1
|
Montresor S, Pigazzini ML, Baskaran S, Sleiman M, Adhikari G, Basilicata L, Secker L, Jacob N, Ehlert Y, Kelkar A, Kalsi GK, Kulkarni N, Spellerberg P, Kirstein J. HSP110 is a modulator of amyloid beta (Aβ) aggregation and proteotoxicity. J Neurochem 2025; 169:e16214. [PMID: 39180255 PMCID: PMC11657929 DOI: 10.1111/jnc.16214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/01/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024]
Abstract
Chaperones safeguard protein homeostasis by promoting folding and preventing aggregation. HSP110 is a cytosolic chaperone that functions as a nucleotide exchange factor for the HSP70 cycle. Together with HSP70 and a J-domain protein (JDP), HSP110 maintains protein folding and resolubilizes aggregates. Interestingly, HSP110 is vital for the HSP70/110/JDP-mediated disaggregation of amyloidogenic proteins implicated in neurodegenerative diseases (i.e., α-synuclein, HTT, and tau). However, despite its abundance, HSP110 remains still an enigmatic chaperone, and its functional spectrum is not very well understood. Of note, the disaggregation activity of neurodegenerative disease-associated amyloid fibrils showed both beneficial and detrimental outcomes in vivo. To gain a more comprehensive understanding of the chaperone HSP110 in vivo, we analyzed its role in neuronal proteostasis and neurodegeneration in C. elegans. Specifically, we investigated the role of HSP110 in the regulation of amyloid beta peptide (Aβ) aggregation using an established Aβ-C. elegans model that mimics Alzheimer's disease pathology. We generated a novel C. elegans model that over-expresses hsp-110 pan-neuronally, and we also depleted hsp-110 by RNAi-mediated knockdown. We assessed Aβ aggregation in vivo and in situ by fluorescence lifetime imaging. We found that hsp-110 over-expression exacerbated Aβ aggregation and appeared to reduce the conformational variability of the Aβ aggregates, whereas hsp-110 depletion reduced aggregation more significantly in the IL2 neurons, which marked the onset of Aβ aggregation. HSP-110 also plays a central role in growth and fertility as its over-expression compromises nematode physiology. In addition, we found that HSP-110 modulation affects the autophagy pathway. While hsp-110 over-expression impairs the autophagic flux, a depletion enhances it. Thus, HSP-110 regulates multiple nodes of the proteostasis network to control amyloid protein aggregation, disaggregation, and autophagic clearance.
Collapse
Affiliation(s)
| | - Maria Lucia Pigazzini
- Leibniz Institute for Molecular PharmacologyBerlinGermany
- Present address:
EMBL HeidelbergMeyerhofstrasse 169117HeidelbergGermany
| | | | - Mira Sleiman
- Department of Cell BiologyUniversity of BremenBremenGermany
- Leibniz Institute on Aging—Fritz‐Lipmann‐InstituteJenaGermany
| | | | | | - Luca Secker
- Department of Cell BiologyUniversity of BremenBremenGermany
| | - Natascha Jacob
- Department of Cell BiologyUniversity of BremenBremenGermany
| | - Yara Ehlert
- Department of Cell BiologyUniversity of BremenBremenGermany
| | | | | | - Niraj Kulkarni
- Department of Cell BiologyUniversity of BremenBremenGermany
| | | | - Janine Kirstein
- Leibniz Institute on Aging—Fritz‐Lipmann‐InstituteJenaGermany
- Friedrich‐Schiller‐Universität, Institute for Biochemistry & BiophysicsJenaGermany
| |
Collapse
|
2
|
Lei J, Cao L, Li Y, Kan Q, Yang L, Dai W, Liu G, Fu J, Chen Y, Huang Q, Ho CT, Cao Y, Wen L. Physiological evaluation and transcriptomic and proteomic analyses to reveal the anti-aging and reproduction-promoting mechanisms of glycitein in Caenorhabditis elegans. Food Funct 2024; 15:9849-9862. [PMID: 39240213 DOI: 10.1039/d4fo02271h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Soy isoflavones from soy sauce residues have important biological activities. However, the anti-aging and reproduction-promoting effects of glycitein are still rarely reported. Here, we systematically evaluated and explored the anti-aging and reproduction-promoting effects of glycitein in Caenorhabditis elegans (C. elegans). Firstly, we analyzed the effects of glycitein on the lifespan under normal and heat stress, reproduction, locomotion, and reactive oxygen species (ROS) levels of C. elegans. The results showed that 100 μmol L-1 glycitein increased the anti-stress ability of nematodes and activated the antioxidant defense system. Secondly, transcriptomic and proteomic technologies were further used to explore in-depth the anti-aging and reproduction-promoting mechanisms of glycitein in C. elegans. The results showed that both differentially expressed proteins (DEPs) including PDE-2 and MSRA-1 and differentially expressed genes (DEGs) including skpo-2 and cytochrome P450 (cyp-35A3, cyp-35A5, cyp-35C1, cyp-35D1) were associated with the extension of the lifespan and the exertion of antioxidant capacity. VIT-1, plx-2, and Y73F8A.35 were related to promoting reproduction. ASP-1, DNJ-10, and abu-1 were related to the anti-stress ability of glycitein. Pathway analysis revealed that the longevity regulation pathway and FOXO signaling pathway were regulated by the changes in genes and proteins to improve the lifespan of the nematode. Moreover, hydrogenase regulation, longevity regulation, and lipid metabolism were regulated by the changes in genes and proteins to promote the reproduction of nematodes. This study not only demonstrates a viable strategy for utilizing soy sauce residues, but also provides a theoretical foundation and developmental insights for the future application of glycitein.
Collapse
Affiliation(s)
- Jianping Lei
- WENS Foodstuff Group Co., Ltd, Yunfu, 527400, China
| | - Longbifei Cao
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, USA
- Guangzhou Fenghuan Biotechnology Co., Ltd, Guangzhou, 510555, China
| | - Yifeng Li
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Huiertai Biotechnology Co., Ltd, Guangzhou, 510000, China
| | - Qixin Kan
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Lixin Yang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Weijie Dai
- Guangdong Huiertai Biotechnology Co., Ltd, Guangzhou, 510000, China
| | - Guo Liu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China
| | - Jiangyan Fu
- Guangdong Meiweixian Flavoring Foods Co., Ltd, Zhongshan, 528437, China
| | - Yunjiao Chen
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Qingrong Huang
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, USA
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, USA
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Linfeng Wen
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
3
|
Velasco‐Carneros L, Bernardo‐Seisdedos G, Maréchal J, Millet O, Moro F, Muga A. Pseudophosphorylation of single residues of the J-domain of DNAJA2 regulates the holding/folding balance of the Hsc70 system. Protein Sci 2024; 33:e5105. [PMID: 39012012 PMCID: PMC11249846 DOI: 10.1002/pro.5105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/20/2024] [Accepted: 06/23/2024] [Indexed: 07/17/2024]
Abstract
The Hsp70 system is essential for maintaining protein homeostasis and comprises a central Hsp70 and two accessory proteins that belong to the J-domain protein (JDP) and nucleotide exchange factor families. Posttranslational modifications offer a means to tune the activity of the system. We explore how phosphorylation of specific residues of the J-domain of DNAJA2, a class A JDP, regulates Hsc70 activity using biochemical and structural approaches. Among these residues, we find that pseudophosphorylation of Y10 and S51 enhances the holding/folding balance of the Hsp70 system, reducing cochaperone collaboration with Hsc70 while maintaining the holding capacity. Truly phosphorylated J domains corroborate phosphomimetic variant effects. Notably, distinct mechanisms underlie functional impacts of these DNAJA2 variants. Pseudophosphorylation of Y10 induces partial disordering of the J domain, whereas the S51E substitution weakens essential DNAJA2-Hsc70 interactions without a large structural reorganization of the protein. S51 phosphorylation might be class-specific, as all cytosolic class A human JDPs harbor a phosphorylatable residue at this position.
Collapse
Affiliation(s)
- Lorea Velasco‐Carneros
- Instituto Biofisika (UPV/EHU, CSIC)University of Basque CountryLeioaSpain
- Department of Biochemistry and Molecular Biology, Faculty of Science and TechnologyUniversity of the Basque Country (UPV/EHU)LeioaSpain
| | - Ganeko Bernardo‐Seisdedos
- Precision Medicine and Metabolism LabCIC bioGUNEDerioSpain
- Department of Medicine, Faculty of Health SciencesUniversity of DeustoBilbaoSpain
| | - Jean‐Didier Maréchal
- Insilichem, Departament de QuímicaUniversitat Autònoma de Barcelona (UAB)Bellaterra (Barcelona)Spain
| | - Oscar Millet
- Precision Medicine and Metabolism LabCIC bioGUNEDerioSpain
| | - Fernando Moro
- Instituto Biofisika (UPV/EHU, CSIC)University of Basque CountryLeioaSpain
- Department of Biochemistry and Molecular Biology, Faculty of Science and TechnologyUniversity of the Basque Country (UPV/EHU)LeioaSpain
| | - Arturo Muga
- Instituto Biofisika (UPV/EHU, CSIC)University of Basque CountryLeioaSpain
- Department of Biochemistry and Molecular Biology, Faculty of Science and TechnologyUniversity of the Basque Country (UPV/EHU)LeioaSpain
| |
Collapse
|
4
|
Mahto FK, Bhattacharya A, Bhattacharya S. Molecular dynamics simulations shows real-time lid opening in Hsp70 chaperone. J Mol Graph Model 2024; 129:108726. [PMID: 38377794 DOI: 10.1016/j.jmgm.2024.108726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/12/2023] [Accepted: 02/08/2024] [Indexed: 02/22/2024]
Abstract
The stress-inducible mammalian heat shock protein Hsp70 and its bacterial orthologue DnaK are highly conserved molecular chaperones and a crucial part of the machinery responsible for protein folding and homeostasis. Hsp70 is a three-domain, 70 kDa protein that cycles between an ATP-bound state in which all three domains are securely coupled into one unit and an ADP-bound state in which they are loosely attached via a flexible interdomain linker. The Hsp70 presents an alluring novel therapeutic target since it is crucial for maintaining cellular proteostasis and is particularly crucial to cancer cells. We have performed molecular dynamics simulations of the SBD (substrate binding domain) along with the Lid domain in response to experimental efforts to identify small molecule inhibitors that impair the functioning of Hsp70. Our intent has been to characterize the motion of the SBD/Lid allosteric machinery and in, addition, to identify the effect of the PET16 molecule on this motion. Interestingly, we noticed the opening of the entire Lid domain in the apo-form of the dimer. The configuration of the open structure was very different from previously published structures (PDB 4JN4) of the open and docked conformation of the ATP bound form. MD simulations revealed the Lid to be capable of far greater dynamical excursions than has been anticipated by experimental structural biology. This is of value in future drug discovery efforts targeted to modulating Hsp70 activity. The PET16 molecule appears to be weakly bound and its effect on the dynamics of the complex is yet to be elucidated.
Collapse
Affiliation(s)
- Farindra Kumar Mahto
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| | | | - Swati Bhattacharya
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| |
Collapse
|
5
|
Flis Ł, Malewski T, Dobosz R. Temperature Effects on Expression Levels of hsp Genes in Eggs and Second-Stage Juveniles of Meloidogyne hapla Chitwood, 1949. Int J Mol Sci 2024; 25:4867. [PMID: 38732085 PMCID: PMC11084963 DOI: 10.3390/ijms25094867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
Meloidogyne hapla is one of the most important nematode pathogens. It is a sedentary, biotrophic parasite of plants that overwinters in the soil or in diseased roots. The development of M. hapla is temperature dependent. Numerous studies have been performed on the effect of temperature on the development of M. hapla, but only a few of them analyzed the heat shock protein (hsp) genes. The aim of the study was to perform expression profiling of eight hsp genes (Mh-hsp90, Mh-hsp1, Mh-hsp4, Mh-hsp6, Mh-hsp60, Mh-dnj19, Mh-hsp43, and Mh-hsp12.2) at two development stages of M. hapla, i.e., in eggs and second-stage juveniles (J2). The eggs and J2 were incubated under cold stress (5 °C), heat stress (35 °C, 40 °C), and non-stress (10 °C, 20 °C, and 30 °C) conditions. Expression profiling was performed by qPCR. It was demonstrated that only two genes, Mh-hsp60 and Mh-dnj19, have been upregulated by heat and cold stress at both development stages. Heat stress upregulated the expression of more hsp genes than cold stress did. The level of upregulation of most hsp genes was more marked in J2 than in eggs. The obtained results suggest that the Mh-hsp90 and Mh-hsp1 genes can be used as bioindicators of environmental impacts on nematodes of the Meloidogyne genus.
Collapse
Affiliation(s)
- Łukasz Flis
- Museum and Institute of Zoology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warsaw, Poland;
| | - Tadeusz Malewski
- Museum and Institute of Zoology, Polish Academy of Sciences, Twarda 51/55, 00-818 Warsaw, Poland;
| | - Renata Dobosz
- Department of Entomology and Animal Pests, Institute of Plant Protection-National Research Institute, Węgorka 20, 60-318 Poznan, Poland;
| |
Collapse
|
6
|
Wentink A, Rosenzweig R. Protein disaggregation machineries in the human cytosol. Curr Opin Struct Biol 2023; 83:102735. [PMID: 38000128 DOI: 10.1016/j.sbi.2023.102735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023]
Abstract
Proteins carry out the vast majority of functions in cells, but can only do so when properly folded. Following stress or mutation, proteins can lose their proper fold, resulting in misfolding, inactivity, and aggregation-posing a threat to cellular health. In order to counteract protein aggregation, cells have evolved a remarkable subset of molecular chaperones, called protein disaggregases, which collaboratively possess the ability to forcibly untangle protein aggregates. Here, we review the different chaperone disaggregation machineries present in the human cytosol and their mechanisms of action. Understanding, how these disaggregases function, is both universally and clinically important, as protein aggregation has been linked to multiple, debilitating neurodegenerative diseases.
Collapse
Affiliation(s)
- Anne Wentink
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, Netherlands.
| | - Rina Rosenzweig
- Chemical and Structural Biology Department, Weizmann Institute of Science, Rehovot, 761000, Israel.
| |
Collapse
|
7
|
Velasco-Carneros L, Cuéllar J, Dublang L, Santiago C, Maréchal JD, Martín-Benito J, Maestro M, Fernández-Higuero JÁ, Orozco N, Moro F, Valpuesta JM, Muga A. The self-association equilibrium of DNAJA2 regulates its interaction with unfolded substrate proteins and with Hsc70. Nat Commun 2023; 14:5436. [PMID: 37670029 PMCID: PMC10480186 DOI: 10.1038/s41467-023-41150-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 08/24/2023] [Indexed: 09/07/2023] Open
Abstract
J-domain proteins tune the specificity of Hsp70s, engaging them in precise functions. Despite their essential role, the structure and function of many J-domain proteins remain largely unknown. We explore human DNAJA2, finding that it reversibly forms highly-ordered, tubular structures that can be dissociated by Hsc70, the constitutively expressed Hsp70 isoform. Cryoelectron microscopy and mutational studies reveal that different domains are involved in self-association. Oligomer dissociation into dimers potentiates its interaction with unfolded client proteins. The J-domains are accessible to Hsc70 within the tubular structure. They allow binding of closely spaced Hsc70 molecules that could be transferred to the unfolded substrate for its cooperative remodelling, explaining the efficient recovery of DNAJA2-bound clients. The disordered C-terminal domain, comprising the last 52 residues, regulates its holding activity and productive interaction with Hsc70. These in vitro findings suggest that the association equilibrium of DNAJA2 could regulate its interaction with client proteins and Hsc70.
Collapse
Affiliation(s)
- Lorea Velasco-Carneros
- Biofisika Institute (CSIC, UPV/EHU), University of the Basque Country, 48940, Leioa, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Jorge Cuéllar
- Department of Macromolecular Structure, National Centre for Biotechnology (CNB-CSIC), 28049, Madrid, Spain
| | - Leire Dublang
- Biofisika Institute (CSIC, UPV/EHU), University of the Basque Country, 48940, Leioa, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - César Santiago
- Department of Macromolecular Structure, National Centre for Biotechnology (CNB-CSIC), 28049, Madrid, Spain
| | - Jean-Didier Maréchal
- Insilichem, Departament de Química, Universitat Autònoma de Barcelona, (UAB), 08193, Bellaterra (Barcelona), Spain
| | - Jaime Martín-Benito
- Department of Macromolecular Structure, National Centre for Biotechnology (CNB-CSIC), 28049, Madrid, Spain
| | - Moisés Maestro
- Department of Macromolecular Structure, National Centre for Biotechnology (CNB-CSIC), 28049, Madrid, Spain
| | - José Ángel Fernández-Higuero
- Biofisika Institute (CSIC, UPV/EHU), University of the Basque Country, 48940, Leioa, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Natalia Orozco
- Biofisika Institute (CSIC, UPV/EHU), University of the Basque Country, 48940, Leioa, Spain
| | - Fernando Moro
- Biofisika Institute (CSIC, UPV/EHU), University of the Basque Country, 48940, Leioa, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - José María Valpuesta
- Department of Macromolecular Structure, National Centre for Biotechnology (CNB-CSIC), 28049, Madrid, Spain.
| | - Arturo Muga
- Biofisika Institute (CSIC, UPV/EHU), University of the Basque Country, 48940, Leioa, Spain.
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain.
| |
Collapse
|
8
|
van Oosten-Hawle P. Exploiting inter-tissue stress signaling mechanisms to preserve organismal proteostasis during aging. Front Physiol 2023; 14:1228490. [PMID: 37469564 PMCID: PMC10352849 DOI: 10.3389/fphys.2023.1228490] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/26/2023] [Indexed: 07/21/2023] Open
Abstract
Aging results in a decline of cellular proteostasis capacity which culminates in the accumulation of phototoxic material, causing the onset of age-related maladies and ultimately cell death. Mechanisms that regulate proteostasis such as cellular stress response pathways sense disturbances in the proteome. They are activated to increase the expression of protein quality control components that counteract cellular damage. Utilizing invertebrate model organisms such as Caenorhabditis elegans, it has become increasingly evident that the regulation of proteostasis and the activation of cellular stress responses is not a cell autonomous process. In animals, stress responses are orchestrated by signals coming from other tissues, including the nervous system, the intestine and the germline that have a profound impact on determining the aging process. Genetic pathways discovered in C. elegans that facilitate cell nonautonomous regulation of stress responses are providing an exciting feeding ground for new interventions. In this review I will discuss cell nonautonomous proteostasis mechanisms and their impact on aging as well as ongoing research and clinical trials that can increase organismal proteostasis to lengthen health- and lifespan.
Collapse
|
9
|
Kohler V, Andréasson C. Reversible protein assemblies in the proteostasis network in health and disease. Front Mol Biosci 2023; 10:1155521. [PMID: 37021114 PMCID: PMC10067754 DOI: 10.3389/fmolb.2023.1155521] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/09/2023] [Indexed: 04/07/2023] Open
Abstract
While proteins populating their native conformations constitute the functional entities of cells, protein aggregates are traditionally associated with cellular dysfunction, stress and disease. During recent years, it has become clear that large aggregate-like protein condensates formed via liquid-liquid phase separation age into more solid aggregate-like particles that harbor misfolded proteins and are decorated by protein quality control factors. The constituent proteins of the condensates/aggregates are disentangled by protein disaggregation systems mainly based on Hsp70 and AAA ATPase Hsp100 chaperones prior to their handover to refolding and degradation systems. Here, we discuss the functional roles that condensate formation/aggregation and disaggregation play in protein quality control to maintain proteostasis and why it matters for understanding health and disease.
Collapse
Affiliation(s)
- Verena Kohler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Claes Andréasson
- Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden
| |
Collapse
|
10
|
Zhang R, Malinverni D, Cyr DM, Rios PDL, Nillegoda NB. J-domain protein chaperone circuits in proteostasis and disease. Trends Cell Biol 2023; 33:30-47. [PMID: 35729039 PMCID: PMC9759622 DOI: 10.1016/j.tcb.2022.05.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 12/27/2022]
Abstract
The J-domain proteins (JDP) form the largest protein family among cellular chaperones. In cooperation with the Hsp70 chaperone system, these co-chaperones orchestrate a plethora of distinct functions, including those that help maintain cellular proteostasis and development. JDPs evolved largely through the fusion of a J-domain with other protein subdomains. The highly conserved J-domain facilitates the binding and activation of Hsp70s. How JDPs (re)wire Hsp70 chaperone circuits and promote functional diversity remains insufficiently explained. Here, we discuss recent advances in our understanding of the JDP family with a focus on the regulation built around J-domains to ensure correct pairing and assembly of JDP-Hsp70 machineries that operate on different clientele under various cellular growth conditions.
Collapse
Affiliation(s)
- Ruobing Zhang
- Australian Regenerative Medicine Institute (ARMI), Monash University, Melbourne, Victoria, Australia
| | - Duccio Malinverni
- MRC Laboratory of Molecular Biology, Cambridge, UK; Department of Structural Biology and Center for Data Driven Discovery, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Douglas M Cyr
- Department of Cell Biology and Physiology and the Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Paolo De Los Rios
- Institute of Physics, School of Basic Sciences and Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nadinath B Nillegoda
- Australian Regenerative Medicine Institute (ARMI), Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
11
|
Shrivastava A, Sandhof CA, Reinle K, Jawed A, Ruger-Herreros C, Schwarz D, Creamer D, Nussbaum-Krammer C, Mogk A, Bukau B. The cytoprotective sequestration activity of small heat shock proteins is evolutionarily conserved. J Cell Biol 2022; 221:213447. [PMID: 36069810 PMCID: PMC9458469 DOI: 10.1083/jcb.202202149] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/21/2022] [Accepted: 08/16/2022] [Indexed: 11/22/2022] Open
Abstract
The chaperone-mediated sequestration of misfolded proteins into inclusions is a pivotal cellular strategy to maintain proteostasis in Saccharomyces cerevisiae, executed by small heat shock proteins (sHsps) Hsp42 and Btn2. Direct homologs of Hsp42 and Btn2 are absent in other organisms, questioning whether sequestration represents a conserved proteostasis strategy and, if so, which factors are involved. We examined sHsps from Escherchia coli, Caenorhabditis elegans, and humans for their ability to complement the defects of yeast sequestrase mutants. We show that sequestration of misfolded proteins is an original and widespread activity among sHsps executed by specific family members. Sequestrase positive C. elegans' sHsps harbor specific sequence features, including a high content of aromatic and methionine residues in disordered N-terminal extensions. Those sHsps buffer limitations in Hsp70 capacity in C. elegans WT animals and are upregulated in long-lived daf-2 mutants, contributing to lifespan extension. Cellular protection by sequestration of misfolded proteins is, therefore, an evolutionarily conserved activity of the sHsp family.
Collapse
Affiliation(s)
- Aseem Shrivastava
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carl Alexander Sandhof
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kevin Reinle
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Areeb Jawed
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carmen Ruger-Herreros
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominic Schwarz
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Declan Creamer
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Carmen Nussbaum-Krammer
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Axel Mogk
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernd Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
12
|
Costa MD, Maciel P. Modifier pathways in polyglutamine (PolyQ) diseases: from genetic screens to drug targets. Cell Mol Life Sci 2022; 79:274. [PMID: 35503478 PMCID: PMC11071829 DOI: 10.1007/s00018-022-04280-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/14/2022] [Accepted: 03/30/2022] [Indexed: 12/17/2022]
Abstract
Polyglutamine (PolyQ) diseases include a group of inherited neurodegenerative disorders caused by unstable expansions of CAG trinucleotide repeats in the coding region of specific genes. Such genetic alterations produce abnormal proteins containing an unusually long PolyQ tract that renders them more prone to aggregate and cause toxicity. Although research in the field in the last years has contributed significantly to the knowledge of the biological mechanisms implicated in these diseases, effective treatments are still lacking. In this review, we revisit work performed in models of PolyQ diseases, namely the yeast Saccharomyces cerevisiae, the nematode worm Caenorhabditis elegans and the fruit fly Drosophila melanogaster, and provide a critical overview of the high-throughput unbiased genetic screens that have been performed using these systems to identify novel genetic modifiers of PolyQ diseases. These approaches have revealed a wide variety of cellular processes that modulate the toxicity and aggregation of mutant PolyQ proteins, reflecting the complexity of these disorders and demonstrating how challenging the development of therapeutic strategies can be. In addition to the unbiased large-scale genetic screenings in non-vertebrate models, complementary studies in mammalian systems, closer to humans, have contributed with novel genetic modifiers of PolyQ diseases, revealing neuronal function and inflammation as key disease modulators. A pathway enrichment analysis, using the human orthologues of genetic modifiers of PolyQ diseases clustered modifier genes into major themes translatable to the human disease context, such as protein folding and transport as well as transcription regulation. Innovative genetic strategies of genetic manipulation, together with significant advances in genomics and bioinformatics, are taking modifier genetic studies to more realistic disease contexts. The characterization of PolyQ disease modifier pathways is of extreme relevance to reveal novel therapeutic possibilities to delay disease onset and progression in patients.
Collapse
Affiliation(s)
- Marta Daniela Costa
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, 4710-057, Braga, Portugal
- ICVS/3Bs-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Maciel
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, 4710-057, Braga, Portugal.
- ICVS/3Bs-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
13
|
Alagar Boopathy LR, Jacob-Tomas S, Alecki C, Vera M. Mechanisms tailoring the expression of heat shock proteins to proteostasis challenges. J Biol Chem 2022; 298:101796. [PMID: 35248532 PMCID: PMC9065632 DOI: 10.1016/j.jbc.2022.101796] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 12/14/2022] Open
Abstract
All cells possess an internal stress response to cope with environmental and pathophysiological challenges. Upon stress, cells reprogram their molecular functions to activate a survival mechanism known as the heat shock response, which mediates the rapid induction of molecular chaperones such as the heat shock proteins (HSPs). This potent production overcomes the general suppression of gene expression and results in high levels of HSPs to subsequently refold or degrade misfolded proteins. Once the damage or stress is repaired or removed, cells terminate the production of HSPs and resume regular functions. Thus, fulfillment of the stress response requires swift and robust coordination between stress response activation and completion that is determined by the status of the cell. In recent years, single-cell fluorescence microscopy techniques have begun to be used in unravelling HSP-gene expression pathways, from DNA transcription to mRNA degradation. In this review, we will address the molecular mechanisms in different organisms and cell types that coordinate the expression of HSPs with signaling networks that act to reprogram gene transcription, mRNA translation, and decay and ensure protein quality control.
Collapse
|
14
|
Kumari D, Fisher EA, Brodsky JL. Hsp40s play distinct roles during the initial stages of apolipoprotein B biogenesis. Mol Biol Cell 2021; 33:ar15. [PMID: 34910568 PMCID: PMC9236142 DOI: 10.1091/mbc.e21-09-0436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Apolipoprotein B (ApoB) is the primary component of atherogenic lipoproteins, which transport serum fats and cholesterol. Therefore, elevated levels of circulating ApoB are a primary risk factor for cardiovascular disease. During ApoB biosynthesis in the liver and small intestine under nutrient-rich conditions, ApoB cotranslationally translocates into the endoplasmic reticulum (ER) and is lipidated and ultimately secreted. Under lipid-poor conditions, ApoB is targeted for ER Associated Degradation (ERAD). Although prior work identified select chaperones that regulate ApoB biogenesis, the contributions of cytoplasmic Hsp40s are undefined. To this end, we screened ApoB-expressing yeast and determined that a class A ER-associated Hsp40, Ydj1, associates with and facilitates the ERAD of ApoB. Consistent with these results, a homologous Hsp40, DNAJA1, functioned similarly in rat hepatoma cells. DNAJA1 deficient cells also secreted hyperlipidated lipoproteins, in accordance with attenuated ERAD. In contrast to the role of DNAJA1 during ERAD, DNAJB1-a class B Hsp40-helped stabilize ApoB. Depletion of DNAJA1 and DNAJB1 also led to opposing effects on ApoB ubiquitination. These data represent the first example in which different Hsp40s exhibit disparate effects during regulated protein biogenesis in the ER, and highlight distinct roles that chaperones can play on a single ERAD substrate.
Collapse
Affiliation(s)
- Deepa Kumari
- Department of Biological Sciences, A320 Langley Hall, Fifth & Ruskin Ave, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Edward A Fisher
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, United States
| | - Jeffrey L Brodsky
- Department of Biological Sciences, A320 Langley Hall, Fifth & Ruskin Ave, University of Pittsburgh, Pittsburgh, PA 15260 USA
| |
Collapse
|
15
|
Guédon R, Maremonti E, Armant O, Galas S, Brede DA, Lecomte-Pradines C. A systems biology analysis of reproductive toxicity effects induced by multigenerational exposure to ionizing radiation in C. elegans. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 225:112793. [PMID: 34544019 DOI: 10.1016/j.ecoenv.2021.112793] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 06/13/2023]
Abstract
Understanding the effects of chronic exposure to pollutants over generations is of primary importance for the protection of humans and the environment; however, to date, knowledge on the molecular mechanisms underlying multigenerational adverse effects is scarce. We employed a systems biology approach to analyze effects of chronic exposure to gamma radiation at molecular, tissue and individual levels in the nematode Caenorhabditis elegans. Our data show a decrease of 23% in the number of offspring on the first generation F0 and more than 40% in subsequent generations F1, F2 and F3. To unveil the impact on the germline, an in-depth analysis of reproductive processes involved in gametes formation was performed for all four generations. We measured a decrease in the number of mitotic germ cells accompanied by increased cell-cycle arrest in the distal part of the gonad. Further impact on the germline was manifested by decreased sperm quantity and quality. In order to obtain insight in the molecular mechanisms leading to decreased fecundity, gene expression was investigated via whole genome RNA sequencing. The transcriptomic analysis revealed modulation of transcription factors, as well as genes involved in stress response, unfolded protein response, lipid metabolism and reproduction. Furthermore, a drastic increase in the number of differentially expressed genes involved in defense response was measured in the last two generations, suggesting a cumulative stress effect of ionizing radiation exposure. Transcription factor binding site enrichment analysis and the use of transgenic strain identified daf-16/FOXO as a master regulator of genes differentially expressed in response to radiation. The presented data provide new knowledge with respect to the molecular mechanisms involved in reproductive toxic effects and accumulated stress resulting from multigenerational exposure to ionizing radiation.
Collapse
Affiliation(s)
- Rémi Guédon
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PRP-ENV, SRTE, Laboratoire d'ECOtoxicologie des radionucléides (LECO), Cadarache, France
| | - Erica Maremonti
- Centre for Environmental Radioactivity (CERAD), Faculty of Environmental Sciences and Natural Resource Management (MINA), Norwegian University of Life Sciences (NMBU), 1432 Ås, Norway
| | - Olivier Armant
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PRP-ENV, SRTE, Laboratoire d'ECOtoxicologie des radionucléides (LECO), Cadarache, France
| | - Simon Galas
- Institut des biomolecules Max Mousseron (IBMM), University of Montpellier, Centre National de Recherche Scientifique (CNRS), ENSCM, Montpellier, France
| | - Dag Anders Brede
- Centre for Environmental Radioactivity (CERAD), Faculty of Environmental Sciences and Natural Resource Management (MINA), Norwegian University of Life Sciences (NMBU), 1432 Ås, Norway
| | - Catherine Lecomte-Pradines
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PRP-ENV, SRTE, Laboratoire d'ECOtoxicologie des radionucléides (LECO), Cadarache, France.
| |
Collapse
|
16
|
Unzipping the Secrets of Amyloid Disassembly by the Human Disaggregase. Cells 2021; 10:cells10102745. [PMID: 34685723 PMCID: PMC8534776 DOI: 10.3390/cells10102745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 01/08/2023] Open
Abstract
Neurodegenerative diseases (NDs) are increasingly positioned as leading causes of global deaths. The accelerated aging of the population and its strong relationship with neurodegeneration forecast these pathologies as a huge global health problem in the upcoming years. In this scenario, there is an urgent need for understanding the basic molecular mechanisms associated with such diseases. A major molecular hallmark of most NDs is the accumulation of insoluble and toxic protein aggregates, known as amyloids, in extracellular or intracellular deposits. Here, we review the current knowledge on how molecular chaperones, and more specifically a ternary protein complex referred to as the human disaggregase, deals with amyloids. This machinery, composed of the constitutive Hsp70 (Hsc70), the class B J-protein DnaJB1 and the nucleotide exchange factor Apg2 (Hsp110), disassembles amyloids of α-synuclein implicated in Parkinson’s disease as well as of other disease-associated proteins such as tau and huntingtin. We highlight recent studies that have led to the dissection of the mechanism used by this chaperone system to perform its disaggregase activity. We also discuss whether this chaperone-mediated disassembly mechanism could be used to solubilize other amyloidogenic substrates. Finally, we evaluate the implications of the chaperone system in amyloid clearance and associated toxicity, which could be critical for the development of new therapies.
Collapse
|
17
|
Joshi P, Perni M, Limbocker R, Mannini B, Casford S, Chia S, Habchi J, Labbadia J, Dobson CM, Vendruscolo M. Two human metabolites rescue a C. elegans model of Alzheimer's disease via a cytosolic unfolded protein response. Commun Biol 2021; 4:843. [PMID: 34234268 PMCID: PMC8263720 DOI: 10.1038/s42003-021-02218-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
Age-related changes in cellular metabolism can affect brain homeostasis, creating conditions that are permissive to the onset and progression of neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Although the roles of metabolites have been extensively studied with regard to cellular signaling pathways, their effects on protein aggregation remain relatively unexplored. By computationally analysing the Human Metabolome Database, we identified two endogenous metabolites, carnosine and kynurenic acid, that inhibit the aggregation of the amyloid beta peptide (Aβ) and rescue a C. elegans model of Alzheimer's disease. We found that these metabolites act by triggering a cytosolic unfolded protein response through the transcription factor HSF-1 and downstream chaperones HSP40/J-proteins DNJ-12 and DNJ-19. These results help rationalise previous observations regarding the possible anti-ageing benefits of these metabolites by providing a mechanism for their action. Taken together, our findings provide a link between metabolite homeostasis and protein homeostasis, which could inspire preventative interventions against neurodegenerative disorders.
Collapse
Affiliation(s)
- Priyanka Joshi
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK ,grid.47840.3f0000 0001 2181 7878Present Address: The California Institute for Quantitative Biosciences (QB3-Berkeley), University of California, Berkeley, CA USA
| | - Michele Perni
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Ryan Limbocker
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK ,grid.419884.80000 0001 2287 2270Present Address: Department of Chemistry and Life Science, United States Military Academy, West Point, NY USA
| | - Benedetta Mannini
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Sam Casford
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Sean Chia
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Johnny Habchi
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Johnathan Labbadia
- grid.83440.3b0000000121901201Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK
| | - Christopher M. Dobson
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Michele Vendruscolo
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| |
Collapse
|
18
|
Mathangasinghe Y, Fauvet B, Jane SM, Goloubinoff P, Nillegoda NB. The Hsp70 chaperone system: distinct roles in erythrocyte formation and maintenance. Haematologica 2021; 106:1519-1534. [PMID: 33832207 PMCID: PMC8168490 DOI: 10.3324/haematol.2019.233056] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Indexed: 01/14/2023] Open
Abstract
Erythropoiesis is a tightly regulated cell differentiation process in which specialized oxygen- and carbon dioxide-carrying red blood cells are generated in vertebrates. Extensive reorganization and depletion of the erythroblast proteome leading to the deterioration of general cellular protein quality control pathways and rapid hemoglobin biogenesis rates could generate misfolded/aggregated proteins and trigger proteotoxic stresses during erythropoiesis. Such cytotoxic conditions could prevent proper cell differentiation resulting in premature apoptosis of erythroblasts (ineffective erythropoiesis). The heat shock protein 70 (Hsp70) molecular chaperone system supports a plethora of functions that help maintain cellular protein homeostasis (proteostasis) and promote red blood cell differentiation and survival. Recent findings show that abnormalities in the expression, localization and function of the members of this chaperone system are linked to ineffective erythropoiesis in multiple hematological diseases in humans. In this review, we present latest advances in our understanding of the distinct functions of this chaperone system in differentiating erythroblasts and terminally differentiated mature erythrocytes. We present new insights into the protein repair-only function(s) of the Hsp70 system, perhaps to minimize protein degradation in mature erythrocytes to warrant their optimal function and survival in the vasculature under healthy conditions. The work also discusses the modulatory roles of this chaperone system in a wide range of hematological diseases and the therapeutic gain of targeting Hsp70.
Collapse
Affiliation(s)
| | - Bruno Fauvet
- Department of Plant Molecular Biology, Lausanne University, Lausanne
| | - Stephen M Jane
- Central Clinical School, Monash University, Prahran, Victoria, Australia; Department of Hematology, Alfred Hospital, Monash University, Prahran, Victoria
| | | | - Nadinath B Nillegoda
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria.
| |
Collapse
|
19
|
Mesenchymal stem cell secretome protects against alpha-synuclein-induced neurodegeneration in a Caenorhabditis elegans model of Parkinson's disease. Cytotherapy 2021; 23:894-901. [PMID: 34059421 DOI: 10.1016/j.jcyt.2021.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/17/2021] [Accepted: 04/04/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND AIMS The capacity of the secretome from bone marrow-derived mesenchymal stem cells (BMSCs) to prevent dopaminergic neuron degeneration caused by overexpression of alpha-synuclein (α-syn) was explored using two Caenorhabditis elegans models of Parkinson's disease (PD). METHODS First, a more predictive model of PD that overexpresses α-syn in dopamine neurons was subjected to chronic treatment with secretome. This strain displays progressive dopaminergic neurodegeneration that is age-dependent. Following chronic treatment with secretome, the number of intact dopaminergic neurons was determined. Following these initial experiments, a C. elegans strain that overexpresses α-syn in body wall muscle cells was used to determine the impact of hBMSC secretome on α-syn inclusions. Lastly, in silico analysis of the components that constitute the secretome was performed. RESULTS The human BMSC (hBMSC) secretome induced a neuroprotective effect, leading to reduced dopaminergic neurodegeneration. Moreover, in animals submitted to chronic treatment with secretome, the number of α-syn inclusions was reduced, indicating that the secretome of MSCs was possibly contributing to the degradation of those structures. In silico analysis identified possible suppressors of α-syn proteotoxicity, including growth factors and players in the neuronal protein quality control mechanisms. CONCLUSIONS The present findings indicate that hBMSC secretome has the potential to be used as a disease-modifying strategy in future PD regenerative medicine approaches.
Collapse
|
20
|
Interactome Mapping Provides a Network of Neurodegenerative Disease Proteins and Uncovers Widespread Protein Aggregation in Affected Brains. Cell Rep 2021; 32:108050. [PMID: 32814053 DOI: 10.1016/j.celrep.2020.108050] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 02/15/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Interactome maps are valuable resources to elucidate protein function and disease mechanisms. Here, we report on an interactome map that focuses on neurodegenerative disease (ND), connects ∼5,000 human proteins via ∼30,000 candidate interactions and is generated by systematic yeast two-hybrid interaction screening of ∼500 ND-related proteins and integration of literature interactions. This network reveals interconnectivity across diseases and links many known ND-causing proteins, such as α-synuclein, TDP-43, and ATXN1, to a host of proteins previously unrelated to NDs. It facilitates the identification of interacting proteins that significantly influence mutant TDP-43 and HTT toxicity in transgenic flies, as well as of ARF-GEP100 that controls misfolding and aggregation of multiple ND-causing proteins in experimental model systems. Furthermore, it enables the prediction of ND-specific subnetworks and the identification of proteins, such as ATXN1 and MKL1, that are abnormally aggregated in postmortem brains of Alzheimer's disease patients, suggesting widespread protein aggregation in NDs.
Collapse
|
21
|
Radzinski M, Oppenheim T, Metanis N, Reichmann D. The Cys Sense: Thiol Redox Switches Mediate Life Cycles of Cellular Proteins. Biomolecules 2021; 11:469. [PMID: 33809923 PMCID: PMC8004198 DOI: 10.3390/biom11030469] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
Protein homeostasis is an essential component of proper cellular function; however, sustaining protein health is a challenging task, especially during the aerobic lifestyle. Natural cellular oxidants may be involved in cell signaling and antibacterial defense; however, imbalanced levels can lead to protein misfolding, cell damage, and death. This merges together the processes of protein homeostasis and redox regulation. At the heart of this process are redox-regulated proteins or thiol-based switches, which carefully mediate various steps of protein homeostasis across folding, localization, quality control, and degradation pathways. In this review, we discuss the "redox code" of the proteostasis network, which shapes protein health during cell growth and aging. We describe the sources and types of thiol modifications and elaborate on diverse strategies of evolving antioxidant proteins in proteostasis networks during oxidative stress conditions. We also highlight the involvement of cysteines in protein degradation across varying levels, showcasing the importance of cysteine thiols in proteostasis at large. The individual examples and mechanisms raised open the door for extensive future research exploring the interplay between the redox and protein homeostasis systems. Understanding this interplay will enable us to re-write the redox code of cells and use it for biotechnological and therapeutic purposes.
Collapse
Affiliation(s)
- Meytal Radzinski
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem, Jerusalem 91904, Israel; (M.R.); (T.O.)
| | - Tal Oppenheim
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem, Jerusalem 91904, Israel; (M.R.); (T.O.)
| | - Norman Metanis
- Institute of Chemistry, Safra Campus Givat Ram, The Hebrew University of Jerusalem, Jerusalem 91904, Israel;
| | - Dana Reichmann
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem, Jerusalem 91904, Israel; (M.R.); (T.O.)
| |
Collapse
|
22
|
Interaction between non-coding RNAs and JNK in human disorders. Biomed Pharmacother 2021; 138:111497. [PMID: 33735819 DOI: 10.1016/j.biopha.2021.111497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/06/2021] [Accepted: 03/09/2021] [Indexed: 12/31/2022] Open
Abstract
Jun N-terminal Kinase (JNK) signaling pathway is a conserved cascade among species with particular roles in diverse processes during embryogenesis and normal life. These kinases regulate functions of neurons and the immune system by affecting the expression of genes, modulating the arrangement of cytoskeletal proteins, and regulating apoptosis/survival pathways. They are also involved in carcinogenesis. Several miRNAs and lncRNAs have a functional relationship with JNKs. This interaction contributes to the pathogenesis of traumatic brain injury, ulcerative colitis, hepatic ischemia/ reperfusion injury, acute myocardial infarction, and a number of other disorders. Lung cancer, hepatocellular carcinoma, gall bladder cancer, melanoma, and colon cancer are among malignant conditions in which JNK-related miRNAs/ lncRNAs contribute. The current review aims at depicting the functional interaction between JNKs and lncRNAs/ miRNAs and describing the role of these regulatory transcripts in the pathobiology of human disorders.
Collapse
|
23
|
Hedgehog Signaling Modulates Glial Proteostasis and Lifespan. Cell Rep 2021; 30:2627-2643.e5. [PMID: 32101741 DOI: 10.1016/j.celrep.2020.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/11/2019] [Accepted: 01/31/2020] [Indexed: 12/18/2022] Open
Abstract
The conserved Hedgehog signaling pathway has well-established roles in development. However, its function during adulthood remains largely unknown. Here, we investigated whether the Hedgehog signaling pathway is active during adult life in Drosophila melanogaster, and we uncovered a protective function for Hedgehog signaling in coordinating correct proteostasis in glial cells. Adult-specific depletion of Hedgehog reduces lifespan, locomotor activity, and dopaminergic neuron integrity. Conversely, increased expression of Hedgehog extends lifespan and improves fitness. Moreover, Hedgehog pathway activation in glia rescues the lifespan and age-associated defects of hedgehog mutants. The Hedgehog pathway regulates downstream chaperones, whose overexpression in glial cells was sufficient to rescue the shortened lifespan and proteostasis defects of hedgehog mutants. Finally, we demonstrate the protective ability of Hedgehog signaling in a Drosophila Alzheimer's disease model expressing human amyloid beta in the glia. Overall, we propose that Hedgehog signaling is requisite for lifespan determination and correct proteostasis in glial cells.
Collapse
|
24
|
Ayala Mariscal SM, Kirstein J. J-domain proteins interaction with neurodegenerative disease-related proteins. Exp Cell Res 2021; 399:112491. [PMID: 33460589 DOI: 10.1016/j.yexcr.2021.112491] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 12/28/2022]
Abstract
HSP70 chaperones, J-domain proteins (JDPs) and nucleotide exchange factors (NEF) form functional networks that have the ability to prevent and reverse the aggregation of proteins associated with neurodegenerative diseases. JDPs can interact with specific substrate proteins, hold them in a refolding-competent conformation and target them to specific HSP70 chaperones for remodeling. Thereby, JDPs select specific substrates and constitute an attractive target for pharmacological intervention of neurodegenerative diseases. This, under the condition that the exact mechanism of JDPs interaction with specific substrates is unveiled. In this review, we provide an overview of the structural and functional variety of JDPs that interact with neurodegenerative disease-associated proteins and we highlight those studies that identified specific residues, domains or regions of JDPs that are crucial for substrate binding.
Collapse
Affiliation(s)
- Sara María Ayala Mariscal
- Leibniz Research Institute for Molecular Pharmacology Im Forschungsverbund Berlin e.V., R.-Roessle-Strasse 10, 13125, Berlin, Germany
| | - Janine Kirstein
- Leibniz Research Institute for Molecular Pharmacology Im Forschungsverbund Berlin e.V., R.-Roessle-Strasse 10, 13125, Berlin, Germany; University of Bremen, Faculty 2, Cell Biology, Leobener Strasse, 28359, Bremen, Germany.
| |
Collapse
|
25
|
Zhang Y, Zhu X, Qiao X, Sun L, Xia T, Liu C. Clinical Implications of HSC70 Expression in Clear Cell Renal Cell Carcinoma. Int J Med Sci 2021; 18:239-244. [PMID: 33390792 PMCID: PMC7738978 DOI: 10.7150/ijms.43100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 10/07/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose: The role of heat shock protein 70 (HSC70) in the progression of clear cell renal cell carcinoma (ccRCC) is unclear. This study explored the effect of the HSC70 on the survival of ccRCC patients. Methods: Immunohistochemical analysis was performed to determine HSC70 expression in samples obtained from 121 ccRCC patients with at least 5 years of follow-up. We also analyzed the association between HSC70 expression and clinicopathological characteristics. Furthermore, the association of overall survival (OS) with HSC70 expression was analyzed using Kaplan-Meier curves. Finally, we used the Oncomine and CCLE databases to determine the effects of HSC70 mRNA expression on ccRCC. Results: HSC70 expression was associated with distant metastasis and death of ccRCC patients. HSC70 was expressed in the nucleus and/or cytoplasm of ccRCC cells. The incidence of distant organ metastasis and death was higher in patients with HSC70 expression than in those without it. Survival analysis revealed that patients with HSC70 expression had significantly shorter OS. Oncomine analyses also showed that the HSC70 mRNA was significantly upregulated in ccRCC tissues. Conclusions: HSC70 expression was related to adverse prognosis, and patients with HSC70 expression had a worse prognosis than those without HSC70 expression. HSC70 may thus serve as a potential therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Yixiao Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Xudong Zhu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Xinbo Qiao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Lisha Sun
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Tianhui Xia
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Caigang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| |
Collapse
|
26
|
Määttä TA, Rettel M, Sridharan S, Helm D, Kurzawa N, Stein F, Savitski MM. Aggregation and disaggregation features of the human proteome. Mol Syst Biol 2020; 16:e9500. [PMID: 33022891 PMCID: PMC7538195 DOI: 10.15252/msb.20209500] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 08/25/2020] [Accepted: 09/09/2020] [Indexed: 12/15/2022] Open
Abstract
Protein aggregates have negative implications in disease. While reductionist experiments have increased our understanding of aggregation processes, the systemic view in biological context is still limited. To extend this understanding, we used mass spectrometry-based proteomics to characterize aggregation and disaggregation in human cells after non-lethal heat shock. Aggregation-prone proteins were enriched in nuclear proteins, high proportion of intrinsically disordered regions, high molecular mass, high isoelectric point, and hydrophilic amino acids. During recovery, most aggregating proteins disaggregated with a rate proportional to the aggregation propensity: larger loss in solubility was counteracted by faster disaggregation. High amount of intrinsically disordered regions were associated with faster disaggregation. However, other characteristics enriched in aggregating proteins did not correlate with the disaggregation rates. In addition, we analyzed changes in protein thermal stability after heat shock. Soluble remnants of aggregated proteins were more thermally stable compared with control condition. Therefore, our results provide a rich resource of heat stress-related protein solubility data and can foster further studies related to protein aggregation diseases.
Collapse
Affiliation(s)
- Tomi A Määttä
- Genome Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- Faculty of BiosciencesCollaboration for Joint PhD Degree between EMBL and Heidelberg UniversityHeidelbergGermany
| | - Mandy Rettel
- Proteomics Core FacilityEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Sindhuja Sridharan
- Genome Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Dominic Helm
- Proteomics Core FacilityEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Nils Kurzawa
- Genome Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- Faculty of BiosciencesCollaboration for Joint PhD Degree between EMBL and Heidelberg UniversityHeidelbergGermany
| | - Frank Stein
- Proteomics Core FacilityEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Mikhail M Savitski
- Genome Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- Proteomics Core FacilityEuropean Molecular Biology LaboratoryHeidelbergGermany
| |
Collapse
|
27
|
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease and the leading cause of dementia in the elderly. Recent decades have been marked by considerable advances in our understanding of genetic and environmental risk factors and also of the AD mechanism(s) of action. Nonetheless, there is still no cure and the myriad ways AD affects the brain is overwhelmingly complex. Such complexity is manifest in part by the fact that genetic background interacts with the environment, including traffic-derived particulate air pollution, to greatly exacerbate AD risk. Determining the mechanisms by which particulate air pollution acts as an AD risk factor has the potential to reveal yet unknown aspects of AD pathology. This review carefully peels back the layers of complexity to discern whether a unifying disease model, one with proteostasis imbalance at its core, holds up to scrutiny in light of the recent literature. While the data are compelling, it is now time for carefully designed studies to definitively determine whether particulate air pollution acts with ageing, genetic background and other sources of proteotoxic stress to disrupt the delicate proteostasis balance.
Collapse
Affiliation(s)
- Elise A Kikis
- Biology Department, the University of the South, 735 University Avenue, Sewanee, TN 37383, USA
| |
Collapse
|
28
|
Tittelmeier J, Sandhof CA, Ries HM, Druffel-Augustin S, Mogk A, Bukau B, Nussbaum-Krammer C. The HSP110/HSP70 disaggregation system generates spreading-competent toxic α-synuclein species. EMBO J 2020; 39:e103954. [PMID: 32449565 PMCID: PMC7327497 DOI: 10.15252/embj.2019103954] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 04/19/2020] [Accepted: 04/23/2020] [Indexed: 12/21/2022] Open
Abstract
The accumulation and prion-like propagation of α-synuclein and other amyloidogenic proteins are associated with devastating neurodegenerative diseases. Metazoan heat shock protein HSP70 and its co-chaperones DNAJB1 and HSP110 constitute a disaggregation machinery that is able to disassemble α-synuclein fibrils in vitro, but its physiological effects on α-synuclein toxicity are unknown. Here, we depleted Caenorhabditis elegans HSP-110 and monitored the consequences on α-synuclein-related pathological phenotypes such as misfolding, intercellular spreading, and toxicity in C. elegans in vivo models. Depletion of HSP-110 impaired HSP70 disaggregation activity, prevented resolubilization of amorphous aggregates, and compromised the overall cellular folding capacity. At the same time, HSP-110 depletion reduced α-synuclein foci formation, cell-to-cell transmission, and toxicity. These data demonstrate that the HSP70 disaggregation activity constitutes a double-edged sword, as it is essential for maintaining cellular proteostasis but also involved in the generation of toxic amyloid-type protein species.
Collapse
Affiliation(s)
- Jessica Tittelmeier
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Carl Alexander Sandhof
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Heidrun Maja Ries
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Silke Druffel-Augustin
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Axel Mogk
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Bernd Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Carmen Nussbaum-Krammer
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| |
Collapse
|
29
|
Villali J, Dark J, Brechtel TM, Pei F, Sindi SS, Serio TR. Nucleation seed size determines amyloid clearance and establishes a barrier to prion appearance in yeast. Nat Struct Mol Biol 2020; 27:540-549. [PMID: 32367069 PMCID: PMC7293557 DOI: 10.1038/s41594-020-0416-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 03/18/2020] [Indexed: 12/14/2022]
Abstract
Amyloid appearance is a rare event that is promoted in the presence of
other aggregated proteins. These aggregates were thought to act by templating
the formation of an assembly-competent nucleation seed, but we find an
unanticipated role for them in enhancing the persistence of amyloid after it
arises. Specifically, Saccharoymyces cerevisiae Rnq1 amyloid
reduces chaperone-mediated disassembly of Sup35 amyloid, promoting its
persistence in yeast. Mathematical modeling and corresponding in
vivo experiments link amyloid persistence to the conformationally
defined size of the Sup35 nucleation seed and suggest that amyloid is actively
cleared by disassembly below this threshold to suppress appearance of the
[PSI+] prion in vivo.
Remarkably, this framework resolves multiple known inconsistencies in the
appearance and curing of yeast prions. Thus, our observations establish the size
of the nucleation seed as a previously unappreciated characteristic of prion
variants that is key to understanding transitions between prion states.
Collapse
Affiliation(s)
- Janice Villali
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.,Relay Therapeutics, Cambridge, MA, USA
| | - Jason Dark
- Department of Applied Mathematics, University of California, Merced, Merced, CA, USA
| | - Teal M Brechtel
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, USA
| | - Fen Pei
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, USA.,BioLegend, San Diego, CA, USA
| | - Suzanne S Sindi
- Department of Applied Mathematics, University of California, Merced, Merced, CA, USA.
| | - Tricia R Serio
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, USA.
| |
Collapse
|
30
|
Skuodas S, Clemons A, Hayes M, Goll A, Zora B, Weeks DL, Phillips BT, Fassler JS. The ABCF gene family facilitates disaggregation during animal development. Mol Biol Cell 2020; 31:1324-1345. [PMID: 32320318 PMCID: PMC7353142 DOI: 10.1091/mbc.e19-08-0443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Protein aggregation, once believed to be a harbinger and/or consequence of stress, age, and pathological conditions, is emerging as a novel concept in cellular regulation. Normal versus pathological aggregation may be distinguished by the capacity of cells to regulate the formation, modification, and dissolution of aggregates. We find that Caenorhabditis elegans aggregates are observed in large cells/blastomeres (oocytes, embryos) and in smaller, further differentiated cells (primordial germ cells), and their analysis using cell biological and genetic tools is straightforward. These observations are consistent with the hypothesis that aggregates are involved in normal development. Using cross-platform analysis in Saccharomyces cerevisiae, C. elegans, and Xenopus laevis, we present studies identifying a novel disaggregase family encoded by animal genomes and expressed embryonically. Our initial analysis of yeast Arb1/Abcf2 in disaggregation and animal ABCF proteins in embryogenesis is consistent with the possibility that members of the ABCF gene family may encode disaggregases needed for aggregate processing during the earliest stages of animal development.
Collapse
Affiliation(s)
- Sydney Skuodas
- Department of Biology, University of Iowa, Iowa City, IA 52242
| | - Amy Clemons
- Department of Biology, University of Iowa, Iowa City, IA 52242
| | - Michael Hayes
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242
| | - Ashley Goll
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242
| | - Betul Zora
- Department of Biology, University of Iowa, Iowa City, IA 52242
| | - Daniel L Weeks
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242
| | | | - Jan S Fassler
- Department of Biology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
31
|
Tan K, Deng D, Ma X, Cui Y, Tian Z. Pediococcus acidilactici P25 Protected Caenorhabditis elegans against Enterotoxigenic Escherichia coli K88 Infection and Transcriptomic Analysis of Its Potential Mechanisms. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7340312. [PMID: 32337270 PMCID: PMC7150717 DOI: 10.1155/2020/7340312] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/11/2020] [Indexed: 02/07/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) K88 is a zoonotic pathogen. Previous studies have shown that lactic acid bacteria (LAB) have great potential in promoting health and resisting pathogenic infections; however, relatively little research has been done on the Pediococcus genus of LAB. This study is aimed at exploring the mechanisms imparted by Pediococcus acidilactici P25 against ETEC K88 in Caenorhabditis elegans. The probiotic performance of P25 was investigated in vitro. Colonization of K88 in the intestinal tract of C. elegans and abundance of enterotoxin genes were measured. In addition, the transcriptome of C. elegans infected by K88 was analyzed. The result showed that P25 possessed the ability to produce acid, as well as high tolerances to acidic and high bile salt concentrations. Coculture revealed that the growth of ETEC K88 was significantly inhibited by the presence of P25. The median survival of C. elegans fed P25 was 2 days longer than the group infected with K88 alone (P < 0.01). At the same time, the number of colonizing K88 and the abundances of estB and elt were reduced by up to 71.70% and 2.17 times, respectively, by P25. Transcriptome data indicated that P25 affected expression of genes relative to innate immune response and upregulated the abundance of genes in multiple pathways of C. elegans, including peroxisome, longevity, and mitogen-activated protein kinase (MAPK) pathways. These results demonstrated that in the presence of P25, K88 colonization and their expression of enterotoxin genes were reduced. This was accomplished through the alteration of environmental parameters (pH and bile salt) as well as through the promotion of the innate immune response processes, increased longevity, and increased antipathogenic bacteria-related pathways. This work highlights the potential application of P. acidilactici P25 as a probiotic resistant to ETEC K88.
Collapse
Affiliation(s)
- Keqin Tan
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, The Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China 510640
| | - Dun Deng
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, The Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China 510640
| | - Xianyong Ma
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, The Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China 510640
| | - Yiyan Cui
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, The Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China 510640
| | - Zhimei Tian
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, The Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Engineering Technology Research Center of Animal Meat Quality and Safety Control and Evaluation, Guangzhou, China 510640
| |
Collapse
|
32
|
Rosenzweig R, Nillegoda NB, Mayer MP, Bukau B. The Hsp70 chaperone network. Nat Rev Mol Cell Biol 2020; 20:665-680. [PMID: 31253954 DOI: 10.1038/s41580-019-0133-3] [Citation(s) in RCA: 699] [Impact Index Per Article: 139.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The 70-kDa heat shock proteins (Hsp70s) are ubiquitous molecular chaperones that act in a large variety of cellular protein folding and remodelling processes. They function virtually at all stages of the life of proteins from synthesis to degradation and are thus crucial for maintaining protein homeostasis, with direct implications for human health. A large set of co-chaperones comprising J-domain proteins and nucleotide exchange factors regulate the ATPase cycle of Hsp70s, which is allosterically coupled to substrate binding and release. Moreover, Hsp70s cooperate with other cellular chaperone systems including Hsp90, Hsp60 chaperonins, small heat shock proteins and Hsp100 AAA+ disaggregases, together constituting a dynamic and functionally versatile network for protein folding, unfolding, regulation, targeting, aggregation and disaggregation, as well as degradation. In this Review we describe recent advances that have increased our understanding of the molecular mechanisms and working principles of the Hsp70 network. This knowledge showcases how the Hsp70 chaperone system controls diverse cellular functions, and offers new opportunities for the development of chemical compounds that modulate disease-related Hsp70 activities.
Collapse
Affiliation(s)
- Rina Rosenzweig
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Nadinath B Nillegoda
- Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,DKFZ-ZMBH Alliance, Heidelberg, Germany.,Australian Regenerative Medicine Institute (ARMI), Monash University, Clayton, VIC, Australia
| | - Matthias P Mayer
- Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany.,DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Bernd Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany. .,German Cancer Research Center (DKFZ), Heidelberg, Germany. .,DKFZ-ZMBH Alliance, Heidelberg, Germany.
| |
Collapse
|
33
|
Deane CAS, Brown IR. Intracellular Targeting of Heat Shock Proteins in Differentiated Human Neuronal Cells Following Proteotoxic Stress. J Alzheimers Dis 2019; 66:1295-1308. [PMID: 30412487 DOI: 10.3233/jad-180536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
HSPA6 (Hsp70B') is an inducible member of the Hsp70 (HSPA) family of heat shock proteins that is present in the human genome and not found in mouse and rat. Hence it is lacking in current animal models of neurodegenerative diseases. To advance knowledge of the little studied HSPA6, differentiated human neuronal SH-SY5Y cells were treated with the proteotoxic stress-inducing agent MG132. A robust induction of HSPA6 was apparent which localized to the periphery of MG132-induced protein aggregates in the neuronal cytoplasm. Components of the protein disaggregation/refolding machine that co-operate with Hsp70 also targeted the periphery of cytoplasmic protein aggregates, including DNAJB1 (Hsp40-1), HSPH1 (Hsp105α), and HSPB1 (Hsp27). These data suggest that HSPA6 is involved in the response of human neuronal cells to proteotoxic stress that is a feature of neurodegenerative diseases which have been characterized as protein misfolding disorders. Constitutively expressed HSPA8 (Hsc70) also localized tothe periphery of cytoplasmic protein aggregates following the treatment of differentiated human neuronal cells with MG132. HSPA8 could provide a rapid response to proteotoxic stress in neuronal cells, circumventing the time required to upregulate inducible Hsps.
Collapse
Affiliation(s)
- Catherine A S Deane
- Department of Biological Sciences, Centre for the Neurobiology of Stress, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Ian R Brown
- Department of Biological Sciences, Centre for the Neurobiology of Stress, University of Toronto Scarborough, Toronto, Ontario, Canada
| |
Collapse
|
34
|
Papsdorf K, Sima S, Schmauder L, Peter S, Renner L, Hoffelner P, Richter K. head-bent resistant Hsc70 variants show reduced Hsp40 affinity and altered protein folding activity. Sci Rep 2019; 9:11955. [PMID: 31420580 PMCID: PMC6697693 DOI: 10.1038/s41598-019-48109-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 07/25/2019] [Indexed: 01/19/2023] Open
Abstract
The molecular chaperone Hsc70 performs essential tasks by folding proteins. Hsc70 is driven by the hydrolysis of ATP and tuned by the association with various co-chaperones. One such cofactor is the nematode nucleotide exchange factor UNC-23, whose mutation disrupts muscle attachment and induces a severe head-bent phenotype in C.elegans. Interestingly, four mutations in Hsc70 can suppress this phenotype, but the molecular mechanism underlying this suppression is unknown. Here we characterize these four suppressor variants, Hsc70 D233N, S321F, A379V and D384N. In vitro only Hsc70 S321F shows reduced stability and altered nucleotide interaction, but all mutations affect the ATPase stimulation. In particular, Hsc70 D233N and Hsc70 A379V show strongly reduced interactions with DNJ-12 and DNJ-13. Nucleotide exchange factor binding instead is barely influenced in Hsc70 D233N, A379V and D384N and their chaperone activity is preserved. Molecular dynamics simulations suggest that effects in Hsc70 S321F and Hsc70 A379V originate from steric clashes in the vicinity of the mutation site, while D233N disrupts a salt bridge that contributes to Hsc70’s nucleotide-induced conformational changes. In summary, the analyzed mutants show altered ATPase and refolding activity caused by changes in Hsp40 binding.
Collapse
Affiliation(s)
- Katharina Papsdorf
- Center for integrated protein research at the Department of Chemie, Technische Universität München, Lichtenbergstr. 4, 85748, Garching, Germany.,Stanford University School of Medicine, Department of Genetics, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Siyuan Sima
- Center for integrated protein research at the Department of Chemie, Technische Universität München, Lichtenbergstr. 4, 85748, Garching, Germany
| | - Lukas Schmauder
- Center for integrated protein research at the Department of Chemie, Technische Universität München, Lichtenbergstr. 4, 85748, Garching, Germany
| | - Sebastian Peter
- Center for integrated protein research at the Department of Chemie, Technische Universität München, Lichtenbergstr. 4, 85748, Garching, Germany
| | - Lisa Renner
- Center for integrated protein research at the Department of Chemie, Technische Universität München, Lichtenbergstr. 4, 85748, Garching, Germany
| | - Patrica Hoffelner
- Center for integrated protein research at the Department of Chemie, Technische Universität München, Lichtenbergstr. 4, 85748, Garching, Germany
| | - Klaus Richter
- Center for integrated protein research at the Department of Chemie, Technische Universität München, Lichtenbergstr. 4, 85748, Garching, Germany.
| |
Collapse
|
35
|
Wentink A, Nussbaum-Krammer C, Bukau B. Modulation of Amyloid States by Molecular Chaperones. Cold Spring Harb Perspect Biol 2019; 11:a033969. [PMID: 30755450 PMCID: PMC6601462 DOI: 10.1101/cshperspect.a033969] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aberrant protein aggregation is a defining feature of most neurodegenerative diseases. During pathological aggregation, key proteins transition from their native state to alternative conformations, which are prone to oligomerize into highly ordered fibrillar states. As part of the cellular quality control machinery, molecular chaperones can intervene at many stages of the aggregation process to inhibit or reverse aberrant protein aggregation or counteract the toxicity associated with amyloid species. Although the action of chaperones is considered cytoprotective, essential housekeeping functions can be hijacked for the propagation and spreading of protein aggregates, suggesting the cellular protein quality control system constitutes a double-edged sword in neurodegeneration. Here, we discuss the various mechanisms used by chaperones to influence protein aggregation into amyloid fibrils to understand how the interplay of these activities produces specific cellular outcomes and to define mechanisms that may be targeted by pharmacological agents for the treatment of neurodegenerative conditions.
Collapse
Affiliation(s)
- Anne Wentink
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Carmen Nussbaum-Krammer
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Bernd Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| |
Collapse
|
36
|
Feleciano DR, Juenemann K, Iburg M, Brás IC, Holmberg CI, Kirstein J. Crosstalk Between Chaperone-Mediated Protein Disaggregation and Proteolytic Pathways in Aging and Disease. Front Aging Neurosci 2019; 11:9. [PMID: 30760997 PMCID: PMC6361847 DOI: 10.3389/fnagi.2019.00009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/11/2019] [Indexed: 12/19/2022] Open
Abstract
A functional protein quality control machinery is crucial to maintain cellular and organismal physiology. Perturbation in the protein homeostasis network can lead to the formation of misfolded and aggregated proteins that are a hallmark of protein conformational disorders and aging. Protein aggregation is counteracted by the action of chaperones that can resolubilize aggregated proteins. An alternative protein aggregation clearance strategy is the elimination by proteolysis employing the ubiquitin proteasome system (UPS) or autophagy. Little is known how these three protein aggregate clearance strategies are regulated and coordinated in an organism with the progression of aging or upon expression of disease-associated proteins. To unravel the crosstalk between the protein aggregate clearance options, we investigated how autophagy and the UPS respond to perturbations in protein disaggregation capacity. We found that autophagy is induced as a potential compensatory mechanism, whereas the UPS exhibits reduced capacity upon depletion of disaggregating chaperones in C. elegans and HEK293 cells. The expression of amyloid proteins Aβ3-42 and Q40 result in an impairment of autophagy as well as the UPS within the same and even across tissues. Our data indicate a tight coordination between the different nodes of the proteostasis network (PN) with the progression of aging and upon imbalances of the capacity of each clearance mechanism.
Collapse
Affiliation(s)
- Diogo R Feleciano
- Leibniz Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin, Berlin, Germany
| | - Katrin Juenemann
- Leibniz Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin, Berlin, Germany
| | - Manuel Iburg
- Leibniz Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin, Berlin, Germany
| | - Inês C Brás
- Department of Experimental Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Carina I Holmberg
- Research Programs Unit, Translational Cancer Biology Program, and Medicum, Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
| | - Janine Kirstein
- Leibniz Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin, Berlin, Germany
| |
Collapse
|
37
|
Cellular Handling of Protein Aggregates by Disaggregation Machines. Mol Cell 2019; 69:214-226. [PMID: 29351843 DOI: 10.1016/j.molcel.2018.01.004] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/15/2017] [Accepted: 01/02/2018] [Indexed: 11/20/2022]
Abstract
Both acute proteotoxic stresses that unfold proteins and expression of disease-causing mutant proteins that expose aggregation-prone regions can promote protein aggregation. Protein aggregates can interfere with cellular processes and deplete factors crucial for protein homeostasis. To cope with these challenges, cells are equipped with diverse folding and degradation activities to rescue or eliminate aggregated proteins. Here, we review the different chaperone disaggregation machines and their mechanisms of action. In all these machines, the coating of protein aggregates by Hsp70 chaperones represents the conserved, initializing step. In bacteria, fungi, and plants, Hsp70 recruits and activates Hsp100 disaggregases to extract aggregated proteins. In the cytosol of metazoa, Hsp70 is empowered by a specific cast of J-protein and Hsp110 co-chaperones allowing for standalone disaggregation activity. Both types of disaggregation machines are supported by small Hsps that sequester misfolded proteins.
Collapse
|
38
|
Kampinga HH, Andreasson C, Barducci A, Cheetham ME, Cyr D, Emanuelsson C, Genevaux P, Gestwicki JE, Goloubinoff P, Huerta-Cepas J, Kirstein J, Liberek K, Mayer MP, Nagata K, Nillegoda NB, Pulido P, Ramos C, De Los Rios P, Rospert S, Rosenzweig R, Sahi C, Taipale M, Tomiczek B, Ushioda R, Young JC, Zimmermann R, Zylicz A, Zylicz M, Craig EA, Marszalek J. Function, evolution, and structure of J-domain proteins. Cell Stress Chaperones 2019; 24:7-15. [PMID: 30478692 PMCID: PMC6363617 DOI: 10.1007/s12192-018-0948-4] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2018] [Indexed: 01/06/2023] Open
Abstract
Hsp70 chaperone systems are very versatile machines present in nearly all living organisms and in nearly all intracellular compartments. They function in many fundamental processes through their facilitation of protein (re)folding, trafficking, remodeling, disaggregation, and degradation. Hsp70 machines are regulated by co-chaperones. J-domain containing proteins (JDPs) are the largest family of Hsp70 co-chaperones and play a determining role functionally specifying and directing Hsp70 functions. Many features of JDPs are not understood; however, a number of JDP experts gathered at a recent CSSI-sponsored workshop in Gdansk (Poland) to discuss various aspects of J-domain protein function, evolution, and structure. In this report, we present the main findings and the consensus reached to help direct future developments in the field of Hsp70 research.
Collapse
Affiliation(s)
- Harm H Kampinga
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Claes Andreasson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91, Stockholm, Sweden
| | - Alessandro Barducci
- Inserm, U1054, CNRS, UMR 5048, Centre de Biochimie Structurale, Universite de Montpellier, Montpellier, France
| | | | - Douglas Cyr
- University of North Carolina, Chapel Hill, NC, USA
| | - Cecilia Emanuelsson
- Center for Molecular Protein Sciences, CMPS, Dept. Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Pierre Genevaux
- Laboratoire de Microbiologie et de Génétique Moléculaires, Centre de Biologie Intégrative (CBI), CNRS-Université de Toulouse, 118 route de Narbonne, 31062, Toulouse Cedex 9, France
| | - Jason E Gestwicki
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, USA
| | - Pierre Goloubinoff
- Department of Plant Molecular Biology, University of Lausanne, Lausanne, Switzerland
| | | | - Janine Kirstein
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie im Forschungsverbund Berlin e.V. (FMP), Berlin, Germany
| | - Krzysztof Liberek
- Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307, Gdansk, Poland
| | - Matthias P Mayer
- Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany
| | - Kazuhiro Nagata
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, 603-8555, Japan
| | - Nadinath B Nillegoda
- Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany
- Australian Regenerative Medicine Institute (ARMI), Monash University, 15 Innovative Walk, Wellington Road, Clayton, VIC, 3800, Australia
| | - Pablo Pulido
- Plant Molecular Biology, Faculty of Biology, Ludwig-Maximilians-University, Planegg-Martinsried, 82152, Munich, Germany
| | - Carlos Ramos
- Institute of Chemistry, University of Campinas UNICAMP, Campinas, SP, Brazil
| | - Paolo De Los Rios
- EPFL SB IPHYS LBS BSP 723 (Cubotron UNIL), Rte de la Sorge, CH-1015, Lausanne, Switzerland
| | - Sabine Rospert
- Institut fur Biochemie und Molekularbiologie, Universitat Freiburg, Freiburg, Germany
| | | | - Chandan Sahi
- Indian Institute of Science Education and Research Bhopal, Bhauri Bhopal, Madhya Pradesh, 462 066, India
| | - Mikko Taipale
- Donnelly Centre for Cellular and Biomolecular Research, Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Bratłomiej Tomiczek
- Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307, Gdansk, Poland
| | - Ryo Ushioda
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, 603-8555, Japan
| | - Jason C Young
- Department of Biochemistry, McGill University, Montreal, Canada
| | - Richard Zimmermann
- Medical Biochemistry and Molecular Biology, Saarland University, 66421, Homburg, Germany
| | - Alicja Zylicz
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Maciej Zylicz
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Elizabeth A Craig
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jaroslaw Marszalek
- Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307, Gdansk, Poland
| |
Collapse
|
39
|
Mayer MP, Gierasch LM. Recent advances in the structural and mechanistic aspects of Hsp70 molecular chaperones. J Biol Chem 2018; 294:2085-2097. [PMID: 30455352 DOI: 10.1074/jbc.rev118.002810] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hsp70 chaperones are central hubs of the protein quality control network and collaborate with co-chaperones having a J-domain (an ∼70-residue-long helical hairpin with a flexible loop and a conserved His-Pro-Asp motif required for ATP hydrolysis by Hsp70s) and also with nucleotide exchange factors to facilitate many protein-folding processes that (re)establish protein homeostasis. The Hsp70s are highly dynamic nanomachines that modulate the conformation of their substrate polypeptides by transiently binding to short, mostly hydrophobic stretches. This interaction is regulated by an intricate allosteric mechanism. The J-domain co-chaperones target Hsp70 to their polypeptide substrates, and the nucleotide exchange factors regulate the lifetime of the Hsp70-substrate complexes. Significant advances in recent years are beginning to unravel the molecular mechanism of this chaperone machine and how they treat their substrate proteins.
Collapse
Affiliation(s)
- Matthias P Mayer
- From the Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH-Alliance, 69120 Heidelberg, Germany and
| | - Lila M Gierasch
- the Departments of Biochemistry and Molecular Biology and.,Chemistry, University of Massachusetts, Amherst, Massachusetts 01003
| |
Collapse
|
40
|
Rowarth NM, MacRae TH. Post-diapause synthesis of ArHsp40-2, a type 2 J-domain protein from Artemia franciscana, is developmentally regulated and induced by stress. PLoS One 2018; 13:e0201477. [PMID: 30048537 PMCID: PMC6062144 DOI: 10.1371/journal.pone.0201477] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/16/2018] [Indexed: 12/16/2022] Open
Abstract
Post-diapause cysts of Artemia franciscana undergo a well-defined developmental process whereby internal differentiation leads to rupture of the cyst shell, release of membrane-enclosed nauplii and hatching to yield swimming larvae. The post-diapause development of A. franciscana has been examined at biochemical and molecular levels, yet little is known about molecular chaperone function during this process. In addressing this we recently described ArHsp40, a type 1 J-domain protein in post-diapause A. franciscana cysts and larvae. The current report describes ArHsp40-2, a second J-domain protein from A. franciscana. ArHsp40-2 is a type 2 J-domain protein, lacking a zinc binding domain but containing other domains characteristic of these proteins. Notably, ArHsp40-2 possesses a double barrel β-domain structure in its substrate binding region, as does ArHsp40. qPCR revealed a relatively low amount of ArHsp40-2 mRNA in 0 h cysts which increased significantly until the E1 stage, most likely as a result of enhanced transcription, after which it declined. An antibody specific to ArHsp40-2 was produced and used to show that like its mRNA, ArHsp40-2 accumulated until the E1 stage and then decreased to amounts lower than those in 0 h cysts. The synthesis of ArHsp40-2 was induced by heat shock indicating that ArHsp40-2 is involved in stress resistance in cysts and nauplii. Accumulation in cysts during early post-diapause development followed by its sharp decline suggests a role in protein disaggregation/refolding, a function of Hsp40s from other organisms, where ArHsp40-2 assists in the rescue of proteins sequestered during diapause by p26, an abundant small heat shock protein (sHsp) in A. franciscana cysts.
Collapse
Affiliation(s)
| | - Thomas H. MacRae
- Department of Biology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
41
|
Nillegoda NB, Wentink AS, Bukau B. Protein Disaggregation in Multicellular Organisms. Trends Biochem Sci 2018; 43:285-300. [PMID: 29501325 DOI: 10.1016/j.tibs.2018.02.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/29/2018] [Accepted: 02/01/2018] [Indexed: 12/13/2022]
Abstract
Protein aggregates are formed in cells with profoundly perturbed proteostasis, where the generation of misfolded proteins exceeds the cellular refolding and degradative capacity. They are a hallmark of protein conformational disorders and aged and/or environmentally stressed cells. Protein aggregation is a reversible process in vivo, which counteracts proteotoxicities derived from aggregate persistence, but the chaperone machineries involved in protein disaggregation in Metazoa were uncovered only recently. Here we highlight recent advances in the mechanistic understanding of the major protein disaggregation machinery mediated by the Hsp70 chaperone system and discuss emerging alternative disaggregation activities in multicellular organisms.
Collapse
Affiliation(s)
- Nadinath B Nillegoda
- Center for Molecular Biology of Heidelberg University (ZMBH), Im Neuenheimer Feld 282, 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany.
| | - Anne S Wentink
- Center for Molecular Biology of Heidelberg University (ZMBH), Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | - Bernd Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH), Im Neuenheimer Feld 282, 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany.
| |
Collapse
|
42
|
Scior A, Buntru A, Arnsburg K, Ast A, Iburg M, Juenemann K, Pigazzini ML, Mlody B, Puchkov D, Priller J, Wanker EE, Prigione A, Kirstein J. Complete suppression of Htt fibrilization and disaggregation of Htt fibrils by a trimeric chaperone complex. EMBO J 2017; 37:282-299. [PMID: 29212816 DOI: 10.15252/embj.201797212] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 10/19/2017] [Accepted: 10/26/2017] [Indexed: 02/06/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by an expanded CAG trinucleotide repeat in the huntingtin gene (HTT). Molecular chaperones have been implicated in suppressing or delaying the aggregation of mutant Htt. Using in vitro and in vivo assays, we have identified a trimeric chaperone complex (Hsc70, Hsp110, and J-protein) that completely suppresses fibrilization of HttExon1Q48 The composition of this chaperone complex is variable as recruitment of different chaperone family members forms distinct functional complexes. The trimeric chaperone complex is also able to resolubilize Htt fibrils. We confirmed the biological significance of these findings in HD patient-derived neural cells and on an organismal level in Caenorhabditis elegans Among the proteins in this chaperone complex, the J-protein is the concentration-limiting factor. The single overexpression of DNAJB1 in HEK293T cells is sufficient to profoundly reduce HttExon1Q97 aggregation and represents a target of future therapeutic avenues for HD.
Collapse
Affiliation(s)
- Annika Scior
- Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin, Berlin, Germany
| | | | - Kristin Arnsburg
- Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin, Berlin, Germany
| | - Anne Ast
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Manuel Iburg
- Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin, Berlin, Germany
| | - Katrin Juenemann
- Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin, Berlin, Germany
| | - Maria Lucia Pigazzini
- Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin, Berlin, Germany.,Charité - Universitätsmedizin and NeuroCure Cluster of Excellence, Berlin, Germany
| | - Barbara Mlody
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Dmytro Puchkov
- Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin, Berlin, Germany
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charite Universitätsmedizin Berlin, Berlin, Germany
| | - Erich E Wanker
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | | | - Janine Kirstein
- Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin, Berlin, Germany
| |
Collapse
|