute for Cardiovascular Research & Development (EHKI), Bad Oeynhausen, Germany
| | - Lech Paluszkiewicz
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, Erich and Hanna Klessmann Institute for Cardiovascular Research & Development (EHKI), Bad Oeynhausen, Germany
| | - Hendrik Milting
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, Erich and Hanna Klessmann Institute for Cardiovascular Research & Development (EHKI), Bad Oeynhausen, Germany
| |
Collapse
|
59
|
Kimura H, Eguchi S, Sasaki J, Kuba K, Nakanishi H, Takasuga S, Yamazaki M, Goto A, Watanabe H, Itoh H, Imai Y, Suzuki A, Mizushima N, Sasaki T. Vps34 regulates myofibril proteostasis to prevent hypertrophic cardiomyopathy. JCI Insight 2017; 2:e89462. [PMID: 28097232 DOI: 10.1172/jci.insight.89462] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a common heart disease with a prevalence of 1 in 500 in the general population. Several mutations in genes encoding cardiac proteins have been found in HCM patients, but these changes do not predict occurrence or prognosis and the molecular mechanisms underlying HCM remain largely elusive. Here we show that cardiac expression of vacuolar protein sorting 34 (Vps34) is reduced in a subset of HCM patients. In a mouse model, muscle-specific loss of Vps34 led to HCM-like manifestations and sudden death. Vps34-deficient hearts exhibited abnormal histopathologies, including myofibrillar disarray and aggregates containing αB-crystallin (CryAB). These features result from a block in the ESCRT-mediated proteolysis that normally degrades K63-polyubiquitinated CryAB. CryAB deposition was also found in myocardial specimens from a subset of HCM patients whose hearts showed decreased Vps34. Our results identify disruption of the previously unknown Vps34-CryAB axis as a potentially novel etiology of HCM.
Collapse
Affiliation(s)
- Hirotaka Kimura
- Research Center for Biosignaling, Department of.,Medical Biology
| | | | | | - Keiji Kuba
- Biological Informatics and Experimental Therapeutics Pathology
| | | | | | | | | | - Hiroyuki Watanabe
- Cardiovascular and Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroshi Itoh
- Cardiovascular and Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Yumiko Imai
- Biological Informatics and Experimental Therapeutics Pathology
| | - Akira Suzuki
- Department of Molecular Genetics, Division of Cancer Genetics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Takehiko Sasaki
- Research Center for Biosignaling, Department of.,Medical Biology
| |
Collapse
|
60
|
Lim EMF, Musa A, Frederick A, Ousman SS. AlphaB-crystallin expression correlates with aging deficits in the peripheral nervous system. Neurobiol Aging 2017; 53:138-149. [PMID: 28185662 DOI: 10.1016/j.neurobiolaging.2017.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 01/03/2017] [Accepted: 01/04/2017] [Indexed: 11/17/2022]
Abstract
In an effort to identify factors that contribute to age-related deficits in the undamaged and injured peripheral nervous system (PNS), we noted that Brady and colleagues found that mice null for a small heat shock protein called alphaB-crystallin (αBC) developed abnormalities early in life that are reminiscent of aging pathologies. Because of our observation that αBC protein levels markedly reduce as wild-type mice age, we investigated whether the crystallin plays a role in modulating age-related deficits in the uninjured and damaged PNS. We show here that the presence of αBC correlates with maintenance of myelin sheath thickness, reducing macrophage presence, sustaining lipid metabolism, and promoting remyelination following peripheral nerve injury in an age-dependent manner. More specifically, animals null for αBC displayed a higher frequency of thinly myelinated axons, enhanced presence of Iba1+ macrophages, and fewer immunoreactive profiles of the cholesterol biosynthesis enzyme, squalene monooxygenase, before and after sciatic nerve crush injury. These findings thus suggest that αBC plays a protective and beneficial role in the aging PNS.
Collapse
Affiliation(s)
- Erin-Mai F Lim
- Department of Neuroscience, University of Calgary and the Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Alim Musa
- Department of Clinical Neurosciences, University of Calgary and the Hotchkiss Brain Institute, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary and the Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Ariana Frederick
- Department of Clinical Neurosciences, University of Calgary and the Hotchkiss Brain Institute, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary and the Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Shalina S Ousman
- Department of Clinical Neurosciences, University of Calgary and the Hotchkiss Brain Institute, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary and the Hotchkiss Brain Institute, Calgary, Alberta, Canada.
| |
Collapse
|
61
|
Shiliaev NG, Selivanova OM, Galzitskaya OV. Search for conserved amino acid residues of the α-crystallin proteins of vertebrates. J Bioinform Comput Biol 2016; 14:1641004. [PMID: 26972563 DOI: 10.1142/s0219720016410043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
[Formula: see text]-crystallin is the major eye lens protein and a member of the small heat-shock protein (sHsp) family. [Formula: see text]-crystallins have been shown to support lens clarity by preventing the aggregation of lens proteins. We performed the bioinformatics analysis of [Formula: see text]-crystallin sequences from vertebrates to find conserved amino acid residues as the three-dimensional (3D) structure of [Formula: see text]-crystallin is not identified yet. We are the first who demonstrated that the N-terminal region is conservative along with the central domain for vertebrate organisms. We have found that there is correlation between the conserved and structured regions. Moreover, amyloidogenic regions also correspond to the structured regions. We analyzed the amino acid composition of [Formula: see text]-crystallin A and B chains. Analyzing the occurrence of each individual amino acid residue, we have found that such amino acid residues as leucine, serine, lysine, proline, phenylalanine, histidine, isoleucine, glutamic acid, and valine change their content simultaneously in A and B chains in different classes of vertebrates. Aromatic amino acids occur more often in [Formula: see text]-crystallins from vertebrates than on the average in proteins among 17 animal proteomes. We obtained that the identity between A and B chains in the mammalian group is 0.35, which is lower than the published 0.60.
Collapse
Affiliation(s)
- Nikita G Shiliaev
- 1 Institute of Protein Research, Russian Academy of Sciences, Institutskaya str., 4 Pushchino, Moscow Region 142290, Russia
| | - Olga M Selivanova
- 1 Institute of Protein Research, Russian Academy of Sciences, Institutskaya str., 4 Pushchino, Moscow Region 142290, Russia
| | - Oxana V Galzitskaya
- 1 Institute of Protein Research, Russian Academy of Sciences, Institutskaya str., 4 Pushchino, Moscow Region 142290, Russia
| |
Collapse
|
62
|
Bakthisaran R, Akula KK, Tangirala R, Rao CM. Phosphorylation of αB-crystallin: Role in stress, aging and patho-physiological conditions. Biochim Biophys Acta Gen Subj 2015; 1860:167-82. [PMID: 26415747 DOI: 10.1016/j.bbagen.2015.09.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 01/18/2023]
Abstract
BACKGROUND αB-crystallin, once thought to be a lenticular protein, is ubiquitous and has critical roles in several cellular processes that are modulated by phosphorylation. Serine residues 19, 45 and 59 of αB-crystallin undergo phosphorylation. Phosphorylation of S45 is mediated by p44/42 MAP kinase, whereas S59 phosphorylation is mediated by MAPKAP kinase-2. Pathway involved in S19 phosphorylation is not known. SCOPE OF REVIEW The review highlights the role of phosphorylation in (i) oligomeric structure, stability and chaperone activity, (ii) cellular processes such as apoptosis, myogenic differentiation, cell cycle regulation and angiogenesis, and (iii) aging, stress, cardiomyopathy-causing αB-crystallin mutants, and in other diseases. MAJOR CONCLUSIONS Depending on the context and extent of phosphorylation, αB-crystallin seems to confer beneficial or deleterious effects. Phosphorylation alters structure, stability, size distribution and dynamics of the oligomeric assembly, thus modulating chaperone activity and various cellular processes. Phosphorylated αB-crystallin has a tendency to partition to the cytoskeleton and hence to the insoluble fraction. Low levels of phosphorylation appear to be protective, while hyperphosphorylation has negative implications. Mutations in αB-crystallin, such as R120G, Q151X and 464delCT, associated with inherited myofibrillar myopathy lead to hyperphosphorylation and intracellular inclusions. An ongoing study in our laboratory with phosphorylation-mimicking mutants indicates that phosphorylation of R120GαB-crystallin increases its propensity to aggregate. GENERAL SIGNIFICANCE Phosphorylation of αB-crystallin has dual role that manifests either beneficial or deleterious consequences depending on the extent of phosphorylation and interaction with cytoskeleton. Considering that disease-causing mutants of αB-crystallin are hyperphosphorylated, moderation of phosphorylation may be a useful strategy in disease management. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
Affiliation(s)
- Raman Bakthisaran
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Kranthi Kiran Akula
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Ramakrishna Tangirala
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Ch Mohan Rao
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India.
| |
Collapse
|
63
|
Su H, Li J, Zhang H, Ma W, Wei N, Liu J, Wang X. COP9 signalosome controls the degradation of cytosolic misfolded proteins and protects against cardiac proteotoxicity. Circ Res 2015; 117:956-66. [PMID: 26383969 DOI: 10.1161/circresaha.115.306783] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 09/17/2015] [Indexed: 01/19/2023]
Abstract
RATIONALE Impaired degradation of misfolded proteins is associated with a large subset of heart diseases. Misfolded proteins are degraded primarily by the ubiquitin-proteasome system, but the ubiquitin ligases responsible for the degradation remain largely unidentified. The cullin deneddylation activity of the COP9 signalosome (CSN) requires all 8 CSN subunits (CSN1 through CSN8) and regulates cullin-RING ligases, thereby controlling ubiquitination of a large number of proteins; however, neither CSN nor cullin-RING ligases is known to regulate the degradation of cytosolic misfolded proteins. OBJECTIVE We sought to investigate the role of CSN8/CSN in misfolded protein degradation and cardiac proteinopathy. METHODS AND RESULTS Cardiac CSN8 knockout causes mouse premature death; hence, CSN8 hypomorphism (CSN8(hypo)) mice were used. Myocardial neddylated forms of cullins were markedly increased, and myocardial capacity of degrading a surrogate misfolded protein was significantly reduced by CSN8 hypomorphism. When introduced into proteinopathic mice in which a bona fide misfolded protein R120G missense mutation of αβ-crystallin (CryAB(R120G)) is overexpressed in the heart, CSN8 hypomorphism aggravated CryAB(R120G)-induced restrictive cardiomyopathy and shortened the lifespan of CryAB(R120G) mice, which was associated with augmented accumulation of protein aggregates, increased neddylated proteins, and reduced levels of total ubiquitinated proteins and LC3-II in the heart. In cultured cardiomyocytes, both CSN8 knockdown and cullin-RING ligase inactivation suppressed the ubiquitination and degradation of CryAB(R120G) but not native CryAB, resulting in accumulation of protein aggregates and exacerbation of CryAB(R120G) cytotoxicity. CONCLUSIONS (1) CSN8/CSN promotes the ubiquitination and degradation of misfolded proteins and protects against cardiac proteotoxicity, and (2) cullin-RING ligases participate in degradation of cytosolic misfolded proteins.
Collapse
Affiliation(s)
- Huabo Su
- From the State Key Laboratory of Respiratory Disease and Department of Pathophysiology, Guangzhou Medical University, Guangzhou, Guangdong, China (H.S., J.Liu, X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (H.S., J.Li, H.Z., J.Liu, X.W.); Vascular Biology Center (H.S., J.Li, W.M.) and Department of Pharmacology and Toxicology (H.S.), Medical College of Georgia, Georgia Regents University, Augusta; and Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT (N.W.)
| | - Jie Li
- From the State Key Laboratory of Respiratory Disease and Department of Pathophysiology, Guangzhou Medical University, Guangzhou, Guangdong, China (H.S., J.Liu, X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (H.S., J.Li, H.Z., J.Liu, X.W.); Vascular Biology Center (H.S., J.Li, W.M.) and Department of Pharmacology and Toxicology (H.S.), Medical College of Georgia, Georgia Regents University, Augusta; and Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT (N.W.)
| | - Hanming Zhang
- From the State Key Laboratory of Respiratory Disease and Department of Pathophysiology, Guangzhou Medical University, Guangzhou, Guangdong, China (H.S., J.Liu, X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (H.S., J.Li, H.Z., J.Liu, X.W.); Vascular Biology Center (H.S., J.Li, W.M.) and Department of Pharmacology and Toxicology (H.S.), Medical College of Georgia, Georgia Regents University, Augusta; and Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT (N.W.)
| | - Wenxia Ma
- From the State Key Laboratory of Respiratory Disease and Department of Pathophysiology, Guangzhou Medical University, Guangzhou, Guangdong, China (H.S., J.Liu, X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (H.S., J.Li, H.Z., J.Liu, X.W.); Vascular Biology Center (H.S., J.Li, W.M.) and Department of Pharmacology and Toxicology (H.S.), Medical College of Georgia, Georgia Regents University, Augusta; and Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT (N.W.)
| | - Ning Wei
- From the State Key Laboratory of Respiratory Disease and Department of Pathophysiology, Guangzhou Medical University, Guangzhou, Guangdong, China (H.S., J.Liu, X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (H.S., J.Li, H.Z., J.Liu, X.W.); Vascular Biology Center (H.S., J.Li, W.M.) and Department of Pharmacology and Toxicology (H.S.), Medical College of Georgia, Georgia Regents University, Augusta; and Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT (N.W.)
| | - Jinbao Liu
- From the State Key Laboratory of Respiratory Disease and Department of Pathophysiology, Guangzhou Medical University, Guangzhou, Guangdong, China (H.S., J.Liu, X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (H.S., J.Li, H.Z., J.Liu, X.W.); Vascular Biology Center (H.S., J.Li, W.M.) and Department of Pharmacology and Toxicology (H.S.), Medical College of Georgia, Georgia Regents University, Augusta; and Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT (N.W.).
| | - Xuejun Wang
- From the State Key Laboratory of Respiratory Disease and Department of Pathophysiology, Guangzhou Medical University, Guangzhou, Guangdong, China (H.S., J.Liu, X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (H.S., J.Li, H.Z., J.Liu, X.W.); Vascular Biology Center (H.S., J.Li, W.M.) and Department of Pharmacology and Toxicology (H.S.), Medical College of Georgia, Georgia Regents University, Augusta; and Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT (N.W.).
| |
Collapse
|
64
|
Anbarasu K, Sivakumar J. Multidimensional significance of crystallin protein-protein interactions and their implications in various human diseases. Biochim Biophys Acta Gen Subj 2015; 1860:222-33. [PMID: 26365509 DOI: 10.1016/j.bbagen.2015.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 08/28/2015] [Accepted: 09/08/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Crystallins are the important structural and functional proteins in the eye lens responsible for refractive index. Post-translational modifications (PTMs) and mutations are major causative factors that affect crystallin structural conformation and functional characteristics thus playing a vital role in the etiology of cataractogenesis. SCOPE OF REVIEW The significance of crystallin protein-protein interactions (PPIs) in the lens and non-lenticular tissues is summarized. MAJOR CONCLUSIONS Aberrancy of PPIs between crystallin, its associated protein and metal ions has been accomplished in various human diseases including cataract. A detailed account on multidimensional structural and functional significance of crystallin PPI in humans must be brought into limelight, in order to understand the biochemical and molecular basis augmenting the aberrancies of such interaction. In this scenario, the present review is focused to shed light on studies which will aid to expand our present understanding on disease pathogenesis related to loss of PPI thereby paving the way for putative future therapeutic targets to curb such diseases. GENERAL SIGNIFICANCE The interactions with α-crystallins always aid to protect their structural and functional characteristics. The up-regulation of αB-crystallin in the non-lenticular tissues always decodes as biomarker for various stress related disorders. For better understanding and treatment of various diseases, PPI studies provide overall outline about the structural and functional characteristics of the proteins. This information not only helps to find out the route of cataractogenesis but also aid to identify potential molecules to inhibit/prevent the further development of such complicated phenomenon. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
Affiliation(s)
- Kumarasamy Anbarasu
- Department of Marine Biotechnology, Bharathidasan University, Tiruchirapalli 620024, Tamil Nadu, India.
| | - Jeyarajan Sivakumar
- Department of Marine Biotechnology, Bharathidasan University, Tiruchirapalli 620024, Tamil Nadu, India
| |
Collapse
|
65
|
Raju M, Santhoshkumar P, Krishna Sharma K. Alpha-crystallin-derived peptides as therapeutic chaperones. Biochim Biophys Acta Gen Subj 2015; 1860:246-51. [PMID: 26141743 DOI: 10.1016/j.bbagen.2015.06.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/19/2015] [Accepted: 06/26/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND The demonstration of chaperone-like activity in peptides (mini-chaperones) derived from α-crystallin's chaperone region has generated significant interest in exploring the therapeutic potential of peptide chaperones in diseases of protein aggregation. Recent studies in experimental animals show that mini-chaperones could reach intended targets and alter the disease phenotype. Although mini-chaperones show potential benefits against protein aggregation diseases, they do tend to form aggregates on storage. There is thus a need to fine-tune peptide chaperones to increase their solubility, pharmacokinetics, and biological efficacy. SCOPE OF REVIEW This review summarizes the properties and the potential therapeutic roles of mini-chaperones in protein aggregation diseases and highlights some of the refinements needed to increase the stability and biological efficacy of mini-chaperones while maintaining or enhancing their chaperone-like activity against precipitation of unfolding proteins. MAJOR CONCLUSIONS Mini-chaperones suppress the aggregation of proteins, block amyloid fibril formation, stabilize mutant proteins, sequester metal ions, and exhibit antiapoptotic properties. Much work must be done to fine-tune mini-chaperones and increase their stability and biological efficacy. Peptide chaperones could have a great therapeutic value in diseases associated with protein aggregation and apoptosis. GENERAL SIGNIFICANCE Accumulation of misfolded proteins is a primary cause for many age-related diseases, including cataract, macular degeneration, and various neurological diseases. Stabilization of native proteins is a logical therapeutic approach for such diseases. Mini-chaperones, with their inherent antiaggregation and antiapoptotic properties, may represent an effective therapeutic molecule to prevent the cascade of protein conformational disorders. Future studies will further uncover the therapeutic potential of mini-chaperones. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
Affiliation(s)
- Murugesan Raju
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Puttur Santhoshkumar
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - K Krishna Sharma
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO 65212, USA; Department of Biochemistry, University of Missouri School of Medicine, Columbia, MO 65212, USA.
| |
Collapse
|
66
|
Panda AK, Nandi SK, Chakraborty A, Nagaraj RH, Biswas A. Differential role of arginine mutations on the structure and functions of α-crystallin. Biochim Biophys Acta Gen Subj 2015; 1860:199-210. [PMID: 26080000 DOI: 10.1016/j.bbagen.2015.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/22/2015] [Accepted: 06/09/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND α-Crystallin is a major protein of the eye lens in vertebrates. It is composed of two subunits, αA- and αB-crystallin. α-Crystallin is an oligomeric protein having these two subunits in 3:1 ratio. It belongs to small heat shock protein family and exhibits molecular chaperone function, which plays an important role in maintaining the lens transparency. Apart from chaperone function, both subunits also exhibit anti-apoptotic property. Comparison of their primary sequences reveals that αA- and αB-crystallin posses 13 and 14 arginine residues, respectively. Several of them undergo mutations which eventually lead to various eye diseases such as congenital cataract, juvenile cataract, and retinal degeneration. Interestingly, many arginine residues of these subunits are modified during glycation and even some are truncated during aging. All these facts indicate the importance of arginine residues in α-crystallin. SCOPE OF REVIEW In this review, we will emphasize the recent in vitro and in vivo findings related to congenital cataract causing arginine mutations in α-crystallin. MAJOR CONCLUSIONS Congenital cataract causing arginine mutations alters the structure and decreases the chaperone function of α-crystallin. These mutations also affect the lens morphology and phenotypes. Interestingly, non-natural arginine mutations (generated for mimicking the glycation and truncation environment) improve the chaperone function of α-crystallin which may play an important role in maintaining the eye lens transparency during aging. GENERAL SIGNIFICANCE The neutralization of positive charge on the guanidino group of arginine residues is not always detrimental to the functionality of α-crystallin. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
Affiliation(s)
- Alok Kumar Panda
- School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Bhubaneswar, Odisha 751013, India
| | - Sandip Kumar Nandi
- School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Bhubaneswar, Odisha 751013, India
| | - Ayon Chakraborty
- School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Bhubaneswar, Odisha 751013, India
| | - Ram H Nagaraj
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Ashis Biswas
- School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Bhubaneswar, Odisha 751013, India.
| |
Collapse
|
67
|
Shao DW, Yang CY, Liu B, Chen W, Wang H, Ru HX, Zhang M, Wang Y. Bioinformatics Analysis of Potential Candidates for Therapy of TDRD7 Deficiency-Induced Congenital Cataract. Ophthalmic Res 2015; 54:10-7. [PMID: 25997407 DOI: 10.1159/000381478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/05/2015] [Indexed: 11/19/2022]
Abstract
AIMS The aim of this study was to identify potential candidates and explore the possible mechanism in congenital cataract induced by tudor domain-containing 7 (TDRD7) deficiency. METHODS The gene expression profile GSE25812 generated from 18 samples was downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between disease and normal groups were identified. Then, gene ontology and pathway enrichment analysis of DEGs were performed. The protein-protein interaction (PPI) network and transcription factor (TF) regulatory network were constructed. The modules in the PPI network were identified. Significant target genes were selected from the TF regulatory network. RESULTS A total of 329 DEGs were obtained, and downregulated DEGs were significantly enriched in biological processes including defense response and immune response. In the PPI network, high-degree genes of complement component 1, q subcomponent, A/B/C chain (C1QA/C1QB/C1QC), lymphocyte antigen 86 (LY86) and neuroblastoma RAS viral oncogene homolog (NRAS) were identified. From the TF regulatory network, the heat shock 27 kDa protein 1 (HSPB1) was the target of the estrogen receptor 1, and LY86 was the target of the v-myc avian myelocytomatosis viral oncogene homolog. CONCLUSION HSPB1, NRAS, immune response, defense response and the related genes LY86, C1QA/C1QB/C1QC may play an important role in the development of congenital cataract induced by TDRD7 deficiency. However, further experiments are still needed.
Collapse
Affiliation(s)
- De-Wang Shao
- Department of Ophthalmology, Air Force General Hospital of PLA, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Hussein RM, Benjamin IJ, Kampinga HH. Rescue of αB Crystallin (HSPB5) Mutants Associated Protein Aggregation by Co-Expression of HSPB5 Partners. PLoS One 2015; 10:e0126761. [PMID: 25961584 PMCID: PMC4427338 DOI: 10.1371/journal.pone.0126761] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 04/07/2015] [Indexed: 11/23/2022] Open
Abstract
HSPB5 (also called αB-crystallin) is a ubiquitously expressed small heat shock protein. Mutations in HSPB5 have been found to cause cataract, but are also associated with a subgroup of myofibrillar myopathies. Cells expressing each of these HSPB5 mutants are characterized by the appearance of protein aggregates of primarily the mutant HSPB5. Like several members of the HSPB family, HSPB5 can form both homo-oligomeric and hetero-oligomeric complexes. Previous studies showed that co-expression of HSPB1 and HSPB8 can prevent the aggregation associated with the HSPB5 (R120G) mutant in cardiomyocytes and in transgenic mice. In this study, we systematically compared the effect of co-expression of each of the members of the human HSPB family (HSPB1-10) on the aggregation of three different HSPB5 mutants (R120G, 450 Δ A, 464 Δ CT). Of all members, co-expression of HSPB1, HSPB4 and HSPB5 itself, most effectively prevent the aggregation of these 3 HSPB5 mutants. HSPB6 and HSPB8 were also active but less, whilst the other 5 HSPB members were ineffective. Co-expression of Hsp70 did not reduce the aggregation of the HSPB5 mutants, suggesting that aggregate formation is most likely not related to a toxic gain of function of the mutants per se, but rather related to a loss of chaperone function of the oligomeric complexes containing the HSPB5 mutants (dominant negative effects). Our data suggest that the rescue of aggregation associated with the HSPB5 mutants is due to competitive incorporation of its partners into hetero-oligomers hereby negating the dominant negative effects of the mutant on the functioning of the hetero-oligomer.
Collapse
Affiliation(s)
- Rasha M. Hussein
- Department of Cell Biology, University Medical Center Groningen, Groningen, The Netherlands
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Salah Salem Street, 62511, Beni-Suef, Egypt
| | - Ivor J. Benjamin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Harm H. Kampinga
- Department of Cell Biology, University Medical Center Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
69
|
Jiao Q, Sanbe A, Zhang X, Liu JP, Minamisawa S. αB-Crystallin R120G variant causes cardiac arrhythmias and alterations in the expression of Ca(2+) -handling proteins and endoplasmic reticulum stress in mice. Clin Exp Pharmacol Physiol 2015; 41:589-99. [PMID: 24825000 DOI: 10.1111/1440-1681.12253] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 04/23/2014] [Accepted: 05/08/2014] [Indexed: 11/29/2022]
Abstract
Mutations of αB-crystallin (CryαB), a small heat shock protein abundantly expressed in cardiac and skeletal muscles, are known to cause desmin-related myopathies. The CryαB R120G allele has been linked to a familial desminopathy and, in transgenic mice, causes a sudden death at about 28 weeks of age. To investigate the mechanisms of the sudden cardiac arrest of CryαB R120G transgenic mice, we prepared protein samples from left ventricular tissues of two different age groups (10 and 28 weeks) and examined Ca(2+) -handling proteins. Expression of sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA) 2, phospholamban, ryanodine receptor 2 and calsequestrin 2 was significantly decreased in 28- versus 10-week-old CryαB R120G transgenic mice. In addition, low heart rate variability, including heart rate, total power and low frequency, was observed and continuous electrocardiogram monitoring revealed cardiac arrhythmias, such as ventricular tachycardia, atrioventricular block and atrial flutter, in 28-week-old CryαB R120G transgenic mice. In contrast, expression of endoplasmic reticulum (ER) degradation enhancing α-mannosidase-like protein, inositol requirement 1 and X-box binding protein 1 were increased significantly in 28- versus 10-week-old CryαBR120G transgenic mice, suggesting that the CryαBR120G transgenic mice exhibit increased ER stress compared with wild-type mice. Together, the data suggest that the CryαB R120G dominant variant induces ER stress and impairs Ca(2+) regulation, leading to ageing-related cardiac dysfunction, arrhythmias and decreased autonomic tone with shortened lifespan.
Collapse
Affiliation(s)
- Qibin Jiao
- Department of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Institute of Ageing Research, School of Medicine, Hangzhou Normal University, Hangzhou, China; Department of Life Science and Medical Bio-Science, Waseda University, Tokyo, Japan; Department of Cell Physiology, Jikei University, Tokyo, Japan
| | | | | | | | | |
Collapse
|
70
|
Abstract
Cataract produces vision loss due to opacification of the lens. Two possible reasons for cataract formation are insolubility of crystallin proteins and fibril (aggregate) formation. Currently, the only available treatment is surgical replacement of the lens with a synthetic one. An alternative treatment that is both economically and technologically accessible is needed in developing countries where untreated cataract and coincident blindness is high, access to trained surgeons is limited, and the cost of surgery may be prohibitive. We present current progress toward the development of a nonsurgical treatment strategy focusing on promoting protein solubility and/or dissolving fibrillar aggregates.
Collapse
|
71
|
Treweek TM, Meehan S, Ecroyd H, Carver JA. Small heat-shock proteins: important players in regulating cellular proteostasis. Cell Mol Life Sci 2015; 72:429-451. [PMID: 25352169 PMCID: PMC11113218 DOI: 10.1007/s00018-014-1754-5] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 09/15/2014] [Accepted: 10/01/2014] [Indexed: 12/13/2022]
Abstract
Small heat-shock proteins (sHsps) are a diverse family of intra-cellular molecular chaperone proteins that play a critical role in mitigating and preventing protein aggregation under stress conditions such as elevated temperature, oxidation and infection. In doing so, they assist in the maintenance of protein homeostasis (proteostasis) thereby avoiding the deleterious effects that result from loss of protein function and/or protein aggregation. The chaperone properties of sHsps are therefore employed extensively in many tissues to prevent the development of diseases associated with protein aggregation. Significant progress has been made of late in understanding the structure and chaperone mechanism of sHsps. In this review, we discuss some of these advances, with a focus on mammalian sHsp hetero-oligomerisation, the mechanism by which sHsps act as molecular chaperones to prevent both amorphous and fibrillar protein aggregation, and the role of post-translational modifications in sHsp chaperone function, particularly in the context of disease.
Collapse
Affiliation(s)
- Teresa M Treweek
- Graduate School of Medicine, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia.
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW, 2522, Australia.
| | - Sarah Meehan
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Heath Ecroyd
- Illawarra Health and Medical Research Institute, Northfields Avenue, Wollongong, NSW, 2522, Australia.
- School of Biological Sciences, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia.
| | - John A Carver
- Research School of Chemistry, The Australian National University, Acton, ACT, 2601, Australia.
| |
Collapse
|
72
|
Arrigo AP, Ducarouge B, Lavial F, Gibert B. Immense Cellular Implications Associated to Small Stress Proteins Expression: Impacts on Human Pathologies. HEAT SHOCK PROTEINS 2015. [DOI: 10.1007/978-3-319-16077-1_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
73
|
|
74
|
Bakthisaran R, Tangirala R, Rao CM. Small heat shock proteins: Role in cellular functions and pathology. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1854:291-319. [PMID: 25556000 DOI: 10.1016/j.bbapap.2014.12.019] [Citation(s) in RCA: 312] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 01/18/2023]
Abstract
Small heat shock proteins (sHsps) are conserved across species and are important in stress tolerance. Many sHsps exhibit chaperone-like activity in preventing aggregation of target proteins, keeping them in a folding-competent state and refolding them by themselves or in concert with other ATP-dependent chaperones. Mutations in human sHsps result in myopathies, neuropathies and cataract. Their expression is modulated in diseases such as Alzheimer's, Parkinson's and cancer. Their ability to bind Cu2+, and suppress generation of reactive oxygen species (ROS) may have implications in Cu2+-homeostasis and neurodegenerative diseases. Circulating αB-crystallin and Hsp27 in the plasma may exhibit immunomodulatory and anti-inflammatory functions. αB-crystallin and Hsp20 exhitbit anti-platelet aggregation: these beneficial effects indicate their use as potential therapeutic agents. sHsps have roles in differentiation, proteasomal degradation, autophagy and development. sHsps exhibit a robust anti-apoptotic property, involving several stages of mitochondrial-mediated, extrinsic apoptotic as well as pro-survival pathways. Dynamic N- and C-termini and oligomeric assemblies of αB-crystallin and Hsp27 are important factors for their functions. We propose a "dynamic partitioning hypothesis" for the promiscuous interactions and pleotropic functions exhibited by sHsps. Stress tolerance and anti-apoptotic properties of sHsps have both beneficial and deleterious consequences in human health and diseases. Conditional and targeted modulation of their expression and/or activity could be used as strategies in treating several human disorders. The review attempts to provide a critical overview of sHsps and their divergent roles in cellular processes particularly in the context of human health and disease.
Collapse
Affiliation(s)
- Raman Bakthisaran
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Ramakrishna Tangirala
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Ch Mohan Rao
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India.
| |
Collapse
|
75
|
McLendon PM, Ferguson BS, Osinska H, Bhuiyan MS, James J, McKinsey TA, Robbins J. Tubulin hyperacetylation is adaptive in cardiac proteotoxicity by promoting autophagy. Proc Natl Acad Sci U S A 2014; 111:E5178-86. [PMID: 25404307 PMCID: PMC4260547 DOI: 10.1073/pnas.1415589111] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Proteinopathy causes cardiac disease, remodeling, and heart failure but the pathological mechanisms remain obscure. Mutated αB-crystallin (CryAB(R120G)), when expressed only in cardiomyocytes in transgenic (TG) mice, causes desmin-related cardiomyopathy, a protein conformational disorder. The disease is characterized by the accumulation of toxic misfolded protein species that present as perinuclear aggregates known as aggresomes. Previously, we have used the CryAB(R120G) model to determine the underlying processes that result in these pathologic accumulations and to explore potential therapeutic windows that might be used to decrease proteotoxicity. We noted that total ventricular protein is hypoacetylated while hyperacetylation of α-tubulin, a substrate of histone deacetylase 6 (HDAC6) occurs. HDAC6 has critical roles in protein trafficking and autophagy, but its function in the heart is obscure. Here, we test the hypothesis that tubulin acetylation is an adaptive process in cardiomyocytes. By modulating HDAC6 levels and/or activity genetically and pharmacologically, we determined the effects of tubulin acetylation on aggregate formation in CryAB(R120G) cardiomyocytes. Increasing HDAC6 accelerated aggregate formation, whereas siRNA-mediated knockdown or pharmacological inhibition ameliorated the process. HDAC inhibition in vivo induced tubulin hyperacetylation in CryAB(R120G) TG hearts, which prevented aggregate formation and significantly improved cardiac function. HDAC6 inhibition also increased autophagic flux in cardiomyocytes, and increased autophagy in the diseased heart correlated with increased tubulin acetylation, suggesting that autophagy induction might underlie the observed cardioprotection. Taken together, our data suggest a mechanistic link between tubulin hyperacetylation and autophagy induction and points to HDAC6 as a viable therapeutic target in cardiovascular disease.
Collapse
Affiliation(s)
- Patrick M McLendon
- The Heart Institute, Department of Pediatrics, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; and
| | - Bradley S Ferguson
- Department of Medicine, Division of Cardiology, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045
| | - Hanna Osinska
- The Heart Institute, Department of Pediatrics, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; and
| | - Md Shenuarin Bhuiyan
- The Heart Institute, Department of Pediatrics, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; and
| | - Jeanne James
- The Heart Institute, Department of Pediatrics, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; and
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045
| | - Jeffrey Robbins
- The Heart Institute, Department of Pediatrics, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; and
| |
Collapse
|
76
|
Banerjee Mustafi S, Grose JH, Zhang H, Pratt GW, Sadoshima J, Christians ES, Benjamin IJ. Aggregate-prone R120GCRYAB triggers multifaceted modifications of the thioredoxin system. Antioxid Redox Signal 2014; 20:2891-906. [PMID: 24180415 PMCID: PMC4039002 DOI: 10.1089/ars.2013.5340] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
AIMS The human mutation R120G in the αB-crystallin (CRYAB) causes a multisystemic disease that is characterized by hypertrophic cardiomyopathy and cytoplasmic protein aggregates. In transgenic mice, human R120GCRYAB (hR120GTg) expression in heart sequentially modifies the REDOX status, in part by the activation of the nuclear factor, erythroid derived 2, like 2 (Nrf2). Thioredoxin system (TS) components are NRF2 target genes, so it could be hypothesized that TS was affected in hR120GTg mice. RESULTS Transgenic hearts overexpressed thioredoxin 1 (Trx1), which was identified by isotope coded affinity tag-mass spectrometry, among hundreds of peptides displaying an increased reduced/oxidized ratio. Coupled to this higher level of reduced cysteines, the activity of thioredoxin reductase 1 (TrxR1) was augmented by 2.5-fold. Combining mutiple experimental approaches, the enzymatic regulation of TrxR1 by a histone deacetylase 3 (HDAC3)-dependent level of acetylation was confirmed. In vitro and in vivo functional tests established that TrxR1 activity is required to mitigate aggregate development, and this could be mediated by Bcl-2-associated athanogene 3 (BAG3) as a potential TS substrate. INNOVATION AND CONCLUSIONS This study uncovers the compartmentalized changes and the involvement of TS in the cardiac stress response elicited by misfolded proteins such as R120GCRYAB. Our work suggests that R120GCRYAB triggers a defensive pathway acting through the newly identified interacting partners HDAC3, TrxR1, and BAG3 to counter aggregate growth. Therefore, those interactors may function as modifier genes contributing to the variable onset and expressivity of such human diseases. Furthermore, our work underscores the potential organismal effects of pharmacological interventions targeting TS and HDAC.
Collapse
Affiliation(s)
- Soumyajit Banerjee Mustafi
- 1 Laboratory of Cardiac Disease, Redox Signaling and Cell Regeneration, Division of Cardiology, University of Utah School of Medicine , Salt Lake City, Utah
| | | | | | | | | | | | | |
Collapse
|
77
|
Kakkar V, Meister-Broekema M, Minoia M, Carra S, Kampinga HH. Barcoding heat shock proteins to human diseases: looking beyond the heat shock response. Dis Model Mech 2014; 7:421-34. [PMID: 24719117 PMCID: PMC3974453 DOI: 10.1242/dmm.014563] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
There are numerous human diseases that are associated with protein misfolding and the formation of toxic protein aggregates. Activating the heat shock response (HSR)--and thus generally restoring the disturbed protein homeostasis associated with such diseases--has often been suggested as a therapeutic strategy. However, most data on activating the HSR or its downstream targets in mouse models of diseases associated with aggregate formation have been rather disappointing. The human chaperonome consists of many more heat shock proteins (HSPs) that are not regulated by the HSR, however, and researchers are now focusing on these as potential therapeutic targets. In this Review, we summarize the existing literature on a set of aggregation diseases and propose that each of them can be characterized or 'barcoded' by a different set of HSPs that can rescue specific types of aggregation. Some of these 'non-canonical' HSPs have demonstrated effectiveness in vivo, in mouse models of protein-aggregation disease. Interestingly, several of these HSPs also cause diseases when mutated--so-called chaperonopathies--which are also discussed in this Review.
Collapse
Affiliation(s)
- Vaishali Kakkar
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Melanie Meister-Broekema
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Melania Minoia
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Serena Carra
- Università degli Studi di Modena e Reggio Emilia, Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, via G. Campi 287, 41125 Modena, Italy
| | - Harm H. Kampinga
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| |
Collapse
|
78
|
Boelens WC. Cell biological roles of αB-crystallin. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 115:3-10. [PMID: 24576798 DOI: 10.1016/j.pbiomolbio.2014.02.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 10/25/2022]
Abstract
αB-crystallin, also called HspB5, is a molecular chaperone able to interact with unfolding proteins. By interacting, it inhibits further unfolding, thereby preventing protein aggregation and allowing ATP-dependent chaperones to refold the proteins. αB-crystallin belongs to the family of small heat-shock proteins (sHsps), which in humans consists of 10 different members. The protein forms large oligomeric complexes, containing up to 40 or more subunits, which in vivo consist of heterooligomeric complexes formed by a mixture of αB-crystallin and other sHsps. αB-crystallin is highly expressed in the lens and to a lesser extent in several other tissues, among which heart, skeletal muscle and brain. αB-crystallin plays a role in several cellular processes, such as signal transduction, protein degradation, stabilization of cytoskeletal structures and apoptosis. Mutations in the αB-crystallin gene can have detrimental effects, leading to pathologies such as cataract and cardiomyopathy. This review describes the biological roles of αB-crystallin, with a special focus on its function in the eye lens, heart muscle and brain. In addition its therapeutic potential is discussed.
Collapse
Affiliation(s)
- Wilbert C Boelens
- Department of Biomolecular Chemistry, Institute for Molecules and Materials and Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
79
|
Dubińska-Magiera M, Jabłońska J, Saczko J, Kulbacka J, Jagla T, Daczewska M. Contribution of small heat shock proteins to muscle development and function. FEBS Lett 2014; 588:517-30. [PMID: 24440355 DOI: 10.1016/j.febslet.2014.01.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/17/2013] [Accepted: 01/02/2014] [Indexed: 12/17/2022]
Abstract
Investigations undertaken over the past years have led scientists to introduce the concept of protein quality control (PQC) systems, which are responsible for polypeptide processing. The PQC system monitors proteostasis and involves activity of different chaperones such as small heat shock proteins (sHSPs). These proteins act during normal conditions as housekeeping proteins regulating cellular processes, and during stress conditions. They also mediate the removal of toxic misfolded polypeptides and thereby prevent development of pathogenic states. It is postulated that sHSPs are involved in muscle development. They could act via modulation of myogenesis or by maintenance of the structural integrity of signaling complexes. Moreover, mutations in genes coding for sHSPs lead to pathological states affecting muscular tissue functioning. This review focuses on the question how sHSPs, still relatively poorly understood proteins, contribute to the development and function of three types of muscle tissue: skeletal, cardiac and smooth.
Collapse
Affiliation(s)
- Magda Dubińska-Magiera
- Department of Animal Developmental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland
| | - Jadwiga Jabłońska
- Department of Animal Developmental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland
| | - Jolanta Saczko
- Department of Medical Biochemistry, Medical University, Chalubinskiego 10, 50-368 Wroclaw, Poland
| | - Julita Kulbacka
- Department of Medical Biochemistry, Medical University, Chalubinskiego 10, 50-368 Wroclaw, Poland
| | - Teresa Jagla
- Institut National de la Santé et de la Recherche Médicale U384, Faculté de Medecine, Clermont-Ferrand, France
| | - Małgorzata Daczewska
- Department of Animal Developmental Biology, University of Wroclaw, 21 Sienkiewicza Street, 50-335 Wroclaw, Poland.
| |
Collapse
|
80
|
Nandi SK, Rehna EAA, Panda AK, Shiburaj S, Dharmalingam K, Biswas A. A S52P mutation in the ‘α-crystallin domain’ ofMycobacterium lepraeHSP18 reduces its oligomeric size and chaperone function. FEBS J 2013; 280:5994-6009. [DOI: 10.1111/febs.12519] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 08/28/2013] [Accepted: 09/04/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Sandip K. Nandi
- School of Basic Sciences; Indian Institute of Technology Bhubaneswar; Orissa India
| | - Elengikal A. A. Rehna
- Department of Genetic Engineering; School of Biotechnology; Madurai Kamraj University; Tamilnadu India
| | - Alok K. Panda
- School of Basic Sciences; Indian Institute of Technology Bhubaneswar; Orissa India
| | - Sugathan Shiburaj
- Jawaharlal Nehru Tropical Botanic Garden and Research Institute; Palode Thiruvananthapuram Kerala India
| | - Kuppamuthu Dharmalingam
- Department of Genetic Engineering; School of Biotechnology; Madurai Kamraj University; Tamilnadu India
| | - Ashis Biswas
- School of Basic Sciences; Indian Institute of Technology Bhubaneswar; Orissa India
| |
Collapse
|
81
|
Simon S, Dimitrova V, Gibert B, Virot S, Mounier N, Nivon M, Kretz-Remy C, Corset V, Mehlen P, Arrigo AP. Analysis of the dominant effects mediated by wild type or R120G mutant of αB-crystallin (HspB5) towards Hsp27 (HspB1). PLoS One 2013; 8:e70545. [PMID: 23950959 PMCID: PMC3741289 DOI: 10.1371/journal.pone.0070545] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 06/19/2013] [Indexed: 12/21/2022] Open
Abstract
Several human small heat shock proteins (sHsps) are phosphorylated oligomeric chaperones that enhance stress resistance. They are characterized by their ability to interact and form polydispersed hetero-oligomeric complexes. We have analyzed the cellular consequences of the stable expression of either wild type HspB5 or its cataracts and myopathies inducing R120G mutant in growing and oxidative stress treated HeLa cells that originally express only HspB1. Here, we describe that wild type and mutant HspB5 induce drastic and opposite effects on cell morphology and oxidative stress resistance. The cellular distribution and phosphorylation of these polypeptides as well as the oligomerization profile of the resulting hetero-oligomeric complexes formed by HspB1 with the two types of exogenous polypeptides revealed the dominant effects induced by HspB5 polypeptides towards HspB1. The R120G mutation enhanced the native size and salt resistance of HspB1-HspB5 complex. However, in oxidative conditions the interaction between HspB1 and mutant HspB5 was drastically modified resulting in the aggregation of both partners. The mutation also induced the redistribution of HspB1 phosphorylated at serine 15, originally observed at the level of the small oligomers that do not interact with wild type HspB5, to the large oligomeric complex formed with mutant HspB5. This phosphorylation stabilized the interaction of HspB1 with mutant HspB5. A dominant negative effect towards HspB1 appears therefore as an important event in the cellular sensitivity to oxidative stress mediated by mutated HspB5 expression. These observations provide novel data that describe how a mutated sHsp can alter the protective activity of another member of this family of chaperones.
Collapse
Affiliation(s)
- Stéphanie Simon
- Hôpital Henri Mondor University, Créteil, France
- CGphiMC, CNRS UMR 5534, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Valeriya Dimitrova
- Department of Clinical Research, Division of Pediatric Hematology/Oncology, Insel Spital, Institute of Pathology, Bern University, Bern, Switzerland
- CGphiMC, CNRS UMR 5534, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Benjamin Gibert
- CGphiMC, CNRS UMR 5534, Claude Bernard University Lyon 1, Villeurbanne, France
- Apoptosis Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Claude Bernard University Lyon 1, Lyon, France
| | - Sophie Virot
- CGphiMC, CNRS UMR 5534, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Nicole Mounier
- CGphiMC, CNRS UMR 5534, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Mathieu Nivon
- CGphiMC, CNRS UMR 5534, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Carole Kretz-Remy
- CGphiMC, CNRS UMR 5534, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Véronique Corset
- Apoptosis Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Claude Bernard University Lyon 1, Lyon, France
| | - Patrick Mehlen
- Apoptosis Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Claude Bernard University Lyon 1, Lyon, France
| | - André-Patrick Arrigo
- CGphiMC, CNRS UMR 5534, Claude Bernard University Lyon 1, Villeurbanne, France
- Apoptosis Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Claude Bernard University Lyon 1, Lyon, France
- * E-mail:
| |
Collapse
|
82
|
Wignes JA, Goldman JW, Weihl CC, Bartley MG, Andley UP. p62 expression and autophagy in αB-crystallin R120G mutant knock-in mouse model of hereditary cataract. Exp Eye Res 2013; 115:263-73. [PMID: 23872361 DOI: 10.1016/j.exer.2013.06.026] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 06/21/2013] [Accepted: 06/25/2013] [Indexed: 01/07/2023]
Abstract
The formation of cataracts is associated with the accumulation of protein aggregates in the ocular lens, suggesting that defective protein degradation plays a role in cataract pathogenesis. Accumulation of the p62 protein has recently been identified as a marker for impaired autophagy in a variety of tissues; however, little information exists on its expression in the ocular lens and in cataracts. In the present study we examined the expression of p62 in the mouse lens and compared its expression in wild-type lenses with that in lenses from knock-in mice with an arginine to glycine mutation in αB-crystallin (αB-R120G) that is known to cause human hereditary cataract. Immunohistochemical, immunoblotting, and transmission electron microscopic analyses of wild type and αB-R120G mutant mice were performed. To assess the effect of increased protein aggregation on autophagy, immunohistochemical staining was performed with an anti-p62 antibody, revealing the presence of p62-positive punctate staining in a band of denucleated cortical fiber cells. The number and size of p62 puncta were significantly greater in αB-R120G homozygous mutant lenses than in wild type and heterozygous mutant lenses. p62 staining was also abundant in lens epithelial cells and was concentrated around the nuclear membrane. Double-membraned structures similar to autophagosomes containing cellular cytoplasmic content were detected in lens epithelial cells by transmission electron microscopy. The autophagosomes in lens epithelial cells from αB-R120G homozygous mutant mice were larger than those in wild type mice. Double-membraned structures that are probably autophagosomes were also detected in cortical fiber cells and were more abundant in the αB-R120G homozygous mutant lens than the wild type lens. This study demonstrates p62 distribution as speckles in the lens fiber cells, altered levels of p62 expression, and the presence of autophagosomes in the ocular lens of αB-R120G mutant mice. We propose that autophagy is inhibited in the αB-R120G mutant lenses because of a defect in protein degradation after autophagosome formation. Further work is necessary to determine the relationship between αB-crystallin function, autophagy, and cataract formation.
Collapse
Affiliation(s)
- Jonathan A Wignes
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
83
|
D'Agostino M, Lemma V, Chesi G, Stornaiuolo M, Cannata Serio M, D'Ambrosio C, Scaloni A, Polishchuk R, Bonatti S. The cytosolic chaperone α-crystallin B rescues folding and compartmentalization of misfolded multispan transmembrane proteins. J Cell Sci 2013; 126:4160-72. [PMID: 23843626 DOI: 10.1242/jcs.125443] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The α-crystallin B chain (CRYAB or HspB5) is a cytosolic chaperone belonging to the small heat shock protein family, which is known to help in the folding of cytosolic proteins. Here we show that CRYAB binds the mutant form of at least two multispan transmembrane proteins (TMPs), exerting an anti-aggregation activity. It rescues the folding of mutant Frizzled4, which is responsible for a rare autosomal dominant form of familial exudative vitreoretinopathy (Fz4-FEVR), and the mutant ATP7B Cu transporter (ATP7B-H1069Q) associated with a common form of Wilson's disease. In the case of Fz4-FEVR, CRYAB prevents the formation of inter-chain disulfide bridges between the lumenal ectodomains of the aggregated mutant chains, which enables correct folding and promotes appropriate compartmentalization on the plasma membrane. ATP7B-H1069Q, with help from CRYAB, folds into the proper conformation, moves to the Golgi complex, and responds to copper overload in the same manner as wild-type ATP7B. These findings strongly suggest that CRYAB plays a pivotal role, previously undetected, in the folding of multispan TMPs and, from the cytosol, is able to orchestrate folding events that take place in the lumen of the ER. Our results contribute to the explanation of the complex scenario behind multispan TMP folding; additionally, they serve to expose interesting avenues for novel therapeutic approaches.
Collapse
Affiliation(s)
- Massimo D'Agostino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Xie HB, Cammarato A, Rajasekaran NS, Zhang H, Suggs JA, Lin HC, Bernstein SI, Benjamin IJ, Golic KG. The NADPH metabolic network regulates human αB-crystallin cardiomyopathy and reductive stress in Drosophila melanogaster. PLoS Genet 2013; 9:e1003544. [PMID: 23818860 PMCID: PMC3688542 DOI: 10.1371/journal.pgen.1003544] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 04/20/2013] [Indexed: 11/18/2022] Open
Abstract
Dominant mutations in the alpha-B crystallin (CryAB) gene are responsible for a number of inherited human disorders, including cardiomyopathy, skeletal muscle myopathy, and cataracts. The cellular mechanisms of disease pathology for these disorders are not well understood. Among recent advances is that the disease state can be linked to a disturbance in the oxidation/reduction environment of the cell. In a mouse model, cardiomyopathy caused by the dominant CryAB(R120G) missense mutation was suppressed by mutation of the gene that encodes glucose 6-phosphate dehydrogenase (G6PD), one of the cell's primary sources of reducing equivalents in the form of NADPH. Here, we report the development of a Drosophila model for cellular dysfunction caused by this CryAB mutation. With this model, we confirmed the link between G6PD and mutant CryAB pathology by finding that reduction of G6PD expression suppressed the phenotype while overexpression enhanced it. Moreover, we find that expression of mutant CryAB in the Drosophila heart impaired cardiac function and increased heart tube dimensions, similar to the effects produced in mice and humans, and that reduction of G6PD ameliorated these effects. Finally, to determine whether CryAB pathology responds generally to NADPH levels we tested mutants or RNAi-mediated knockdowns of phosphogluconate dehydrogenase (PGD), isocitrate dehydrogenase (IDH), and malic enzyme (MEN), the other major enzymatic sources of NADPH, and we found that all are capable of suppressing CryAB(R120G) pathology, confirming the link between NADP/H metabolism and CryAB.
Collapse
Affiliation(s)
- Heng B. Xie
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
| | - Anthony Cammarato
- Department of Biology, San Diego State University, San Diego, California, United States of America
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Namakkal S. Rajasekaran
- Division of Cardiology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Huali Zhang
- Division of Cardiology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Jennifer A. Suggs
- Department of Biology, San Diego State University, San Diego, California, United States of America
| | - Ho-Chen Lin
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
| | - Sanford I. Bernstein
- Department of Biology, San Diego State University, San Diego, California, United States of America
| | - Ivor J. Benjamin
- Division of Cardiology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
- * E-mail: (IJB); (KGG)
| | - Kent G. Golic
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail: (IJB); (KGG)
| |
Collapse
|
85
|
Nefedova VV, Datskevich PN, Sudnitsyna MV, Strelkov SV, Gusev NB. Physico-chemical properties of R140G and K141Q mutants of human small heat shock protein HspB1 associated with hereditary peripheral neuropathies. Biochimie 2013; 95:1582-92. [PMID: 23643870 DOI: 10.1016/j.biochi.2013.04.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 04/22/2013] [Indexed: 01/31/2023]
Abstract
Some physico-chemical properties of R140G and K141Q mutants of human small heat shock protein HspB1 associated with hereditary peripheral neuropathy were analyzed. Mutation K141Q did not affect intrinsic Trp fluorescence and interaction with hydrophobic probe bis-ANS, whereas mutation R140G decreased both intrinsic fluorescence and fluorescence of bis-ANS bound to HspB1. Both mutations decreased thermal stability of HspB1. Mutation R140G increased, whereas mutation K141Q decreased the rate of trypsinolysis of the central part (residues 5-188) of HspB1. Both the wild type HspB1 and its K141Q mutant formed large oligomers with apparent molecular weight ∼560 kDa. The R140G mutant formed two types of oligomers, i.e. large oligomers tending to aggregate and small oligomers with apparent molecular weight ∼70 kDa. The wild type HspB1 formed mixed homooligomers with R140G mutant with apparent molecular weight ∼610 kDa. The R140G mutant was unable to form high molecular weight heterooligomers with HspB6, whereas the K141Q mutant formed two types of heterooligomers with HspB6. In vitro measured chaperone-like activity of the wild type HspB1 was comparable with that of K141Q mutant and was much higher than that of R140G mutant. Mutations of homologous hot-spot Arg (R140G of HspB1 and R120G of αB-crystallin) induced similar changes in the properties of two small heat shock proteins, whereas mutations of two neighboring residues (R140 and K141) induced different changes in the properties of HspB1.
Collapse
Affiliation(s)
- Victoria V Nefedova
- Department of Biochemistry, School of Biology, Moscow State University, Moscow, Russian Federation
| | | | | | | | | |
Collapse
|
86
|
Gibert B, Simon S, Dimitrova V, Diaz-Latoud C, Arrigo AP. Peptide aptamers: tools to negatively or positively modulate HSPB1(27) function. Philos Trans R Soc Lond B Biol Sci 2013; 368:20120075. [PMID: 23530261 DOI: 10.1098/rstb.2012.0075] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Human HSP27 (HSPB1) is a molecular chaperone sensor which, through dynamic changes in its phosphorylation and oligomerization, allows cells to adapt to changes in their physiology and/or mount a protective response to injuries. In pathological conditions, the high level of HSPB1 expression can either be beneficial, such as in diseases characterized by cellular degenerations, or be malignant in cancer cells where it promotes tumourigenesis, metastasis and anti-cancer drug resistance. Structural changes allow HSPB1 to interact with specific client protein partners in order to modulate their folding/activity and/or half-life. Therefore, the search is open for therapeutic compounds aimed at either down- or upregulating HSPB1 activity. In this respect, we have previously described two peptide aptamers (PA11 and PA50) that specifically interact with HSPB1 small oligomers and decrease its anti-apoptotic and tumourigenic activities. A novel analysis of the different HSPB1-interacting aptamers that were isolated earlier revealed that one aptamer (PA23) has the intriguing ability to stimulate the protective activity of HSPB1. We show here that this aptamer abolishes the dominant negative effect induced by the R120G mutant of αB-crystallin (HSPB5) by disrupting its interaction with HSPB1. Hence, developing structure-based interfering strategies could lead to the discovery of HSPB1-based therapeutic drugs.
Collapse
Affiliation(s)
- Benjamin Gibert
- Apoptosis Cancer and Development Laboratory, Lyon Cancer Research Center, Centre Léon Bérard, INSERM U1052-CNRS 5238, University of Lyon, 69008 Lyon, France
| | | | | | | | | |
Collapse
|
87
|
Quinlan RA, Zhang Y, Lansbury A, Williamson I, Pohl E, Sun F. Changes in the quaternary structure and function of MjHSP16.5 attributable to deletion of the IXI motif and introduction of the substitution, R107G, in the α-crystallin domain. Philos Trans R Soc Lond B Biol Sci 2013; 368:20120327. [PMID: 23530263 PMCID: PMC3638399 DOI: 10.1098/rstb.2012.0327] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The archael small heat-shock protein (sHSP), MjHSP16.5, forms a 24-subunit oligomer with octahedral symmetry. Here, we demonstrate that the IXI motif present in the C-terminal domain is necessary for the oligomerization of MjHSP16.5. Removal increased the in vitro chaperone activity with citrate synthase as the client protein. Less predictable were the effects of the R107G substitution in MjHSP16.5 because of the differences in the oligomerization of metazoan and non-metazoan sHSPs. We present the crystal structure for MjHSP16.5 R107G and compare this with an improved (2.5 Å) crystal structure for wild-type (WT) MjHSP16.5. Although no significant structural differences were found in the crystal, using cryo-electron microscopy, we identified two 24mer species with octahedral symmetry for the WT MjHSP16.5 both at room temperature and at 60°C, all showing two major species with the same diameter of 12.4 nm. Similarly, at room temperature, there are also two kinds of 12.4 nm oligomers for R107G MjHSP16.5, but in the 60°C sample, a larger 24mer species with a diameter of 13.6 nm was observed with significant changes in the fourfold symmetry axis and dimer–dimer interface. This highly conserved arginine, therefore, contributes to the quaternary organization of non-metazoan sHSP oligomers. Potentially, the R107G substitution has functional consequences as R107G MjHSP16.5 was far superior to the WT protein in protecting βL-crystallin against heat-induced aggregation.
Collapse
Affiliation(s)
- Roy A Quinlan
- Biophysical Sciences Institute, University of Durham, , South Road, Durham DH1 LE, UK
| | | | | | | | | | | |
Collapse
|
88
|
McGreal RS, Brennan LA, Kantorow WL, Wilcox JD, Wei J, Chauss D, Kantorow M. Chaperone-independent mitochondrial translocation and protection by αB-crystallin in RPE cells. Exp Eye Res 2013; 110:10-7. [PMID: 23466869 DOI: 10.1016/j.exer.2013.02.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 02/12/2013] [Accepted: 02/19/2013] [Indexed: 11/16/2022]
Abstract
αB-crystallin is a small heat shock protein that exhibits chaperone activity and can protect multiple cell types against oxidative stress damage. Altered levels and specific mutations of αB-crystallin are associated with multiple degenerative diseases. We previously found that αB-crystallin translocates to lens and retinal cell mitochondria upon oxidative stress exposure where it provides protection against oxidative stress damage. To date, the role of the chaperone function of αB-crystallin in mitochondrial translocation and protection has not been established. Here, we sought to determine the relationship between the chaperone activity of αB-crystallin and its ability to translocate to and protect retinal cell mitochondria against oxidative stress damage. Our data provide evidence that three forms of αB-crystallin exhibiting different chaperone activity levels including wild-type, R120G (decreased chaperone activity) and M68A (increased chaperone activity) provide comparable levels of mitochondrial translocation and protection to retinal cells exposed to oxidative stress. The results provide evidence that mitochondrial translocation and protection by αB-crystallin is independent of its chaperone activity and that other functions of αB-crystallin may also be independent of its chaperone activity.
Collapse
Affiliation(s)
- Rebecca S McGreal
- Biomedical Sciences Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL 33431, USA
| | | | | | | | | | | | | |
Collapse
|
89
|
Limphong P, Zhang H, Christians E, Liu Q, Riedel M, Ivey K, Cheng P, Mitzelfelt K, Taylor G, Winge D, Srivastava D, Benjamin I. Modeling human protein aggregation cardiomyopathy using murine induced pluripotent stem cells. Stem Cells Transl Med 2013; 2:161-6. [PMID: 23430692 DOI: 10.5966/sctm.2012-0073] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Several mutations in αB-crystallin (CryAB), a heat shock protein with chaperone-like activities, are causally linked to skeletal and cardiac myopathies in humans. To better understand the underlying pathogenic mechanisms, we had previously generated transgenic (TG) mice expressing R120GCryAB, which recapitulated distinguishing features of the myopathic disorder (e.g., protein aggregates, hypertrophic cardiomyopathy). To determine whether induced pluripotent stem cell (iPSC)-derived cardiomyocytes, a new experimental approach for human disease modeling, would be relevant to aggregation-prone disorders, we decided to exploit the existing transgenic mouse model to derive iPSCs from tail tip fibroblasts. Several iPSC lines were generated from TG and non-TG mice and validated for pluripotency. TG iPSC-derived cardiomyocytes contained perinuclear aggregates positive for CryAB staining, whereas CryAB protein accumulated in both detergent-soluble and insoluble fractions. iPSC-derived cardiomyocytes identified by cardiac troponin T staining were significantly larger when expressing R120GCryAB at a high level in comparison with TG low expressor or non-TG cells. Expression of fetal genes such as atrial natriuretic factor, B-type natriuretic peptide, and α-skeletal α-actin, assessed by quantitative reverse transcription-polymerase chain reaction, were increased in TG cardiomyocytes compared with non-TG, indicating the activation of the hypertrophic genetic program in vitro. Our study demonstrates for the first time that differentiation of R120G iPSCs into cardiomyocytes causes protein aggregation and cellular hypertrophy, recapitulating in vitro key pathognomonic hallmarks found in both animal models and patients. Our findings pave the way for further studies exploiting this cell model system for mechanistic and therapeutic investigations.
Collapse
|
90
|
Datskevich PN, Nefedova VV, Sudnitsyna MV, Gusev NB. Mutations of small heat shock proteins and human congenital diseases. BIOCHEMISTRY (MOSCOW) 2013; 77:1500-14. [DOI: 10.1134/s0006297912130081] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
91
|
Shi J, Koteiche HA, McDonald ET, Fox TL, Stewart PL, McHaourab HS. Cryoelectron microscopy analysis of small heat shock protein 16.5 (Hsp16.5) complexes with T4 lysozyme reveals the structural basis of multimode binding. J Biol Chem 2012; 288:4819-30. [PMID: 23277356 DOI: 10.1074/jbc.m112.388132] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Small heat shock proteins (sHSPs) are ubiquitous chaperones that bind and sequester non-native proteins preventing their aggregation. Despite extensive studies of sHSPs chaperone activity, the location of the bound substrate within the sHSP oligomer has not been determined. In this paper, we used cryoelectron microscopy (cryoEM) to visualize destabilized mutants of T4 lysozyme (T4L) bound to engineered variants of the small heat shock protein Hsp16.5. In contrast to wild type Hsp16.5, binding of T4L to these variants does not induce oligomer heterogeneity enabling cryoEM analysis of the complexes. CryoEM image reconstruction reveals the sequestration of T4L in the interior of the Hsp16.5 oligomer primarily interacting with the buried N-terminal domain but also tethered by contacts with the α-crystallin domain shell. Analysis of Hsp16.5-WT/T4L complexes uncovers oligomer expansion as a requirement for high affinity binding. In contrast, a low affinity mode of binding is found to involve T4L binding on the outer surface of the oligomer bridging the formation of large complexes of Hsp16.5. These mechanistic principles were validated by cryoEM analysis of an expanded variant of Hsp16.5 in complex with T4L and Hsp16.5-R107G, which is equivalent to a mutant of human αB-crystallin linked to cardiomyopathy. In both cases, high affinity binding is found to involve conformational changes in the N-terminal region consistent with a central role of this region in substrate recognition.
Collapse
Affiliation(s)
- Jian Shi
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | | | | | |
Collapse
|
92
|
Khan S, Rammeloo AW, Heikkila JJ. Withaferin A induces proteasome inhibition, endoplasmic reticulum stress, the heat shock response and acquisition of thermotolerance. PLoS One 2012; 7:e50547. [PMID: 23226310 PMCID: PMC3511540 DOI: 10.1371/journal.pone.0050547] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 10/25/2012] [Indexed: 01/05/2023] Open
Abstract
In the present study, withaferin A (WA), a steroidal lactone with anti-inflammatory and anti-tumor properties, inhibited proteasome activity and induced endoplasmic reticulum (ER) and cytoplasmic HSP accumulation in Xenopus laevis A6 kidney epithelial cells. Proteasomal inhibition by WA was indicated by an accumulation of ubiquitinated protein and a decrease in chymotrypsin-like activity. Additionally, immunoblot analysis revealed that treatment of cells with WA induced the accumulation of HSPs including ER chaperones, BiP and GRP94, as well as cytoplasmic/nuclear HSPs, HSP70 and HSP30. Furthermore, WA-induced an increase in the relative levels of the protein kinase, Akt, while the levels of actin were unchanged compared to control. Northern blot experiments determined that WA induced an accumulation in bip, hsp70 and hsp30 mRNA but not eIF-1α mRNA. Interestingly, WA acted synergistically with mild heat shock to enhance HSP70 and HSP30 accumulation to a greater extent than the sum of both stressors individually. This latter phenomenon was not observed with BiP or GRP94. Immunocytochemical analysis indicated that WA-induced BiP accumulation occurred mainly in the perinuclear region in a punctate pattern, while HSP30 accumulation occurred primarily in a granular pattern in the cytoplasm with some staining in the nucleus. Prolonged exposure to WA resulted in disorganization of the F-actin cytoskeleton as well as the production of relatively large HSP30 staining structures that co-localized with F-actin. Finally, prior exposure of cells to WA treatment, which induced the accumulation of HSPs conferred a state of thermal protection since it protected the F-actin cytoskeleton against a subsequent cytotoxic thermal challenge.
Collapse
Affiliation(s)
- Saad Khan
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Ashley W. Rammeloo
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - John J. Heikkila
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
- * E-mail:
| |
Collapse
|
93
|
Arrigo AP. Pathology-dependent effects linked to small heat shock proteins expression: an update. SCIENTIFICA 2012; 2012:185641. [PMID: 24278676 PMCID: PMC3820616 DOI: 10.6064/2012/185641] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 09/17/2012] [Indexed: 06/02/2023]
Abstract
Small heat shock proteins (small Hsps) are stress-induced molecular chaperones that act as holdases towards polypeptides that have lost their folding in stress conditions or consequently of mutations in their coding sequence. A cellular protection against the deleterious effects mediated by damaged proteins is thus provided to cells. These chaperones are also highly expressed in response to protein conformational and inflammatory diseases and cancer pathologies. Through specific and reversible modifications in their phospho-oligomeric organization, small Hsps can chaperone appropriate client proteins in order to provide cells with resistance to different types of injuries or pathological conditions. By helping cells to better cope with their pathological status, their expression can be either beneficial, such as in diseases characterized by pathological cell degeneration, or deleterious when they are required for tumor cell survival. Moreover, small Hsps are actively released by cells and can act as immunogenic molecules that have dual effects depending on the pathology. The cellular consequences linked to their expression levels and relationships with other Hsps as well as therapeutic strategies are discussed in view of their dynamic structural organization required to interact with specific client polypeptides.
Collapse
Affiliation(s)
- A.-P. Arrigo
- Apoptosis Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Claude Bernard University Lyon1, 28 Rue Laennec, 69008 Lyon, France
| |
Collapse
|
94
|
Clark AR, Lubsen NH, Slingsby C. sHSP in the eye lens: Crystallin mutations, cataract and proteostasis. Int J Biochem Cell Biol 2012; 44:1687-97. [DOI: 10.1016/j.biocel.2012.02.015] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 02/23/2012] [Indexed: 01/25/2023]
|
95
|
Bax ML, Sayd T, Aubry L, Ferreira C, Viala D, Chambon C, Rémond D, Santé-Lhoutellier V. Muscle composition slightly affects in vitro digestion of aged and cooked meat: identification of associated proteomic markers. Food Chem 2012. [PMID: 23194521 DOI: 10.1016/j.foodchem.2012.09.049] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Meat is an appropriate source of proteins and minerals for human nutrition. Technological treatments modify the physical-chemical properties of proteins, making them liable to decrease the nutritional potential of meat. To counteract this damage, antioxidants and chaperone proteins in muscle cells can prevent oxidation, restore the function of denatured proteins, and thus prevent aggregation. This study aimed to explore the impact of indoor vs outdoor-reared meat protein composition on digestion and to associate protein markers to in vitro digestion parameters. Indoor-reared meat tended to show less oxidation and denaturation than outdoor-reared meat and was characterised by an overexpression of contractile and chaperone proteins. Outdoor-reared meat showed amplification of antioxidant and detoxification metabolism defending against oxidised compounds. Impacts on digestion remained minor. Several protein markers of in vitro digestion parameters were found for aged and cooked meat, linked to the detoxification process and to muscle contraction.
Collapse
Affiliation(s)
- M-L Bax
- INRA, UR 370 QuaPA, F-63122 Saint-Genès-Champanelle, France
| | | | | | | | | | | | | | | |
Collapse
|
96
|
Kurnellas MP, Brownell SE, Su L, Malkovskiy AV, Rajadas J, Dolganov G, Chopra S, Schoolnik GK, Sobel RA, Webster J, Ousman SS, Becker RA, Steinman L, Rothbard JB. Chaperone activity of small heat shock proteins underlies therapeutic efficacy in experimental autoimmune encephalomyelitis. J Biol Chem 2012; 287:36423-34. [PMID: 22955287 DOI: 10.1074/jbc.m112.371229] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To determine whether the therapeutic activity of αB crystallin, small heat shock protein B5 (HspB5), was shared with other human sHsps, a set of seven human family members, a mutant of HspB5 G120 known to exhibit reduced chaperone activity, and a mycobacterial sHsp were expressed and purified from bacteria. Each of the recombinant proteins was shown to be a functional chaperone, capable of inhibiting aggregation of denatured insulin with varying efficiency. When injected into mice at the peak of disease, they were all effective in reducing the paralysis in experimental autoimmune encephalomyelitis. Additional structure activity correlations between chaperone activity and therapeutic function were established when linear regions within HspB5 were examined. A single region, corresponding to residues 73-92 of HspB5, forms amyloid fibrils, exhibited chaperone activity, and was an effective therapeutic for encephalomyelitis. The linkage of the three activities was further established by demonstrating individual substitutions of critical hydrophobic amino acids in the peptide resulted in the loss of all of the functions.
Collapse
Affiliation(s)
- Michael P Kurnellas
- Department Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305-5316, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Mchaourab HS, Lin YL, Spiller BW. Crystal structure of an activated variant of small heat shock protein Hsp16.5. Biochemistry 2012; 51:5105-12. [PMID: 22670769 PMCID: PMC3384710 DOI: 10.1021/bi300525x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
How does the sequence of a single small heat shock protein (sHSP) assemble into oligomers of different sizes? To gain insight into the underlying structural mechanism, we determined the crystal structure of an engineered variant of Methanocaldococcus jannaschii Hsp16.5 wherein a 14 amino acid peptide from human heat shock protein 27 (Hsp27) was inserted at the junction of the N-terminal region and the α-crystallin domain. In response to this insertion, the oligomer shell expands from 24 to 48 subunits while maintaining octahedral symmetry. Oligomer rearrangement does not alter the fold of the conserved α-crystallin domain nor does it disturb the interface holding the dimeric building block together. Rather, the flexible C-terminal tail of Hsp16.5 changes its orientation relative to the α-crystallin domain which enables alternative packing of dimers. This change in orientation preserves a peptide-in-groove interaction of the C-terminal tail with an adjacent β-sandwich, thereby holding the assembly together. The interior of the expanded oligomer, where substrates presumably bind, retains its predominantly nonpolar character relative to the outside surface. New large windows in the outer shell provide increased access to these substrate-binding regions, thus accounting for the higher affinity of this variant to substrates. Oligomer polydispersity regulates sHSPs chaperone activity in vitro and has been implicated in their physiological roles. The structural mechanism of Hsp16.5 oligomer flexibility revealed here, which is likely to be highly conserved across the sHSP superfamily, explains the relationship between oligomer expansion observed in disease-linked mutants and changes in chaperone activity.
Collapse
Affiliation(s)
- Hassane S. Mchaourab
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, TN 37232, USA
| | - Yi-Lun Lin
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, TN 37232, USA
| | - Benjamin W. Spiller
- Department of Pharmacology, Vanderbilt University Medical Center, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, TN 37232, USA
| |
Collapse
|
98
|
McGreal RS, Kantorow WL, Chauss DC, Wei J, Brennan LA, Kantorow M. αB-crystallin/sHSP protects cytochrome c and mitochondrial function against oxidative stress in lens and retinal cells. Biochim Biophys Acta Gen Subj 2012; 1820:921-30. [PMID: 22521365 DOI: 10.1016/j.bbagen.2012.04.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 03/20/2012] [Accepted: 04/05/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND αB-crystallin/sHSP protects cells against oxidative stress damage. Here, we mechanistically examined its ability to preserve mitochondrial function in lens and retinal cells and protect cytochrome c under oxidative stress conditions. METHODS αB-crystallin/sHSP was localized in human lens (HLE-B3) and retinal (ARPE-19) cells. αB-crystallin/sHSP was stably over-expressed and its ability to preserve mitochondrial membrane potential under oxidative stress conditions was monitored. Interactions between αB-crystallin/sHSP and cytochrome c were examined by fluorescent resonance energy transfer (FRET) and by co-immune precipitation. The ability of αB-crystallin/sHSP to protect cytochrome c against methionine-80 oxidation was monitored. RESULTS αB-crystallin/sHSP is present in the mitochondria of lens and retinal cells and is translocated to the mitochondria under oxidative conditions. αB-crystallin/sHSP specifically interacts with cytochrome c in vitro and in vivo and its overexpression preserves mitochondrial membrane potential under oxidative stress conditions. αB-crystallin/sHSP directly protects cytochrome c against oxidation. GENERAL SIGNIFICANCE These data demonstrate that αB-crystallin/sHSP maintains lens and retinal cells under oxidative stress conditions at least in part by preserving mitochondrial function and by protecting cytochrome c against oxidation. Since oxidative stress and loss of mitochondrial function are associated with eye lens cataract and age-related macular degeneration, loss of these αB-crystallin/sHSP functions likely plays a key role in the development of these diseases. αB-crystallin/sHSP is expressed throughout the body and its ability to maintain mitochondrial function is likely important for the prevention of multiple degenerative diseases.
Collapse
Affiliation(s)
- Rebecca S McGreal
- Biomedical Sciences Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | | | | | | | | | | |
Collapse
|
99
|
Boncoraglio A, Minoia M, Carra S. The family of mammalian small heat shock proteins (HSPBs): implications in protein deposit diseases and motor neuropathies. Int J Biochem Cell Biol 2012; 44:1657-69. [PMID: 22484489 DOI: 10.1016/j.biocel.2012.03.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 03/20/2012] [Accepted: 03/20/2012] [Indexed: 12/19/2022]
Abstract
A number of neurological and muscular disorders are characterized by the accumulation of aggregate-prone proteins and are referred to as protein deposit or protein conformation diseases. Besides some sporadic forms, most of them are genetically inherited in an autosomal dominant manner, although recessive forms also exist. Although genetically very heterogeneous, some of these diseases are the result of mutations in some members of the mammalian small heat shock protein family (sHSP/HSPB), which are key players of the protein quality control system and participate, together with other molecular chaperones and co-chaperones, in the maintenance of protein homeostasis. Thus, on one hand upregulation of specific members of the HSPB family can exert protective effects in protein deposit diseases, such as the polyglutamine diseases. On the other hand, mutations in the HSPBs lead to neurological and muscular disorders, which may be due to a loss-of-function in protein quality control and/or to a gain-of-toxic function, resulting from the aggregation-proneness of the mutants. In this review we summarize the current knowledge about some of the best characterized functions of the HSPBs (e.g. role in cytoskeleton stabilization, chaperone function, anti-aggregation and anti-apoptotic activities), also highlighting differences in the properties of the various HSPBs and how these may counteract protein aggregation diseases. We also describe the mutations in the various HSPBs associated with neurological and muscular disorders and we discuss how gain-of-toxic function mechanisms (e.g. due to the mutated HSPB protein instability and aggregation) and/or loss-of-function mechanisms can contribute to HSPB-associated pathologies. This article is part of a Directed Issue entitled: Small HSPs in physiology and pathology.
Collapse
Affiliation(s)
- Alessandra Boncoraglio
- University Medical Center Groningen, Department of Cell Biology, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | | | | |
Collapse
|
100
|
Keduka E, Hayashi YK, Shalaby S, Mitsuhashi H, Noguchi S, Nonaka I, Nishino I. In vivo characterization of mutant myotilins. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1570-80. [PMID: 22349301 DOI: 10.1016/j.ajpath.2011.12.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 12/27/2011] [Accepted: 12/29/2011] [Indexed: 10/28/2022]
Abstract
Myofibrillar myopathy (MFM) is a group of disorders that are pathologically defined by the disorganization of the myofibrillar alignment associated with the intracellular accumulation of Z-disk-associated proteins. MFM is caused by mutations in genes encoding Z-disk-associated proteins, including myotilin. Although a number of MFM mutations have been identified, it has been difficult to elucidate the precise roles of the mutant proteins. Here, we present a useful method for the characterization of mutant proteins associated with MFM. Expression of mutant myotilins in mouse tibialis anterior muscle by in vivo electroporation recapitulated both the pathological changes and the biochemical characteristics observed in patients with myotilinopathy. In mutant myotilin-expressing muscle fibers, myotilin aggregates and is costained with polyubiquitin, and Z-disk-associated proteins and myofibrillar disorganization were commonly seen. In addition, the expressed S60C mutant myotilin protein displayed marked detergent insolubility in electroporated mouse muscle, similar to that observed in human MFM muscle with the same mutation. Thus, in vivo electroporation can be a useful method for evaluating the pathogenicity of mutations identified in MFM.
Collapse
Affiliation(s)
- Etsuko Keduka
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|