1
|
Berkeley R, Plonski AP, Phan TM, Grohe K, Becker L, Wegner S, Herzik MA, Mittal J, Debelouchina GT. Capturing the Conformational Heterogeneity of HSPB1 Chaperone Oligomers at Atomic Resolution. J Am Chem Soc 2025; 147:15181-15194. [PMID: 40146081 PMCID: PMC12063158 DOI: 10.1021/jacs.4c18668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025]
Abstract
Small heat shock proteins (sHSPs), including HSPB1, are essential regulators of cellular proteostasis that interact with unfolded and partially folded proteins to prevent aberrant misfolding and aggregation. These proteins fulfill a similar role in biological condensates, where they interact with intrinsically disordered proteins to modulate their liquid-liquid and liquid-to-solid phase transitions. Characterizing the sHSP structure, dynamics, and client interactions is challenging due to their partially disordered nature, their tendency to form polydisperse oligomers, and their diverse range of clients. In this work, we leverage various biophysical methods, including fast 1H-based magic angle spinning (MAS) NMR spectroscopy, molecular dynamics (MD) simulations, and modeling, to shed new light on the structure and dynamics of HSPB1 oligomers. Using split-intein-mediated segmental labeling, we provide unambiguous evidence that in the oligomer context, the N-terminal domain (NTD) of HSPB1 is rigid and adopts an ensemble of heterogeneous conformations, the α-Crystallin domain (ACD) forms dimers and experiences multiple distinct local environments, while the C-terminal domain (CTD) remains highly dynamic. Our computational models suggest that the NTDs participate in extensive NTD-NTD and NTD-ACD interactions and are sequestered within the oligomer interior. We further demonstrate that HSPB1 higher order oligomers disassemble into smaller oligomeric species in the presence of a client protein and that an accessible NTD is essential for HSPB1 partitioning into condensates and interactions with client proteins. Our integrated approach provides a high-resolution view of the complex oligomeric landscape of HSPB1 and sheds light on the elusive network of interactions that underlies the function of HSPB1 in biological condensates.
Collapse
Affiliation(s)
- Raymond
F. Berkeley
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093, United States
| | - Alexander P. Plonski
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093, United States
| | - Tien M. Phan
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Kristof Grohe
- Bruker
BioSpin GmbH & Co. KG, Ettlingen 76275, Germany
| | - Lukas Becker
- Bruker
BioSpin GmbH & Co. KG, Ettlingen 76275, Germany
| | | | - Mark A. Herzik
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093, United States
| | - Jeetain Mittal
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, United States
- Department
of Chemistry, Texas A&M University, College Station, Texas 77843, United States
- Interdisciplinary
Graduate Program in Genetics and Genomics, Texas A&M University, College
Station, Texas 77843, United States
| | - Galia T. Debelouchina
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
2
|
Racigh V, Rodriguez Sawicki L, Bravo FNE, Fornasari MS. Coevolution in human small Heat Shock Protein 1 is promoted by interactions between the Alpha-Crystallin domain and the disordered regions. PLoS One 2025; 20:e0321163. [PMID: 40323908 PMCID: PMC12052118 DOI: 10.1371/journal.pone.0321163] [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: 12/23/2024] [Accepted: 03/03/2025] [Indexed: 05/07/2025] Open
Abstract
Human small Heat Shock Protein 1 (HSPB1) belongs to the Small Heat Shock Protein (sHSP) superfamily, a group of ATP-independent molecular chaperones essential for cellular stress responses and protein quality control. These proteins share a conserved domain organization, with a structured Alpha-Crystallin domain (ACD) flanked by disordered N-terminal and C-terminal regions (NTR and CTR). While the prevailing evolutionary hypothesis for the sHSP family suggests that the disordered regions evolved independently and at a faster rate than the ACD, this study provides, for the first time, evidence of coevolution between these regions in human HSPB1, introducing new insights into the evolutionary mechanisms that sustain critical regulatory interactions. By integrating evolutionary and structural approaches, we estimated evolutionary rates per region and position, analyzed the composition of key interacting motifs, and employed structural modeling with AlphaFold 2 to assess the prevalence of these interactions. Our findings reveal that while the disordered regions globally evolve faster than the ACD, specific motifs involved in regulatory interactions exhibit lower-than-average evolutionary rates, reflecting evolutionary constraints imposed by their functional importance. This coevolutionary mechanism may also extend to other small Heat Shock Proteins featuring interacting motifs in the NTR, CTR, or both, offering a new perspective for studying their molecular evolution. Furthermore, the analysis presented in this work could be applied to assess coevolution in other proteins with intrinsically disordered regions.
Collapse
Affiliation(s)
- Vanesa Racigh
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Argentina
| | - Luciana Rodriguez Sawicki
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Argentina
- Instituto de Investigaciones Bioquímicas de La Plata, CONICET-UNLP, Facultad de Ciencias Médicas, La Plata, Argentina
| | | | | |
Collapse
|
3
|
Gu J, He Y, He C, Zhang Q, Huang Q, Bai S, Wang R, You Q, Wang L. Advances in the structures, mechanisms and targeting of molecular chaperones. Signal Transduct Target Ther 2025; 10:84. [PMID: 40069202 PMCID: PMC11897415 DOI: 10.1038/s41392-025-02166-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 03/15/2025] Open
Abstract
Molecular chaperones, a class of complex client regulatory systems, play significant roles in the prevention of protein misfolding and abnormal aggregation, the modulation of protein homeostasis, and the protection of cells from damage under constantly changing environmental conditions. As the understanding of the biological mechanisms of molecular chaperones has increased, their link with the occurrence and progression of disease has suggested that these proteins are promising targets for therapeutic intervention, drawing intensive interest. Here, we review recent advances in determining the structures of molecular chaperones and heat shock protein 90 (HSP90) chaperone system complexes. We also describe the features of molecular chaperones and shed light on the complicated regulatory mechanism that operates through interactions with various co-chaperones in molecular chaperone cycles. In addition, how molecular chaperones affect diseases by regulating pathogenic proteins has been thoroughly analyzed. Furthermore, we focus on molecular chaperones to systematically discuss recent clinical advances and various drug design strategies in the preclinical stage. Recent studies have identified a variety of novel regulatory strategies targeting molecular chaperone systems with compounds that act through different mechanisms from those of traditional inhibitors. Therefore, as more novel design strategies are developed, targeting molecular chaperones will significantly contribute to the discovery of new potential drugs.
Collapse
Affiliation(s)
- Jinying Gu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yanyi He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chenxi He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qifei Huang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shangjun Bai
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.
- Jiangsu Provincial TCM Engineering Technology Research Center of Highly Efficient Drug Delivery Systems (DDSs), Nanjing, China.
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China.
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, China.
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
4
|
Sluzala ZB, Hamati A, Fort PE. Key Role of Phosphorylation in Small Heat Shock Protein Regulation via Oligomeric Disaggregation and Functional Activation. Cells 2025; 14:127. [PMID: 39851555 PMCID: PMC11764305 DOI: 10.3390/cells14020127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/08/2025] [Accepted: 01/16/2025] [Indexed: 01/26/2025] Open
Abstract
Heat shock proteins (HSPs) are essential molecular chaperones that protect cells by aiding in protein folding and preventing aggregation under stress conditions. Small heat shock proteins (sHSPs), which include members from HSPB1 to HSPB10, are particularly important for cellular stress responses. These proteins share a conserved α-crystallin domain (ACD) critical for their chaperone function, with flexible N- and C-terminal extensions that facilitate oligomer formation. Phosphorylation, a key post-translational modification (PTM), plays a dynamic role in regulating sHSP structure, oligomeric state, stability, and chaperone function. Unlike other PTMs such as deamidation, oxidation, and glycation-which are often linked to protein destabilization-phosphorylation generally induces structural transitions that enhance sHSP activity. Specifically, phosphorylation promotes the disaggregation of sHSP oligomers into smaller, more active complexes, thereby increasing their efficiency. This disaggregation mechanism is crucial for protecting cells from stress-induced damage, including apoptosis, inflammation, and other forms of cellular dysfunction. This review explores the role of phosphorylation in modulating the function of sHSPs, particularly HSPB1, HSPB4, and HSPB5, and discusses how these modifications influence their protective functions in cellular stress responses.
Collapse
Affiliation(s)
- Zachary B. Sluzala
- Department of Ophthalmology & Visual Sciences, The University of Michigan, Ann Arbor, MI 48109, USA; (Z.B.S.); (A.H.)
| | - Angelina Hamati
- Department of Ophthalmology & Visual Sciences, The University of Michigan, Ann Arbor, MI 48109, USA; (Z.B.S.); (A.H.)
| | - Patrice E. Fort
- Department of Ophthalmology & Visual Sciences, The University of Michigan, Ann Arbor, MI 48109, USA; (Z.B.S.); (A.H.)
- Department of Molecular & Integrative Physiology, The University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
5
|
Peters C, Haslbeck M, Buchner J. Catchers of folding gone awry: a tale of small heat shock proteins. Trends Biochem Sci 2024; 49:1063-1078. [PMID: 39271417 DOI: 10.1016/j.tibs.2024.08.003] [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: 02/07/2024] [Revised: 08/01/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024]
Abstract
Small heat shock proteins (sHsps) are an important part of the cellular system maintaining protein homeostasis under physiological and stress conditions. As molecular chaperones, they form complexes with different non-native proteins in an ATP-independent manner. Many sHsps populate ensembles of energetically similar but different-sized oligomers. Regulation of chaperone activity occurs by changing the equilibrium of these ensembles. This makes sHsps a versatile and adaptive system for trapping non-native proteins in complexes, allowing recycling with the help of ATP-dependent chaperones. In this review, we discuss progress in our understanding of the structural principles of sHsp oligomers and their functional principles, as well as their roles in aging and eye lens transparency.
Collapse
Affiliation(s)
- Carsten Peters
- School of Natural Sciences, and Department Bioscience, Center for Functional Protein Assemblies, Technical University Munich, Ernst-Otto-Fischer Str. 8, 85748 Garching, Germany
| | - Martin Haslbeck
- School of Natural Sciences, and Department Bioscience, Center for Functional Protein Assemblies, Technical University Munich, Ernst-Otto-Fischer Str. 8, 85748 Garching, Germany.
| | - Johannes Buchner
- School of Natural Sciences, and Department Bioscience, Center for Functional Protein Assemblies, Technical University Munich, Ernst-Otto-Fischer Str. 8, 85748 Garching, Germany.
| |
Collapse
|
6
|
Felipe Perez R, Mochi G, Khan A, Woodford M. Mitochondrial Chaperone Code: Just warming up. Cell Stress Chaperones 2024; 29:483-496. [PMID: 38763405 PMCID: PMC11153887 DOI: 10.1016/j.cstres.2024.05.002] [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: 04/17/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024] Open
Abstract
More than 99% of the mitochondrial proteome is encoded by the nucleus and requires refolding following import. Therefore, mitochondrial proteins require the coordinated action of molecular chaperones for their folding and activation. Several heat shock protein (Hsp) molecular chaperones, including members of the Hsp27, Hsp40/70, and Hsp90 families, as well as the chaperonin complex Hsp60/10 have an established role in mitochondrial protein import and folding. The "Chaperone Code" describes the regulation of chaperone activity by dynamic post-translational modifications; however, little is known about the post-translational regulation of mitochondrial chaperones. Dissecting the regulation of chaperone function is essential for understanding their differential regulation in pathogenic conditions and the potential development of efficacious therapeutic strategies. Here, we summarize the recent literature on post-translational regulation of mitochondrial chaperones, the consequences for mitochondrial function, and potential implications for disease.
Collapse
Affiliation(s)
- R Felipe Perez
- Department of Urology, Upstate Medical University, Syracuse, NY, USA
| | - Gianna Mochi
- Department of Urology, Upstate Medical University, Syracuse, NY, USA; Department of Biochemistry & Molecular Biology, Upstate Medical University, Syracuse, NY, USA; Upstate Cancer Center, State University of New York, Upstate Medical University, Syracuse, NY, USA
| | - Ariba Khan
- Department of Urology, Upstate Medical University, Syracuse, NY, USA
| | - Mark Woodford
- Department of Urology, Upstate Medical University, Syracuse, NY, USA; Department of Biochemistry & Molecular Biology, Upstate Medical University, Syracuse, NY, USA; Upstate Cancer Center, State University of New York, Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
7
|
Wang N, Liu X, Liu K, Wang K, Zhang H. Homo-oxidized HSPB1 protects H9c2 cells against oxidative stress via activation of KEAP1/NRF2 signaling pathway. iScience 2023; 26:107443. [PMID: 37575200 PMCID: PMC10415933 DOI: 10.1016/j.isci.2023.107443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 05/02/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023] Open
Abstract
Several heat shock proteins are implicated in the endogenous cardioprotective mechanisms, but little is known about the role of heat shock protein beta-1 (HSPB1). This study aims to investigate the oxidation state and role of HSPB1 in cardiomyocytes undergoing oxidative stress and underlying mechanisms. Here, we demonstrate that hydrogen peroxide (H2O2) promotes the homo-oxidation of HSPB1. Cys137 residue of HSPB1 is not only required for it to protect cardiomyocytes against oxidative injury but also modulates its oxidation, phosphorylation at Ser15, and distribution to insoluble cell components after H2O2 treatment. Moreover, Cys137 residue is indispensable for HSPB1 to interact with KEAP1, thus regulating its oxidation and intracellular distribution, subsequently promoting the nuclear translocation of NRF2, and increasing the transcription of GLCM, HMOX1, and TXNRD1. Altogether, these findings provide evidence that Cys137 residue is indispensable for HSPB1 to maintain its redox state and antioxidant activity via activating KEAP1/NRF2 signaling cascade in cardiomyocytes.
Collapse
Affiliation(s)
- Nian Wang
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, China
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, Hunan 410078, P.R. China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan 410078, P.R. China
| | - Xiehong Liu
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, China
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, Hunan 410078, P.R. China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan 410078, P.R. China
| | - Ke Liu
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, China
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, Hunan 410078, P.R. China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan 410078, P.R. China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, China
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, Hunan 410078, P.R. China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan 410078, P.R. China
| | - Huali Zhang
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, China
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, Hunan 410078, P.R. China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan 410078, P.R. China
| |
Collapse
|
8
|
Gu C, Fan X, Yu W. Functional Diversity of Mammalian Small Heat Shock Proteins: A Review. Cells 2023; 12:1947. [PMID: 37566026 PMCID: PMC10417760 DOI: 10.3390/cells12151947] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
The small heat shock proteins (sHSPs), whose molecular weight ranges from 12∼43 kDa, are members of the heat shock protein (HSP) family that are widely found in all organisms. As intracellular stress resistance molecules, sHSPs play an important role in maintaining the homeostasis of the intracellular environment under various stressful conditions. A total of 10 sHSPs have been identified in mammals, sharing conserved α-crystal domains combined with variable N-terminal and C-terminal regions. Unlike large-molecular-weight HSP, sHSPs prevent substrate protein aggregation through an ATP-independent mechanism. In addition to chaperone activity, sHSPs were also shown to suppress apoptosis, ferroptosis, and senescence, promote autophagy, regulate cytoskeletal dynamics, maintain membrane stability, control the direction of cellular differentiation, modulate angiogenesis, and spermatogenesis, as well as attenuate the inflammatory response and reduce oxidative damage. Phosphorylation is the most significant post-translational modification of sHSPs and is usually an indicator of their activation. Furthermore, abnormalities in sHSPs often lead to aggregation of substrate proteins and dysfunction of client proteins, resulting in disease. This paper reviews the various biological functions of sHSPs in mammals, emphasizing the roles of different sHSPs in specific cellular activities. In addition, we discuss the effect of phosphorylation on the function of sHSPs and the association between sHSPs and disease.
Collapse
Affiliation(s)
- Chaoguang Gu
- Institute of Biochemistry, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Xiasha High-Tech Zone No.2 Road, Hangzhou 310018, China;
| | - Xinyi Fan
- Faculty of Arts and Science, University of Toronto, Toronto, ON M5S1A1, Canada;
| | - Wei Yu
- Institute of Biochemistry, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Xiasha High-Tech Zone No.2 Road, Hangzhou 310018, China;
| |
Collapse
|
9
|
Choi SK, Hwang SY, Jeon S, Yoo H, Lee J, Shin JH, Na Y, Kwon Y, Lee YS. Design, synthesis, and biological evaluation of novel HSP27 inhibitors to sensitize lung cancer cells to clinically available anticancer agents. Bioorg Chem 2023; 130:106260. [PMID: 36410114 DOI: 10.1016/j.bioorg.2022.106260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
Abstract
Expression of heat shock protein (HSP) correlates with the oncogenic status of malignant cells and plays an important role in tumorigenesis. HSP27 is constitutively expressed at specific stages of cancer development, and several clinical trials have reported correlations between HSP27 expression and tumor progression, metastasis, and chemoresistance in various types of cancer cells. These findings indicate that HSP27 is a major drug target, particularly in chemo-resistant cancers. As part of our ongoing efforts to improve the previously identified J2, a HSP27 cross-linker, we, in this study, report the identification of NK16 as a novel inducer of abnormal HSP27 dimers that did not affect the expression of HSP90 in an NCI-H460 lung cancer cell model. When NCI-H460 cells were treated with NK16 in combination with the anticancer drug cisplatin or paclitaxel, cleavage of PARP and caspase-3 was increased compared to administration of cisplatin or paclitaxel alone. Similar results were obtained in an NCI-H460-xenografted mouse model, in which tumor growth was suppressed more by co-administration of NK16 and paclitaxel than by paclitaxel alone. We propose NK16 as a meaningful strategy to improve the anticancer efficacy of cisplatin and paclitaxel.
Collapse
Affiliation(s)
- Seul-Ki Choi
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Soo-Yeon Hwang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seulgi Jeon
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hawon Yoo
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Joohyun Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jae-Ho Shin
- College of Pharmacy, CHA University, Pocheon 11160, Republic of Korea
| | - Younghwa Na
- College of Pharmacy, CHA University, Pocheon 11160, Republic of Korea.
| | - Youngjoo Kwon
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Yun-Sil Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
10
|
Liu WP, Li P, Zhan X, Qu LH, Xiong T, Hou FX, Wang JK, Wei N, Liu FQ. Identification of molecular subtypes of coronary artery disease based on ferroptosis- and necroptosis-related genes. Front Genet 2022; 13:870222. [PMID: 36204316 PMCID: PMC9531137 DOI: 10.3389/fgene.2022.870222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Aim: Coronary artery disease (CAD) is a heterogeneous disorder with high morbidity, mortality, and healthcare costs, representing a major burden on public health. Here, we aimed to improve our understanding of the genetic drivers of ferroptosis and necroptosis and the clustering of gene expression in CAD in order to develop novel personalized therapies to slow disease progression.Methods: CAD datasets were obtained from the Gene Expression Omnibus. The identification of ferroptosis- and necroptosis-related differentially expressed genes (DEGs) and the consensus clustering method including the classification algorithm used km and distance used spearman were performed to differentiate individuals with CAD into two clusters (cluster A and cluster B) based expression matrix of DEGs. Next, we identified four subgroup-specific genes of significant difference between cluster A and B and again divided individuals with CAD into gene cluster A and gene cluster B with same methods. Additionally, we compared differences in clinical information between the subtypes separately. Finally, principal component analysis algorithms were constructed to calculate the cluster-specific gene score for each sample for quantification of the two clusters.Results: In total, 25 ferroptosis- and necroptosis-related DEGs were screened. The genes in cluster A were mostly related to the neutrophil pathway, whereas those in cluster B were mostly related to the B-cell receptor signaling pathway. Moreover, the subgroup-specific gene scores and CAD indices were higher in cluster A and gene cluster A than in cluster B and gene cluster B. We also identified and validated two genes showing upregulation between clusters A and B in a validation dataset.Conclusion: High expression of CBS and TLR4 was related to more severe disease in patients with CAD, whereas LONP1 and HSPB1 expression was associated with delayed CAD progression. The identification of genetic subgroups of patients with CAD may improve clinician knowledge of disease pathogenesis and facilitate the development of methods for disease diagnosis, classification, and prognosis.
Collapse
Affiliation(s)
- Wen-Pan Liu
- Cardiovascular Department, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
- Department of Cardiothoracic Surgery, The First People’s Hospital of Kunming City and Ganmei Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Peng Li
- Department of Surgery, Nanzhao County People’s Hospital, Nanyang, Henan, China
| | - Xu Zhan
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lai-Hao Qu
- Department of Cardiothoracic Surgery, Yan’an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Tao Xiong
- Department of Cardiothoracic Surgery, Yan’an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Fang-Xia Hou
- Cardiovascular Department, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Jun-Kui Wang
- Cardiovascular Department, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
| | - Na Wei
- Cardiovascular Department, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
- *Correspondence: Na Wei, ; Fu-Qiang Liu,
| | - Fu-Qiang Liu
- Cardiovascular Department, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi, China
- *Correspondence: Na Wei, ; Fu-Qiang Liu,
| |
Collapse
|
11
|
Selig EE, Lynn RJ, Zlatic CO, Mok YF, Ecroyd H, Gooley PR, Griffin MDW. The Monomeric α-Crystallin Domain of the Small Heat-shock Proteins αB-crystallin and Hsp27 Binds Amyloid Fibril Ends. J Mol Biol 2022; 434:167711. [PMID: 35777462 DOI: 10.1016/j.jmb.2022.167711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/05/2022] [Accepted: 06/25/2022] [Indexed: 11/25/2022]
Abstract
Small heat-shock proteins (sHSPs) are ubiquitously expressed molecular chaperones present in all kingdoms of life that inhibit protein misfolding and aggregation. Despite their importance in proteostasis, the structure-function relationships of sHSPs remain elusive. Human sHSPs are characterised by a central, highly conserved α-crystallin domain (ACD) and variable-length N- and C-terminal regions. The ACD forms antiparallel homodimers via an extended β-strand, creating a shared β-sheet at the dimer interface. The N- and C-terminal regions mediate formation of higher order oligomers that are thought to act as storage forms for chaperone-active dimers. We investigated the interactions of the ACD of two human sHSPs, αB-crystallin (αB-C) and Hsp27, with apolipoprotein C-II amyloid fibrils using analytical ultracentrifugation and nuclear magnetic resonance spectroscopy. The ACD was found to interact transiently with amyloid fibrils to inhibit fibril elongation and naturally occurring fibril end-to-end joining. This interaction was sensitive to the concentration of fibril ends indicating a 'fibril-capping' interaction. Furthermore, resonances arising from the ACD monomer were attenuated to a greater extent than those of the ACD dimer in the presence of fibrils, suggesting that the monomer may bind fibrils. This hypothesis was supported by mutagenesis studies in which disulfide cross-linked ACD dimers formed by both αB-C and Hsp27 were less effective at inhibiting amyloid fibril elongation and fibril end-to-end joining than ACD constructs lacking disulfide cross-linking. Our results indicate that sHSP monomers inhibit amyloid fibril elongation, highlighting the importance of the dynamic oligomeric nature of sHSPs for client binding.
Collapse
Affiliation(s)
- Emily E Selig
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria 3010, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Roberta J Lynn
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria 3010, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Courtney O Zlatic
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria 3010, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Yee-Foong Mok
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria 3010, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Heath Ecroyd
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.
| | - Paul R Gooley
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria 3010, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Michael D W Griffin
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria 3010, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
12
|
Holguin BA, Hildenbrand ZL, Bernal RA. Insights Into the Role of Heat Shock Protein 27 in the Development of Neurodegeneration. Front Mol Neurosci 2022; 15:868089. [PMID: 35431800 PMCID: PMC9005852 DOI: 10.3389/fnmol.2022.868089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/09/2022] [Indexed: 12/11/2022] Open
Abstract
Small heat shock protein 27 is a critically important chaperone, that plays a key role in several essential and varied physiological processes. These include thermotolerance, apoptosis, cytoskeletal dynamics, cell differentiation, protein folding, among others. Despite its relatively small size and intrinsically disordered termini, it forms large and polydisperse oligomers that are in equilibrium with dimers. This equilibrium is driven by transient interactions between the N-terminal region, the α-crystallin domain, and the C-terminal region. The continuous redistribution of binding partners results in a conformationally dynamic protein that allows it to adapt to different functions where substrate capture is required. However, the intrinsic disorder of the amino and carboxy terminal regions and subsequent conformational variability has made structural investigations challenging. Because heat shock protein 27 is critical for so many key cellular functions, it is not surprising that it also has been linked to human disease. Charcot-Marie-Tooth and distal hereditary motor neuropathy are examples of neurodegenerative disorders that arise from single point mutations in heat shock protein 27. The development of possible treatments, however, depends on our understanding of its normal function at the molecular level so we might be able to understand how mutations manifest as disease. This review will summarize recent reports describing investigations into the structurally elusive regions of Hsp27. Recent insights begin to provide the required context to explain the relationship between a mutation and the resulting loss or gain of function that leads to Charcot-Marie Tooth disease and distal hereditary motor neuropathy.
Collapse
|
13
|
Tedesco B, Cristofani R, Ferrari V, Cozzi M, Rusmini P, Casarotto E, Chierichetti M, Mina F, Galbiati M, Piccolella M, Crippa V, Poletti A. Insights on Human Small Heat Shock Proteins and Their Alterations in Diseases. Front Mol Biosci 2022; 9:842149. [PMID: 35281256 PMCID: PMC8913478 DOI: 10.3389/fmolb.2022.842149] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
The family of the human small Heat Shock Proteins (HSPBs) consists of ten members of chaperones (HSPB1-HSPB10), characterized by a low molecular weight and capable of dimerization and oligomerization forming large homo- or hetero-complexes. All HSPBs possess a highly conserved centrally located α-crystallin domain and poorly conserved N- and C-terminal domains. The main feature of HSPBs is to exert cytoprotective functions by preserving proteostasis, assuring the structural maintenance of the cytoskeleton and acting in response to cellular stresses and apoptosis. HSPBs take part in cell homeostasis by acting as holdases, which is the ability to interact with a substrate preventing its aggregation. In addition, HSPBs cooperate in substrates refolding driven by other chaperones or, alternatively, promote substrate routing to degradation. Notably, while some HSPBs are ubiquitously expressed, others show peculiar tissue-specific expression. Cardiac muscle, skeletal muscle and neurons show high expression levels for a wide variety of HSPBs. Indeed, most of the mutations identified in HSPBs are associated to cardiomyopathies, myopathies, and motor neuropathies. Instead, mutations in HSPB4 and HSPB5, which are also expressed in lens, have been associated with cataract. Mutations of HSPBs family members encompass base substitutions, insertions, and deletions, resulting in single amino acid substitutions or in the generation of truncated or elongated proteins. This review will provide an updated overview of disease-related mutations in HSPBs focusing on the structural and biochemical effects of mutations and their functional consequences.
Collapse
Affiliation(s)
- B. Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - R. Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - V. Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Cozzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - P. Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - E. Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - F. Mina
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - V. Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - A. Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
- *Correspondence: A. Poletti,
| |
Collapse
|
14
|
Rada CC, Mejia-Pena H, Grimsey NJ, Cordova IC, Olson J, Wozniak J, Gonzalez DJ, Nizet V, Trejo J. Heat shock protein 27 activity is linked to endothelial barrier recovery after proinflammatory GPCR-induced disruption. Sci Signal 2021; 14:eabc1044. [PMID: 34516752 PMCID: PMC8538426 DOI: 10.1126/scisignal.abc1044] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Vascular inflammation causes endothelial barrier disruption and tissue edema. Several inflammatory mediators act through G protein–coupled receptors (GPCRs), including protease-activated receptor-1 (PAR1), to elicit inflammatory responses. The activation of PAR1 by its ligand thrombin stimulates proinflammatory, p38 mitogen-activated protein kinase (MAPK) signaling that promotes endothelial barrier disruption. Through mass spectrometry phosphoproteomics, we identified heat shock protein 27 (HSP27), which exists as a large oligomer that binds to actin, as a promising candidate for the p38-mediated regulation of barrier integrity. Depletion of HSP27 by siRNA enhanced endothelial cell barrier permeability and slowed recovery after thrombin stimulation. We further showed that two effector kinases of p38 MAPK, MAPKAPK2 (MK2) and MAPKAPK3 (MK3), differentially phosphorylated HSP27 at Ser15, Ser78, and Ser82. Whereas inhibition of thrombin-stimulated p38 activation blocked HSP27 phosphorylation at all three sites, inhibition of MK2 reduced the phosphorylation of only Ser15 and Ser78. Inhibition of both MK2 and MK3 was necessary to attenuate Ser82 phosphorylation. Thrombin-stimulated p38-MK2-MK3 signaling induced HSP27 oligomer disassembly. However, a phosphorylation-deficient mutant of HSP27 exhibited defective oligomer disassembly and altered the dynamics of barrier recovery after thrombin stimulation. Moreover, blocking HSP27 oligomer reassembly with the small-molecule inhibitor J2 enhanced endothelial barrier permeability in vitro and vascular leakage in vivo in response to PAR1 activation. These studies reveal the distinct regulation of HSP27 phosphorylation and function induced by the GPCR-stimulated p38-MK2-MK3 signaling axis that controls the dynamics of endothelial barrier recovery in vitro and vascular leakage in vivo.
Collapse
Affiliation(s)
- Cara C. Rada
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hilda Mejia-Pena
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Neil J. Grimsey
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30682, USA
| | - Isabel Canto Cordova
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joshua Olson
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jacob Wozniak
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - David J. Gonzalez
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Victor Nizet
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - JoAnn Trejo
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
15
|
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: 21] [Impact Index Per Article: 5.3] [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
|
16
|
Liu X, Xiao W, Jiang Y, Zou L, Chen F, Xiao W, Zhang X, Cao Y, Xu L, Zhu Y. Bmal1 Regulates the Redox Rhythm of HSPB1, and Homooxidized HSPB1 Attenuates the Oxidative Stress Injury of Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5542815. [PMID: 34239687 PMCID: PMC8238613 DOI: 10.1155/2021/5542815] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/05/2021] [Accepted: 05/07/2021] [Indexed: 12/20/2022]
Abstract
Oxidative stress is the main cause of acute myocardial infarction (AMI), which is related to the disorder of the regulation of Bmal1 on the redox state. HSPB1 form homologous-oxidized HSPB1 (homooxidized HSPB1) to resist oxidative damage via S-thiolated modification. However, it is still unclarified whether there is an interaction between the circadian clock and HSPB1 in myocardial injury. A total of 118 AMI patients admitted and treated in our hospital from Sep. 2019 to Sep. 2020 were selected to detect the plasma HSPB1 expression and the redox state. We divided the AMI patients into three subgroups: morning-onset AMI (5 : 00 am to 8 : 00 am; Am-subgroup, n = 38), noon-onset AMI (12 : 00 pm to 15 : 00; Pm-subgroup, n = 45), and night-onset AMI (20 : 00 pm to 23 : 00 pm; Eve-subgroup, n = 35) according to the circadian rhythm of onset. The Am-subgroup had remarkably higher cardiac troponin I (cTnI), creatine kinase MB (CK-MB), and B-type natriuretic peptide (BNP) but lower left ventricular ejection fraction (LVEF) than the Pm-subgroup and Eve-subgroup. Patients complicated with cardiogenic shock were significantly higher in the Am-subgroup than in the other two groups. The homooxidized HSPB1 in plasma markedly decreased in the Am-subgroup. The HSPB1C141S mutant accelerated H9c2 cell apoptosis, increased reactive oxygen species (ROS), and decreased reduced-glutathione (GSH) and the ratio of reduced-GSH and GSSG during oxidative stress. Importantly, we found that the redox state of HSPB1 was consistent with the oscillatory rhythm of Bmal1 expression in normal C57B/L mice. The circadian rhythm disorder contributed to decrease Bmal1 and homooxidized HSPB1 in cardiomyocytes of C57BL/6 mice. In addition, Bmal1 and homooxidized HSPB1 decreased in neonatal rat cardiomyocytes exposed to H2O2. Knockdown of Bmal1 led to significant attenuation in homooxidized HSPB1 expression, whereas overexpression of Bmal1 increased homooxidized HSPB1 expression in response to H2O2. Our findings indicated that the homooxidized HSPB1 reduced probably the AMI patients' risk of shock and target organ damage, which was associated with Bmal1 regulating the redox state of HSPB1.
Collapse
Affiliation(s)
- Xiehong Liu
- Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Institute of Emergency Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, China
| | - Wen Xiao
- Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Institute of Emergency Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, China
- Emergency Department, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Yu Jiang
- Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Institute of Emergency Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, China
| | - Lianhong Zou
- Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Institute of Emergency Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, China
| | - Fang Chen
- Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Institute of Emergency Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, China
- Emergency Department, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Weiwei Xiao
- Emergency Department, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Xingwen Zhang
- Emergency Department, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Yan Cao
- Emergency Department, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Lei Xu
- Public Health Clinical Center, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Yimin Zhu
- Hunan Provincial Key Laboratory of Emergency and Critical Care Metabonomics, Institute of Emergency Medicine, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan, China
| |
Collapse
|
17
|
Reid Alderson T, Adriaenssens E, Asselbergh B, Pritišanac I, Van Lent J, Gastall HY, Wälti MA, Louis JM, Timmerman V, Baldwin AJ, LP Benesch J. A weakened interface in the P182L variant of HSP27 associated with severe Charcot-Marie-Tooth neuropathy causes aberrant binding to interacting proteins. EMBO J 2021; 40:e103811. [PMID: 33644875 PMCID: PMC8047445 DOI: 10.15252/embj.2019103811] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/10/2021] [Accepted: 01/14/2021] [Indexed: 01/18/2023] Open
Abstract
HSP27 is a human molecular chaperone that forms large, dynamic oligomers and functions in many aspects of cellular homeostasis. Mutations in HSP27 cause Charcot-Marie-Tooth (CMT) disease, the most common inherited disorder of the peripheral nervous system. A particularly severe form of CMT disease is triggered by the P182L mutation in the highly conserved IxI/V motif of the disordered C-terminal region, which interacts weakly with the structured core domain of HSP27. Here, we observed that the P182L mutation disrupts the chaperone activity and significantly increases the size of HSP27 oligomers formed in vivo, including in motor neurons differentiated from CMT patient-derived stem cells. Using NMR spectroscopy, we determined that the P182L mutation decreases the affinity of the HSP27 IxI/V motif for its own core domain, leaving this binding site more accessible for other IxI/V-containing proteins. We identified multiple IxI/V-bearing proteins that bind with higher affinity to the P182L variant due to the increased availability of the IxI/V-binding site. Our results provide a mechanistic basis for the impact of the P182L mutation on HSP27 and suggest that the IxI/V motif plays an important, regulatory role in modulating protein-protein interactions.
Collapse
Affiliation(s)
- T Reid Alderson
- Chemistry Research LaboratoryUniversity of OxfordOxfordUK
- Laboratory of Chemical PhysicsNational Institutes of HealthBethesdaMDUSA
- Present address:
Department of BiochemistryTorontoONCanada
| | - Elias Adriaenssens
- Peripheral Neuropathy Research GroupDepartment of Biomedical SciencesInstitute Born BungeUniversity of AntwerpAntwerpenBelgium
| | - Bob Asselbergh
- Neuromics Support FacilityVIB Center for Molecular NeurologyVIBAntwerpenBelgium
- Neuromics Support Facility, Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
| | - Iva Pritišanac
- Molecular Medicine ProgramThe Hospital for Sick ChildrenTorontoONCanada
| | - Jonas Van Lent
- Peripheral Neuropathy Research GroupDepartment of Biomedical SciencesInstitute Born BungeUniversity of AntwerpAntwerpenBelgium
| | | | - Marielle A Wälti
- Laboratory of Chemical PhysicsNational Institutes of HealthBethesdaMDUSA
| | - John M Louis
- Laboratory of Chemical PhysicsNational Institutes of HealthBethesdaMDUSA
| | - Vincent Timmerman
- Peripheral Neuropathy Research GroupDepartment of Biomedical SciencesInstitute Born BungeUniversity of AntwerpAntwerpenBelgium
| | | | | |
Collapse
|
18
|
Makley LN, Johnson OT, Ghanakota P, Rauch JN, Osborn D, Wu TS, Cierpicki T, Carlson HA, Gestwicki JE. Chemical validation of a druggable site on Hsp27/HSPB1 using in silico solvent mapping and biophysical methods. Bioorg Med Chem 2021; 34:115990. [PMID: 33549906 PMCID: PMC7968374 DOI: 10.1016/j.bmc.2020.115990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 11/26/2022]
Abstract
Destabilizing mutations in small heat shock proteins (sHsps) are linked to multiple diseases; however, sHsps are conformationally dynamic, lack enzymatic function and have no endogenous chemical ligands. These factors render sHsps as classically "undruggable" targets and make it particularly challenging to identify molecules that might bind and stabilize them. To explore potential solutions, we designed a multi-pronged screening workflow involving a combination of computational and biophysical ligand-discovery platforms. Using the core domain of the sHsp family member Hsp27/HSPB1 (Hsp27c) as a target, we applied mixed solvent molecular dynamics (MixMD) to predict three possible binding sites, which we confirmed using NMR-based solvent mapping. Using this knowledge, we then used NMR spectroscopy to carry out a fragment-based drug discovery (FBDD) screen, ultimately identifying two fragments that bind to one of these sites. A medicinal chemistry effort improved the affinity of one fragment by ~50-fold (16 µM), while maintaining good ligand efficiency (~0.32 kcal/mol/non-hydrogen atom). Finally, we found that binding to this site partially restored the stability of disease-associated Hsp27 variants, in a redox-dependent manner. Together, these experiments suggest a new and unexpected binding site on Hsp27, which might be exploited to build chemical probes.
Collapse
Affiliation(s)
- Leah N Makley
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California at San Francisco, San Francisco, CA 94158, United States; Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, United States
| | - Oleta T Johnson
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California at San Francisco, San Francisco, CA 94158, United States
| | - Phani Ghanakota
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, United States
| | - Jennifer N Rauch
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California at San Francisco, San Francisco, CA 94158, United States
| | - Delaney Osborn
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California at San Francisco, San Francisco, CA 94158, United States
| | - Taia S Wu
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California at San Francisco, San Francisco, CA 94158, United States
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Heather A Carlson
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, United States
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California at San Francisco, San Francisco, CA 94158, United States.
| |
Collapse
|
19
|
Proteinaceous Transformers: Structural and Functional Variability of Human sHsps. Int J Mol Sci 2020; 21:ijms21155448. [PMID: 32751672 PMCID: PMC7432308 DOI: 10.3390/ijms21155448] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 02/04/2023] Open
Abstract
The proteostasis network allows organisms to support and regulate the life cycle of proteins. Especially regarding stress, molecular chaperones represent the main players within this network. Small heat shock proteins (sHsps) are a diverse family of ATP-independent molecular chaperones acting as the first line of defense in many stress situations. Thereby, the promiscuous interaction of sHsps with substrate proteins results in complexes from which the substrates can be refolded by ATP-dependent chaperones. Particularly in vertebrates, sHsps are linked to a broad variety of diseases and are needed to maintain the refractive index of the eye lens. A striking key characteristic of sHsps is their existence in ensembles of oligomers with varying numbers of subunits. The respective dynamics of these molecules allow the exchange of subunits and the formation of hetero-oligomers. Additionally, these dynamics are closely linked to the chaperone activity of sHsps. In current models a shift in the equilibrium of the sHsp ensemble allows regulation of the chaperone activity, whereby smaller oligomers are commonly the more active species. Different triggers reversibly change the oligomer equilibrium and regulate the activity of sHsps. However, a finite availability of high-resolution structures of sHsps still limits a detailed mechanistic understanding of their dynamics and the correlating recognition of substrate proteins. Here we summarize recent advances in understanding the structural and functional relationships of human sHsps with a focus on the eye-lens αA- and αB-crystallins.
Collapse
|
20
|
Alderson TR, Ying J, Bax A, Benesch JLP, Baldwin AJ. Conditional Disorder in Small Heat-shock Proteins. J Mol Biol 2020; 432:3033-3049. [PMID: 32081587 PMCID: PMC7245567 DOI: 10.1016/j.jmb.2020.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/27/2020] [Accepted: 02/09/2020] [Indexed: 12/31/2022]
Abstract
Small heat-shock proteins (sHSPs) are molecular chaperones that respond to cellular stresses to combat protein aggregation. HSP27 is a critical human sHSP that forms large, dynamic oligomers whose quaternary structures and chaperone activities depend on environmental factors. Upon exposure to cellular stresses, such as heat shock or acidosis, HSP27 oligomers can dissociate into dimers and monomers, which leads to significantly enhanced chaperone activity. The structured core of the protein, the α-crystallin domain (ACD), forms dimers and can prevent the aggregation of substrate proteins to a similar degree as the full-length protein. When the ACD dimer dissociates into monomers, it partially unfolds and exhibits enhanced activity. Here, we used solution-state NMR spectroscopy to characterize the structure and dynamics of the HSP27 ACD monomer. Web show that the monomer is stabilized at low pH and that its backbone chemical shifts, 15N relaxation rates, and 1H-15N residual dipolar couplings suggest structural changes and rapid motions in the region responsible for dimerization. By analyzing the solvent accessible and buried surface areas of sHSP structures in the context of a database of dimers that are known to dissociate into disordered monomers, we predict that ACD dimers from sHSPs across all kingdoms of life may partially unfold upon dissociation. We propose a general model in which conditional disorder-the partial unfolding of ACDs upon monomerization-is a common mechanism for sHSP activity.
Collapse
Affiliation(s)
- T Reid Alderson
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK; Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jinfa Ying
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ad Bax
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Justin L P Benesch
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK.
| | - Andrew J Baldwin
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK.
| |
Collapse
|
21
|
Hsp27 chaperones FUS phase separation under the modulation of stress-induced phosphorylation. Nat Struct Mol Biol 2020; 27:363-372. [PMID: 32231288 DOI: 10.1038/s41594-020-0399-3] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 02/24/2020] [Indexed: 12/13/2022]
Abstract
Protein phase separation drives the assembly of membraneless organelles, but little is known about how these membraneless organelles are maintained in a metastable liquid- or gel-like phase rather than proceeding to solid aggregation. Here, we find that human small heat-shock protein 27 (Hsp27), a canonical chaperone that localizes to stress granules (SGs), prevents FUS from undergoing liquid-liquid phase separation (LLPS) via weak interactions with the FUS low complexity (LC) domain. Remarkably, stress-induced phosphorylation of Hsp27 alters its activity, leading Hsp27 to partition with FUS LC to preserve the liquid phase against amyloid fibril formation. NMR spectroscopy demonstrates that Hsp27 uses distinct structural mechanisms for both functions. Our work reveals a fine-tuned regulation of Hsp27 for chaperoning FUS into either a polydispersed state or a LLPS state and suggests an essential role for Hsp27 in stabilizing the dynamic phase of stress granules.
Collapse
|
22
|
Neuromuscular Diseases Due to Chaperone Mutations: A Review and Some New Results. Int J Mol Sci 2020; 21:ijms21041409. [PMID: 32093037 PMCID: PMC7073051 DOI: 10.3390/ijms21041409] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle and the nervous system depend on efficient protein quality control, and they express chaperones and cochaperones at high levels to maintain protein homeostasis. Mutations in many of these proteins cause neuromuscular diseases, myopathies, and hereditary motor and sensorimotor neuropathies. In this review, we cover mutations in DNAJB6, DNAJB2, αB-crystallin (CRYAB, HSPB5), HSPB1, HSPB3, HSPB8, and BAG3, and discuss the molecular mechanisms by which they cause neuromuscular disease. In addition, previously unpublished results are presented, showing downstream effects of BAG3 p.P209L on DNAJB6 turnover and localization.
Collapse
|
23
|
Baughman HER, Pham THT, Adams CS, Nath A, Klevit RE. Release of a disordered domain enhances HspB1 chaperone activity toward tau. Proc Natl Acad Sci U S A 2020; 117:2923-2929. [PMID: 31974309 PMCID: PMC7022203 DOI: 10.1073/pnas.1915099117] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Small heat shock proteins (sHSPs) are a class of ATP-independent molecular chaperones that play vital roles in maintaining protein solubility and preventing aberrant protein aggregation. They form highly dynamic, polydisperse oligomeric ensembles and contain long intrinsically disordered regions. Experimental challenges posed by these properties have greatly impeded our understanding of sHSP structure and mechanism of action. Here we characterize interactions between the human sHSP HspB1 (Hsp27) and microtubule-associated protein tau, which is implicated in multiple dementias, including Alzheimer's disease. We show that tau binds both to a well-known binding groove within the structured alpha-crystallin domain (ACD) and to sites within the enigmatic, disordered N-terminal region (NTR) of HspB1. However, only interactions involving the NTR lead to productive chaperone activity, whereas ACD binding is uncorrelated with chaperone function. The tau-binding groove in the ACD also binds short hydrophobic regions within HspB1 itself, and HspB1 mutations that disrupt these intrinsic ACD-NTR interactions greatly enhance chaperone activity toward tau. This leads to a mechanism in which the release of the disordered NTR from a binding groove on the ACD enhances chaperone activity toward tau. The study advances understanding of the mechanisms by which sHSPs achieve their chaperone activity against amyloid-forming clients and how cells defend against pathological tau aggregation. Furthermore, the resulting mechanistic model points to ways in which sHSP chaperone activity may be increased, either by native factors within the cell or by therapeutic intervention.
Collapse
Affiliation(s)
- Hannah E R Baughman
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA 98195
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA 98195
| | - Thanh-Hau T Pham
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA 98195
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA 98195
| | - Chloe S Adams
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA 98195
| | - Abhinav Nath
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA 98195
| | - Rachel E Klevit
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA 98195;
| |
Collapse
|
24
|
Clouser AF, Baughman HER, Basanta B, Guttman M, Nath A, Klevit RE. Interplay of disordered and ordered regions of a human small heat shock protein yields an ensemble of 'quasi-ordered' states. eLife 2019; 8:e50259. [PMID: 31573509 PMCID: PMC6791718 DOI: 10.7554/elife.50259] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022] Open
Abstract
Small heat shock proteins (sHSPs) are nature's 'first responders' to cellular stress, interacting with affected proteins to prevent their aggregation. Little is known about sHSP structure beyond its structured α-crystallin domain (ACD), which is flanked by disordered regions. In the human sHSP HSPB1, the disordered N-terminal region (NTR) represents nearly 50% of the sequence. Here, we present a hybrid approach involving NMR, hydrogen-deuterium exchange mass spectrometry, and modeling to provide the first residue-level characterization of the NTR. The results support a model in which multiple grooves on the ACD interact with specific NTR regions, creating an ensemble of 'quasi-ordered' NTR states that can give rise to the known heterogeneity and plasticity of HSPB1. Phosphorylation-dependent interactions inform a mechanism by which HSPB1 is activated under stress conditions. Additionally, we examine the effects of disease-associated NTR mutations on HSPB1 structure and dynamics, leveraging our emerging structural insights.
Collapse
Affiliation(s)
- Amanda F Clouser
- Department of BiochemistryUniversity of WashingtonSeattleUnited States
| | - Hannah ER Baughman
- Department of BiochemistryUniversity of WashingtonSeattleUnited States
- Department of Medicinal ChemistryUniversity of WashingtonSeattleUnited States
| | - Benjamin Basanta
- Department of BiochemistryUniversity of WashingtonSeattleUnited States
| | - Miklos Guttman
- Department of Medicinal ChemistryUniversity of WashingtonSeattleUnited States
| | - Abhinav Nath
- Department of Medicinal ChemistryUniversity of WashingtonSeattleUnited States
| | - Rachel E Klevit
- Department of BiochemistryUniversity of WashingtonSeattleUnited States
| |
Collapse
|
25
|
Janowska MK, Baughman HER, Woods CN, Klevit RE. Mechanisms of Small Heat Shock Proteins. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a034025. [PMID: 30833458 DOI: 10.1101/cshperspect.a034025] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Small heat shock proteins (sHSPs) are ATP-independent chaperones that delay formation of harmful protein aggregates. sHSPs' role in protein homeostasis has been appreciated for decades, but their mechanisms of action remain poorly understood. This gap in understanding is largely a consequence of sHSP properties that make them recalcitrant to detailed study. Multiple stress-associated conditions including pH acidosis, oxidation, and unusual availability of metal ions, as well as reversible stress-induced phosphorylation can modulate sHSP chaperone activity. Investigations of sHSPs reveal that sHSPs can engage in transient or long-lived interactions with client proteins depending on solution conditions and sHSP or client identity. Recent advances in the field highlight both the diversity of function within the sHSP family and the exquisite sensitivity of individual sHSPs to cellular and experimental conditions. Here, we will present and highlight current understanding, recent progress, and future challenges.
Collapse
Affiliation(s)
- Maria K Janowska
- Department of Biochemistry, University of Washington, Seattle, Washington 98195
| | - Hannah E R Baughman
- Department of Biochemistry, University of Washington, Seattle, Washington 98195
| | - Christopher N Woods
- Department of Biochemistry, University of Washington, Seattle, Washington 98195
| | - Rachel E Klevit
- Department of Biochemistry, University of Washington, Seattle, Washington 98195
| |
Collapse
|
26
|
Chiu MH, Shi C, Rosin M, Batulan Z, O'Brien ER. Biophysical analyses and functional implications of the interaction between Heat Shock Protein 27 and antibodies to HSP27. Biochim Biophys Acta Gen Subj 2019; 1863:1536-1546. [PMID: 31136785 DOI: 10.1016/j.bbagen.2019.05.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 03/28/2019] [Accepted: 05/23/2019] [Indexed: 10/26/2022]
Abstract
Heat Shock Protein 27 (HSP27) is a small molecular chaperone that reduces the development of atherosclerosis by lowering plasma cholesterol levels as well as inflammation. Human studies show an inverse correlation between atherosclerotic burden and HSP27 expression, and are supported by murine models in which augmenting HSP27 levels curbs experimental atherogenesis. Natural HSP27 auto-antibodies (AAb) are found in human plasma, however their role in modulating the athero-protective effects of HSP27 is unknown. The purpose of this study is to characterize the biophysical interaction between human recombinant HSP27 and AAb. A validated polyclonal anti-HSP27 IgG antibody (PAb) was used to mimic natural AAb. Homology modeling and secondary structure prediction tools facilitated the design of HSP27 truncation and phosphorylation mutants. Secondary structural changes were identified using Circular Dichroism (CD) and Dynamic Light Scattering (DLS). Similar to prior structural investigations of HSP27, there was a predominance of α-helical content in the N-terminal truncation and dephosphorylation ("AA") mutants. The α-crystallin domain (ACD) predominantly consists of β-strands, with the addition of the N-terminal increasing helical content and the C-terminal maintaining β structure. With increasing ratios of PAb to HSP27 β structure abundance and particle size increased, with a similar trend observed with the N-terminus, C-terminus and ACD peptides but an opposite trend with the phosphorylation peptides. Taken together, these studies provide insights into the interaction of HSP27 and its AAb that ultimately may aid in optimizing the design of HSP27 peptidomimetics with anti-atherogenic potential.
Collapse
Affiliation(s)
- Michael H Chiu
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Chunhua Shi
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Matthew Rosin
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Zarah Batulan
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Edward R O'Brien
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada.
| |
Collapse
|
27
|
Collier MP, Alderson TR, de Villiers CP, Nicholls D, Gastall HY, Allison TM, Degiacomi MT, Jiang H, Mlynek G, Fürst DO, van der Ven PFM, Djinovic-Carugo K, Baldwin AJ, Watkins H, Gehmlich K, Benesch JLP. HspB1 phosphorylation regulates its intramolecular dynamics and mechanosensitive molecular chaperone interaction with filamin C. SCIENCE ADVANCES 2019; 5:eaav8421. [PMID: 31131323 PMCID: PMC6530996 DOI: 10.1126/sciadv.aav8421] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 04/16/2019] [Indexed: 05/13/2023]
Abstract
Mechanical force-induced conformational changes in proteins underpin a variety of physiological functions, typified in muscle contractile machinery. Mutations in the actin-binding protein filamin C (FLNC) are linked to musculoskeletal pathologies characterized by altered biomechanical properties and sometimes aggregates. HspB1, an abundant molecular chaperone, is prevalent in striated muscle where it is phosphorylated in response to cues including mechanical stress. We report the interaction and up-regulation of both proteins in three mouse models of biomechanical stress, with HspB1 being phosphorylated and FLNC being localized to load-bearing sites. We show how phosphorylation leads to increased exposure of the residues surrounding the HspB1 phosphosite, facilitating their binding to a compact multidomain region of FLNC proposed to have mechanosensing functions. Steered unfolding of FLNC reveals that its extension trajectory is modulated by the phosphorylated region of HspB1. This may represent a posttranslationally regulated chaperone-client protection mechanism targeting over-extension during mechanical stress.
Collapse
Affiliation(s)
- Miranda P. Collier
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, UK
| | - T. Reid Alderson
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, UK
| | - Carin P. de Villiers
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Headington, Oxford OX3 9DU, UK
| | - Daisy Nicholls
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, UK
| | - Heidi Y. Gastall
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, UK
| | - Timothy M. Allison
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, UK
- Biomolecular Interaction Centre and School of Physical and Chemical Sciences, University of Canterbury, Christchurch 8140, New Zealand
| | - Matteo T. Degiacomi
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, UK
- Department of Chemistry, Durham University, South Road, Durham DH1 3LE, UK
| | - He Jiang
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Headington, Oxford OX3 9DU, UK
| | - Georg Mlynek
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Dieter O. Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Peter F. M. van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Kristina Djinovic-Carugo
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
- Department of Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, 1000 Ljubljana, Slovenia
| | - Andrew J. Baldwin
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Headington, Oxford OX3 9DU, UK
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Headington, Oxford OX3 9DU, UK
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Corresponding author. (J.L.P.B.); (K.G.)
| | - Justin L. P. Benesch
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, UK
- Corresponding author. (J.L.P.B.); (K.G.)
| |
Collapse
|
28
|
Local unfolding of the HSP27 monomer regulates chaperone activity. Nat Commun 2019; 10:1068. [PMID: 30842409 PMCID: PMC6403371 DOI: 10.1038/s41467-019-08557-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 12/20/2018] [Indexed: 12/21/2022] Open
Abstract
The small heat-shock protein HSP27 is a redox-sensitive molecular chaperone that is expressed throughout the human body. Here, we describe redox-induced changes to the structure, dynamics, and function of HSP27 and its conserved α-crystallin domain (ACD). While HSP27 assembles into oligomers, we show that the monomers formed upon reduction are highly active chaperones in vitro, but are susceptible to self-aggregation. By using relaxation dispersion and high-pressure nuclear magnetic resonance (NMR) spectroscopy, we observe that the pair of β-strands that mediate dimerisation partially unfold in the monomer. We note that numerous HSP27 mutations associated with inherited neuropathies cluster to this dynamic region. High levels of sequence conservation in ACDs from mammalian sHSPs suggest that the exposed, disordered interface present in free monomers or oligomeric subunits may be a general, functional feature of sHSPs. The small heat-shock protein HSP27 occurs predominantly in oligomeric forms, which makes its structural characterisation challenging. Here the authors employ CPMG and high-pressure NMR with native mass spectrometry and biophysical assays to show that the active monomeric form of HSP27 is substantially disordered and highly chaperone-active.
Collapse
|
29
|
Gliniewicz EF, Chambers KM, De Leon ER, Sibai D, Campbell HC, McMenimen KA. Chaperone-like activity of the N-terminal region of a human small heat shock protein and chaperone-functionalized nanoparticles. Proteins 2019; 87:401-415. [PMID: 30684363 DOI: 10.1002/prot.25662] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/03/2019] [Accepted: 01/22/2019] [Indexed: 11/10/2022]
Abstract
Small heat shock proteins (sHsps) are molecular chaperones employed to interact with a diverse range of substrates as the first line of defense against cellular protein aggregation. The N-terminal region (NTR) is implicated in defining features of sHsps; notably in their ability to form dynamic and polydisperse oligomers, and chaperone activity. The physiological relevance of oligomerization and chemical-scale mode(s) of chaperone function remain undefined. We present novel chemical tools to investigate chaperone activity and substrate specificity of human HspB1 (B1NTR), through isolation of B1NTR and development of peptide-conjugated gold nanoparticles (AuNPs). We demonstrate that B1NTR exhibits chaperone capacity for some substrates, determined by anti-aggregation assays and size-exclusion chromatography. The importance of protein dynamics and multivalency on chaperone capacity was investigated using B1NTR-conjugated AuNPs, which exhibit concentration-dependent chaperone activity for some substrates. Our results implicate sHsp NTRs in chaperone activity, and demonstrate the therapeutic potential of sHsp-AuNPs in rescuing aberrant protein aggregation.
Collapse
Affiliation(s)
- Emily F Gliniewicz
- Department of Chemistry, Mount Holyoke College, South Hadley, Massachusetts
| | - Kelly M Chambers
- Department of Chemistry, Mount Holyoke College, South Hadley, Massachusetts
| | | | - Diana Sibai
- Department of Chemistry, Mount Holyoke College, South Hadley, Massachusetts
| | - Helen C Campbell
- Department of Chemistry, Mount Holyoke College, South Hadley, Massachusetts
| | | |
Collapse
|
30
|
Competing protein-protein interactions regulate binding of Hsp27 to its client protein tau. Nat Commun 2018; 9:4563. [PMID: 30385828 PMCID: PMC6212398 DOI: 10.1038/s41467-018-07012-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/08/2018] [Indexed: 01/17/2023] Open
Abstract
Small heat shock proteins (sHSPs) are a class of oligomeric molecular chaperones that limit protein aggregation. However, it is often not clear where sHSPs bind on their client proteins or how these protein-protein interactions (PPIs) are regulated. Here, we map the PPIs between human Hsp27 and the microtubule-associated protein tau (MAPT/tau). We find that Hsp27 selectively recognizes two aggregation-prone regions of tau, using the conserved β4-β8 cleft of its alpha-crystallin domain. The β4-β8 region is also the site of Hsp27–Hsp27 interactions, suggesting that competitive PPIs may be an important regulatory paradigm. Indeed, we find that each of the individual PPIs are relatively weak and that competition for shared sites seems to control both client binding and Hsp27 oligomerization. These findings highlight the importance of multiple, competitive PPIs in the function of Hsp27 and suggest that the β4-β8 groove acts as a tunable sensor for clients. Small heat shock proteins (sHSPs) limit the aggregation of proteins, such as tau. Here the authors show that Hsp27 recognizes two aggregation-prone regions of tau and that this interaction competes with Hsp27 oligomerization.
Collapse
|
31
|
Haslbeck M, Weinkauf S, Buchner J. Small heat shock proteins: Simplicity meets complexity. J Biol Chem 2018; 294:2121-2132. [PMID: 30385502 DOI: 10.1074/jbc.rev118.002809] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Small heat shock proteins (sHsps) are a ubiquitous and ancient family of ATP-independent molecular chaperones. A key characteristic of sHsps is that they exist in ensembles of iso-energetic oligomeric species differing in size. This property arises from a unique mode of assembly involving several parts of the subunits in a flexible manner. Current evidence suggests that smaller oligomers are more active chaperones. Thus, a shift in the equilibrium of the sHsp ensemble allows regulating the chaperone activity. Different mechanisms have been identified that reversibly change the oligomer equilibrium. The promiscuous interaction with non-native proteins generates complexes that can form aggregate-like structures from which native proteins are restored by ATP-dependent chaperones such as Hsp70 family members. In recent years, this basic paradigm has been expanded, and new roles and new cofactors, as well as variations in structure and regulation of sHsps, have emerged.
Collapse
Affiliation(s)
- Martin Haslbeck
- From the Department of Chemie and Center for Integrated Protein Science, Technische Universität München, Lichtenbergstrasse 4, 85 748 Garching, Germany
| | - Sevil Weinkauf
- From the Department of Chemie and Center for Integrated Protein Science, Technische Universität München, Lichtenbergstrasse 4, 85 748 Garching, Germany
| | - Johannes Buchner
- From the Department of Chemie and Center for Integrated Protein Science, Technische Universität München, Lichtenbergstrasse 4, 85 748 Garching, Germany
| |
Collapse
|
32
|
Freilich R, Arhar T, Abrams JL, Gestwicki JE. Protein-Protein Interactions in the Molecular Chaperone Network. Acc Chem Res 2018; 51:940-949. [PMID: 29613769 DOI: 10.1021/acs.accounts.8b00036] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Molecular chaperones play a central role in protein homeostasis (a.k.a. proteostasis) by balancing protein folding, quality control, and turnover. To perform these diverse tasks, chaperones need the malleability to bind nearly any "client" protein and the fidelity to detect when it is misfolded. Remarkably, these activities are carried out by only ∼180 dedicated chaperones in humans. How do a relatively small number of chaperones maintain cellular and organismal proteostasis for an entire proteome? Furthermore, once a chaperone binds a client, how does it "decide" what to do with it? One clue comes from observations that individual chaperones engage in protein-protein interactions (PPIs)-both with each other and with their clients. These physical links coordinate multiple chaperones into organized, functional complexes and facilitate the "handoff" of clients between them. PPIs also link chaperones and their clients to other cellular pathways, such as those that mediate trafficking (e.g., cytoskeleton) and degradation (e.g., proteasome). The PPIs of the chaperone network have a wide range of affinity values (nanomolar to micromolar) and involve many distinct types of domain modules, such as J domains, zinc fingers, and tetratricopeptide repeats. Many of these motifs have the same binding surfaces on shared partners, such that members of one chaperone class often compete for the same interactions. Somehow, this collection of PPIs draws together chaperone families and creates multiprotein subnetworks that are able to make the "decisions" of protein quality control. The key to understanding chaperone-mediated proteostasis might be to understand how PPIs are regulated. This Account will discuss the efforts of our group and others to map, measure, and chemically perturb the PPIs within the molecular chaperone network. Structural biology methods, including X-ray crystallography, NMR spectroscopy, and electron microscopy, have all played important roles in visualizing the chaperone PPIs. Guided by these efforts and -omics approaches to measure PPIs, new advances in high-throughput chemical screening that are specially designed to account for the challenges of this system have emerged. Indeed, chemical biology has played a particularly important role in this effort, as molecules that either promote or inhibit specific PPIs have proven to be invaluable research probes in cells and animals. In addition, these molecules have provided leads for the potential treatment of protein misfolding diseases. One of the major products of this research field has been the identification of putative PPI drug targets within the chaperone network, which might be used to change chaperone "decisions" and rebalance proteostasis.
Collapse
Affiliation(s)
- Rebecca Freilich
- Department of Pharmaceutical Chemistry and The Institute for Neurodegenerative Disease, University of California—San Francisco, San Francisco, California 94158, United States
| | - Taylor Arhar
- Department of Pharmaceutical Chemistry and The Institute for Neurodegenerative Disease, University of California—San Francisco, San Francisco, California 94158, United States
| | - Jennifer L. Abrams
- Department of Pharmaceutical Chemistry and The Institute for Neurodegenerative Disease, University of California—San Francisco, San Francisco, California 94158, United States
| | - Jason E. Gestwicki
- Department of Pharmaceutical Chemistry and The Institute for Neurodegenerative Disease, University of California—San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
33
|
He L, Desai JX, Gao J, Hazeltine LB, Lian Z, Calley JN, Frye CC. Elucidating the Impact of CHO Cell Culture Media on Tryptophan Oxidation of a Monoclonal Antibody Through Gene Expression Analyses. Biotechnol J 2018. [DOI: 10.1002/biot.201700254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Luhong He
- Bioprocess Research and Development, Eli Lilly and Company; Indianapolis 46285 IN USA
| | - Jairav X. Desai
- Informatics Capabilities − Research IT, Eli Lilly and Company; Indianapolis 46285 IN USA
| | - Jinxin Gao
- Statistics- Discovery/Development, Eli Lilly and Company; Indianapolis 46285 IN USA
| | - Laurie B. Hazeltine
- Bioprocess Research and Development, Eli Lilly and Company; Indianapolis 46285 IN USA
| | - Zhirui Lian
- Bioprocess Research and Development, Eli Lilly and Company; Indianapolis 46285 IN USA
| | - John N. Calley
- Tailored Therapeutics Bioinformatics, Eli Lilly and Company; Indianapolis 46285 IN USA
| | - Christopher C. Frye
- Bioprocess Research and Development, Eli Lilly and Company; Indianapolis 46285 IN USA
| |
Collapse
|
34
|
Arbach H, Butler C, McMenimen KA. Chaperone activity of human small heat shock protein-GST fusion proteins. Cell Stress Chaperones 2017; 22:503-515. [PMID: 28130664 PMCID: PMC5465028 DOI: 10.1007/s12192-017-0764-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/09/2017] [Accepted: 01/11/2017] [Indexed: 12/17/2022] Open
Abstract
Small heat shock proteins (sHsps) are a ubiquitous part of the machinery that maintains cellular protein homeostasis by acting as molecular chaperones. sHsps bind to and prevent the aggregation of partially folded substrate proteins in an ATP-independent manner. sHsps are dynamic, forming an ensemble of structures from dimers to large oligomers through concentration-dependent equilibrium dissociation. Based on structural studies and mutagenesis experiments, it is proposed that the dimer is the smallest active chaperone unit, while larger oligomers may act as storage depots for sHsps or play additional roles in chaperone function. The complexity and dynamic nature of their structural organization has made elucidation of their chaperone function challenging. HspB1 and HspB5 are two canonical human sHsps that vary in sequence and are expressed in a wide variety of tissues. In order to determine the role of the dimer in chaperone activity, glutathione-S-transferase (GST) was genetically linked as a fusion protein to the N-terminus regions of both HspB1 and HspB5 (also known as Hsp27 and αB-crystallin, respectively) proteins in order to constrain oligomer formation of HspB1 and HspB5, by using GST, since it readily forms a dimeric structure. We monitored the chaperone activity of these fusion proteins, which suggest they primarily form dimers and monomers and function as active molecular chaperones. Furthermore, the two different fusion proteins exhibit different chaperone activity for two model substrate proteins, citrate synthase (CS) and malate dehydrogenase (MDH). GST-HspB1 prevents more aggregation of MDH compared to GST-HspB5 and wild type HspB1. However, when CS is the substrate, both GST-HspB1 and GST-HspB5 are equally effective chaperones. Furthermore, wild type proteins do not display equal activity toward the substrates, suggesting that each sHsp exhibits different substrate specificity. Thus, substrate specificity, as described here for full-length GST fusion proteins with MDH and CS, is modulated by both sHsp oligomeric conformation and by variations of sHsp sequences.
Collapse
Affiliation(s)
- Hannah Arbach
- Department of Chemistry, Mount Holyoke College, 50 College Street, South Hadley, MA, 01075, USA
| | - Caley Butler
- Department of Chemistry, Mount Holyoke College, 50 College Street, South Hadley, MA, 01075, USA
| | - Kathryn A McMenimen
- Department of Chemistry, Mount Holyoke College, 50 College Street, South Hadley, MA, 01075, USA.
| |
Collapse
|
35
|
Clouser AF, Klevit RE. pH-dependent structural modulation is conserved in the human small heat shock protein HSBP1. Cell Stress Chaperones 2017; 22:569-575. [PMID: 28332148 PMCID: PMC5465033 DOI: 10.1007/s12192-017-0783-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/23/2017] [Accepted: 02/24/2017] [Indexed: 10/19/2022] Open
Abstract
The holdase activity and oligomeric propensity of human small heat shock proteins (sHSPs) are regulated by environmental factors. However, atomic-level details are lacking for the mechanisms by which stressors alter sHSP responses. We previously demonstrated that regulation of HSPB5 is mediated by a single conserved histidine over a physiologically relevant pH range of 6.5-7.5. Here, we demonstrate that HSPB1 responds to pH via a similar mechanism through pH-dependent structural changes that are induced via protonation of the structurally analogous histidine. Results presented here show that acquisition of a positive charge, either by protonation of His124 or its substitution by lysine, reduces the stability of the dimer interface of the α-crystallin domain, increases oligomeric size, and modestly increases chaperone activity. Our results suggest a conserved mechanism of pH-dependent structural regulation among the human sHSPs that possess the conserved histidine, although the functional consequences of the structural modulations vary for different sHSPs.
Collapse
Affiliation(s)
- Amanda F Clouser
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Rachel E Klevit
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
36
|
Alderson TR, Benesch JLP, Baldwin AJ. Proline isomerization in the C-terminal region of HSP27. Cell Stress Chaperones 2017; 22:639-651. [PMID: 28547731 PMCID: PMC5465039 DOI: 10.1007/s12192-017-0791-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 12/12/2022] Open
Abstract
In mammals, small heat-shock proteins (sHSPs) typically assemble into interconverting, polydisperse oligomers. The dynamic exchange of sHSP oligomers is regulated, at least in part, by molecular interactions between the α-crystallin domain and the C-terminal region (CTR). Here we report solution-state nuclear magnetic resonance (NMR) spectroscopy investigations of the conformation and dynamics of the disordered and flexible CTR of human HSP27, a systemically expressed sHSP. We observed multiple NMR signals for residues in the vicinity of proline 194, and we determined that, while all observed forms are highly disordered, the extra resonances arise from cis-trans peptidyl-prolyl isomerization about the G193-P194 peptide bond. The cis-P194 state is populated to near 15% at physiological temperatures, and, although both cis- and trans-P194 forms of the CTR are flexible and dynamic, both states show a residual but differing tendency to adopt β-strand conformations. In NMR spectra of an isolated CTR peptide, we observed similar evidence for isomerization involving proline 182, found within the IPI/V motif. Collectively, these data indicate a potential role for cis-trans proline isomerization in regulating the oligomerization of sHSPs.
Collapse
Affiliation(s)
- T Reid Alderson
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Justin L P Benesch
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK.
| | - Andrew J Baldwin
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK.
| |
Collapse
|
37
|
Arrigo AP. Mammalian HspB1 (Hsp27) is a molecular sensor linked to the physiology and environment of the cell. Cell Stress Chaperones 2017; 22:517-529. [PMID: 28144778 PMCID: PMC5465029 DOI: 10.1007/s12192-017-0765-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/11/2017] [Accepted: 01/14/2017] [Indexed: 12/11/2022] Open
Abstract
Constitutively expressed small heat shock protein HspB1 regulates many fundamental cellular processes and plays major roles in many human pathological diseases. In that regard, this chaperone has a huge number of apparently unrelated functions that appear linked to its ability to recognize many client polypeptides that are subsequently modified in their activity and/or half-life. A major parameter to understand how HspB1 is dedicated to interact with particular clients in defined cellular conditions relates to its complex oligomerization and phosphorylation properties. Indeed, HspB1 structural organization displays dynamic and complex rearrangements in response to changes in the cellular environment or when the cell physiology is modified. These structural modifications probably reflect the formation of structural platforms aimed at recognizing specific client polypeptides. Here, I have reviewed data from the literature and re-analyzed my own studies to describe and discuss these fascinating changes in HspB1 structural organization.
Collapse
Affiliation(s)
- André-Patrick Arrigo
- Apoptosis, Cancer and Development Laboratory, Lyon Cancer Research Center, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, 28 rue Laennec, Lyon, 69008, France.
| |
Collapse
|
38
|
Batulan Z, Pulakazhi Venu VK, Li Y, Koumbadinga G, Alvarez-Olmedo DG, Shi C, O'Brien ER. Extracellular Release and Signaling by Heat Shock Protein 27: Role in Modifying Vascular Inflammation. Front Immunol 2016; 7:285. [PMID: 27507972 PMCID: PMC4960997 DOI: 10.3389/fimmu.2016.00285] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/14/2016] [Indexed: 12/31/2022] Open
Abstract
Heat shock protein 27 (HSP27) is traditionally viewed as an intracellular chaperone protein with anti-apoptotic properties. However, recent data indicate that a number of heat shock proteins, including HSP27, are also found in the extracellular space where they may signal via membrane receptors to alter gene transcription and cellular function. Therefore, there is increasing interest in better understanding how HSP27 is released from cells, its levels and composition in the extracellular space, and the cognate cell membrane receptors involved in effecting cell signaling. In this paper, the knowledge to date, as well as some emerging paradigms about the extracellular function of HSP27 is presented. Of particular interest is the role of HSP27 in attenuating atherogenesis by modifying lipid uptake and inflammation in the plaque. Moreover, the abundance of HSP27 in serum is an emerging new biomarker for ischemic events. Finally, HSP27 replacement therapy may represent a novel therapeutic opportunity for chronic inflammatory disorders, such as atherosclerosis.
Collapse
Affiliation(s)
- Zarah Batulan
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Vivek Krishna Pulakazhi Venu
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Yumei Li
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Geremy Koumbadinga
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Daiana Gisela Alvarez-Olmedo
- Oncology Laboratory, Institute for Experimental Medicine and Biology of Cuyo (IMBECU), CCT CONICET , Mendoza , Argentina
| | - Chunhua Shi
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| | - Edward R O'Brien
- Vascular Biology Laboratory, Health Research Innovation Centre, Libin Cardiovascular Institute of Alberta, University of Calgary Cumming School of Medicine , Calgary, AB , Canada
| |
Collapse
|