1
|
Sarkar S, Pandey A, Kumar Yadav S, Haris Siddiqui M, Pant AB, Yadav S. Differentiated and mature neurons are more responsive to neurotoxicant exposure at both transcriptional and translational levels. Neuroscience 2025; 564:110-125. [PMID: 39571964 DOI: 10.1016/j.neuroscience.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/13/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024]
Abstract
SH-SY5Y human neuroblastoma cells have been extensively used as an in vitro model system in a diverse range of studies involving neurodevelopment, neurotoxicity, neurodegeneration, and neuronal ageing. Both naïve and differentiated phenotypes of SH-SY5Y cells are utilized to model human neurons under in vitro conditions. The process of differentiation causes extensive remodeling of neuronal cells at multiple omic levels, including the epigenome and proteome. In the present investigation, the miRNAome and proteome profiles of arsenic-treated naïve and differentiated SH-SY5Y cells were generated using the miRNA OpenArray technology and high-resolution mass spectrometry. Our findings demonstrated that differentiation dramatically affected the response of SH-SY5Y cells to toxicant exposure, as indicated by increased tolerance of differentiated cells against arsenic exposure compared to naïve cells in cell viability assay. Arsenic-exposed naïve and differentiated SH-SY5Y cells possess distinct miRNA and protein profiles with few similarities. Compared to naïve cells, differentiated cells have undergone higher deregulation in the expression of brain-enriched miRNAs and proteins and have shown a more drastic decrease in oxygen consumption rate, which is a measure of mitochondrial respiration after exposure to arsenic. Proteins identified in arsenic-treated differentiated SH-SY5Y cells were more enriched in pathways underlying multifactorial neurotoxic events. Additionally, more functional regulatory modules have been identified between the miRNAs and proteins differentially expressed in arsenic-treated differentiated SH-SY5Y cells relative to naïve cells. Collectively, our studies have shown that differentiated SH-SY5Y cells displayed alterations in the expression of a greater number of miRNAs and proteins following neurotoxicant exposure, indicating their higher responsivity.
Collapse
Affiliation(s)
- Sana Sarkar
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology (FEST) Division, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India; Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, India.
| | - Anuj Pandey
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology (FEST) Division, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India.
| | - Sanjeev Kumar Yadav
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology (FEST) Division, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India.
| | | | - A B Pant
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology (FEST) Division, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, Lucknow, Uttar Pradesh, India.
| | - Sanjay Yadav
- All India Institute of Medical Sciences (AIIMS), Raebareli, Uttar Pradesh, India.
| |
Collapse
|
2
|
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
|
3
|
Sulatsky MI, Stepanenko OV, Stepanenko OV, Povarova OI, Kuznetsova IM, Turoverov KK, Sulatskaya AI. Broken but not beaten: Challenge of reducing the amyloids pathogenicity by degradation. J Adv Res 2024:S2090-1232(24)00161-9. [PMID: 38642804 DOI: 10.1016/j.jare.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND The accumulation of ordered protein aggregates, amyloid fibrils, accompanies various neurodegenerative diseases (such as Parkinson's, Huntington's, Alzheimer's, etc.) and causes a wide range of systemic and local amyloidoses (such as insulin, hemodialysis amyloidosis, etc.). Such pathologies are usually diagnosed when the disease is already irreversible and a large amount of amyloid plaques have accumulated. In recent years, new drugs aimed at reducing amyloid levels have been actively developed. However, although clinical trials have demonstrated a reduction in amyloid plaque size with these drugs, their effect on disease progression has been controversial and associated with significant side effects, the reasons of which are not fully understood. AIM OF REVIEW The purpose of this review is to summarize extensive array of data on the effect of exogenous and endogenous factors (physico-mechanical effects, chemical effects of low molecular weight compounds, macromolecules and their complexes) on the structure and pathogenicity of mature amyloids for proposing future directions of the development of effective and safe anti-amyloid therapeutics. KEY SCIENTIFIC CONCEPTS OF REVIEW Our analysis show that destruction of amyloids is in most cases incomplete and degradation products often retain the properties of amyloids (including high and sometimes higher than fibrils, cytotoxicity), accelerate amyloidogenesis and promote the propagation of amyloids between cells. Probably, the appearance of protein aggregates, polymorphic in structure and properties (such as amorphous aggregates, fibril fragments, amyloid oligomers, etc.), formed because of uncontrolled degradation of amyloids, may be one of the reasons for the ambiguous effectiveness and serious side effects of the anti-amyloid drugs. This means that all medications that are supposed to be used both for degradation and slow down the fibrillogenesis must first be tested on mature fibrils: the mechanism of drug action and cytotoxic, seeding, and infectious activity of the degradation products must be analyzed.
Collapse
Affiliation(s)
- Maksim I Sulatsky
- Laboratory of Cell Morphology, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia
| | - Olga V Stepanenko
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia
| | - Olesya V Stepanenko
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia
| | - Olga I Povarova
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia
| | - Irina M Kuznetsova
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia
| | - Konstantin K Turoverov
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia
| | - Anna I Sulatskaya
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky ave., 194064 St. Petersburg, Russia.
| |
Collapse
|
4
|
Ren W, Ding B, Dong W, Yue Y, Long X, Zhou Z. Unveiling HSP40/60/70/90/100 gene families and abiotic stress response in Jerusalem artichoke. Gene 2024; 893:147912. [PMID: 37863300 DOI: 10.1016/j.gene.2023.147912] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/28/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
Heat shock proteins (HSPs) are essential for plant growth, development, and stress adaptation. However, their roles in Jerusalem artichoke are largely unexplored. Using bioinformatics, we classified 143 HSP genes into distinct families: HSP40 (82 genes), HSP60 (22 genes), HSP70 (29 genes), HSP90 (6 genes), and HSP100 (4 genes). Our analysis covered their traits, evolution, and structures. Using RNA-seq data, we uncovered unique expression patterns of these HSP genes across growth stages and tissues. Notably, HSP40, HSP60, HSP70, HSP90, and HSP100 families each had specific roles. We also studied how these gene families responded to various stresses, from extreme temperatures to drought and salinity, revealing intricate expression dynamics. Remarkably, HSP40 showed remarkable flexibility, while HSP60, HSP70, HSP90, and HSP100 responded specifically to stress types. Moreover, our analysis unveiled significant correlations between gene pairs under stress, implying cooperative interactions. qRT-PCR validation underscored the significance of particular genes such as HtHSP60-7, HtHSP90-5, HtHSP100-2, and HtHSP100-3 in responding to stress. In summary, our study advances the understanding of how HSP gene families collectively manage stresses in Jerusalem artichoke. This provides insights into specific gene functions and broader plant stress responses.
Collapse
Affiliation(s)
- Wencai Ren
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Baishui Ding
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Wenhan Dong
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Yang Yue
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaohua Long
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhaosheng Zhou
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
5
|
Farley FW, McCully RR, Maslo PB, Yu L, Sheff MA, Sadeghi H, Elion EA. Effects of HSP70 chaperones Ssa1 and Ssa2 on Ste5 scaffold and the mating mitogen-activated protein kinase (MAPK) pathway in Saccharomyces cerevisiae. PLoS One 2023; 18:e0289339. [PMID: 37851593 PMCID: PMC10584130 DOI: 10.1371/journal.pone.0289339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 07/17/2023] [Indexed: 10/20/2023] Open
Abstract
Ste5 is a prototype of scaffold proteins that regulate activation of mitogen-activated protein kinase (MAPK) cascades in all eukaryotes. Ste5 associates with many proteins including Gβγ (Ste4), Ste11 MAPKKK, Ste7 MAPKK, Fus3 and Kss1 MAPKs, Bem1, Cdc24. Here we show that Ste5 also associates with heat shock protein 70 chaperone (Hsp70) Ssa1 and that Ssa1 and its ortholog Ssa2 are together important for Ste5 function and efficient mating responses. The majority of purified overexpressed Ste5 associates with Ssa1. Loss of Ssa1 and Ssa2 has deleterious effects on Ste5 abundance, integrity, and localization particularly when Ste5 is expressed at native levels. The status of Ssa1 and Ssa2 influences Ste5 electrophoresis mobility and formation of high molecular weight species thought to be phosphorylated, ubiquitinylated and aggregated and lower molecular weight fragments. A Ste5 VWA domain mutant with greater propensity to form punctate foci has reduced predicted propensity to bind Ssa1 near the mutation sites and forms more punctate foci when Ssa1 Is overexpressed, supporting a dynamic protein quality control relationship between Ste5 and Ssa1. Loss of Ssa1 and Ssa2 reduces activation of Fus3 and Kss1 MAPKs and FUS1 gene expression and impairs mating shmoo morphogenesis. Surprisingly, ssa1, ssa2, ssa3 and ssa4 single, double and triple mutants can still mate, suggesting compensatory mechanisms exist for folding. Additional analysis suggests Ssa1 is the major Hsp70 chaperone for the mating and invasive growth pathways and reveals several Hsp70-Hsp90 chaperone-network proteins required for mating morphogenesis.
Collapse
Affiliation(s)
- Francis W. Farley
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, United States of America
| | - Ryan R. McCully
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, United States of America
| | - Paul B. Maslo
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, United States of America
| | - Lu Yu
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, United States of America
| | - Mark A. Sheff
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, United States of America
| | - Homayoun Sadeghi
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, United States of America
| | - Elaine A. Elion
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
6
|
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
|
7
|
The role of Atg16 in autophagy, anthocyanin biosynthesis, and programmed cell death in leaves of the lace plant (Aponogeton madagascariensis). PLoS One 2023; 18:e0281668. [PMID: 36795694 PMCID: PMC9934333 DOI: 10.1371/journal.pone.0281668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/29/2023] [Indexed: 02/17/2023] Open
Abstract
Aponogeton madagascariensis, commonly known as the lace plant, produces leaves that form perforations by programmed cell death (PCD). Leaf development is divided into several stages beginning with "pre-perforation" furled leaves enriched with red pigmentation from anthocyanins. The leaf blade is characterized by a series of grids known as areoles bounded by veins. As leaves develop into the "window stage", anthocyanins recede from the center of the areole towards the vasculature creating a gradient of pigmentation and cell death. Cells in the middle of the areole that lack anthocyanins undergo PCD (PCD cells), while cells that retain anthocyanins (non-PCD cells) maintain homeostasis and persist in the mature leaf. Autophagy has reported roles in survival or PCD promotion across different plant cell types. However, the direct involvement of autophagy in PCD and anthocyanin levels during lace plant leaf development has not been determined. Previous RNA sequencing analysis revealed the upregulation of autophagy-related gene Atg16 transcripts in pre-perforation and window stage leaves, but how Atg16 affects PCD in lace plant leaf development is unknown. In this study, we investigated the levels of Atg16 in lace plant PCD by treating whole plants with either an autophagy promoter rapamycin or inhibitors concanamycin A (ConA) or wortmannin. Following treatments, window and mature stage leaves were harvested and analyzed using microscopy, spectrophotometry, and western blotting. Western blotting showed significantly higher Atg16 levels in rapamycin-treated window leaves, coupled with lower anthocyanin levels. Wortmannin-treated leaves had significantly lower Atg16 protein and higher anthocyanin levels compared to the control. Mature leaves from rapamycin-treated plants generated significantly fewer perforations compared to control, while wortmannin had the opposite effect. However, ConA treatment did not significantly change Atg16 levels, nor the number of perforations compared to the control, but anthocyanin levels did increase significantly in window leaves. We propose autophagy plays a dual role in promoting cell survival in NPCD cells by maintaining optimal anthocyanin levels and mediating a timely cell death in PCD cells in developing lace plant leaves. How autophagy specifically affects anthocyanin levels remained unexplained.
Collapse
|
8
|
Almeida ZL, Brito RMM. Amyloid Disassembly: What Can We Learn from Chaperones? Biomedicines 2022; 10:3276. [PMID: 36552032 PMCID: PMC9776232 DOI: 10.3390/biomedicines10123276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 12/23/2022] Open
Abstract
Protein aggregation and subsequent accumulation of insoluble amyloid fibrils with cross-β structure is an intrinsic characteristic of amyloid diseases, i.e., amyloidoses. Amyloid formation involves a series of on-pathway and off-pathway protein aggregation events, leading to mature insoluble fibrils that eventually accumulate in multiple tissues. In this cascade of events, soluble oligomeric species are formed, which are among the most cytotoxic molecular entities along the amyloid cascade. The direct or indirect action of these amyloid soluble oligomers and amyloid protofibrils and fibrils in several tissues and organs lead to cell death in some cases and organ disfunction in general. There are dozens of different proteins and peptides causing multiple amyloid pathologies, chief among them Alzheimer's, Parkinson's, Huntington's, and several other neurodegenerative diseases. Amyloid fibril disassembly is among the disease-modifying therapeutic strategies being pursued to overcome amyloid pathologies. The clearance of preformed amyloids and consequently the arresting of the progression of organ deterioration may increase patient survival and quality of life. In this review, we compiled from the literature many examples of chemical and biochemical agents able to disaggregate preformed amyloids, which have been classified as molecular chaperones, chemical chaperones, and pharmacological chaperones. We focused on their mode of action, chemical structure, interactions with the fibrillar structures, morphology and toxicity of the disaggregation products, and the potential use of disaggregation agents as a treatment option in amyloidosis.
Collapse
Affiliation(s)
| | - Rui M. M. Brito
- Chemistry Department and Coimbra Chemistry Centre—Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
9
|
Cabrera Y, Bernardo-Seisdedos G, Dublang L, Albesa-Jové D, Orozco N, Rosa Viguera A, Millet O, Muga A, Moro F. Fine-tuning of the Hsc70-based human protein disaggregase machinery by the distinctive C-terminal extension of Apg2. J Mol Biol 2022; 434:167841. [PMID: 36167183 DOI: 10.1016/j.jmb.2022.167841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/14/2022] [Accepted: 09/21/2022] [Indexed: 10/31/2022]
Abstract
Apg2, one of the three cytosolic Hsp110 chaperones in humans, supports reactivation of unordered and ordered protein aggregates by Hsc70 (HspA8). Together with DnaJB1, Apg2 serves to nucleate Hsc70 molecules into sites where productive entropic pulling forces can be developed. During aggregate reactivation, Apg2 performs as a specialized nucleotide exchange factor, but the origin of its specialization is poorly defined. Here we report on the role of the distinctive C-terminal extension present in Apg2 and other metazoan homologs. We found that the first part of this Apg2 subdomain with propensity to adopt α-helical structure interacts with the nucleotide binding domain of Hsc70 in a nucleotide-dependent manner, contributing significantly to the stability of the Hsc70:Apg2 complex. Moreover, the second intrinsically disordered segment of Apg2 C-terminal extension plays an important role as a downregulator of nucleotide exchange. An NMR analysis showed that the interaction with Hsc70 nucleotide binding domain modifies the chemical environment of residues located in important functional sites such as the interface between lobe I and II and the nucleotide binding site. Our data indicate that Apg2 C-terminal extension is a fine-tuner of human Hsc70 activity that optimizes the substrate remodeling ability of the chaperone system.
Collapse
Affiliation(s)
- Yovana Cabrera
- Instituto Biofisika (UPV/EHU, CSIC) y Dpto. de Bioquímica y Biología Molecular, Facultad de Ciencia y Tecnología, Universidad del País Vasco, Barrio Sarriena S/N, 48490 Leioa, Spain; Department of Chemistry and Molecular Biology, University of Gothenburg, Medicinaregatan 9C, 41390 Gothenburg, Sweden
| | | | - Leire Dublang
- Instituto Biofisika (UPV/EHU, CSIC) y Dpto. de Bioquímica y Biología Molecular, Facultad de Ciencia y Tecnología, Universidad del País Vasco, Barrio Sarriena S/N, 48490 Leioa, Spain
| | - David Albesa-Jové
- Instituto Biofisika (UPV/EHU, CSIC) y Dpto. de Bioquímica y Biología Molecular, Facultad de Ciencia y Tecnología, Universidad del País Vasco, Barrio Sarriena S/N, 48490 Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Natalia Orozco
- Fundación Biofísica Bizkaia, Barrio Sarriena S/N, 48940 Leioa, Spain
| | - Ana Rosa Viguera
- Instituto Biofisika (UPV/EHU, CSIC) y Dpto. de Bioquímica y Biología Molecular, Facultad de Ciencia y Tecnología, Universidad del País Vasco, Barrio Sarriena S/N, 48490 Leioa, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Spain
| | - Arturo Muga
- Instituto Biofisika (UPV/EHU, CSIC) y Dpto. de Bioquímica y Biología Molecular, Facultad de Ciencia y Tecnología, Universidad del País Vasco, Barrio Sarriena S/N, 48490 Leioa, Spain
| | - Fernando Moro
- Instituto Biofisika (UPV/EHU, CSIC) y Dpto. de Bioquímica y Biología Molecular, Facultad de Ciencia y Tecnología, Universidad del País Vasco, Barrio Sarriena S/N, 48490 Leioa, Spain.
| |
Collapse
|
10
|
Pal A, Paripati A, Deolal P, Chatterjee A, Prasad PR, Adla P, Sepuri NBV. Eisosome protein Pil1 regulates mitochondrial morphology, mitophagy, and cell death in Saccharomyces cerevisiae. J Biol Chem 2022; 298:102533. [PMID: 36162502 PMCID: PMC9619184 DOI: 10.1016/j.jbc.2022.102533] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 12/06/2022] Open
Abstract
Mitochondrial morphology and dynamics maintain mitochondrial integrity by regulating its size, shape, distribution, and connectivity, thereby modulating various cellular processes. Several studies have established a functional link between mitochondrial dynamics, mitophagy, and cell death, but further investigation is needed to identify specific proteins involved in mitochondrial dynamics. Any alteration in the integrity of mitochondria has severe ramifications that include disorders like cancer and neurodegeneration. In this study, we used budding yeast as a model organism and found that Pil1, the major component of the eisosome complex, also localizes to the periphery of mitochondria. Interestingly, the absence of Pil1 causes the branched tubular morphology of mitochondria to be abnormally fused or aggregated, whereas its overexpression leads to mitochondrial fragmentation. Most importantly, pil1Δ cells are defective in mitophagy and bulk autophagy, resulting in elevated levels of reactive oxygen species and protein aggregates. In addition, we show that pil1Δ cells are more prone to cell death. Yeast two-hybrid analysis and co-immunoprecipitations show the interaction of Pil1 with two major proteins in mitochondrial fission, Fis1 and Dnm1. Additionally, our data suggest that the role of Pil1 in maintaining mitochondrial shape is dependent on Fis1 and Dnm1, but it functions independently in mitophagy and cell death pathways. Together, our data suggest that Pil1, an eisosome protein, is a novel regulator of mitochondrial morphology, mitophagy, and cell death.
Collapse
Affiliation(s)
- Amita Pal
- Department of Biochemistry, University of Hyderabad, Prof. C.R Rao Road, Gachibowli, Hyderabad, TS -500046
| | - Arunkumar Paripati
- Department of Biochemistry, University of Hyderabad, Prof. C.R Rao Road, Gachibowli, Hyderabad, TS -500046
| | - Pallavi Deolal
- Department of Biochemistry, University of Hyderabad, Prof. C.R Rao Road, Gachibowli, Hyderabad, TS -500046
| | - Arpan Chatterjee
- Department of Biochemistry, University of Hyderabad, Prof. C.R Rao Road, Gachibowli, Hyderabad, TS -500046
| | - Pushpa Rani Prasad
- Department of Biochemistry, University of Hyderabad, Prof. C.R Rao Road, Gachibowli, Hyderabad, TS -500046
| | - Priyanka Adla
- Department of Biochemistry, University of Hyderabad, Prof. C.R Rao Road, Gachibowli, Hyderabad, TS -500046
| | - Naresh Babu V Sepuri
- Department of Biochemistry, University of Hyderabad, Prof. C.R Rao Road, Gachibowli, Hyderabad, TS -500046.
| |
Collapse
|
11
|
Joshi A, Ito T, Picard D, Neckers L. The Mitochondrial HSP90 Paralog TRAP1: Structural Dynamics, Interactome, Role in Metabolic Regulation, and Inhibitors. Biomolecules 2022; 12:biom12070880. [PMID: 35883436 PMCID: PMC9312948 DOI: 10.3390/biom12070880] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
The HSP90 paralog TRAP1 was discovered more than 20 years ago; yet, a detailed understanding of the function of this mitochondrial molecular chaperone remains elusive. The dispensable nature of TRAP1 in vitro and in vivo further complicates an understanding of its role in mitochondrial biology. TRAP1 is more homologous to the bacterial HSP90, HtpG, than to eukaryotic HSP90. Lacking co-chaperones, the unique structural features of TRAP1 likely regulate its temperature-sensitive ATPase activity and shed light on the alternative mechanisms driving the chaperone’s nucleotide-dependent cycle in a defined environment whose physiological temperature approaches 50 °C. TRAP1 appears to be an important bioregulator of mitochondrial respiration, mediating the balance between oxidative phosphorylation and glycolysis, while at the same time promoting mitochondrial homeostasis and displaying cytoprotective activity. Inactivation/loss of TRAP1 has been observed in several neurodegenerative diseases while TRAP1 expression is reported to be elevated in multiple cancers and, as with HSP90, evidence of addiction to TRAP1 has been observed. In this review, we summarize what is currently known about this unique HSP90 paralog and why a better understanding of TRAP1 structure, function, and regulation is likely to enhance our understanding of the mechanistic basis of mitochondrial homeostasis.
Collapse
Affiliation(s)
- Abhinav Joshi
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA; (A.J.); (T.I.)
| | - Takeshi Ito
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA; (A.J.); (T.I.)
| | - Didier Picard
- Department of Molecular and Cellular Biology, Université de Genève, Sciences III, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland;
| | - Len Neckers
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA; (A.J.); (T.I.)
- Correspondence: ; Tel.: +1-240-858-3918
| |
Collapse
|
12
|
Di Domenico F, Lanzillotta C. The disturbance of protein synthesis/degradation homeostasis is a common trait of age-related neurodegenerative disorders. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 132:49-87. [PMID: 36088079 DOI: 10.1016/bs.apcsb.2022.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Protein homeostasis or "proteostasis" represent the process that regulates the balance of the intracellular functional and "healthy" proteins. Proteostasis is fundamental to preserve physiological metabolic processes in the cell and it allow to respond to any given stimulus as the expression of components of the proteostasis network is customized according to the proteomic demands of different cellular environments. In conditions that promote unfolding/misfolding of proteins chaperones act as signaling molecules inducing extreme measures to either fix the problem or destroy unfolded proteins. When the chaperone machinery fails under pathological insults unfolded proteins induce the endoplasmic reticulum (ER) stress activating the unfolded protein response (UPR) machinery. The activation of the UPR restores ER proteostasis primarily through the transcriptional remodeling of ER protein folding, trafficking, and degradation pathways, such as the ubiquitin proteasome system (UPS). If these mechanisms do not manage to clear the aberrant proteins, proteasome overload and become defective, and misfolded proteins may form aggregates thus extending the UPR mechanism. These aggregates are then attempted to be cleared by macroautophagy. Impaired proteostasis promote the accumulation of misfolded proteins that exacerbate the damage to chaperones, surveillance systems and/or degradative activities. Remarkably, the removal of toxic misfolded proteins is critical for all cells, but it is especially significant in neurons since these cannot be readily replaced. In neurons, the maintenance of efficient proteostasis is essential to healthy aging since the dysregulation of the proteostasis network can lead to neurodegenerative disease. Each of these brain pathologies is characterized by the repeated misfolding of one of more peculiar proteins, which evade both the protein folding machinery and cellular degradation mechanisms and begins to form aggregates that nucleate out into large fibrillar aggregates. In this chapter we describe the mechanisms, associated with faulty proteostasis, that promote the formation of protein aggregates, amyloid fibrils, intracellular, and extracellular inclusions in the most common nondegenerative disorders also referred to as protein misfolding disorders.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy.
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
13
|
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
|
14
|
Sorokina I, Mushegian AR, Koonin EV. Is Protein Folding a Thermodynamically Unfavorable, Active, Energy-Dependent Process? Int J Mol Sci 2022; 23:521. [PMID: 35008947 PMCID: PMC8745595 DOI: 10.3390/ijms23010521] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
The prevailing current view of protein folding is the thermodynamic hypothesis, under which the native folded conformation of a protein corresponds to the global minimum of Gibbs free energy G. We question this concept and show that the empirical evidence behind the thermodynamic hypothesis of folding is far from strong. Furthermore, physical theory-based approaches to the prediction of protein folds and their folding pathways so far have invariably failed except for some very small proteins, despite decades of intensive theory development and the enormous increase of computer power. The recent spectacular successes in protein structure prediction owe to evolutionary modeling of amino acid sequence substitutions enhanced by deep learning methods, but even these breakthroughs provide no information on the protein folding mechanisms and pathways. We discuss an alternative view of protein folding, under which the native state of most proteins does not occupy the global free energy minimum, but rather, a local minimum on a fluctuating free energy landscape. We further argue that ΔG of folding is likely to be positive for the majority of proteins, which therefore fold into their native conformations only through interactions with the energy-dependent molecular machinery of living cells, in particular, the translation system and chaperones. Accordingly, protein folding should be modeled as it occurs in vivo, that is, as a non-equilibrium, active, energy-dependent process.
Collapse
Affiliation(s)
| | - Arcady R. Mushegian
- Division of Molecular and Cellular Biosciences, National Science Foundation, Alexandria, VA 22314, USA;
- Clare Hall College, University of Cambridge, Cambridge CB3 9AL, UK
| | - Eugene V. Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| |
Collapse
|
15
|
Hosfelt J, Richards A, Zheng M, Adura C, Nelson B, Yang A, Fay A, Resager W, Ueberheide B, Glickman JF, Lupoli TJ. An allosteric inhibitor of bacterial Hsp70 chaperone potentiates antibiotics and mitigates resistance. Cell Chem Biol 2021; 29:854-869.e9. [PMID: 34818532 DOI: 10.1016/j.chembiol.2021.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/20/2021] [Accepted: 11/02/2021] [Indexed: 12/23/2022]
Abstract
DnaK is the bacterial homolog of Hsp70, an ATP-dependent chaperone that helps cofactor proteins to catalyze nascent protein folding and salvage misfolded proteins. In the pathogen Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), DnaK and its cofactors are proposed antimycobacterial targets, yet few small-molecule inhibitors or probes exist for these families of proteins. Here, we describe the repurposing of a drug called telaprevir that is able to allosterically inhibit the ATPase activity of DnaK and to prevent chaperone function by mimicking peptide substrates. In mycobacterial cells, telaprevir disrupts DnaK- and cofactor-mediated cellular proteostasis, resulting in enhanced efficacy of aminoglycoside antibiotics and reduced resistance to the frontline TB drug rifampin. Hence, this work contributes to a small but growing collection of protein chaperone inhibitors, and it demonstrates that these molecules disrupt bacterial mechanisms of survival in the presence of different antibiotic classes.
Collapse
Affiliation(s)
- Jordan Hosfelt
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Aweon Richards
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Meng Zheng
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Carolina Adura
- High-Throughput and Spectroscopy Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Brock Nelson
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Amy Yang
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Allison Fay
- Immunology Program, Sloan Kettering Insitute, New York, NY 10065, USA
| | - William Resager
- Departments of Biochemistry and Molecular Pharmacology, Neurology and Director Proteomics Lab, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Beatrix Ueberheide
- Departments of Biochemistry and Molecular Pharmacology, Neurology and Director Proteomics Lab, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - J Fraser Glickman
- High-Throughput and Spectroscopy Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Tania J Lupoli
- Department of Chemistry, New York University, New York, NY 10003, USA.
| |
Collapse
|
16
|
Fassler JS, Skuodas S, Weeks DL, Phillips BT. Protein Aggregation and Disaggregation in Cells and Development. J Mol Biol 2021; 433:167215. [PMID: 34450138 PMCID: PMC8530975 DOI: 10.1016/j.jmb.2021.167215] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/01/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022]
Abstract
Protein aggregation is a feature of numerous neurodegenerative diseases. However, regulated, often reversible, formation of protein aggregates, also known as condensates, helps control a wide range of cellular activities including stress response, gene expression, memory, cell development and differentiation. This review presents examples of aggregates found in biological systems, how they are used, and cellular strategies that control aggregation and disaggregation. We include features of the aggregating proteins themselves, environmental factors, co-aggregates, post-translational modifications and well-known aggregation-directed activities that influence their formation, material state, stability and dissolution. We highlight the emerging roles of biomolecular condensates in early animal development, and disaggregation processing proteins that have recently been shown to play key roles in gametogenesis and embryogenesis.
Collapse
Affiliation(s)
- Jan S Fassler
- Department of Biology, University of Iowa, Iowa City, IA 52242, United States.
| | - Sydney Skuodas
- Department of Biology, University of Iowa, Iowa City, IA 52242, United States. https://twitter.com/@sskuodas
| | - Daniel L Weeks
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242, United States
| | - Bryan T Phillips
- Department of Biology, University of Iowa, Iowa City, IA 52242, United States. https://twitter.com/@bt4phillips
| |
Collapse
|
17
|
Schneider MM, Gautam S, Herling TW, Andrzejewska E, Krainer G, Miller AM, Trinkaus VA, Peter QAE, Ruggeri FS, Vendruscolo M, Bracher A, Dobson CM, Hartl FU, Knowles TPJ. The Hsc70 disaggregation machinery removes monomer units directly from α-synuclein fibril ends. Nat Commun 2021; 12:5999. [PMID: 34650037 PMCID: PMC8516981 DOI: 10.1038/s41467-021-25966-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 08/25/2021] [Indexed: 02/02/2023] Open
Abstract
Molecular chaperones contribute to the maintenance of cellular protein homoeostasis through assisting de novo protein folding and preventing amyloid formation. Chaperones of the Hsp70 family can further disaggregate otherwise irreversible aggregate species such as α-synuclein fibrils, which accumulate in Parkinson's disease. However, the mechanisms and kinetics of this key functionality are only partially understood. Here, we combine microfluidic measurements with chemical kinetics to study α-synuclein disaggregation. We show that Hsc70 together with its co-chaperones DnaJB1 and Apg2 can completely reverse α-synuclein aggregation back to its soluble monomeric state. This reaction proceeds through first-order kinetics where monomer units are removed directly from the fibril ends with little contribution from intermediate fibril fragmentation steps. These findings extend our mechanistic understanding of the role of chaperones in the suppression of amyloid proliferation and in aggregate clearance, and inform on possibilities and limitations of this strategy in the development of therapeutics against synucleinopathies.
Collapse
Affiliation(s)
- Matthias M. Schneider
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW UK
| | - Saurabh Gautam
- grid.418615.f0000 0004 0491 845XDepartment of Cellular Biochemistry, Max-Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany ,Present Address: ViraTherapeutics GmbH, 6063 Rum, Austria
| | - Therese W. Herling
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW UK
| | - Ewa Andrzejewska
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW UK
| | - Georg Krainer
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW UK
| | - Alyssa M. Miller
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW UK
| | - Victoria A. Trinkaus
- grid.418615.f0000 0004 0491 845XDepartment of Cellular Biochemistry, Max-Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Quentin A. E. Peter
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW UK
| | - Francesco Simone Ruggeri
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW UK
| | - Michele Vendruscolo
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW UK
| | - Andreas Bracher
- grid.418615.f0000 0004 0491 845XDepartment of Cellular Biochemistry, Max-Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Christopher M. Dobson
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW UK
| | - F. Ulrich Hartl
- grid.418615.f0000 0004 0491 845XDepartment of Cellular Biochemistry, Max-Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Tuomas P. J. Knowles
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW UK ,grid.5335.00000000121885934Department of Physics, Cavendish Laboratory, University of Cambridge, JJ Thomson Road, Cambridge, CB3 0HE UK
| |
Collapse
|
18
|
Teshima H, Watanabe H, Yasutake R, Ikeda Y, Yonezu Y, Okamoto N, Kakihana A, Yuki R, Nakayama Y, Saito Y. Functional differences between Hsp105/110 family proteins in cell proliferation, cell division, and drug sensitivity. J Cell Biochem 2021; 122:1958-1967. [PMID: 34617313 DOI: 10.1002/jcb.30158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/08/2021] [Accepted: 09/16/2021] [Indexed: 11/07/2022]
Abstract
The mammalian HSP105/110 family consists of four members, including Hsp105 and Apg-1, which function as molecular chaperones. Recently, we reported that Hsp105 knockdown increases sensitivity to the DNA-damaging agent Adriamycin but decreases sensitivity to the microtubule-targeting agent paclitaxel. However, whether the other Hsp105/110 family proteins have the same functional property is unknown. Here, we show that Apg-1 has different roles from Hsp105 in cell proliferation, cell division, and drug sensitivity. We generated the Apg-1-knockdown HeLa S3 cells by lentiviral expression of Apg-1-targeting short hairpin RNA. Knockdown of Apg-1 but not Hsp105 decreased cell proliferation. Apg-1 knockdown increased cell death upon Adriamycin treatment without affecting paclitaxel sensitivity. The cell synchronization experiment suggests that Apg-1 functions in mitotic progression at a different mitotic subphase from Hsp105, which cause difference in paclitaxel sensitivity. Since Apg-1 is overexpressed in certain types of tumors, Apg-1 may become a potential therapeutic target for cancer treatment without causing resistance to the microtubule-targeting agents.
Collapse
Affiliation(s)
- Hiroko Teshima
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Hiroko Watanabe
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Ryuji Yasutake
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yuki Ikeda
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yukiko Yonezu
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Namiko Okamoto
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Ayana Kakihana
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Ryuzaburo Yuki
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yuji Nakayama
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Youhei Saito
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
19
|
Gonçalves CC, Sharon I, Schmeing TM, Ramos CHI, Young JC. The chaperone HSPB1 prepares protein aggregates for resolubilization by HSP70. Sci Rep 2021; 11:17139. [PMID: 34429462 PMCID: PMC8384840 DOI: 10.1038/s41598-021-96518-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/11/2021] [Indexed: 01/22/2023] Open
Abstract
In human cells under stress conditions, misfolded polypeptides can form potentially cytotoxic insoluble aggregates. To eliminate aggregates, the HSP70 chaperone machinery extracts and resolubilizes polypeptides for triage to refolding or degradation. Yeast and bacterial chaperones of the small heat-shock protein (sHSP) family can bind substrates at early stages of misfolding, during the aggregation process. The co-aggregated sHSPs then facilitate downstream disaggregation by HSP70. Because it is unknown whether a human sHSP has this activity, we investigated the disaggregation role of human HSPB1. HSPB1 co-aggregated with unfolded protein substrates, firefly luciferase and mammalian lactate dehydrogenase. The co-aggregates formed with HSPB1 were smaller and more regularly shaped than those formed in its absence. Importantly, co-aggregation promoted the efficient disaggregation and refolding of the substrates, led by HSP70. HSPB1 itself was also extracted during disaggregation, and its homo-oligomerization ability was not required. Therefore, we propose that a human sHSP is an integral part of the chaperone network for protein disaggregation.
Collapse
Affiliation(s)
- Conrado C Gonçalves
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Room 900, Montreal, QC, H3G 1Y6, Canada
| | - Itai Sharon
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, Room 457, Montreal, QC, H3G 0B1, Canada
| | - T Martin Schmeing
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, Room 457, Montreal, QC, H3G 0B1, Canada
| | - Carlos H I Ramos
- Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, 13083-970, Brazil
| | - Jason C Young
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Room 900, Montreal, QC, H3G 1Y6, Canada.
| |
Collapse
|
20
|
Arkan S, Ljungberg M, Kirik D, Hansen C. DNAJB6 suppresses alpha-synuclein induced pathology in an animal model of Parkinson's disease. Neurobiol Dis 2021; 158:105477. [PMID: 34390836 DOI: 10.1016/j.nbd.2021.105477] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/05/2021] [Accepted: 08/09/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND α-synuclein (α-syn) aggregation can lead to degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) as invariably observed in patients with Parkinson's Disease (PD). The co-chaperone DNAJB6 has previously been found to be expressed at higher levels in PD patients than in control subjects and was also found in Lewy bodies. Our previous experiments showed that knock out of DNAJB6 induced α-syn aggregation in cellular level. However, effects of overexpression of DNAJB6 against α-syn aggregation remains to be investigated. METHODS We used a α-syn CFP/YFP HEK293 FRET cell line to investigate the effects of overexpression of DNAJB6 in cellular level. α-syn aggregation was induced by transfection α-syn preformed fibrils (PPF), then was measured FRET analysis. We proceeded to investigate if DNAJB6b can impair α-syn aggregation and toxicity in an animal model and used adeno associated vira (AAV6) designed to overexpress of human wt α-syn, GFP-DNAJB6 or GFP in rats. These vectors were injected into the SNpc of the rats, unilaterally. Rats injected with vira to express α-syn along with GFP in the SNpc where compared to rats expressing α-syn and GFP-DNAJB6. We evaluated motor functions, dopaminergic cell death, and axonal degeneration in striatum. RESULTS We show that DNAJB6 prevent α-syn aggregation induced by α-syn PFF's, in a cell culture model. In addition, we observed α-syn overexpression caused dopaminergic cell death and that this was strongly reduced by co-expression of DNAJB6b. The lesion caused by α-syn overexpression resulted in behavior deficits, which increased over time as seen in stepping test, which was rescued by co-expression of DNAJB6b. CONCLUSION We here demonstrate for the first time that DNAJB6 is a strong suppressor of α-syn aggregation in cells and in animals and that this results in a suppression of dopaminergic cell death and PD related motor deficits in an animal model of PD.
Collapse
Affiliation(s)
- Sertan Arkan
- Lund University, Molecular Neurobiology, Department of Experimental Medical Science, BMC B11, 221 84 Lund, Sweden.
| | - Mårten Ljungberg
- Lund University, Molecular Neurobiology, Department of Experimental Medical Science, BMC B11, 221 84 Lund, Sweden
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems, Department of Experimental Medical Science, Lund University, BMC D11, Lund 22184, Sweden.
| | - Christian Hansen
- Lund University, Molecular Neurobiology, Department of Experimental Medical Science, BMC B11, 221 84 Lund, Sweden; Department of Technology, University College Copenhagen, Denmark.
| |
Collapse
|
21
|
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
|
22
|
Pingale T, Gupta GL. Current and emerging therapeutic targets for Parkinson's disease. Metab Brain Dis 2021; 36:13-27. [PMID: 33090348 DOI: 10.1007/s11011-020-00636-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) is characterized by gradual neurodegeneration and forfeiture of dopamine neurons in substantia nigra pars compacta which ultimately leads to depletion of dopamine levels. PD patients not only display motor features such as rigidity, tremor, and bradykinesia but also non-motor features such as depression, anxiety, etc. Various treatments are available for PD patients such as dopamine replacement are well established but it is only partially or transiently effective. As these therapies not able to restore dopaminergic neurons and delay the development of Parkinson's disease, therefore, the need for an effective therapeutic approach is crucial. The present review discusses a comprehensive overview of current novel targets for PD which includes molecular chaperone, neuroinflammation, mitochondrial dysfunction, neuromelanin, Ubiquitin-proteasome system, protein Abelson, Synaptic vesicle glycoprotein 2C, and Cocaine-amphetamine-regulated transcript, etc. These approaches will help to identify new targets for the treatment of disease and may provide a ray of hope for PD patient treatment. Graphical abstract.
Collapse
Affiliation(s)
- Tanvi Pingale
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400 056, Maharashtra, India
| | - Girdhari Lal Gupta
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400 056, Maharashtra, India.
- Department of Pharmacology, School of Pharmacy & Technology Management, SVKM'S NMIMS, Shirpur, 425 405, Maharashtra, India.
| |
Collapse
|
23
|
Nguyen P, Hess K, Smulders L, Le D, Briseno C, Chavez CM, Nikolaidis N. Origin and Evolution of the Human Bcl2-Associated Athanogene-1 (BAG-1). Int J Mol Sci 2020; 21:ijms21249701. [PMID: 33353252 PMCID: PMC7766421 DOI: 10.3390/ijms21249701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/13/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023] Open
Abstract
Molecular chaperones, particularly the 70-kDa heat shock proteins (Hsp70s), are key orchestrators of the cellular stress response. To perform their critical functions, Hsp70s require the presence of specific co-chaperones, which include nucleotide exchange factors containing the BCL2-associated athanogene (BAG) domain. BAG-1 is one of these proteins that function in a wide range of cellular processes, including apoptosis, protein refolding, and degradation, as well as tumorigenesis. However, the origin of BAG-1 proteins and their evolution between and within species are mostly uncharacterized. This report investigated the macro- and micro-evolution of BAG-1 using orthologous sequences and single nucleotide polymorphisms (SNPs) to elucidate the evolution and understand how natural variation affects the cellular stress response. We first collected and analyzed several BAG-1 sequences across animals, plants, and fungi; mapped intron positions and phases; reconstructed phylogeny; and analyzed protein characteristics. These data indicated that BAG-1 originated before the animals, plants, and fungi split, yet most extant fungal species have lost BAG-1. Furthermore, although BAG-1's structure has remained relatively conserved, kingdom-specific conserved differences exist at sites of known function, suggesting functional specialization within each kingdom. We then analyzed SNPs from the 1000 genomes database to determine the evolutionary patterns within humans. These analyses revealed that the SNP density is unequally distributed within the BAG1 gene, and the ratio of non-synonymous/synonymous SNPs is significantly higher than 1 in the BAG domain region, which is an indication of positive selection. To further explore this notion, we performed several biochemical assays and found that only one out of five mutations tested altered the major co-chaperone properties of BAG-1. These data collectively suggest that although the co-chaperone functions of BAG-1 are highly conserved and can probably tolerate several radical mutations, BAG-1 might have acquired specialized and potentially unexplored functions during the evolutionary process.
Collapse
Affiliation(s)
- Peter Nguyen
- Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, USA; (P.N.); (L.S.); (D.L.); (C.B.); (C.M.C.)
| | - Kyle Hess
- Department of Genome Sciences, Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA;
| | - Larissa Smulders
- Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, USA; (P.N.); (L.S.); (D.L.); (C.B.); (C.M.C.)
| | - Dat Le
- Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, USA; (P.N.); (L.S.); (D.L.); (C.B.); (C.M.C.)
| | - Carolina Briseno
- Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, USA; (P.N.); (L.S.); (D.L.); (C.B.); (C.M.C.)
| | - Christina M. Chavez
- Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, USA; (P.N.); (L.S.); (D.L.); (C.B.); (C.M.C.)
| | - Nikolas Nikolaidis
- Center for Applied Biotechnology Studies, and Center for Computational and Applied Mathematics, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92834-6850, USA; (P.N.); (L.S.); (D.L.); (C.B.); (C.M.C.)
- Correspondence: ; Tel.: +1-657-278-4526
| |
Collapse
|
24
|
Hervás R, Oroz J. Mechanistic Insights into the Role of Molecular Chaperones in Protein Misfolding Diseases: From Molecular Recognition to Amyloid Disassembly. Int J Mol Sci 2020; 21:ijms21239186. [PMID: 33276458 PMCID: PMC7730194 DOI: 10.3390/ijms21239186] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/29/2020] [Accepted: 11/29/2020] [Indexed: 12/14/2022] Open
Abstract
Age-dependent alterations in the proteostasis network are crucial in the progress of prevalent neurodegenerative diseases, such as Alzheimer’s, Parkinson’s, or amyotrophic lateral sclerosis, which are characterized by the presence of insoluble protein deposits in degenerating neurons. Because molecular chaperones deter misfolded protein aggregation, regulate functional phase separation, and even dissolve noxious aggregates, they are considered major sentinels impeding the molecular processes that lead to cell damage in the course of these diseases. Indeed, members of the chaperome, such as molecular chaperones and co-chaperones, are increasingly recognized as therapeutic targets for the development of treatments against degenerative proteinopathies. Chaperones must recognize diverse toxic clients of different orders (soluble proteins, biomolecular condensates, organized protein aggregates). It is therefore critical to understand the basis of the selective chaperone recognition to discern the mechanisms of action of chaperones in protein conformational diseases. This review aimed to define the selective interplay between chaperones and toxic client proteins and the basis for the protective role of these interactions. The presence and availability of chaperone recognition motifs in soluble proteins and in insoluble aggregates, both functional and pathogenic, are discussed. Finally, the formation of aberrant (pro-toxic) chaperone complexes will also be disclosed.
Collapse
Affiliation(s)
- Rubén Hervás
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA;
| | - Javier Oroz
- Rocasolano Institute for Physical Chemistry, Spanish National Research Council (IQFR-CSIC), Serrano 119, E-28006 Madrid, Spain
- Correspondence: ; Tel.: +34-915619400
| |
Collapse
|
25
|
Yakubu UM, Catumbela CSG, Morales R, Morano KA. Understanding and exploiting interactions between cellular proteostasis pathways and infectious prion proteins for therapeutic benefit. Open Biol 2020; 10:200282. [PMID: 33234071 PMCID: PMC7729027 DOI: 10.1098/rsob.200282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Several neurodegenerative diseases of humans and animals are caused by the misfolded prion protein (PrPSc), a self-propagating protein infectious agent that aggregates into oligomeric, fibrillar structures and leads to cell death by incompletely understood mechanisms. Work in multiple biological model systems, from simple baker's yeast to transgenic mouse lines, as well as in vitro studies, has illuminated molecular and cellular modifiers of prion disease. In this review, we focus on intersections between PrP and the proteostasis network, including unfolded protein stress response pathways and roles played by the powerful regulators of protein folding known as protein chaperones. We close with analysis of promising therapeutic avenues for treatment enabled by these studies.
Collapse
Affiliation(s)
- Unekwu M Yakubu
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth, Houston, TX USA.,MD Anderson UTHealth Graduate School at UTHealth, Houston, TX USA
| | - Celso S G Catumbela
- MD Anderson UTHealth Graduate School at UTHealth, Houston, TX USA.,Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at UTHealth, Houston, TX USA
| | - Rodrigo Morales
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at UTHealth, Houston, TX USA.,Centro integrativo de biología y química aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Kevin A Morano
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth, Houston, TX USA
| |
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
|
From Seeds to Fibrils and Back: Fragmentation as an Overlooked Step in the Propagation of Prions and Prion-Like Proteins. Biomolecules 2020; 10:biom10091305. [PMID: 32927676 PMCID: PMC7563560 DOI: 10.3390/biom10091305] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Many devastating neurodegenerative diseases are driven by the misfolding of normal proteins into a pathogenic abnormal conformation. Examples of such protein misfolding diseases include Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and prion diseases. The misfolded proteins involved in these diseases form self-templating oligomeric assemblies that recruit further correctly folded protein and induce their conversion. Over time, this leads to the formation of high molecular and mostly fibrillar aggregates that are increasingly inefficient at converting normal protein. Evidence from a multitude of in vitro models suggests that fibrils are fragmented to form new seeds, which can convert further normal protein and also spread to neighboring cells as observed in vivo. While fragmentation and seed generation were suggested as crucial steps in aggregate formation decades ago, the biological pathways involved remain largely unknown. Here, we show that mechanisms of aggregate clearance—namely the mammalian Hsp70–Hsp40–Hsp110 tri-chaperone system, macro-autophagy, and the proteasome system—may not only be protective, but also play a role in fragmentation. We further review the challenges that exist in determining the precise contribution of these mechanisms to protein misfolding diseases and suggest future directions to resolve these issues.
Collapse
|
28
|
Serrano A, Qiao X, Matos JO, Farley L, Cilenti L, Chen B, Tatulian SA, Teter K. Reversal of Alpha-Synuclein Fibrillization by Protein Disulfide Isomerase. Front Cell Dev Biol 2020; 8:726. [PMID: 32850841 PMCID: PMC7406567 DOI: 10.3389/fcell.2020.00726] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022] Open
Abstract
Aggregates of α-synuclein contribute to the etiology of Parkinson's Disease. Protein disulfide isomerase (PDI), a chaperone and oxidoreductase, blocks the aggregation of α-synuclein. An S-nitrosylated form of PDI that cannot function as a chaperone is associated with elevated levels of aggregated α-synuclein and is found in brains afflicted with Parkinson's Disease. The protective role of PDI in Parkinson's Disease and other neurodegenerative disorders is linked to its chaperone function, yet the mechanism of neuroprotection remains unclear. Using Thioflavin-T fluorescence and transmission electron microscopy, we show here for the first time that PDI can break down nascent fibrils of α-synuclein. Mature fibrils were not affected by PDI. Another PDI family member, ERp57, could prevent but not reverse α-synuclein aggregation. The disaggregase activity of PDI was effective at a 1:50 molar ratio of PDI:α-synuclein and was blocked by S-nitrosylation. PDI could not reverse the aggregation of malate dehydrogenase, which indicated its disaggregase activity does not operate on all substrates. These findings establish a previously unrecognized disaggregase property of PDI that could underlie its neuroprotective function.
Collapse
Affiliation(s)
- Albert Serrano
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Xin Qiao
- Department of Physics, College of Sciences, University of Central Florida, Orlando, FL, United States
| | - Jason O Matos
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Lauren Farley
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Lucia Cilenti
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Bo Chen
- Department of Physics, College of Sciences, University of Central Florida, Orlando, FL, United States
| | - Suren A Tatulian
- Department of Physics, College of Sciences, University of Central Florida, Orlando, FL, United States
| | - Ken Teter
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
29
|
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
|
30
|
Mróz D, Wyszkowski H, Szablewski T, Zawieracz K, Dutkiewicz R, Bury K, Wortmann SB, Wevers RA, Ziętkiewicz S. CLPB (caseinolytic peptidase B homolog), the first mitochondrial protein refoldase associated with human disease. Biochim Biophys Acta Gen Subj 2020; 1864:129512. [DOI: 10.1016/j.bbagen.2020.129512] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 12/23/2019] [Accepted: 01/02/2020] [Indexed: 11/16/2022]
|
31
|
Elmatboly AM, Sherif AM, Deeb DA, Benmelouka A, Bin-Jumah MN, Aleya L, Abdel-Daim MM. The impact of proteostasis dysfunction secondary to environmental and genetic causes on neurodegenerative diseases progression and potential therapeutic intervention. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:11461-11483. [PMID: 32072427 DOI: 10.1007/s11356-020-07914-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/28/2020] [Indexed: 06/10/2023]
Abstract
Aggregation of particular proteins in the form of inclusion bodies or plaques followed by neuronal death is a hallmark of neurodegenerative proteopathies such as primary Parkinsonism, Alzheimer's disease, Lou Gehrig's disease, and Huntington's chorea. Complex polygenic and environmental factors implicated in these proteopathies. Accumulation of proteins in these disorders indicates a substantial disruption in protein homeostasis (proteostasis). Proteostasis or cellular proteome homeostasis is attained by the synchronization of a group of cellular mechanisms called the proteostasis network (PN), which is responsible for the stability of the proteome and achieves the equilibrium between synthesis, folding, and degradation of proteins. In this review, we will discuss the different types of PN and the impact of PN component dysfunction on the four major neurodegenerative diseases mentioned earlier. Graphical abstract.
Collapse
Affiliation(s)
| | - Ahmed M Sherif
- Faculty of Medicine, Zagazig University, El-Sharkia, Egypt
| | - Dalia A Deeb
- Faculty of Medicine, Zagazig University, El-Sharkia, Egypt
| | - Amira Benmelouka
- Faculty of Medicine, University of Algiers, Sidi M'Hamed, Algeria
| | - May N Bin-Jumah
- Biology Department, College Of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon Cedex, France
| | - Mohamed M Abdel-Daim
- Department of Zoology, Science College, King Saud University, Riyadh, 11451, Saudi Arabia.
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
32
|
Proteostasis Failure in Neurodegenerative Diseases: Focus on Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5497046. [PMID: 32308803 PMCID: PMC7140146 DOI: 10.1155/2020/5497046] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 03/03/2020] [Indexed: 12/11/2022]
Abstract
Protein homeostasis or proteostasis is an essential balance of cellular protein levels mediated through an extensive network of biochemical pathways that regulate different steps of the protein quality control, from the synthesis to the degradation. All proteins in a cell continuously turn over, contributing to development, differentiation, and aging. Due to the multiple interactions and connections of proteostasis pathways, exposure to stress conditions may cause various types of protein damage, altering cellular homeostasis and disrupting the entire network with additional cellular stress. Furthermore, protein misfolding and/or alterations during protein synthesis results in inactive or toxic proteins, which may overload the degradation mechanisms. The maintenance of a balanced proteome, preventing the formation of impaired proteins, is accomplished by two major catabolic routes: the ubiquitin proteasomal system (UPS) and the autophagy-lysosomal system. The proteostasis network is particularly important in nondividing, long-lived cells, such as neurons, as its failure is implicated with the development of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. These neurological disorders share common risk factors such as aging, oxidative stress, environmental stress, and protein dysfunction, all of which alter cellular proteostasis, suggesting that general mechanisms controlling proteostasis may underlay the etiology of these diseases. In this review, we describe the major pathways of cellular proteostasis and discuss how their disruption contributes to the onset and progression of neurodegenerative diseases, focusing on the role of oxidative stress.
Collapse
|
33
|
Terrab L, Wipf P. Hsp70 and the Unfolded Protein Response as a Challenging Drug Target and an Inspiration for Probe Molecule Development. ACS Med Chem Lett 2020; 11:232-236. [PMID: 32184949 DOI: 10.1021/acsmedchemlett.9b00583] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The unfolded protein response (UPR) is a cellular stress response mechanism that is critical for cell survival. Pharmacological modulation of the ATPase activity of the chaperone Hsp70 can trigger UPR-mediated cell death, thus removing pathogenic cells in human malignancies, or, alternatively, stimulate survival, thereby preventing apoptosis in neuronal cells and slowing the progress of inflammation, neurodegeneration, and aging. This Viewpoint highlights the complexity of the protein homeostasis network and discusses different approaches for modulating Hsp70 activity, including the use of a chemical reaction development-inspired library of Hsp70 agonists and antagonists.
Collapse
Affiliation(s)
- Leila Terrab
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
34
|
Davis AK, Pratt WB, Lieberman AP, Osawa Y. Targeting Hsp70 facilitated protein quality control for treatment of polyglutamine diseases. Cell Mol Life Sci 2020; 77:977-996. [PMID: 31552448 PMCID: PMC7137528 DOI: 10.1007/s00018-019-03302-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/26/2019] [Accepted: 09/16/2019] [Indexed: 12/14/2022]
Abstract
The polyglutamine (polyQ) diseases are a group of nine fatal, adult-onset neurodegenerative disorders characterized by the misfolding and aggregation of mutant proteins containing toxic expansions of CAG/polyQ tracts. The heat shock protein 90 and 70 (Hsp90/Hsp70) chaperone machinery is a key component of cellular protein quality control, playing a role in the regulation of folding, aggregation, and degradation of polyQ proteins. The ability of Hsp70 to facilitate disaggregation and degradation of misfolded proteins makes it an attractive therapeutic target in polyQ diseases. Genetic studies have demonstrated that manipulation of Hsp70 and related co-chaperones can enhance the disaggregation and/or degradation of misfolded proteins in models of polyQ disease. Therefore, the development of small molecules that enhance Hsp70 activity is of great interest. However, it is still unclear if currently available Hsp70 modulators can selectively enhance disaggregation or degradation of misfolded proteins without perturbing other Hsp70 functions essential for cellular homeostasis. This review discusses the multifaceted role of Hsp70 in protein quality control and the opportunities and challenges Hsp70 poses as a potential therapeutic target in polyQ disease.
Collapse
Affiliation(s)
- Amanda K Davis
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - William B Pratt
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Yoichi Osawa
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
Doonan LM, Guerriero CJ, Preston GM, Buck TM, Khazanov N, Fisher EA, Senderowitz H, Brodsky JL. Hsp104 facilitates the endoplasmic-reticulum-associated degradation of disease-associated and aggregation-prone substrates. Protein Sci 2019; 28:1290-1306. [PMID: 31050848 DOI: 10.1002/pro.3636] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022]
Abstract
Misfolded proteins in the endoplasmic reticulum (ER) are selected for ER-associated degradation (ERAD). More than 60 disease-associated proteins are substrates for the ERAD pathway due to the presence of missense or nonsense mutations. In yeast, the Hsp104 molecular chaperone disaggregates detergent-insoluble ERAD substrates, but the spectrum of disease-associated ERAD substrates that may be aggregation prone is unknown. To determine if Hsp104 recognizes aggregation-prone ERAD substrates associated with human diseases, we developed yeast expression systems for a hydrophobic lipid-binding protein, apolipoprotein B (ApoB), along with a chimeric protein harboring a nucleotide-binding domain from the cystic fibrosis transmembrane conductance regulator (CFTR) into which disease-causing mutations were introduced. We discovered that Hsp104 facilitates the degradation of ER-associated ApoB as well as a truncated CFTR chimera in which a premature stop codon corresponds to a disease-causing mutation. Chimeras containing a wild-type version of the CFTR domain or a different mutation were stable and thus Hsp104 independent. We also discovered that the detergent solubility of the unstable chimera was lower than the stable chimeras, and Hsp104 helped retrotranslocate the unstable chimera from the ER, consistent with disaggregase activity. To determine why the truncated chimera was unstable, we next performed molecular dynamics simulations and noted significant unraveling of the CFTR nucleotide-binding domain. Because human cells lack Hsp104, these data indicate that an alternate disaggregase or mechanism facilitates the removal of aggregation-prone, disease-causing ERAD substrates in their native environments.
Collapse
Affiliation(s)
- Lynley M Doonan
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260
| | - Christopher J Guerriero
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260
| | - G Michael Preston
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260
| | - Teresa M Buck
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260
| | - Netaly Khazanov
- Department of Chemistry, Bar Ilan University, Ramat Gan, 5290002, Israel
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine and Cell Biology, New York University, New York, New York, 10016
| | - Hanoch Senderowitz
- Department of Chemistry, Bar Ilan University, Ramat Gan, 5290002, Israel
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260
| |
Collapse
|
38
|
The Physics of Entropic Pulling: A Novel Model for the Hsp70 Motor Mechanism. Int J Mol Sci 2019; 20:ijms20092334. [PMID: 31083504 PMCID: PMC6539501 DOI: 10.3390/ijms20092334] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 01/04/2023] Open
Abstract
Hsp70s use ATP to generate forces that disassemble protein complexes and aggregates, and that translocate proteins into organelles. Entropic pulling has been proposed as a novel mechanism, distinct from the more familiar power-stroke and Brownian ratchet models, for how Hsp70s generate these forces. Experimental evidence supports entropic pulling, but this model may not be well understood among scientists studying these systems. In this review we address persistent misconceptions regarding the dynamics of proteins in solution that contribute to this lack of understanding, and we clarify the basic physics of entropic pulling with some simple analogies. We hope that increased understanding of the entropic pulling mechanism will inform future efforts to characterize how Hsp70s function as motors, and how they coordinate with their regulatory cochaperones in mechanochemical cycles that transduce the energy of ATP hydrolysis into physical changes in their protein substrates.
Collapse
|
39
|
Fang CT, Kuo HH, Hsu SC, Yih LH. HSP70 is required for the proper assembly of pericentriolar material and function of mitotic centrosomes. Cell Div 2019; 14:4. [PMID: 31110557 PMCID: PMC6511203 DOI: 10.1186/s13008-019-0047-7] [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: 02/26/2019] [Accepted: 04/30/2019] [Indexed: 12/03/2022] Open
Abstract
Background At the onset of mitosis, the centrosome expands and matures, acquiring enhanced activities for microtubule nucleation and assembly of a functional bipolar mitotic spindle. However, the mechanisms that regulate centrosome expansion and maturation are largely unknown. Previously, we demonstrated in an immortalized human cell line CGL2 and cancer cell line HeLa that the inducible form of heat shock protein 70 (HSP70) accumulates at the mitotic centrosome and is required for centrosome maturation and bipolar spindle assembly. Results In this study, we further show that HSP70 accumulated at the spindle pole in a PLK1-dependent manner. HSP70 colocalized with pericentrin (PCNT), CEP215 and γ-tubulin at the spindle pole and was required for the 3D assembly of these three proteins, which supports mitotic centrosome function. Loss of HSP70 disrupted mitotic centrosome structure, reduced pericentriolar material recruitment and induced fragmentation of spindle poles. In addition, HSP70 was necessary for the interaction between PCNT and CEP215 and also facilitated PLK1 accumulation and function at the spindle pole. Furthermore, we found that HSP70 chaperone activity is required for PCNT accumulation at the mitotic centrosome and assembly of mitotic spindles. Conclusion Our current results demonstrate that HSP70 is required for the accurate assembly of the pericentriolar material and proper functioning of mitotic centrosomes.
Collapse
Affiliation(s)
- Chieh-Ting Fang
- 1Department of Life Science, National Taiwan University, Taipei, Taiwan.,2Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115 Taiwan
| | - Hsiao-Hui Kuo
- 2Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115 Taiwan
| | - Shao-Chun Hsu
- 2Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115 Taiwan
| | - Ling-Huei Yih
- 2Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115 Taiwan
| |
Collapse
|
40
|
Mayer MP. Intra-molecular pathways of allosteric control in Hsp70s. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0183. [PMID: 29735737 DOI: 10.1098/rstb.2017.0183] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2018] [Indexed: 12/17/2022] Open
Abstract
The 70 kDa heat-shock protein (Hsp70) is undoubtedly the most versatile of all molecular chaperones. Hsp70 is involved in numerous cellular protein folding processes, accompanying proteins throughout their lifespan from de novo folding at the ribosome to degradation at the proteasome, surveilling protein stability and functionality. Several properties of this ATP-dependent chaperone constitute the molecular basis for this versatility. With its substrate binding domain (SBD), Hsp70 transiently interacts with a short degenerative linear sequence motif found practically in all proteins and, in addition, with more folded protein conformers. Binding to polypeptides is tightly regulated by ATP binding and hydrolysis in the nucleotide binding domain, which is coupled to the SBD by an intricate allosteric mechanism. Hsp70 is regulated by a host of J-cochaperones, which act as targeting factors by regulating the ATPase activity of Hsp70 in synergism with the substrates themselves, and by several families of nucleotide exchange factors. In this review, I focus on the allosteric mechanism, which allows Hsp70s to interact with substrates with ultrahigh affinity through a non-equilibrium mode of action and summarize what mutagenesis and structural studies have taught us about the pathways and mechanics of interdomain communication.This article is part of a discussion meeting issue 'Allostery and molecular machines'.
Collapse
Affiliation(s)
- Matthias P Mayer
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH-Alliance, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| |
Collapse
|
41
|
Regulation of Human Hsc70 ATPase and Chaperone Activities by Apg2: Role of the Acidic Subdomain. J Mol Biol 2018; 431:444-461. [PMID: 30521813 DOI: 10.1016/j.jmb.2018.11.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/30/2018] [Accepted: 11/26/2018] [Indexed: 12/28/2022]
Abstract
Protein aggregate reactivation in metazoans is accomplished by the combined activity of Hsp70, Hsp40 and Hsp110 chaperones. Hsp110s support the refolding of aggregated polypeptides acting as specialized nucleotide exchange factors of Hsp70. We have studied how Apg2, one of the three human Hsp110s, regulates the activity of Hsc70 (HspA8), the constitutive Hsp70 in our cells. Apg2 shows a biphasic behavior: at low concentration, it stimulates the ATPase cycle of Hsc70, binding of the chaperone to protein aggregates and the refolding activity of the system, while it inhibits these three processes at high concentration. When the acidic subdomain of Apg2, a characteristic sequence present in the substrate binding domain of all Hsp110s, is deleted, the detrimental effects occur at lower concentration and are more pronounced, which concurs with an increase in the affinity of the Apg2 mutant for Hsc70. Our data support a mechanism in which Apg2 arrests the chaperone cycle through an interaction with Hsc70(ATP) that might lead to premature ATP dissociation before hydrolysis. In this line, the acidic subdomain might serve as a conformational switch to support dissociation of the Hsc70:Apg2 complex.
Collapse
|
42
|
Batista FA, Dores-Silva PR, Borges JC. Molecular Chaperones Involved in Protein Recovery from Aggregates are Present in Protozoa Causative of Malaria and Leishmaniasis. CURR PROTEOMICS 2018. [DOI: 10.2174/1570164615666180626123823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Molecular chaperones have several critical functions in protein metabolism. Among them,
some are involved in processes that culminate in the extraction of entangled polypeptides from protein
aggregates, releasing unfolded structures prone to be refolded or directed to degradation. This action
avoids the effect of toxic aggregates on cells and tissues. Molecular chaperones belonging to the
Hsp100 family are widely distributed from unicellular and sessile organisms up to fungi and plants,
exerting key functions related to the reduction of the effects caused by different forms of stress. The
Hsp100 proteins belong to the AAA+ (ATPases Associated with diverse cellular Activities) family and
form multichaperone systems with Hsp70 and small Hsp chaperones families. However, Hsp100 are
absent in metazoan, where protein disaggregation action is performed by a system involving the Hsp70
family, including Hsp110 and J-protein co-chaperones. Here, the structural and functional aspects of
these protein disaggregation systems will be reviewed and discussed in the perspective of the Hsp100
system absent in the metazoan kingdom. This feature focuses on Hsp100 as a hot spot for drug discovery
against human infectious diseases such as leishmaniasis and malaria, as Hsp100 is critical for microorganisms.
The current data available for Hsp100 in Leishmania spp. and Plasmodium spp. are also
reviewed.
Collapse
Affiliation(s)
- Fernanda A.H. Batista
- Instituto de Quimica de Sao Carlos, Universidade de Sao Paulo, Sao Carlos, SP, Brazil
| | - Paulo R. Dores-Silva
- Instituto de Quimica de Sao Carlos, Universidade de Sao Paulo, Sao Carlos, SP, Brazil
| | - Júlio C. Borges
- Instituto de Quimica de Sao Carlos, Universidade de Sao Paulo, Sao Carlos, SP, Brazil
| |
Collapse
|
43
|
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: 169] [Impact Index Per Article: 24.1] [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
|
44
|
Allosteric landscapes of eukaryotic cytoplasmic Hsp70s are shaped by evolutionary tuning of key interfaces. Proc Natl Acad Sci U S A 2018; 115:11970-11975. [PMID: 30397123 DOI: 10.1073/pnas.1811105115] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The 70-kDa heat shock proteins (Hsp70s) are molecular chaperones that perform a wide range of critical cellular functions. They assist in the folding of newly synthesized proteins, facilitate assembly of specific protein complexes, shepherd proteins across membranes, and prevent protein misfolding and aggregation. Hsp70s perform these functions by a conserved mechanism that relies on allosteric cycles of nucleotide-modulated binding and release of client proteins. Current models for Hsp70 allostery have come from extensive study of the bacterial Hsp70, DnaK. Extending our understanding to eukaryotic Hsp70s is extremely important not only in providing a likely common mechanistic framework but also because of their central roles in cellular physiology. In this study, we examined the allosteric behaviors of the eukaryotic cytoplasmic Hsp70s, HspA1 and Hsc70, and found significant differences from that of DnaK. We found that HspA1 and Hsc70 favor a state in which the nucleotide-binding domain (NBD) and substrate-binding domain (SBD) are intimately docked significantly more as compared to DnaK. Past work established that the NBD-SBD interface and the helical lid-β-SBD interface govern the allosteric landscape of DnaK. Here, we identified sites on these interfaces that differ between eukaryotic cytoplasmic Hsp70s and DnaK. Our mutational analysis has revealed key evolutionary variations that account for the population shifts between the docked and undocked conformations. These results underline the tunability of Hsp70 functions by modulation of allosteric interfaces through evolutionary diversification and also suggest sites where the binding of small-molecule modulators could influence Hsp70 function.
Collapse
|
45
|
Validation of Internal Control Genes for Quantitative Real-Time PCR Gene Expression Analysis in Morchella. Molecules 2018; 23:molecules23092331. [PMID: 30213125 PMCID: PMC6225436 DOI: 10.3390/molecules23092331] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/14/2018] [Accepted: 09/08/2018] [Indexed: 01/07/2023] Open
Abstract
The reliability of qRT-PCR results depend on the stability of reference genes used for normalization, suggesting the necessity of identification of reference genes before gene expression analysis. Morels are edible mushrooms well-known across the world and highly prized by many culinary kitchens. Here, several candidate genes were selected and designed according to the Morchella importuna transcriptome data. The stability of the candidate genes was evaluated with geNorm and NormFinder under three different experimental conditions, and several genes with excellent stability were selected. The extensive adaptability of the selected genes was tested in ten Morchella species. Results from the three experimental conditions revealed that ACT1 and INTF7 were the most prominent genes in Morchella, CYC3 was the most stable gene in different development stages, INTF4/AEF3 were the top-ranked genes across carbon sources, while INTF3/CYC3 pair showed the robust stability for temperature stress treatment. We suggest using ACT1, AEF3, CYC3, INTF3, INTF4 and INTF7 as reference genes for gene expression analysis studies for any of the 10 Morchella strains tested in this study. The stability and practicality of the gene, vacuolar protein sorting (INTF3), vacuolar ATP synthase (INTF4) and14-3-3 protein (INTF7) involving the basic biological processes were validated for the first time as the candidate reference genes for quantitative PCR. Furthermore, the stability of the reference genes was found to vary under the three different experimental conditions, indicating the importance of identifying specific reference genes for particular conditions.
Collapse
|
46
|
Ghosh DK, Roy A, Ranjan A. The ATPase VCP/p97 functions as a disaggregase against toxic Huntingtin-exon1 aggregates. FEBS Lett 2018; 592:2680-2692. [PMID: 30069866 DOI: 10.1002/1873-3468.13213] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/23/2018] [Accepted: 07/27/2018] [Indexed: 11/12/2022]
Abstract
Intracellular protein aggregation is characterized by accumulation of misfolded proteins. Chaperones, degradation machineries, and quality-control mechanisms counteract protein aggregation. In this study, we report that the ATPase valosin-containing protein (VCP/p97) acts as a functional disaggregase that disassembles Huntingtin-exon1 aggregates in vitro and in HeLa cells. The N-terminal part of VCP (Cdc48_N domain) interacts with the N-terminal 17-amino acid region of Huntingtin-exon1. We show that VCP has properties of a disaggregase, since it is capable of reducing preformed protein aggregates and displays increased ATPase activity in the presence of protein aggregates. However, VCP shows high divergence/disparity from other disaggregases. Taken together, our studies show the novel function of VCP/p97 as a disaggregase which detangles protein aggregates to probably channelize their degradation.
Collapse
Affiliation(s)
- Debasish Kumar Ghosh
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad, India.,Graduate Studies, Manipal Academy of Higher Education, Karnataka, India
| | - Ajit Roy
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad, India
| | - Akash Ranjan
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad, India
| |
Collapse
|
47
|
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
|
48
|
Griffith AA, Boutin J, Holmes W. A highly efficient, one-step purification of the Hsp70 chaperone Ssa1. Protein Expr Purif 2018; 152:56-63. [PMID: 30030046 DOI: 10.1016/j.pep.2018.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/13/2018] [Accepted: 07/14/2018] [Indexed: 11/16/2022]
Abstract
Chaperone proteins are required to maintain the overall fold and function of proteins in the cell. As part of the Hsp70 family, Ssa1 acts to maintain cellular proteostasis through a variety of diverse pathways aimed to preserve the native conformation of target proteins, thereby preventing aggregation and future states of cellular toxicity. Studying the structural dynamics of Ssa1 in vitro is essential to determining their precise mechanisms and requires the development of purification methods that result in highly pure chaperones. Current methods of expressing and purifying Ssa1 utilize affinity tagged constructs expressed in Escherichia coli, however, expression in an exogenous source produces proteins that lack post-translational modifications leading to undesired structural and functional effects. Current protocols to purify Ssa1 from Saccharomyces cerevisiae require large amounts of starting material, multiple steps of chromatography, and result in low yield. Our objective was to establish a small-scale purification of Ssa1 expressed from its endogenous source, Saccharomyces cerevisiae, with significant yield and purity. We utilized a protein A affinity tag that was previously used to purify large protein complexes from yeast, combined with magnetic Dynabeads that are conjugated with rabbit immunoglobulin G (IgG). Our results show that we can produce native, highly pure, active Ssa1 via this one-step purification with minimal amounts of starting material, and this Ssa1-protein A fusion does not alter cellular phenotypes. This methodology is a significant improvement in Ssa1 purification and will facilitate future experiments that will elucidate the biochemical and biophysical properties of Hsp70 chaperones.
Collapse
|
49
|
Jores T, Lawatscheck J, Beke V, Franz-Wachtel M, Yunoki K, Fitzgerald JC, Macek B, Endo T, Kalbacher H, Buchner J, Rapaport D. Cytosolic Hsp70 and Hsp40 chaperones enable the biogenesis of mitochondrial β-barrel proteins. J Cell Biol 2018; 217:3091-3108. [PMID: 29930205 PMCID: PMC6122992 DOI: 10.1083/jcb.201712029] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 05/04/2018] [Accepted: 05/31/2018] [Indexed: 11/22/2022] Open
Abstract
Mitochondrial β-barrel proteins are imported from the cytosol into the organelle. Jores et al. provide new insights into the early events of this process by describing an array of cytosolic chaperones and cochaperones that associate with newly synthesized β-barrel proteins and assure their optimal biogenesis. Mitochondrial β-barrel proteins are encoded in the nucleus, translated by cytosolic ribosomes, and then imported into the organelle. Recently, a detailed understanding of the intramitochondrial import pathway of β-barrel proteins was obtained. In contrast, it is still completely unclear how newly synthesized β-barrel proteins reach the mitochondrial surface in an import-competent conformation. In this study, we show that cytosolic Hsp70 chaperones and their Hsp40 cochaperones Ydj1 and Sis1 interact with newly synthesized β-barrel proteins. These interactions are highly relevant for proper biogenesis, as inhibiting the activity of the cytosolic Hsp70, preventing its docking to the mitochondrial receptor Tom70, or depleting both Ydj1 and Sis1 resulted in a significant reduction in the import of such substrates into mitochondria. Further experiments demonstrate that the interactions between β-barrel proteins and Hsp70 chaperones and their importance are conserved also in mammalian cells. Collectively, this study outlines a novel mechanism in the early events of the biogenesis of mitochondrial outer membrane β-barrel proteins.
Collapse
Affiliation(s)
- Tobias Jores
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Jannis Lawatscheck
- Center for Integrated Protein Science, Department Chemie, Technische Universität München, Garching, Germany
| | - Viktor Beke
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Mirita Franz-Wachtel
- Proteome Center Tübingen, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Kaori Yunoki
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Julia C Fitzgerald
- Department of Neurodegenerative Diseases and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Boris Macek
- Proteome Center Tübingen, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Toshiya Endo
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Hubert Kalbacher
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Johannes Buchner
- Center for Integrated Protein Science, Department Chemie, Technische Universität München, Garching, Germany
| | - Doron Rapaport
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| |
Collapse
|
50
|
Wilkening A, Rüb C, Sylvester M, Voos W. Analysis of heat-induced protein aggregation in human mitochondria. J Biol Chem 2018; 293:11537-11552. [PMID: 29895621 DOI: 10.1074/jbc.ra118.002122] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/01/2018] [Indexed: 12/30/2022] Open
Abstract
Proteins in mammalian cells exhibit optimal stability at physiological temperatures, and even small temperature variations may cause unfolding and nonspecific aggregation. Because this process leads to a loss of function of the affected polypeptides and to cytotoxic stress, formation of protein aggregates has been recognized as a major pathogenic factor in human diseases. In this study, we determined the impact of physiological heat stress on mitochondria isolated from HeLa cells. We found that the heat-stressed mitochondria had lower membrane potential and ATP level and exhibited a decreased production of reactive oxygen species. An analysis of the mitochondrial proteome by 2D PAGE showed that the overall solubility of endogenous proteins was only marginally affected by elevated temperatures. However, a small subset of polypeptides exhibited an high sensitivity to heat stress. The mitochondrial translation elongation factor Tu (Tufm), a protein essential for organellar protein biosynthesis, was highly aggregation-prone and lost its solubility already under mild heat-stress conditions. Moreover, mitochondrial translation and the import of cytosolic proteins were defective in the heat-stressed mitochondria. Both types of nascent polypeptides, produced by translation or imported into the mitochondria, exhibited a strong tendency to aggregate in the heat-exposed mitochondria. We propose that a fast and specific inactivation of elongation factors may prevent the accumulation of misfolded nascent polypeptides and may thereby attenuate proteotoxicity under heat stress.
Collapse
Affiliation(s)
- Anne Wilkening
- Institute for Biochemistry and Molecular Biology, Friedrich Wilhelm University, 53115 Bonn, Germany
| | - Cornelia Rüb
- Institute for Biochemistry and Molecular Biology, Friedrich Wilhelm University, 53115 Bonn, Germany
| | - Marc Sylvester
- Institute for Biochemistry and Molecular Biology, Friedrich Wilhelm University, 53115 Bonn, Germany
| | - Wolfgang Voos
- Institute for Biochemistry and Molecular Biology, Friedrich Wilhelm University, 53115 Bonn, Germany.
| |
Collapse
|