1
|
Arefnezhd R, Chahardehi AM, Asadi A, Shadravan MM, Shariati A, Rezaee A, Radmanesh M, Nazarian M, Helfi M, Soleimani Meigoli MS, Motedayyen H, Rezaei-Tazangi F, Tavakoli MR. The function of chaperones in the radioresistance of glioblastoma: a new insight into the current knowledge. Brain Tumor Pathol 2025:10.1007/s10014-025-00501-7. [PMID: 40259161 DOI: 10.1007/s10014-025-00501-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/27/2025] [Indexed: 04/23/2025]
Abstract
Radiotherapy remains a cornerstone of brain tumor treatment; however, its effectiveness is frequently undermined by the development of radioresistance. This review highlights the pivotal role of molecular chaperones in promoting radioresistance and explores the potential to increase radioresistance in brain cancers, particularly glioblastoma (GBM). Among chaperones, heat shock proteins (HSPs), such as HSP70 and HSP90, have been identified as key contributors to radioresistance, acting through mechanisms that include the maintenance of protein homeostasis, enhancement of DNA repair processes, and protection of cancer stem cells. Specifically, HSP70 and HSP90 are crucial in stabilizing oncogenic proteins and preventing apoptosis, thus enabling tumor survival during radiotherapy. Also, HSP27 and GRP78 are involved in the radioresistance of brain tumors mainly by suppressing cell death and enhancing tumor stem cell propagation. Emerging evidence also suggests that targeting these chaperones, in combination with radiotherapy, can enhance tumor radiosensitivity, offering promising therapeutic strategies. Recent studies have revealed novel aspects of chaperone-mediated autophagy and interaction with non-coding RNAs, providing deeper insights into the molecular mechanisms underlying radioresistance. This review also addresses the potential of combining chaperone-targeted therapies, such as HSP90 inhibitors, with radiotherapy to overcome resistance. Ultimately, understanding these mechanisms may pave the way for innovative clinical applications and personalized therapeutic approaches in brain tumor treatment.
Collapse
Affiliation(s)
- Reza Arefnezhd
- Coenzyme R Research Institute, Tehran, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Amirmasoud Asadi
- Department of Medical Physics, School of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | | | | | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrsa Radmanesh
- Faculty of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Mohammadreza Nazarian
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Helfi
- Department of Medical Physics, School of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | | | - Hossein Motedayyen
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran.
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| | - Marziye Ranjbar Tavakoli
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
2
|
Xu SM, Liu XZ, Wang L, Huang WH, Hu YT, Chen SB, Huang ZS, Huang SL. Synergistic anticancer activity of HSP70 and HSF1 inhibitors in colorectal cancer cells: A new strategy for combination therapy. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167630. [PMID: 39675530 DOI: 10.1016/j.bbadis.2024.167630] [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: 08/02/2024] [Revised: 11/30/2024] [Accepted: 12/13/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND The heat shock response (HSR) is a highly conserved mechanism that maintains intracellular homeostasis in response to various environmental and physiological stresses. Heat shock proteins (HSPs), particularly HSP70, play a pivotal role in this process as molecular chaperones. Although HSP70 inhibitors have demonstrated anti-cancer activity, their therapeutic potential has been limited by the negative feedback mechanism between HSP70 and heat shock factor 1 (HSF1). The combination of HSP70 inhibitors with HSF1 inhibitors has been proposed to overcome this limitation and enhance anti-cancer effects. METHODS We combined HSP70 inhibitors (VER-155008 and YK-5) with an HSF1 inhibitor (DTHIB) in CRC cells and evaluated their effects on cell survival, apoptosis, and protein homeostasis. RESULTS Strong synergistic effects were observed (combination index <0.5, ZIP score > 10) with the combination treatment, leading to decreased cell survival and increased apoptosis in CRC cells. Mechanistic studies revealed that HSP70 inhibitors activated the phosphorylation of HSF1, inducing HSP70 expression, and that the combination therapy resulted in more pronounced HSR inhibition and protein homeostasis disturbances. CONCLUSION The combination therapy of HSP 70 and HSF 1 inhibitors showed significant synergistic antitumor activity. GENERAL SIGNIFICANCE Combining HSP70 and HSF1 inhibitors may be a promising anti-cancer strategy, offering a potential solution to overcome the negative feedback mechanism and enhance anti-cancer effects.
Collapse
Affiliation(s)
- Shu-Min Xu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Xing-Zi Liu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Lu Wang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Wei-Hao Huang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Yu-Tao Hu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Shuo-Bin Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhi-Shu Huang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China.
| | - Shi-Liang Huang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
3
|
Garcia AM, Davis AK, Martinez-Ramos C, Morishima Y, Lau M, Xu E, Sunil A, Zhang H, Alt A, Lieberman AP, Osawa Y. High-throughput screening identifies a novel small-molecule modulator of Hsp70 that selectively enhances ubiquitination and degradation of misfolded neuronal NO synthase. Mol Pharmacol 2025; 107:100008. [PMID: 40023517 PMCID: PMC11934283 DOI: 10.1016/j.molpha.2024.100008] [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: 07/23/2024] [Accepted: 11/19/2024] [Indexed: 03/04/2025] Open
Abstract
The Hsp90 and Hsp70 chaperones act as a protein quality control system for several hundred client proteins, including many implicated in neurodegenerative disorders. Hsp90 and Hsp70 are widely thought to be important drug targets. Although many structurally distinct compounds have been developed to target Hsp90, relatively few are known to target Hsp70 and even fewer have been tested in protein quality control systems. To address this, we describe a high-throughput thermal shift-based screen to find compounds that bind and stabilize Hsp70 and then employ assays with misfolded forms of a well-established client protein, neuronal NO synthase (nNOS), to identify compounds that enhance ubiquitination of client proteins. The ubiquitination assay employed a quantitative ELISA method to measure Hsp70:CHIP-dependent ubiquitination of heme-deficient nNOS, which is a model of a misfolded client, in reaction mixtures containing purified E1, E2, Hsp70, CHIP, and ubiquitin. We screened 44,447 molecules from the Maybridge and ChemDiv libraries and found one compound, protein folding disease compound 15 (PFD-15), that enhanced in vitro nNOS ubiquitination with an EC50 of approximately 8 μM. PFD-15 was tested in human embryonic kidney 293 cells stably transfected with a C331A nNOS, a mutation that makes nNOS a preferred client protein for ubiquitination. In this model, PFD-15 decreased steady-state levels of C331A nNOS, but not the wild-type nNOS, in a time- and concentration-dependent manner by a process attenuated by lactacystin, an inhibitor to the proteasome. PFD-15 appears to enhance binding of Hsp70 and CHIP to client proteins without interference of protein quality control mechanisms, enabling the selective clearance of misfolded proteins. SIGNIFICANCE STATEMENT: There are few treatment options for neurodegenerative diseases, which are widely thought to be caused by formation of toxic misfolded proteins. One novel approach is to enhance the Hsp90/Hsp70 protein quality control machinery to remove these misfolded proteins. Targeting Hsp70 may have advantages over targeting Hsp90, but fewer compounds targeting Hsp70 have been developed relative to those for Hsp90. The current study provides a novel approach to enhance the number of compounds targeting the Hsp70's role in protein quality control.
Collapse
Affiliation(s)
- Anthony M Garcia
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Amanda K Davis
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| | | | - Yoshihiro Morishima
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Miranda Lau
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Emily Xu
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Arya Sunil
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Haoming Zhang
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Andrew Alt
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan; Center for Chemical Genomics, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Yoichi Osawa
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
4
|
Pokhrel S, Devi S, Gestwicki JE. Chaperone-dependent and chaperone-independent functions of carboxylate clamp tetratricopeptide repeat (CC-TPR) proteins. Trends Biochem Sci 2025; 50:121-133. [PMID: 39706778 DOI: 10.1016/j.tibs.2024.11.004] [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: 08/09/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 12/23/2024]
Abstract
The molecular chaperones HSP70 and HSP90 play key roles in proteostasis by acting as adapters; they bind to a 'client' protein, often with the assistance of cochaperones, and then recruit additional cochaperones that promote specific fates (e.g., folding or degradation). One family of cochaperones contains a region termed the tetratricopeptide repeat with carboxylate clamps (CC-TPRs) domain. These domains bind to an EEVD motif at the C-termini of cytoplasmic HSP70 and HSP90 proteins, bringing them into proximity to chaperone-bound clients. It has recently become clear that CC-TPR proteins also bind to 'EEVD-like' motifs in non-chaperone proteins, circumventing the need for HSP70s or HSP90s. We provide an overview of the chaperone-dependent and -independent roles of CC-TPR proteins and discuss how, together, they shape proteostasis.
Collapse
Affiliation(s)
- Saugat Pokhrel
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Diseases, University of California San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Shweta Devi
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Diseases, University of California San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Diseases, University of California San Francisco (UCSF), San Francisco, CA 94158, USA.
| |
Collapse
|
5
|
Jami KM, Farb DC, Osumi KM, Shafer CC, Criscione S, Murray DT. Small heat shock protein HSPB8 interacts with a pre-fibrillar TDP43 low complexity domain species to delay fibril formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635368. [PMID: 39974920 PMCID: PMC11838303 DOI: 10.1101/2025.01.28.635368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The loss of cellular proteostasis through aberrant stress granule formation is implicated in neurodegenerative diseases. Stress granules are formed by biomolecular condensation involving protein-protein and protein-RNA interactions. These assemblies are protective, but can rigidify, leading to amyloid-like fibril formation, a hallmark of the disease pathology. Key proteins dictating stress granule formation and disassembly, such as TDP43, contain low-complexity (LC) domains that drive fibril formation. HSPB8, a small heat shock protein, plays a critical role modulating stress granule fluidity, preventing aggregation and promoting degradation of misfolded proteins. We examined the interaction between HSPB8 and the TDP43 LC using thioflavin T (ThT) and fluorescence polarization (FP) aggregation assays, fluorescence microscopy and photobleaching experiments, and crosslinking mass spectrometry (XL-MS). Our results indicate that HSPB8 delays TDP43 LC aggregation through domain-specific interactions with fibril nucleating species, without affecting fibril elongation rates. These findings provide mechanistic insight into how ATP-independent chaperones mediate LC domain aggregation and provide a basis for investigating how the TDP43 LC subverts chaperone activity in neurodegenerative disease.
Collapse
Affiliation(s)
- Khaled M. Jami
- Department of Chemistry, University of California, Davis, California, United States of America
| | - Daniel C. Farb
- Department of Chemistry, University of California, Davis, California, United States of America
| | - Kayla M. Osumi
- Department of Chemistry, University of California, Davis, California, United States of America
| | - Catelynn C. Shafer
- Department of Chemistry, University of California, Davis, California, United States of America
| | - Sophie Criscione
- Department of Chemistry, University of California, Davis, California, United States of America
| | - Dylan T. Murray
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, United States of America
| |
Collapse
|
6
|
Braxton JR, Shao H, Tse E, Gestwicki JE, Southworth DR. Asymmetric apical domain states of mitochondrial Hsp60 coordinate substrate engagement and chaperonin assembly. Nat Struct Mol Biol 2024; 31:1848-1858. [PMID: 38951622 PMCID: PMC11638070 DOI: 10.1038/s41594-024-01352-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/07/2024] [Indexed: 07/03/2024]
Abstract
The mitochondrial chaperonin, mitochondrial heat shock protein 60 (mtHsp60), promotes the folding of newly imported and transiently misfolded proteins in the mitochondrial matrix, assisted by its co-chaperone mtHsp10. Despite its essential role in mitochondrial proteostasis, structural insights into how this chaperonin progresses through its ATP-dependent client folding cycle are not clear. Here, we determined cryo-EM structures of a hyperstable disease-associated human mtHsp60 mutant, V72I. Client density is identified in three distinct states, revealing interactions with the mtHsp60 apical domains and C termini that coordinate client positioning in the folding chamber. We further identify an asymmetric arrangement of the apical domains in the ATP state, in which an alternating up/down configuration positions interaction surfaces for simultaneous recruitment of mtHsp10 and client retention. Client is then fully encapsulated in mtHsp60-10, revealing prominent contacts at two discrete sites that potentially support maturation. These results identify distinct roles for the apical domains in coordinating client capture and progression through the chaperone cycle, supporting a conserved mechanism of group I chaperonin function.
Collapse
Affiliation(s)
- Julian R Braxton
- Graduate Program in Chemistry and Chemical Biology, University of California San Francisco, San Francisco, CA, USA
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Hao Shao
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Eric Tse
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Jason E Gestwicki
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA.
| | - Daniel R Southworth
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Mitra P, Deshmukh AS. Proteostasis is a key driver of the pathogenesis in Apicomplexa. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119824. [PMID: 39168412 DOI: 10.1016/j.bbamcr.2024.119824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Proteostasis, including protein folding mediated by molecular chaperones, protein degradation, and stress response pathways in organelles like ER (unfolded protein response: UPR), are responsible for cellular protein quality control. This is essential for cell survival as it regulates and reprograms cellular processes. Here, we underscore the role of the proteostasis pathway in Apicomplexan parasites with respect to their well-characterized roles as well as potential roles in many parasite functions, including survival, multiplication, persistence, and emerging drug resistance. In addition to the diverse physiological importance of proteostasis in Apicomplexa, we assess the potential of the pathway's components as chemotherapeutic targets.
Collapse
Affiliation(s)
- Pallabi Mitra
- BRIC-Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.
| | | |
Collapse
|
8
|
Hu YT, Lin YW, Guo SY, Jiang Z, Xu SM, Su Z, Zhang JM, Rao Y, Chen SB, Huang ZS. Disrupting the protein-protein interaction network of Hsp72 inhibits adipogenic differentiation and lipid synthesis in adipocytes. Cell Signal 2024; 124:111431. [PMID: 39312987 DOI: 10.1016/j.cellsig.2024.111431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
The biological function against obesity of heat shock protein Hsp72 in adipose tissue has remained unclear. Our findings demonstrated that the expression levels of Hsp72 increased during the triglyceride (TG) accumulation process both in adipose tissue and 3T3-L1 cells. A significant decrease in adipogenic gene expression and TG levels was observed upon Hsp72 knockdown in 3T3-L1 cells, suggesting that Hsp72 promoted adipogenic differentiation and lipid synthesis processes. Encouraged by these findings, we further confirmed the allosteric Hsp72 inhibitors YK5 and MKT-077 also exhibited inhibition of both these processes. Further evaluation revealed that Hsp72 played a key role in interacting with numerous novel metabolic and cytomorphologic-related client proteins, thereby mediating the adipogenesis and lipogenesis process. Hsp72 inhibitors had the potential to disrupt these interactions, leading to the downregulation of adipogenic and lipogenic gene expression, as well as the suppression of TG accumulation. These findings suggested that inhibiting Hsp72 to disrupt adipogenic differentiation and lipid synthesis in adipocytes may be a promising anti-obesity strategy.
Collapse
Affiliation(s)
- Yu-Tao Hu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Yu-Wei Lin
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Shi-Yao Guo
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhi Jiang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Shu-Min Xu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Zheng Su
- The Division of Plastic and Reconstructive of Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510235, China
| | - Jin-Ming Zhang
- The Division of Plastic and Reconstructive of Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510235, China
| | - Yong Rao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570200, China.
| | - Shuo-Bin Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China.
| | - Zhi-Shu Huang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
9
|
Nussinov R, Jang H. The value of protein allostery in rational anticancer drug design: an update. Expert Opin Drug Discov 2024; 19:1071-1085. [PMID: 39068599 PMCID: PMC11390313 DOI: 10.1080/17460441.2024.2384467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION Allosteric drugs are advantageous. However, they still face hurdles, including identification of allosteric sites that will effectively alter the active site. Current strategies largely focus on identifying pockets away from the active sites into which the allosteric ligand will dock and do not account for exactly how the active site is altered. Favorable allosteric inhibitors dock into sites that are nearby the active sites and follow nature, mimicking diverse allosteric regulation strategies. AREAS COVERED The following article underscores the immense significance of allostery in drug design, describes current allosteric strategies, and especially offers a direction going forward. The article concludes with the authors' expert perspectives on the subject. EXPERT OPINION To select a productive venue in allosteric inhibitor development, we should learn from nature. Currently, useful strategies follow this route. Consider, for example, the mechanisms exploited in relieving autoinhibition and in harnessing allosteric degraders. Mimicking compensatory, or rescue mutations may also fall into such a thesis, as can molecular glues that capture features of scaffolding proteins. Capturing nature and creatively tailoring its mimicry can continue to innovate allosteric drug discovery.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
10
|
Xu P, Yang JC, Chen B, Ning S, Zhang X, Wang L, Nip C, Shen Y, Johnson OT, Grigorean G, Phinney B, Liu L, Wei Q, Corey E, Tepper CG, Chen HW, Evans CP, Dall'Era MA, Gao AC, Gestwicki JE, Liu C. Proteostasis perturbation of N-Myc leveraging HSP70 mediated protein turnover improves treatment of neuroendocrine prostate cancer. Nat Commun 2024; 15:6626. [PMID: 39103353 PMCID: PMC11300456 DOI: 10.1038/s41467-024-50459-x] [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: 11/30/2023] [Accepted: 07/11/2024] [Indexed: 08/07/2024] Open
Abstract
N-Myc is a key driver of neuroblastoma and neuroendocrine prostate cancer (NEPC). One potential way to circumvent the challenge of undruggable N-Myc is to target the protein homeostasis (proteostasis) system that maintains N-Myc levels. Here, we identify heat shock protein 70 (HSP70) as a top partner of N-Myc, which binds a conserved "SELILKR" motif and prevents the access of E3 ubiquitin ligase, STIP1 homology and U-box containing protein 1 (STUB1), possibly through steric hindrance. When HSP70's dwell time on N-Myc is increased by treatment with the HSP70 allosteric inhibitor, STUB1 is in close proximity with N-Myc and becomes functional to promote N-Myc ubiquitination on the K416 and K419 sites and forms polyubiquitination chains linked by the K11 and K63 sites. Notably, HSP70 inhibition significantly suppressed NEPC tumor growth, increased the efficacy of aurora kinase A (AURKA) inhibitors, and limited the expression of neuroendocrine-related pathways.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Joy C Yang
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Bo Chen
- Department of Urologic Surgery, University of California, Davis, CA, USA
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Shu Ning
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Xiong Zhang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA
| | - Leyi Wang
- Department of Urologic Surgery, University of California, Davis, CA, USA
- Graduate Group in Integrative Pathobiology, University of California, Davis, CA, USA
| | - Christopher Nip
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Yuqiu Shen
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Oleta T Johnson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | | | - Brett Phinney
- Proteomics Core Facility, University of California, Davis, CA, USA
| | - Liangren Liu
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Eva Corey
- Department of Urology, University of Washington, Washington, WA, USA
| | - Clifford G Tepper
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Christopher P Evans
- Department of Urologic Surgery, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Marc A Dall'Era
- Department of Urologic Surgery, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Allen C Gao
- Department of Urologic Surgery, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Chengfei Liu
- Department of Urologic Surgery, University of California, Davis, CA, USA.
- Graduate Group in Integrative Pathobiology, University of California, Davis, CA, USA.
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA.
| |
Collapse
|
11
|
Frasnetti E, Magni A, Castelli M, Serapian SA, Moroni E, Colombo G. Structures, dynamics, complexes, and functions: From classic computation to artificial intelligence. Curr Opin Struct Biol 2024; 87:102835. [PMID: 38744148 DOI: 10.1016/j.sbi.2024.102835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/14/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Computational approaches can provide highly detailed insight into the molecular recognition processes that underlie drug binding, the assembly of protein complexes, and the regulation of biological functional processes. Classical simulation methods can bridge a wide range of length- and time-scales typically involved in such processes. Lately, automated learning and artificial intelligence methods have shown the potential to expand the reach of physics-based approaches, ushering in the possibility to model and even design complex protein architectures. The synergy between atomistic simulations and AI methods is an emerging frontier with a huge potential for advances in structural biology. Herein, we explore various examples and frameworks for these approaches, providing select instances and applications that illustrate their impact on fundamental biomolecular problems.
Collapse
Affiliation(s)
- Elena Frasnetti
- Department of Chemistry, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Andrea Magni
- Department of Chemistry, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Matteo Castelli
- Department of Chemistry, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Stefano A Serapian
- Department of Chemistry, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | | | - Giorgio Colombo
- Department of Chemistry, University of Pavia, via Taramelli 12, 27100 Pavia, Italy.
| |
Collapse
|
12
|
Jiang M, Zhang H, Song Y, Yin F, Hu Z, Li X, Wang Y, Wang Z, Li Y, Wang Z, Zhang Y, Wang S, Lu S, Xu G, Song T, Wang Z, Zhang Z. Discovery of Biphenyl Derivatives to Target Hsp70-Bim Protein-Protein Interaction in Chronic Myeloid Leukemia by Scaffold Hopping Strategy. J Med Chem 2024; 67:12068-12084. [PMID: 39012838 DOI: 10.1021/acs.jmedchem.4c00780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Hsp70-Bim protein-protein interaction (PPI) is the most recently identified specific target in chronic myeloid leukemia (CML) therapy. Herein, we developed a new class of Hsp70-Bim PPI inhibitors via scaffold hopping of S1g-10, the most potent Hsp70-Bim PPI inhibitor thus far. Through structure-activity relationship (SAR) study, we obtained a biphenyl scaffold compound JL-15 with a 5.6-fold improvement in Hsp70-Bim PPI suppression (Kd = 123 vs 688 nM) and a 4-fold improvement in water solubility (29.42 vs 7.19 μg/mL) compared to S1g-10. It maintains comparable apoptosis induction capability with S1g-10 against both TKI-sensitive and TKI-resistant CML cell lines in an Hsp70-Bim-dependent manner. Additionally, through SAR, 1H-15N TRSOY-NMR, and molecular docking, we revealed that Lys319 is a "hot spot" in the Hsp70-Bim PPI interface. Collectively, these results provide a novel chemical scaffold and structural insights for the rational design of Hsp70-Bim PPI inhibitors.
Collapse
MESH Headings
- Humans
- HSP70 Heat-Shock Proteins/metabolism
- HSP70 Heat-Shock Proteins/antagonists & inhibitors
- HSP70 Heat-Shock Proteins/chemistry
- Biphenyl Compounds/pharmacology
- Biphenyl Compounds/chemistry
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Structure-Activity Relationship
- Molecular Docking Simulation
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Bcl-2-Like Protein 11/metabolism
- Cell Line, Tumor
- Apoptosis/drug effects
- Protein Binding
- Drug Discovery
Collapse
Affiliation(s)
- Maojun Jiang
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Hong Zhang
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Yang Song
- Department of Hematology, Central Hospital of Dalian University of Technology, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Fangkui Yin
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Zhiyuan Hu
- School of Life Science and Technology, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Xin Li
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Yuying Wang
- School of Life Science and Technology, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Zheming Wang
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Yitong Li
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Zihan Wang
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Yanxin Zhang
- School of Life Science and Technology, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Siyao Wang
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Shaohua Lu
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Guanghong Xu
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Ting Song
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Ziqian Wang
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Zhichao Zhang
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| |
Collapse
|
13
|
Devi S, Charvat A, Millbern Z, Vinueza N, Gestwicki JE. Exploration of the binding determinants of protein phosphatase 5 (PP5) reveals a chaperone-independent activation mechanism. J Biol Chem 2024; 300:107435. [PMID: 38830406 PMCID: PMC11259706 DOI: 10.1016/j.jbc.2024.107435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/10/2024] [Accepted: 05/23/2024] [Indexed: 06/05/2024] Open
Abstract
The protein phosphatase 5 (PP5) is normally recruited to its substrates by the molecular chaperones, heat shock protein 70 (Hsp70) and heat shock protein 90 (Hsp90). This interaction requires the tetratricopeptide repeat (TPR) domain of PP5, which binds to an EEVD motif at the extreme C termini of cytosolic Hsp70 and Hsp90 isoforms. In addition to bringing PP5 into proximity with chaperone-bound substrates, this interaction also relieves autoinhibition in PP5's catalytic domain, promoting its phosphatase activity. To better understand the molecular determinants of this process, we screened a large, pentapeptide library for binding to PP5. This screen identified the amino acid preferences at each position, which we validated by showing that the optimal sequences bind 4- to 7-fold tighter than the natural EEVD motifs and stimulate PP5's enzymatic activity. The enhanced affinity for PP5's TPR domain was confirmed using a protein-adaptive differential scanning fluorimetry assay. Using this increased knowledge of structure-activity relationships, we re-examined affinity proteomics results to look for potential EEVD-like motifs in the C termini of known PP5-binding partners. This search identified elongator acetyltransferase complex subunit 1 (IKBKAP) as a putative partner, and indeed, we found that its C-terminal sequence, LSLLD, binds directly to PP5's TPR domain in vitro. Consistent with this idea, mutation of elongator acetyltransferase complex subunit 1's terminal aspartate was sufficient to interrupt the interaction with PP5 in vitro and in cells. Together, these findings reveal the sequence preferences of PP5's TPR domain and expand the scope of PP5's functions to include chaperone-independent complexes.
Collapse
Affiliation(s)
- Shweta Devi
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, California, USA
| | - Annemarie Charvat
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, California, USA
| | - Zoe Millbern
- Department of Textile Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Nelson Vinueza
- Department of Textile Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
14
|
Zhao P, Wang C, Sun S, Wang X, Balch WE. Tracing genetic diversity captures the molecular basis of misfolding disease. Nat Commun 2024; 15:3333. [PMID: 38637533 PMCID: PMC11026414 DOI: 10.1038/s41467-024-47520-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 04/04/2024] [Indexed: 04/20/2024] Open
Abstract
Genetic variation in human populations can result in the misfolding and aggregation of proteins, giving rise to systemic and neurodegenerative diseases that require management by proteostasis. Here, we define the role of GRP94, the endoplasmic reticulum Hsp90 chaperone paralog, in managing alpha-1-antitrypsin deficiency on a residue-by-residue basis using Gaussian process regression-based machine learning to profile the spatial covariance relationships that dictate protein folding arising from sequence variants in the population. Covariance analysis suggests a role for the ATPase activity of GRP94 in controlling the N- to C-terminal cooperative folding of alpha-1-antitrypsin responsible for the correction of liver aggregation and lung-disease phenotypes of alpha-1-antitrypsin deficiency. Gaussian process-based spatial covariance profiling provides a standard model built on covariant principles to evaluate the role of proteostasis components in guiding information flow from genome to proteome in response to genetic variation, potentially allowing us to intervene in the onset and progression of complex multi-system human diseases.
Collapse
Affiliation(s)
- Pei Zhao
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA
| | - Chao Wang
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA.
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
| | - Shuhong Sun
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Institute for Brain Tumors, Collaborative Innovation Center for Cancer Personalized Medicine, and Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Xi Wang
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - William E Balch
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA.
| |
Collapse
|
15
|
Song T, Zhang H, Zhao Q, Hu Z, Wang Z, Song Y, Zhang Z. Small molecule inhibitor targeting the Hsp70-Bim protein-protein interaction in estrogen receptor-positive breast cancer overcomes tamoxifen resistance. Breast Cancer Res 2024; 26:33. [PMID: 38409088 PMCID: PMC10895875 DOI: 10.1186/s13058-024-01790-0] [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: 08/24/2023] [Accepted: 02/15/2024] [Indexed: 02/28/2024] Open
Abstract
INTRODUCTION Estrogen receptor (ER) positive patients compromise about 70% of breast cancers. Tamoxifen, an antagonist of ERα66 (the classic ER), is the most effective and the standard first-line drug. However, its efficacy is limited by the development of acquired resistance. METHODS A specific inhibitor of Hsp70-Bim protein-protein interaction (PPI), S1g-2, together with an inhibitor of Hsp70-Bag3 PPI, MKT-077 and an ATP-competitive inhibitor VER155008, were used as chemical tools. Cell viability assays, co-immunoprecipitation and gene knockdown were used to investigate the role of Hsp70 in tamoxifen resistance. A xenograft model was established in which tamoxifen-resistant breast cancer (MCF-7/TAM-R) cells maintained in the presence of 5 μM tamoxifen were subcutaneously inoculated. The anti-tumor efficiency of S1g-2 was measured after a daily injection of 0.8 mg/kg for 14 days. RESULTS It was revealed that Hsp70-Bim PPI protects ERα-positive breast cancer from tamoxifen-induced apoptosis through binding and stabilizing ERα36, rather than ERα66, resulting in sustained EGFR mRNA and protein expression. Disruption of Hsp70-Bim PPI and downregulation of ERα36 expression in tumor samples are consistent with the in vitro functions of S1g-2, resulting in about a three-fold reduction in tumor volume. CONCLUSIONS The in vivo activity and safety of S1g-2 illustrated that it is a potential strategy for Hsp70-Bim disruption to overcome tamoxifen-resistant ER-positive breast cancer.
Collapse
Affiliation(s)
- Ting Song
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning, China.
| | - Hong Zhang
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning, China
| | - Qicheng Zhao
- Cancer Rehabilitation Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tong Ji University, Shanghai, China
| | - Zhiyuan Hu
- School of Life Science and Technology, Dalian University of Technology, Dalian, Liaoning, China
| | - Ziqian Wang
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning, China
| | - Yang Song
- Central Hospital of Dalian University of Technology, Dalian, Liaoning, China
| | - Zhichao Zhang
- Cancer Hospital of Dalian University of Technology, School of Chemistry, Dalian University of Technology, Dalian, Liaoning, China.
| |
Collapse
|
16
|
Ambrose AJ, Zerio CJ, Sivinski J, Zhu X, Godek J, Sanchez JL, Khanna M, Khanna R, Lairson L, Zhang DD, Chapman E. Human Hsp70 Substrate-Binding Domains Recognize Distinct Client Proteins. Biochemistry 2024; 63:251-263. [PMID: 38243804 DOI: 10.1021/acs.biochem.3c00531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2024]
Abstract
The 13 Hsp70 proteins in humans act on unique sets of substrates with diversity often being attributed to J-domain-containing protein (Hsp40 or JDP) cofactors. We were therefore surprised to find drastically different binding affinities for Hsp70-peptide substrates, leading us to probe substrate specificity among the 8 canonical Hsp70s from humans. We used peptide arrays to characterize Hsp70 binding and then mined these data using machine learning to develop an algorithm for isoform-specific prediction of Hsp70 binding sequences. The results of this algorithm revealed recognition patterns not predicted based on local sequence alignments. We then showed that none of the human isoforms can complement heat-shocked DnaK knockout Escherichia coli cells. However, chimeric Hsp70s consisting of the human nucleotide-binding domain and the substrate-binding domain of DnaK complement during heat shock, providing further evidence in vivo of the divergent function of the Hsp70 substrate-binding domains. We also demonstrated that the differences in heat shock complementation among the chimeras are not due to loss of DnaJ binding. Although we do not exclude JDPs as additional specificity factors, our data demonstrate substrate specificity among the Hsp70s, which has important implications for inhibitor development in cancer and neurodegeneration.
Collapse
Affiliation(s)
- Andrew J Ambrose
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721, United States
| | - Christopher J Zerio
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721, United States
| | - Jared Sivinski
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721, United States
| | - Xiaoyi Zhu
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721, United States
| | - Jack Godek
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721, United States
| | - Jonathan L Sanchez
- Department of Pharmacology, College of Medicine, The University of Arizona Health Sciences, Tucson, Arizona 85424, United States
| | - May Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York 10010, United States
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York 10010, United States
| | - Luke Lairson
- Department of Chemistry, Scripps Research, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721, United States
| | - Eli Chapman
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
17
|
Li Y, Yu Y, Li Y, Wang H, Li Q. Molecular evolution of the heat shock protein family and the role of HSP30 in immune response and wound healing in lampreys (Lethenteron reissneri). FISH & SHELLFISH IMMUNOLOGY 2024; 145:109323. [PMID: 38147915 DOI: 10.1016/j.fsi.2023.109323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/02/2023] [Accepted: 12/19/2023] [Indexed: 12/28/2023]
Abstract
Heat shock proteins (HSPs) are molecular chaperones that ubiquitously exist in various organisms and play essential roles in protein folding, transport, and expression. While most HSPs are highly conserved across species, a few HSPs are evolutionarily distinct in some species and may have unique functions. To explore the evolutionary history of the vertebrate HSP family, we identify members of the HSP family at the genome-wide level in lampreys (Lethenteron reissneri), a living representative of jawless vertebrates diverged from jawed vertebrates over 500 million years ago. The phylogenetic analysis reveals that the lamprey HSP family contains HSP90a1, HSP90a2, HSC70, HSP60, HSP30, HSP27, HSP17, and HSP10, which have a primitive status in the molecular evolution of vertebrate HSPs. Transcriptome analysis reveals the expression distribution of members of the HSP family in various tissues of lampreys. It is shown that HSP30, normally found in birds, amphibians, and fish, is also present in lampreys, with remarkable expansion of HSP30 gene copies in the lamprey genome. The transcription of HSP30 is significantly induced in leukocytes and heart of lampreys during various pathogens or poly(I:C) stimulation, indicating that HSP30 may be involved in the immune defense of lampreys in response to bacterial or viral infection. Immunohistochemistry demonstrates significantly increased HSP30 expression in subcutaneous muscle tissue after skin injury in lamprey models of wound repair. Furthermore, transcriptome analysis shows that ectopic expression of HSP30 in 3T3-L1 fibroblasts affect the expression of genes related to the PI3K-AKT signaling pathway, suggesting that HSP30 could serves as a negative regulator of fibrosis. These results indicate that HSP30 may play a critical role in facilitating the process of lamprey skin repair following injury. This study provides new insights into the origin and evolution of the HSP gene family in vertebrates and offers valuable clues to reveal the important role of HSP30 in immune defense and wound healing of lampreys.
Collapse
Affiliation(s)
- Yao Li
- School of Life Science, Liaoning Normal University, Dalian, China; Lamprey Research Center, Liaoning Normal University, Dalian, China; College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Yang Yu
- Department of Urology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Yuting Li
- School of Life Science, Liaoning Normal University, Dalian, China
| | - Hao Wang
- School of Life Science, Liaoning Normal University, Dalian, China; Lamprey Research Center, Liaoning Normal University, Dalian, China.
| | - Qingwei Li
- School of Life Science, Liaoning Normal University, Dalian, China; Lamprey Research Center, Liaoning Normal University, Dalian, China.
| |
Collapse
|
18
|
Wang Z, Zhang H, Li X, Song Y, Wang Y, Hu Z, Gao Q, Jiang M, Yin F, Yuan L, Liu J, Song T, Lu S, Xu G, Zhang Z. Exploiting the "Hot-Spots" of Hsp70 -Bim Protein -Protein Interaction to Optimize the 1-Oxo-1 H-phenalene-2,3-dicarbonitrile Analogues as Specific Hsp70 -Bim Inhibitors. J Med Chem 2023; 66:16377-16387. [PMID: 38011535 DOI: 10.1021/acs.jmedchem.3c01783] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Selectively targeting the cancer-specific protein-protein interaction (PPI) between Hsp70 and Bim has been discovered as a promising strategy for treating chronic myeloid leukemia (CML). The first Hsp70-Bim PPI inhibitor, S1g-2, has been identified to overcome the on-target toxicity of known Hsp70 inhibitors when it induces apoptosis of CML cells. Herein, we carried out a hit-to-lead optimization of S1g-2, yielding S1g-10, which exhibited a 10-fold increase in Hsp70/Bim suppressing potency. Furthermore, S1g-10 not only exhibited a 5- to 10-fold stronger antitumor activity in the sub-μM range against CML cells than S1g-2 in vitro, but it also overcame BCR-ABL-independent tyrosine kinase inhibitor resistance in CML in vivo depending on the Hsp70-Bim signaling pathway. Moreover, through structure-activity relationship analysis, TROSY-HSQC NMR, molecular dynamics simulation, and point mutation validation, two hydrophobic pockets composed of eight key residues were demonstrated to produce predominant interactions with either Bim or S1g-10, regarded as the "hot-spots" in the Hsp70-Bim PPI interface.
Collapse
Affiliation(s)
- Ziqian Wang
- School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Hong Zhang
- School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Xin Li
- School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Yang Song
- Department of Hematology, Central Hospital of Dalian University of Technology, Dalian, Liaoning 116023, China
| | - Yuying Wang
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Zhiyuan Hu
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Qishuang Gao
- School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Maojun Jiang
- School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Fangkui Yin
- School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Linjie Yuan
- School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Jingjing Liu
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Ting Song
- School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Shaohua Lu
- School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Guanghong Xu
- School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Zhichao Zhang
- School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116024, China
| |
Collapse
|
19
|
Richards A, Lupoli TJ. Peptide-based molecules for the disruption of bacterial Hsp70 chaperones. Curr Opin Chem Biol 2023; 76:102373. [PMID: 37516006 PMCID: PMC11217992 DOI: 10.1016/j.cbpa.2023.102373] [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: 04/27/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/31/2023]
Abstract
DnaK is a chaperone that aids in nascent protein folding and the maintenance of proteome stability across bacteria. Due to the importance of DnaK in cellular proteostasis, there have been efforts to generate molecules that modulate its function. In nature, both protein substrates and antimicrobial peptides interact with DnaK. However, many of these ligands interact with other cellular machinery as well. Recent work has sought to modify these peptide scaffolds to create DnaK-selective and species-specific probes. Others have reported protein domain mimics of interaction partners to disrupt cellular DnaK function and high-throughput screening approaches to discover clinically-relevant peptidomimetics that inhibit DnaK. The described work provides a foundation for the design of new assays and molecules to regulate DnaK activity.
Collapse
Affiliation(s)
- Aweon Richards
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Tania J Lupoli
- Department of Chemistry, New York University, New York, NY 10003, USA.
| |
Collapse
|
20
|
Thompson AD, Wagner MJ, Rodriguez J, Malhotra A, Vander Roest S, Lilienthal U, Shao H, Vignesh M, Weber K, Yob JM, Prosser BL, Helms AS, Gestwicki JE, Ginsburg D, Day SM. An Unbiased Screen Identified the Hsp70-BAG3 Complex as a Regulator of Myosin-Binding Protein C3. JACC Basic Transl Sci 2023; 8:1198-1211. [PMID: 37791314 PMCID: PMC10544073 DOI: 10.1016/j.jacbts.2023.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 10/05/2023]
Abstract
Variants in the gene myosin-binding protein C3 (MYBPC3) account for approximately 50% of familial hypertrophic cardiomyopathy (HCM), leading to reduced levels of myosin-binding protein C3 (MyBP-C), the protein product made by gene MYBPC3. Elucidation of the pathways that regulate MyBP-C protein homeostasis could uncover new therapeutic strategies. Toward this goal, we screened a library of 2,426 bioactive compounds and identified JG98, an allosteric modulator of heat shock protein 70 that inhibits interaction with Bcl-2-associated athanogene (BAG) domain co-chaperones. JG98 reduces MyBP-C protein levels. Furthermore, genetic reduction of BAG3 phenocopies treatment with JG-98 by reducing MYBP-C protein levels.. Thus, an unbiased compound screen identified the heat shock protein 70-BAG3 complex as a regulator of MyBP-C stability.
Collapse
Affiliation(s)
- Andrea D. Thompson
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Marcus J. Wagner
- Department of Internal Medicine, Division of Cardiovascular Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Juliani Rodriguez
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alok Malhotra
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Steve Vander Roest
- Center for Chemical Genomics, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Ulla Lilienthal
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hao Shao
- Institute for Neurodegenerative Diseases and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Mathav Vignesh
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Keely Weber
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jaime M. Yob
- Department of Internal Medicine, Division of Cardiovascular Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Benjamin L. Prosser
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adam S. Helms
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason E. Gestwicki
- Institute for Neurodegenerative Diseases and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - David Ginsburg
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
- The Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Sharlene M. Day
- Department of Internal Medicine, Division of Cardiovascular Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Patwardhan CA, Kommalapati VK, Llbiyi T, Singh D, Alfa E, Horuzsko A, Korkaya H, Panda S, Reilly CA, Popik V, Chadli A. Capsaicin binds the N-terminus of Hsp90, induces lysosomal degradation of Hsp70, and enhances the anti-tumor effects of 17-AAG (Tanespimycin). Sci Rep 2023; 13:13790. [PMID: 37612326 PMCID: PMC10447550 DOI: 10.1038/s41598-023-40933-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023] Open
Abstract
Heat shock protein 90 (Hsp90) and its co-chaperones promote cancer, and targeting Hsp90 holds promise for cancer treatment. Most of the efforts to harness this potential have focused on targeting the Hsp90 N-terminus ATP binding site. Although newer-generation inhibitors have shown improved efficacy in aggressive cancers, induction of the cellular heat shock response (HSR) by these inhibitors is thought to limit their clinical efficacy. Therefore, Hsp90 inhibitors with novel mechanisms of action and that do not trigger the HSR would be advantageous. Here, we investigated the mechanism by which capsaicin inhibits Hsp90. Through mutagenesis, chemical modifications, and proteomic studies, we show that capsaicin binds to the N-terminus of Hsp90 and inhibits its ATPase activity. Consequently, capsaicin and its analogs inhibit Hsp90 ATPase-dependent progesterone receptor reconstitution in vitro. Capsaicin did not induce the HSR, instead, it promoted the degradation of Hsp70 through the lysosome-autophagy pathway. Remarkably, capsaicin did not induce degradation of the constitutively expressed cognate Hsc70, indicating selectivity for Hsp70. Combined treatments of capsaicin and the Hsp90 inhibitor 17-AAG improved the anti-tumor efficacy of 17-AAG in cell culture and tridimensional tumor spheroid growth assays using breast and prostate cancer models. Consistent with this, in silico docking studies revealed that capsaicin binding to the ATP binding site of Hsp90 was distinct from classical N-terminus Hsp90 inhibitors, indicating a novel mechanism of action. Collectively, these findings support the use of capsaicin as a chemical scaffold to develop novel Hsp90 N-terminus inhibitors as well as its ability to be a potential cancer co-therapeutic.
Collapse
Affiliation(s)
- Chaitanya A Patwardhan
- Georgia Cancer Center at Augusta University (Formerly Medical College of Georgia), 1410 Laney Walker Blvd, CN-3313, Augusta, GA, 30912, USA
| | - Vamsi Krishna Kommalapati
- Georgia Cancer Center at Augusta University (Formerly Medical College of Georgia), 1410 Laney Walker Blvd, CN-3313, Augusta, GA, 30912, USA
| | - Taoufik Llbiyi
- Georgia Cancer Center at Augusta University (Formerly Medical College of Georgia), 1410 Laney Walker Blvd, CN-3313, Augusta, GA, 30912, USA
| | - Digvijay Singh
- Georgia Cancer Center at Augusta University (Formerly Medical College of Georgia), 1410 Laney Walker Blvd, CN-3313, Augusta, GA, 30912, USA
| | - Eyad Alfa
- Georgia Cancer Center at Augusta University (Formerly Medical College of Georgia), 1410 Laney Walker Blvd, CN-3313, Augusta, GA, 30912, USA
| | - Anatolij Horuzsko
- Georgia Cancer Center at Augusta University (Formerly Medical College of Georgia), 1410 Laney Walker Blvd, CN-3313, Augusta, GA, 30912, USA
| | - Hasan Korkaya
- Georgia Cancer Center at Augusta University (Formerly Medical College of Georgia), 1410 Laney Walker Blvd, CN-3313, Augusta, GA, 30912, USA
| | - Siva Panda
- Department of Chemistry and Biochemistry, Augusta University, Augusta, GA, 30912, USA
| | - Christopher A Reilly
- Department of Pharmacology and Toxicology, Center for Human Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, 84112, USA
| | - Vladimir Popik
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Ahmed Chadli
- Georgia Cancer Center at Augusta University (Formerly Medical College of Georgia), 1410 Laney Walker Blvd, CN-3313, Augusta, GA, 30912, USA.
| |
Collapse
|
22
|
Braxton JR, Shao H, Tse E, Gestwicki JE, Southworth DR. Asymmetric apical domain states of mitochondrial Hsp60 coordinate substrate engagement and chaperonin assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540872. [PMID: 37293102 PMCID: PMC10245740 DOI: 10.1101/2023.05.15.540872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The mitochondrial chaperonin, mtHsp60, promotes the folding of newly imported and transiently misfolded proteins in the mitochondrial matrix, assisted by its co-chaperone mtHsp10. Despite its essential role in mitochondrial proteostasis, structural insights into how this chaperonin binds to clients and progresses through its ATP-dependent reaction cycle are not clear. Here, we determined cryo-electron microscopy (cryo-EM) structures of a hyperstable disease-associated mtHsp60 mutant, V72I, at three stages in this cycle. Unexpectedly, client density is identified in all states, revealing interactions with mtHsp60's apical domains and C-termini that coordinate client positioning in the folding chamber. We further identify a striking asymmetric arrangement of the apical domains in the ATP state, in which an alternating up/down configuration positions interaction surfaces for simultaneous recruitment of mtHsp10 and client retention. Client is then fully encapsulated in mtHsp60/mtHsp10, revealing prominent contacts at two discrete sites that potentially support maturation. These results identify a new role for the apical domains in coordinating client capture and progression through the cycle, and suggest a conserved mechanism of group I chaperonin function.
Collapse
Affiliation(s)
- Julian R. Braxton
- Graduate Program in Chemistry and Chemical Biology; University of California, San Francisco; San Francisco, CA 94158, USA
- Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry; University of California, San Francisco; San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Hao Shao
- Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Eric Tse
- Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Jason E. Gestwicki
- Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
- Department of Pharmaceutical Chemistry; University of California, San Francisco; San Francisco, CA 94158, USA
| | - Daniel R. Southworth
- Institute for Neurodegenerative Diseases; University of California, San Francisco; San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics; University of California, San Francisco; San Francisco, CA 94158, USA
| |
Collapse
|
23
|
Alhasan B, Mikeladze M, Guzhova I, Margulis B. Autophagy, molecular chaperones, and unfolded protein response as promoters of tumor recurrence. Cancer Metastasis Rev 2023; 42:217-254. [PMID: 36723697 DOI: 10.1007/s10555-023-10085-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/16/2023] [Indexed: 02/02/2023]
Abstract
Tumor recurrence is a paradoxical function of a machinery, whereby a small proportion of the cancer cell population enters a resistant, dormant state, persists long-term in this condition, and then transitions to proliferation. The dormant phenotype is typical of cancer stem cells, tumor-initiating cells, disseminated tumor cells, and drug-tolerant persisters, which all demonstrate similar or even equivalent properties. Cancer cell dormancy and its conversion to repopulation are regulated by several protein signaling systems that inhibit or induce cell proliferation and provide optimal interrelations between cancer cells and their special niche; these systems act in close connection with tumor microenvironment and immune response mechanisms. During dormancy and reawakening periods, cell proteostasis machineries, autophagy, molecular chaperones, and the unfolded protein response are recruited to protect refractory tumor cells from a wide variety of stressors and therapeutic insults. Proteostasis mechanisms functionally or even physically interfere with the main regulators of tumor relapse, and the significance of these interactions and implications in the tumor recurrence phases are discussed in this review.
Collapse
Affiliation(s)
- Bashar Alhasan
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia.
| | - Marina Mikeladze
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| | - Irina Guzhova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| | - Boris Margulis
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| |
Collapse
|
24
|
Wilson MR, Satapathy S, Vendruscolo M. Extracellular protein homeostasis in neurodegenerative diseases. Nat Rev Neurol 2023; 19:235-245. [PMID: 36828943 DOI: 10.1038/s41582-023-00786-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2023] [Indexed: 02/26/2023]
Abstract
The protein homeostasis (proteostasis) system encompasses the cellular processes that regulate protein synthesis, folding, concentration, trafficking and degradation. In the case of intracellular proteostasis, the identity and nature of these processes have been extensively studied and are relatively well known. By contrast, the mechanisms of extracellular proteostasis are yet to be fully elucidated, although evidence is accumulating that their age-related progressive impairment might contribute to neuronal death in neurodegenerative diseases. Constitutively secreted extracellular chaperones are emerging as key players in processes that operate to protect neurons and other brain cells by neutralizing the toxicity of extracellular protein aggregates and promoting their safe clearance and disposal. Growing evidence indicates that these extracellular chaperones exert multiple effects to promote cell viability and protect neurons against pathologies arising from the misfolding and aggregation of proteins in the synaptic space and interstitial fluid. In this Review, we outline the current knowledge of the mechanisms of extracellular proteostasis linked to neurodegenerative diseases, and we examine the latest understanding of key molecules and processes that protect the brain from the pathological consequences of extracellular protein aggregation and proteotoxicity. Finally, we contemplate possible therapeutic opportunities for neurodegenerative diseases on the basis of this emerging knowledge.
Collapse
Affiliation(s)
- Mark R Wilson
- School of Chemistry and Molecular Bioscience, Molecular Horizons Research Institute, University of Wollongong, Wollongong, New South Wales, Australia.
| | - Sandeep Satapathy
- Blavatnik Institute of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
25
|
HSP70 mediates a crosstalk between the estrogen and the heat shock response pathways. J Biol Chem 2023; 299:102872. [PMID: 36610605 PMCID: PMC9926311 DOI: 10.1016/j.jbc.2023.102872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 12/24/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Cells respond to multiple signals from the environment simultaneously, which often creates crosstalk between pathways affecting the capacity to adapt to the changing environment. Chaperones are an important component in the cellular integration of multiple responses to environmental signals, often implicated in negative feedback and inactivation mechanisms. These mechanisms include the stabilization of steroid hormone nuclear receptors in the cytoplasm in the absence of their ligand. Here, we show using immunofluorescence, chromatin immunoprecipitation, and nascent transcripts production that the heat shock protein 70 (HSP70) chaperone plays a central role in a new crosstalk mechanism between the steroid and heat shock response pathways. HSP70-dependent feedback mechanisms are required to inactivate the heat shock factor 1 (HSF1) after activation. Interestingly, a steroid stimulation leads to faster accumulation of HSF1 in inactive foci following heat shock. Our results further show that in the presence of estrogen, HSP70 accumulates at HSF1-regulated noncoding regions, leading to deactivation of HSF1 and the abrogation of the heat shock transcriptional response. Using an HSP70 inhibitor, we demonstrate that the crosstalk between both pathways is dependent on the chaperone activity. These results suggest that HSP70 availability is a key determinant in the transcriptional integration of multiple external signals. Overall, these results offer a better understanding of the crosstalk between the heat shock and steroid responses, which are salient in neurodegenerative disorders and cancers.
Collapse
|
26
|
Schneider M, Antes I. Comparison of allosteric signaling in DnaK and BiP using mutual information between simulated residue conformations. Proteins 2023; 91:237-255. [PMID: 36111439 DOI: 10.1002/prot.26425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/06/2022] [Accepted: 09/13/2022] [Indexed: 01/13/2023]
Abstract
The heat shock protein 70 kDa (Hsp70) chaperone system serves as a critical component of protein quality control across a wide range of prokaryotic and eukaryotic organisms. Divergent evolution and specialization to particular organelles have produced numerous Hsp70 variants which share similarities in structure and general function, but differ substantially in regulatory aspects, including conformational dynamics and activity modulation by cochaperones. The human Hsp70 variant BiP (also known as GRP78 or HSPA5) is of therapeutic interest in the context of cancer, neurodegenerative diseases, and viral infection, including for treatment of the pandemic virus SARS-CoV-2. Due to the complex conformational rearrangements and high sequential variance within the Hsp70 protein family, it is in many cases poorly understood which amino acid mutations are responsible for biochemical differences between protein variants. In this study, we predicted residues associated with conformational regulation of human BiP and Escherichia coli DnaK. Based on protein structure networks obtained from molecular dynamics simulations, we analyzed the shared information between interaction timelines to highlight residue positions with strong conformational coupling to their environment. Our predictions, which focus on the binding processes of the chaperone's substrate and cochaperones, indicate residues filling potential signaling roles specific to either DnaK or BiP. By combining predictions of individual residues into conformationally coupled chains connecting ligand binding sites, we predict a BiP specific secondary signaling pathway associated with substrate binding. Our study sheds light on mechanistic differences in signaling and regulation between Hsp70 variants, which provide insights relevant to therapeutic applications of these proteins.
Collapse
Affiliation(s)
- Markus Schneider
- TUM Center for Functional Protein Assemblies and TUM School of Life Sciences, Technische Universität München, Freising, Bavaria, Germany
| | - Iris Antes
- TUM Center for Functional Protein Assemblies and TUM School of Life Sciences, Technische Universität München, Freising, Bavaria, Germany
| |
Collapse
|
27
|
Kuzuoglu-Ozturk D, Aksoy O, Schmidt C, Lea R, Larson JD, Phelps RRL, Nasholm N, Holt M, Contreras A, Huang M, Wong-Michalak S, Shao H, Wechsler-Reya R, Phillips JJ, Gestwicki JE, Ruggero D, Weiss WA. N-myc-Mediated Translation Control Is a Therapeutic Vulnerability in Medulloblastoma. Cancer Res 2023; 83:130-140. [PMID: 36264168 PMCID: PMC9812901 DOI: 10.1158/0008-5472.can-22-0945] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/17/2022] [Accepted: 10/18/2022] [Indexed: 02/03/2023]
Abstract
Deregulation of neuroblastoma-derived myc (N-myc) is a leading cause of malignant brain tumors in children. To target N-myc-driven medulloblastoma, most research has focused on identifying genomic alterations or on the analysis of the medulloblastoma transcriptome. Here, we have broadly characterized the translatome of medulloblastoma and shown that N-myc unexpectedly drives selective translation of transcripts that promote protein homeostasis. Cancer cells are constantly exposed to proteotoxic stress associated with alterations in protein production or folding. It remains poorly understood how cancers cope with proteotoxic stress to promote their growth. Here, our data revealed that N-myc regulates the expression of specific components (∼5%) of the protein folding machinery at the translational level through the major cap binding protein, eukaryotic initiation factor eIF4E. Reducing eIF4E levels in mouse models of medulloblastoma blocked tumorigenesis. Importantly, targeting Hsp70, a protein folding chaperone translationally regulated by N-myc, suppressed tumor growth in mouse and human medulloblastoma xenograft models. These findings reveal a previously hidden molecular program that promotes medulloblastoma formation and identify new therapies that may have impact in the clinic. SIGNIFICANCE Translatome analysis in medulloblastoma shows that N-myc drives selective translation of transcripts that promote protein homeostasis and that represent new therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Duygu Kuzuoglu-Ozturk
- Department of Urology, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Ozlem Aksoy
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Neurology, University of California, San Francisco, California
| | - Christin Schmidt
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Neurology, University of California, San Francisco, California
| | - Robin Lea
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Neurology, University of California, San Francisco, California
| | - Jon D Larson
- Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Ryan R L Phelps
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Neurological Surgery, University of California, San Francisco, California
- Department of Neurological Surgery, Stanford University, Stanford, California
| | - Nicole Nasholm
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Neurology, University of California, San Francisco, California
| | - Megan Holt
- Department of Urology, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Adrian Contreras
- Department of Urology, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Miller Huang
- Children's Hospital Los Angeles, Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology and Blood & Marrow Transplantation, and The Saban Research Institute, Los Angeles, California
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Shannon Wong-Michalak
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Neurology, University of California, San Francisco, California
| | - Hao Shao
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, California
| | - Robert Wechsler-Reya
- Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Joanna J Phillips
- Department of Neurological Surgery, University of California, San Francisco, California
- Division of Neuropathology, Department of Pathology, University of California, San Francisca, San Francisco, California
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, California
| | - Davide Ruggero
- Department of Urology, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California
| | - William A Weiss
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
- Department of Neurology, University of California, San Francisco, California
- Department of Neurological Surgery, University of California, San Francisco, California
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| |
Collapse
|
28
|
Backe SJ, Woodford MR, Ahanin E, Sager RA, Bourboulia D, Mollapour M. Impact of Co-chaperones and Posttranslational Modifications Toward Hsp90 Drug Sensitivity. Subcell Biochem 2023; 101:319-350. [PMID: 36520312 PMCID: PMC10077965 DOI: 10.1007/978-3-031-14740-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Posttranslational modifications (PTMs) regulate myriad cellular processes by modulating protein function and protein-protein interaction. Heat shock protein 90 (Hsp90) is an ATP-dependent molecular chaperone whose activity is responsible for the stabilization and maturation of more than 300 client proteins. Hsp90 is a substrate for numerous PTMs, which have diverse effects on Hsp90 function. Interestingly, many Hsp90 clients are enzymes that catalyze PTM, demonstrating one of the several modes of regulation of Hsp90 activity. Approximately 25 co-chaperone regulatory proteins of Hsp90 impact structural rearrangements, ATP hydrolysis, and client interaction, representing a second layer of influence on Hsp90 activity. A growing body of literature has also established that PTM of these co-chaperones fine-tune their activity toward Hsp90; however, many of the identified PTMs remain uncharacterized. Given the critical role of Hsp90 in supporting signaling in cancer, clinical evaluation of Hsp90 inhibitors is an area of great interest. Interestingly, differential PTM and co-chaperone interaction have been shown to impact Hsp90 binding to its inhibitors. Therefore, understanding these layers of Hsp90 regulation will provide a more complete understanding of the chaperone code, facilitating the development of new biomarkers and combination therapies.
Collapse
Affiliation(s)
- Sarah J Backe
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Mark R Woodford
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Elham Ahanin
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Rebecca A Sager
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Dimitra Bourboulia
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Mehdi Mollapour
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA. .,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA. .,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
29
|
Shao H, Taguwa S, Gilbert L, Shkedi A, Sannino S, Guerriero CJ, Gale-Day ZJ, Young ZT, Brodsky JL, Weissman J, Gestwicki JE, Frydman J. A campaign targeting a conserved Hsp70 binding site uncovers how subcellular localization is linked to distinct biological activities. Cell Chem Biol 2022; 29:1303-1316.e3. [PMID: 35830852 PMCID: PMC9513760 DOI: 10.1016/j.chembiol.2022.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 02/20/2022] [Accepted: 06/20/2022] [Indexed: 12/14/2022]
Abstract
The potential of small molecules to localize within subcellular compartments is rarely explored. To probe this question, we measured the localization of Hsp70 inhibitors using fluorescence microscopy. We found that even closely related analogs had dramatically different distributions, with some residing predominantly in the mitochondria and others in the ER. CRISPRi screens supported this idea, showing that different compounds had distinct chemogenetic interactions with Hsp70s of the ER (HSPA5/BiP) and mitochondria (HSPA9/mortalin) and their co-chaperones. Moreover, localization seemed to determine function, even for molecules with conserved binding sites. Compounds with distinct partitioning have distinct anti-proliferative activity in breast cancer cells compared with anti-viral activity in cellular models of Dengue virus replication, likely because different sets of Hsp70s are required in these processes. These findings highlight the contributions of subcellular partitioning and chemogenetic interactions to small molecule activity, features that are rarely explored during medicinal chemistry campaigns.
Collapse
Affiliation(s)
- Hao Shao
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA; College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Shuhei Taguwa
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan; Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Luke Gilbert
- Department of Urology and Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Arielle Shkedi
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Sara Sannino
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Zachary J Gale-Day
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Zapporah T Young
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan Weissman
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA.
| | - Judith Frydman
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
30
|
Gomez-Sintes R, Xin Q, Jimenez-Loygorri JI, McCabe M, Diaz A, Garner TP, Cotto-Rios XM, Wu Y, Dong S, Reynolds CA, Patel B, de la Villa P, Macian F, Boya P, Gavathiotis E, Cuervo AM. Targeting retinoic acid receptor alpha-corepressor interaction activates chaperone-mediated autophagy and protects against retinal degeneration. Nat Commun 2022; 13:4220. [PMID: 35864098 PMCID: PMC9304322 DOI: 10.1038/s41467-022-31869-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/30/2022] [Indexed: 11/09/2022] Open
Abstract
Chaperone-mediated autophagy activity, essential in the cellular defense against proteotoxicity, declines with age, and preventing this decline in experimental genetic models has proven beneficial. Here, we have identified the mechanism of action of selective chaperone-mediated autophagy activators previously developed by our group and have leveraged that information to generate orally bioavailable chaperone-mediated autophagy activators with favorable brain exposure. Chaperone-mediated autophagy activating molecules stabilize the interaction between retinoic acid receptor alpha - a known endogenous inhibitor of chaperone-mediated autophagy - and its co-repressor, nuclear receptor corepressor 1, resulting in changes of a discrete subset of the retinoic acid receptor alpha transcriptional program that leads to selective chaperone-mediated autophagy activation. Chaperone-mediated autophagy activators molecules activate this pathway in vivo and ameliorate retinal degeneration in a retinitis pigmentosa mouse model. Our findings reveal a mechanism for pharmacological targeting of chaperone-mediated autophagy activation and suggest a therapeutic strategy for retinal degeneration.
Collapse
Affiliation(s)
- Raquel Gomez-Sintes
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, 28040, Spain
| | - Qisheng Xin
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Juan Ignacio Jimenez-Loygorri
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, 28040, Spain
| | - Mericka McCabe
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Antonio Diaz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Thomas P Garner
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Xiomaris M Cotto-Rios
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yang Wu
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Shuxian Dong
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Cara A Reynolds
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Bindi Patel
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Pedro de la Villa
- Department; of System Biology, Universidad de Alcalá, Madrid, Spain and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, 28801, Spain
| | - Fernando Macian
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, 28040, Spain.
| | - Evripidis Gavathiotis
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
31
|
Johnson OT, Gestwicki JE. Multivalent protein-protein interactions are pivotal regulators of eukaryotic Hsp70 complexes. Cell Stress Chaperones 2022; 27:397-415. [PMID: 35670950 PMCID: PMC9346034 DOI: 10.1007/s12192-022-01281-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 11/26/2022] Open
Abstract
Heat shock protein 70 (Hsp70) is a molecular chaperone and central regulator of protein homeostasis (proteostasis). Paramount to this role is Hsp70's binding to client proteins and co-chaperones to produce distinct complexes, such that understanding the protein-protein interactions (PPIs) of Hsp70 is foundational to describing its function and dysfunction in disease. Mounting evidence suggests that these PPIs include both "canonical" interactions, which are universally conserved, and "non-canonical" (or "secondary") contacts that seem to have emerged in eukaryotes. These two categories of interactions involve discrete binding surfaces, such that some clients and co-chaperones engage Hsp70 with at least two points of contact. While the contributions of canonical interactions to chaperone function are becoming increasingly clear, it can be challenging to deconvolute the roles of secondary interactions. Here, we review what is known about non-canonical contacts and highlight examples where their contributions have been parsed, giving rise to a model in which Hsp70's secondary contacts are not simply sites of additional avidity but are necessary and sufficient to impart unique functions. From this perspective, we propose that further exploration of non-canonical contacts will generate important insights into the evolution of Hsp70 systems and inspire new approaches for developing small molecules that tune Hsp70-mediated proteostasis.
Collapse
Affiliation(s)
- Oleta T Johnson
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
32
|
Eronina TB, Mikhaylova VV, Chebotareva NA, Tugaeva KV, Kurganov BI. Effect of Betaine and Arginine on Interaction of αB-Crystallin with Glycogen Phosphorylase b. Int J Mol Sci 2022; 23:3816. [PMID: 35409175 PMCID: PMC8998655 DOI: 10.3390/ijms23073816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Protein-protein interactions (PPIs) play an important role in many biological processes in a living cell. Among them chaperone-client interactions are the most important. In this work PPIs of αB-crystallin and glycogen phosphorylase b (Phb) in the presence of betaine (Bet) and arginine (Arg) at 48 °C and ionic strength of 0.15 M were studied using methods of dynamic light scattering, differential scanning calorimetry, and analytical ultracentrifugation. It was shown that Bet enhanced, while Arg reduced both the stability of αB-crystallin and its adsorption capacity (AC0) to the target protein at the stage of aggregate growth. Thus, the anti-aggregation activity of αB-crystallin increased in the presence of Bet and decreased under the influence of Arg, which resulted in inhibition or acceleration of Phb aggregation, respectively. Our data show that chemical chaperones can influence the tertiary and quaternary structure of both the target protein and the protein chaperone. The presence of the substrate protein also affects the quaternary structure of αB-crystallin, causing its disassembly. This is inextricably linked to the anti-aggregation activity of αB-crystallin, which in turn affects its PPI with the target protein. Thus, our studies contribute to understanding the mechanism of interaction between chaperones and proteins.
Collapse
Affiliation(s)
- Tatiana B. Eronina
- Bach Institute of Biochemistry, Federal Research Centre “Fundamentals of Biotechnology” of the Russian Academy of Sciences, Leninsky pr. 33, 119071 Moscow, Russia; (V.V.M.); (N.A.C.); (K.V.T.); (B.I.K.)
| | | | | | | | | |
Collapse
|
33
|
Johnson OT, Nadel CM, Carroll EC, Arhar T, Gestwicki JE. Two distinct classes of cochaperones compete for the EEVD motif in heat shock protein 70 to tune its chaperone activities. J Biol Chem 2022; 298:101697. [PMID: 35148989 PMCID: PMC8913300 DOI: 10.1016/j.jbc.2022.101697] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 12/30/2022] Open
Abstract
Chaperones of the heat shock protein 70 (Hsp70) family engage in protein-protein interactions with many cochaperones. One "hotspot" for cochaperone binding is the EEVD motif, found at the extreme C terminus of cytoplasmic Hsp70s. This motif is known to bind tetratricopeptide repeat domain cochaperones, such as the E3 ubiquitin ligase CHIP. In addition, the EEVD motif also interacts with a structurally distinct domain that is present in class B J-domain proteins, such as DnaJB4. These observations suggest that CHIP and DnaJB4 might compete for binding to Hsp70's EEVD motif; however, the molecular determinants of such competition are not clear. Using a collection of EEVD-derived peptides, including mutations and truncations, we explored which residues are critical for binding to both CHIP and DnaJB4. These results revealed that some features, such as the C-terminal carboxylate, are important for both interactions. However, CHIP and DnaJB4 also had unique preferences, especially at the isoleucine position immediately adjacent to the EEVD. Finally, we show that competition between these cochaperones is important in vitro, as DnaJB4 limits the ubiquitination activity of the Hsp70-CHIP complex, whereas CHIP suppresses the client refolding activity of the Hsp70-DnaJB4 complex. Together, these data suggest that the EEVD motif has evolved to support diverse protein-protein interactions, such that competition between cochaperones may help guide whether Hsp70-bound proteins are folded or degraded.
Collapse
Affiliation(s)
- Oleta T Johnson
- Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, California, USA
| | - Cory M Nadel
- Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, California, USA
| | - Emma C Carroll
- Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, California, USA
| | - Taylor Arhar
- Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, California, USA; Department of Chemistry, Beloit College, Beloit, Wisconsin, USA.
| | - Jason E Gestwicki
- Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco, California, USA; Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
34
|
Triveri A, Sanchez-Martin C, Torielli L, Serapian SA, Marchetti F, D'Acerno G, Pirota V, Castelli M, Moroni E, Ferraro M, Quadrelli P, Rasola A, Colombo G. Protein allostery and ligand design: Computational design meets experiments to discover novel chemical probes. J Mol Biol 2022; 434:167468. [DOI: 10.1016/j.jmb.2022.167468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 10/19/2022]
|
35
|
Helms AS, Thompson AD, Day SM. Translation of New and Emerging Therapies for Genetic Cardiomyopathies. JACC Basic Transl Sci 2022; 7:70-83. [PMID: 35128211 PMCID: PMC8807730 DOI: 10.1016/j.jacbts.2021.07.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 11/05/2022]
Abstract
The primary etiology of a diverse range of cardiomyopathies is now understood to be genetic, creating a new paradigm for targeting treatments on the basis of the underlying molecular cause. This review provides a genetic and etiologic context for the traditional clinical classifications of cardiomyopathy, including molecular subtypes that may exhibit differential responses to existing or emerging treatments. The authors describe several emerging cardiomyopathy treatments, including gene therapy, direct targeting of myofilament function, protein quality control, metabolism, and others. The authors discuss advantages and disadvantages of these approaches and indicate areas of high potential for short- and longer term efficacy.
Collapse
Key Words
- AAV, adeno-associated virus
- ACM, arrhythmogenic cardiomyopathy
- ARVC, arrhythmogenic right ventricular cardiomyopathy
- ATPase, adenosine triphosphatase
- DCM, dilated cardiomyopathy
- DMD, Duchenne muscular dystrophy
- DNA, DNA
- DSP, desmoplakin
- FDA, U.S. Food and Drug Administration
- GRT, gene replacement therapy
- GST, gene silencing therapy
- HCM, hypertrophic cardiomyopathy
- HR, homologous recombination
- LNP, lipid nanoparticle
- LVOT, left ventricular outflow tract
- RNA, RNA
- TTR, transthyretin
- arrhythmogenic cardiomyopathy
- dilated cardiomyopathy
- genetics
- hypertrophic cardiomyopathy
- therapeutics
Collapse
Affiliation(s)
- Adam S. Helms
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrea D. Thompson
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sharlene M. Day
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
36
|
Hosfelt J, Richards A, Zheng M, Adura C, Nelson B, Yang A, Fay A, Resager W, Ueberheide B, Glickman JF, Lupoli TJ. An allosteric inhibitor of bacterial Hsp70 chaperone potentiates antibiotics and mitigates resistance. Cell Chem Biol 2021; 29:854-869.e9. [PMID: 34818532 DOI: 10.1016/j.chembiol.2021.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/20/2021] [Accepted: 11/02/2021] [Indexed: 12/23/2022]
Abstract
DnaK is the bacterial homolog of Hsp70, an ATP-dependent chaperone that helps cofactor proteins to catalyze nascent protein folding and salvage misfolded proteins. In the pathogen Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), DnaK and its cofactors are proposed antimycobacterial targets, yet few small-molecule inhibitors or probes exist for these families of proteins. Here, we describe the repurposing of a drug called telaprevir that is able to allosterically inhibit the ATPase activity of DnaK and to prevent chaperone function by mimicking peptide substrates. In mycobacterial cells, telaprevir disrupts DnaK- and cofactor-mediated cellular proteostasis, resulting in enhanced efficacy of aminoglycoside antibiotics and reduced resistance to the frontline TB drug rifampin. Hence, this work contributes to a small but growing collection of protein chaperone inhibitors, and it demonstrates that these molecules disrupt bacterial mechanisms of survival in the presence of different antibiotic classes.
Collapse
Affiliation(s)
- Jordan Hosfelt
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Aweon Richards
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Meng Zheng
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Carolina Adura
- High-Throughput and Spectroscopy Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Brock Nelson
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Amy Yang
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Allison Fay
- Immunology Program, Sloan Kettering Insitute, New York, NY 10065, USA
| | - William Resager
- Departments of Biochemistry and Molecular Pharmacology, Neurology and Director Proteomics Lab, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Beatrix Ueberheide
- Departments of Biochemistry and Molecular Pharmacology, Neurology and Director Proteomics Lab, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - J Fraser Glickman
- High-Throughput and Spectroscopy Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Tania J Lupoli
- Department of Chemistry, New York University, New York, NY 10003, USA.
| |
Collapse
|
37
|
With or without You: Co-Chaperones Mediate Health and Disease by Modifying Chaperone Function and Protein Triage. Cells 2021; 10:cells10113121. [PMID: 34831344 PMCID: PMC8619055 DOI: 10.3390/cells10113121] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 01/18/2023] Open
Abstract
Heat shock proteins (HSPs) are a family of molecular chaperones that regulate essential protein refolding and triage decisions to maintain protein homeostasis. Numerous co-chaperone proteins directly interact and modify the function of HSPs, and these interactions impact the outcome of protein triage, impacting everything from structural proteins to cell signaling mediators. The chaperone/co-chaperone machinery protects against various stressors to ensure cellular function in the face of stress. However, coding mutations, expression changes, and post-translational modifications of the chaperone/co-chaperone machinery can alter the cellular stress response. Importantly, these dysfunctions appear to contribute to numerous human diseases. Therapeutic targeting of chaperones is an attractive but challenging approach due to the vast functions of HSPs, likely contributing to the off-target effects of these therapies. Current efforts focus on targeting co-chaperones to develop precise treatments for numerous diseases caused by defects in protein quality control. This review focuses on the recent developments regarding selected HSP70/HSP90 co-chaperones, with a concentration on cardioprotection, neuroprotection, cancer, and autoimmune diseases. We also discuss therapeutic approaches that highlight both the utility and challenges of targeting co-chaperones.
Collapse
|
38
|
Molho M, Prasanth KR, Pogany J, Nagy PD. Targeting conserved co-opted host factors to block virus replication: Using allosteric inhibitors of the cytosolic Hsp70s to interfere with tomato bushy stunt virus replication. Virology 2021; 563:1-19. [PMID: 34399236 DOI: 10.1016/j.virol.2021.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 11/21/2022]
Abstract
To further our understanding of the pro-viral roles of the host cytosolic heat shock protein 70 (Hsp70) family, we chose the conserved Arabidopsis thaliana Hsp70-2 and the unique Erd2 (early response to dehydration 2), which contain Hsp70 domains. Based on in vitro studies with purified components, we show that AtHsp70-2 and AtErd2 perform pro-viral functions equivalent to that of the yeast Ssa1 Hsp70. These functions include activation of the tombusvirus RdRp, and stimulation of replicase assembly. Yeast-based complementation studies demonstrate that AtHsp70-2 or AtErd2 are present in the purified tombusvirus replicase. RNA silencing and over-expression studies in Nicotiana benthamiana suggest that both Hsp70-2 and Erd2 are co-opted by tomato bushy stunt virus (TBSV). Moreover, we used allosteric inhibitors of Hsp70s to inhibit replication of TBSV and related plant viruses in plants. Altogether, interfering with the functions of the co-opted Hsp70s could be an effective antiviral approach against tombusviruses in plants.
Collapse
Affiliation(s)
- Melissa Molho
- Department of Plant Pathology, University of Kentucky, Lexington, KY, 40546, USA
| | - K Reddisiva Prasanth
- Department of Plant Pathology, University of Kentucky, Lexington, KY, 40546, USA
| | - Judit Pogany
- Department of Plant Pathology, University of Kentucky, Lexington, KY, 40546, USA
| | - Peter D Nagy
- Department of Plant Pathology, University of Kentucky, Lexington, KY, 40546, USA.
| |
Collapse
|
39
|
Arhar T, Shkedi A, Nadel CM, Gestwicki JE. The interactions of molecular chaperones with client proteins: why are they so weak? J Biol Chem 2021; 297:101282. [PMID: 34624315 PMCID: PMC8567204 DOI: 10.1016/j.jbc.2021.101282] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 12/30/2022] Open
Abstract
The major classes of molecular chaperones have highly variable sequences, sizes, and shapes, yet they all bind to unfolded proteins, limit their aggregation, and assist in their folding. Despite the central importance of this process to protein homeostasis, it has not been clear exactly how chaperones guide this process or whether the diverse families of chaperones use similar mechanisms. For the first time, recent advances in NMR spectroscopy have enabled detailed studies of how unfolded, "client" proteins interact with both ATP-dependent and ATP-independent classes of chaperones. Here, we review examples from four distinct chaperones, Spy, Trigger Factor, DnaK, and HscA-HscB, highlighting the similarities and differences between their mechanisms. One striking similarity is that the chaperones all bind weakly to their clients, such that the chaperone-client interactions are readily outcompeted by stronger, intra- and intermolecular contacts in the folded state. Thus, the relatively weak affinity of these interactions seems to provide directionality to the folding process. However, there are also key differences, especially in the details of how the chaperones release clients and how ATP cycling impacts that process. For example, Spy releases clients in a largely folded state, while clients seem to be unfolded upon release from Trigger Factor or DnaK. Together, these studies are beginning to uncover the similarities and differences in how chaperones use weak interactions to guide protein folding.
Collapse
Affiliation(s)
- Taylor Arhar
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco California, USA
| | - Arielle Shkedi
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco California, USA
| | - Cory M Nadel
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco California, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Disease, University of California San Francisco, San Francisco California, USA.
| |
Collapse
|
40
|
Pharmacological targeting of endoplasmic reticulum stress in disease. Nat Rev Drug Discov 2021; 21:115-140. [PMID: 34702991 DOI: 10.1038/s41573-021-00320-3] [Citation(s) in RCA: 270] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2021] [Indexed: 02/08/2023]
Abstract
The accumulation of misfolded proteins in the endoplasmic reticulum (ER) leads to ER stress, resulting in activation of the unfolded protein response (UPR) that aims to restore protein homeostasis. However, the UPR also plays an important pathological role in many diseases, including metabolic disorders, cancer and neurological disorders. Over the last decade, significant effort has been invested in targeting signalling proteins involved in the UPR and an array of drug-like molecules is now available. However, these molecules have limitations, the understanding of which is crucial for their development into therapies. Here, we critically review the existing ER stress and UPR-directed drug-like molecules, highlighting both their value and their limitations.
Collapse
|
41
|
Small-molecule inhibitor targeting the Hsp70-Bim protein-protein interaction in CML cells overcomes BCR-ABL-independent TKI resistance. Leukemia 2021; 35:2862-2874. [PMID: 34007045 DOI: 10.1038/s41375-021-01283-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/21/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
Herein, we screened a novel inhibitor of the Hsp70-Bim protein-protein interaction (PPI), S1g-2, from a Bcl-2 inhibitor library; this compound specifically disrupted the Hsp70-Bim PPI by direct binding to an unknown site adjacent to that of an allosteric Hsp70 inhibitor MKT-077, showing binding affinity in sub-μM concentration range. S1g-2 exhibited overall 5-10-fold higher apoptosis-inducing activity in CML cells, primary CML blasts, and BCR-ABL-transformed BaF3 cells than other cancer cells, normal lymphocytes, and BaF3 cells, illustrating Hsp70-Bim PPI driven by BCR-ABL protects CML through oncoclient proteins that enriched in three pathways: eIF2 signaling, the regulation of eIF4E and p70S6K signaling, and the mTOR signaling pathways. Moreover, S1g-2 progressively enhanced lethality along with the increase in BCR-ABL-independent TKI resistance in the K562 cell lines and is more effective in primary samples from BCR-ABL-independent TKI-resistant patients than those from TKI-sensitive patients. By comparing the underlying mechanisms of S1g-2, MKT-077, and an ATP-competitive Hsp70 inhibitor VER-155008, the Hsp70-Bim PPI was identified to be a CML-specific target to protect from TKIs through the above three oncogenic signaling pathways. The in vivo activity against CML and low toxicity endows S1g-2 a first-in-class promising drug candidate for both TKI-sensitive and resistant CML.
Collapse
|
42
|
Daniyan MO. Heat Shock Proteins as Targets for Novel Antimalarial Drug Discovery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1340:205-236. [PMID: 34569027 DOI: 10.1007/978-3-030-78397-6_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Plasmodium falciparum, the parasitic agent that is responsible for a severe and dangerous form of human malaria, has a history of long years of cohabitation with human beings with attendant negative consequences. While there have been some gains in the fight against malaria through the application of various control measures and the use of chemotherapeutic agents, and despite the global decline in malaria cases and associated deaths, the continual search for new and effective therapeutic agents is key to achieving sustainable development goals. An important parasite survival strategy, which is also of serious concern to the scientific community, is the rate at which the parasites continually develop resistance to drugs. Among the key players in the parasite's ability to develop resistance, maintain cellular integrity, and survives within an unusual environment of the red blood cells are the molecular chaperones of the heat shock proteins (HSP) family. HSPs constitute a novel avenue for antimalarial drug discovery and by exploring their ubiquitous nature and multifunctional activities, they may be suitable targets for the discovery of multi-targets antimalarial drugs, needed to fight incessant drug resistance. In this chapter, features of selected families of plasmodial HSPs that can be exploited in drug discovery are presented. Also, known applications of HSPs in small molecule screening, their potential usefulness in high throughput drug screening, as well as possible challenges are highlighted.
Collapse
Affiliation(s)
- Michael Oluwatoyin Daniyan
- Department of Pharmacology, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria.
| |
Collapse
|
43
|
Sannino S, Yates ME, Schurdak ME, Oesterreich S, Lee AV, Wipf P, Brodsky JL. Unique integrated stress response sensors regulate cancer cell susceptibility when Hsp70 activity is compromised. eLife 2021; 10:64977. [PMID: 34180400 PMCID: PMC8275131 DOI: 10.7554/elife.64977] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/27/2021] [Indexed: 12/11/2022] Open
Abstract
Molecular chaperones, such as Hsp70, prevent proteotoxicity and maintain homeostasis. This is perhaps most evident in cancer cells, which overexpress Hsp70 and thrive even when harboring high levels of misfolded proteins. To define the response to proteotoxic challenges, we examined adaptive responses in breast cancer cells in the presence of an Hsp70 inhibitor. We discovered that the cells bin into distinct classes based on inhibitor sensitivity. Strikingly, the most resistant cells have higher autophagy levels, and autophagy was maximally activated only in resistant cells upon Hsp70 inhibition. In turn, resistance to compromised Hsp70 function required the integrated stress response transducer, GCN2, which is commonly associated with amino acid starvation. In contrast, sensitive cells succumbed to Hsp70 inhibition by activating PERK. These data reveal an unexpected route through which breast cancer cells adapt to proteotoxic insults and position GCN2 and autophagy as complementary mechanisms to ensure survival when proteostasis is compromised.
Collapse
Affiliation(s)
- Sara Sannino
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, United States
| | - Megan E Yates
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women Research Institute, Pittsburgh, United States.,Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, United States.,Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Mark E Schurdak
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, United States.,University of Pittsburgh Drug Discovery Institute, Pittsburgh, United States
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women Research Institute, Pittsburgh, United States.,Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, United States.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Adrian V Lee
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women Research Institute, Pittsburgh, United States.,Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, United States.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, United States
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, United States
| |
Collapse
|
44
|
Mayer MP. The Hsp70-Chaperone Machines in Bacteria. Front Mol Biosci 2021; 8:694012. [PMID: 34164436 PMCID: PMC8215388 DOI: 10.3389/fmolb.2021.694012] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/20/2021] [Indexed: 12/02/2022] Open
Abstract
The ATP-dependent Hsp70s are evolutionary conserved molecular chaperones that constitute central hubs of the cellular protein quality surveillance network. None of the other main chaperone families (Tig, GroELS, HtpG, IbpA/B, ClpB) have been assigned with a comparable range of functions. Through a multitude of functions Hsp70s are involved in many cellular control circuits for maintaining protein homeostasis and have been recognized as key factors for cell survival. Three mechanistic properties of Hsp70s are the basis for their high versatility. First, Hsp70s bind to short degenerate sequence motifs within their client proteins. Second, Hsp70 chaperones switch in a nucleotide-controlled manner between a state of low affinity for client proteins and a state of high affinity for clients. Third, Hsp70s are targeted to their clients by a large number of cochaperones of the J-domain protein (JDP) family and the lifetime of the Hsp70-client complex is regulated by nucleotide exchange factors (NEF). In this review I will discuss advances in the understanding of the molecular mechanism of the Hsp70 chaperone machinery focusing mostly on the bacterial Hsp70 DnaK and will compare the two other prokaryotic Hsp70s HscA and HscC with DnaK.
Collapse
Affiliation(s)
- Matthias P Mayer
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH-Alliance, Heidelberg, Germany
| |
Collapse
|
45
|
Ambrose AJ, Chapman E. Function, Therapeutic Potential, and Inhibition of Hsp70 Chaperones. J Med Chem 2021; 64:7060-7082. [PMID: 34009983 DOI: 10.1021/acs.jmedchem.0c02091] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hsp70s are among the most highly conserved proteins in all of biology. Through an iterative binding and release of exposed hydrophobic residues on client proteins, Hsp70s can prevent aggregation and promote folding to the native state of their client proteins. The human proteome contains eight canonical Hsp70s. Because Hsp70s are relatively promiscuous they play a role in folding a large proportion of the proteome. Hsp70s are implicated in disease through their ability to regulate protein homeostasis. In recent years, researchers have attempted to develop selective inhibitors of Hsp70 isoforms to better understand the role of individual isoforms in biology and as potential therapeutics. Selective inhibitors have come from rational design, forced localization, and serendipity, but the development of completely selective inhibitors remains elusive. In the present review, we discuss the Hsp70 structure and function, the known Hsp70 client proteins, the role of Hsp70s in disease, and current efforts to discover Hsp70 modulators.
Collapse
Affiliation(s)
- Andrew J Ambrose
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, Tucson, Arizona 85721, United States
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, 1703 East Mabel Street, Tucson, Arizona 85721, United States
| |
Collapse
|
46
|
Wang Z, Song T, Guo Z, Uwituze LB, Guo Y, Zhang H, Wang H, Zhang X, Pan H, Ji T, Yin F, Zhou S, Dai J, Zhang Z. A novel Hsp70 inhibitor specifically targeting the cancer-related Hsp70-Bim protein-protein interaction. Eur J Med Chem 2021; 220:113452. [PMID: 33906046 DOI: 10.1016/j.ejmech.2021.113452] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/29/2021] [Accepted: 04/02/2021] [Indexed: 12/21/2022]
Abstract
Targeting cancer-related Hsp70-Bim protein-protein interactions (PPIs) offers a new strategy for the design of Hsp70 inhibitors. Herein, we discovered a novel Hsp70 inhibitor, S1g-6, based on the established BH3 mimetics. S1g-6 exhibited sub-μM binding affinity toward Hsp70 and selectively disrupted Hsp70-Bim PPI. The target specificity of S1g-6in situ was validated by affinity-based protein profiling, co-immunoprecipitation, and cell-based shRNA assays. S1g-6 specifically antagonized the ATPase activity of Hsp70 upon recruiting Bim and showed selective apoptosis induction in some cancer cell lines over normal ones through suppression of some oncogenic clients of Hsp70, representing a new class of antitumor candidates. Hsp70-Bim PPI exhibited cancer-dependent role as a potential anti-cancer target.
Collapse
Affiliation(s)
- Ziqian Wang
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Ting Song
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| | - Zongwei Guo
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Laura B Uwituze
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Yafei Guo
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Hong Zhang
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Hang Wang
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Xiaodong Zhang
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Hao Pan
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Tong Ji
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Fangkui Yin
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Sheng Zhou
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Jian Dai
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Zhichao Zhang
- State Key Laboratory of Fine Chemicals, Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| |
Collapse
|
47
|
Tavella TA, da Silva NSM, Spillman N, Kayano ACAV, Cassiano GC, Vasconcelos AA, Camargo AP, da Silva DCB, Fontinha D, Salazar Alvarez LC, Ferreira LT, Peralis Tomaz KC, Neves BJ, Almeida LD, Bargieri DY, Lacerda MVGD, Lemos Cravo PV, Sunnerhagen P, Prudêncio M, Andrade CH, Pinto Lopes SC, Carazzolle MF, Tilley L, Bilsland E, Borges JC, Maranhão Costa FT. Violacein-Induced Chaperone System Collapse Underlies Multistage Antiplasmodial Activity. ACS Infect Dis 2021; 7:759-776. [PMID: 33689276 PMCID: PMC8042658 DOI: 10.1021/acsinfecdis.0c00454] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Antimalarial drugs with novel modes of action and wide therapeutic potential are needed to pave the way for malaria eradication. Violacein is a natural compound known for its biological activity against cancer cells and several pathogens, including the malaria parasite, Plasmodium falciparum (Pf). Herein, using chemical genomic profiling (CGP), we found that violacein affects protein homeostasis. Mechanistically, violacein binds Pf chaperones, PfHsp90 and PfHsp70-1, compromising the latter's ATPase and chaperone activities. Additionally, violacein-treated parasites exhibited increased protein unfolding and proteasomal degradation. The uncoupling of the parasite stress response reflects the multistage growth inhibitory effect promoted by violacein. Despite evidence of proteotoxic stress, violacein did not inhibit global protein synthesis via UPR activation-a process that is highly dependent on chaperones, in agreement with the notion of a violacein-induced proteostasis collapse. Our data highlight the importance of a functioning chaperone-proteasome system for parasite development and differentiation. Thus, a violacein-like small molecule might provide a good scaffold for development of a novel probe for examining the molecular chaperone network and/or antiplasmodial drug design.
Collapse
Affiliation(s)
- Tatyana Almeida Tavella
- Laboratory of Tropical Diseases−Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas−UNICAMP, Campinas, SP 13083-970, Brazil
| | - Noeli Soares Melo da Silva
- Biochemistry and Biophysics of Proteins Group−São Carlos Institute of Chemistry−IQSC, University of São Paulo, Trabalhador Sancarlense Avenue, 400, BQ1, S27, São Carlos, SP 13566-590, Brazil
| | - Natalie Spillman
- Department of Biochemistry, Bio 21 Institute, University of Melbourne, 30 Flemington Rd, Parkville, Melbourne,VIC 3052, Australia
| | - Ana Carolina Andrade Vitor Kayano
- Laboratory of Tropical Diseases−Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas−UNICAMP, Campinas, SP 13083-970, Brazil
| | - Gustavo Capatti Cassiano
- Laboratory of Tropical Diseases−Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas−UNICAMP, Campinas, SP 13083-970, Brazil
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, 1099-085 Lisboa, Portugal
| | - Adrielle Ayumi Vasconcelos
- Laboratory of Genomics and BioEnergy, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas−UNICAMP, Campinas, SP 13083-970, Brazil
| | - Antônio Pedro Camargo
- Laboratory of Genomics and BioEnergy, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas−UNICAMP, Campinas, SP 13083-970, Brazil
| | - Djane Clarys Baia da Silva
- Leônidas & Maria Deane Institute, Fundação Oswaldo Cruz−FIOCRUZ, Manaus , AM 69057070, Brazil
- Fundação de Medicina Tropical−Dr. Heitor Vieira Dourado, Manaus, AM 69040-000, Brazil
| | - Diana Fontinha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisboa, Portugal
| | - Luis Carlos Salazar Alvarez
- Laboratory of Tropical Diseases−Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas−UNICAMP, Campinas, SP 13083-970, Brazil
| | - Letícia Tiburcio Ferreira
- Laboratory of Tropical Diseases−Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas−UNICAMP, Campinas, SP 13083-970, Brazil
| | - Kaira Cristina Peralis Tomaz
- Laboratory of Tropical Diseases−Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas−UNICAMP, Campinas, SP 13083-970, Brazil
| | - Bruno Junior Neves
- Laboratory of Molecular Modeling and Drug Design, LabMol, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO 74605-170, Brazil
- LabChem−Laboratory of Cheminformatics, Centro Universitário de Anápolis−UniEVANGÉLICA, Anápolis, GO 75083-515, Brazil
| | - Ludimila Dias Almeida
- Synthetic Biology Laboratory, Department of Structural and Functional Biology, Institute of Biology, UNICAMP, Campinas, SP Brazil
| | - Daniel Youssef Bargieri
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, Cidade Universitária “Armando Salles Oliveira”, São Paulo 05508-000, Brazil
| | | | - Pedro Vitor Lemos Cravo
- LabChem−Laboratory of Cheminformatics, Centro Universitário de Anápolis−UniEVANGÉLICA, Anápolis, GO 75083-515, Brazil
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, 1099-085 Lisboa, Portugal
| | - Per Sunnerhagen
- Department of Chemistry and Molecular Biology, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisboa, Portugal
| | - Carolina Horta Andrade
- Laboratory of Tropical Diseases−Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas−UNICAMP, Campinas, SP 13083-970, Brazil
- Laboratory of Molecular Modeling and Drug Design, LabMol, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO 74605-170, Brazil
| | - Stefanie Costa Pinto Lopes
- Leônidas & Maria Deane Institute, Fundação Oswaldo Cruz−FIOCRUZ, Manaus , AM 69057070, Brazil
- Fundação de Medicina Tropical−Dr. Heitor Vieira Dourado, Manaus, AM 69040-000, Brazil
| | - Marcelo Falsarella Carazzolle
- Laboratory of Genomics and BioEnergy, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas−UNICAMP, Campinas, SP 13083-970, Brazil
| | - Leann Tilley
- Department of Biochemistry, Bio 21 Institute, University of Melbourne, 30 Flemington Rd, Parkville, Melbourne,VIC 3052, Australia
| | - Elizabeth Bilsland
- Synthetic Biology Laboratory, Department of Structural and Functional Biology, Institute of Biology, UNICAMP, Campinas, SP Brazil
| | - Júlio César Borges
- Biochemistry and Biophysics of Proteins Group−São Carlos Institute of Chemistry−IQSC, University of São Paulo, Trabalhador Sancarlense Avenue, 400, BQ1, S27, São Carlos, SP 13566-590, Brazil
| | - Fabio Trindade Maranhão Costa
- Laboratory of Tropical Diseases−Prof. Dr. Luiz Jacinto da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas−UNICAMP, Campinas, SP 13083-970, Brazil
| |
Collapse
|
48
|
Horianopoulos LC, Kronstad JW. Chaperone Networks in Fungal Pathogens of Humans. J Fungi (Basel) 2021; 7:209. [PMID: 33809191 PMCID: PMC7998936 DOI: 10.3390/jof7030209] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
The heat shock proteins (HSPs) function as chaperones to facilitate proper folding and modification of proteins and are of particular importance when organisms are subjected to unfavourable conditions. The human fungal pathogens are subjected to such conditions within the context of infection as they are exposed to human body temperature as well as the host immune response. Herein, the roles of the major classes of HSPs are briefly reviewed and their known contributions in human fungal pathogens are described with a focus on Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus. The Hsp90s and Hsp70s in human fungal pathogens broadly contribute to thermotolerance, morphological changes required for virulence, and tolerance to antifungal drugs. There are also examples of J domain co-chaperones and small HSPs influencing the elaboration of virulence factors in human fungal pathogens. However, there are diverse members in these groups of chaperones and there is still much to be uncovered about their contributions to pathogenesis. These HSPs do not act in isolation, but rather they form a network with one another. Interactions between chaperones define their specific roles and enhance their protein folding capabilities. Recent efforts to characterize these HSP networks in human fungal pathogens have revealed that there are unique interactions relevant to these pathogens, particularly under stress conditions. The chaperone networks in the fungal pathogens are also emerging as key coordinators of pathogenesis and antifungal drug tolerance, suggesting that their disruption is a promising strategy for the development of antifungal therapy.
Collapse
Affiliation(s)
| | - James W. Kronstad
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
| |
Collapse
|
49
|
Dutta T, Singh H, Gestwicki JE, Blatch GL. Exported plasmodial J domain protein, PFE0055c, and PfHsp70-x form a specific co-chaperone-chaperone partnership. Cell Stress Chaperones 2021; 26:355-366. [PMID: 33236291 PMCID: PMC7925779 DOI: 10.1007/s12192-020-01181-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 11/06/2020] [Accepted: 11/13/2020] [Indexed: 12/11/2022] Open
Abstract
Plasmodium falciparum is a unicellular protozoan parasite and causative agent of a severe form of malaria in humans, accounting for very high worldwide fatality rates. At the molecular level, survival of the parasite within the human host is mediated by P. falciparum heat shock proteins (PfHsps) that provide protection during febrile episodes. The ATP-dependent chaperone activity of Hsp70 relies on the co-chaperone J domain protein (JDP), with which it forms a chaperone-co-chaperone complex. The exported P. falciparum JDP (PfJDP), PFA0660w, has been shown to stimulate the ATPase activity of the exported chaperone, PfHsp70-x. Furthermore, PFA0660w has been shown to associate with another exported PfJDP, PFE0055c, and PfHsp70-x in J-dots, highly mobile structures found in the infected erythrocyte cytosol. Therefore, the present study aims to conduct a structural and functional characterization of the full-length exported PfJDP, PFE0055c. Recombinant PFE0055c was successfully expressed and purified and found to stimulate the basal ATPase activity of PfHsp70-x to a greater extent than PFA0660w but, like PFA0660w, did not significantly stimulate the basal ATPase activity of human Hsp70. Small-molecule inhibition assays were conducted to determine the effect of known inhibitors of JDPs (chalcone, C86) and Hsp70 (benzothiazole rhodacyanines, JG231 and JG98) on the basal and PFE0055c-stimulated ATPase activity of PfHsp70-x. In this study, JG231 and JG98 were found to inhibit both the basal and PFE0055c-stimulated ATPase activity of PfHsp70-x. C86 only inhibited the PFE0055c-stimulated ATPase activity of PfHsp70-x, consistent with PFE0055c binding to PfHsp70-x through its J domain. This research has provided further insight into the molecular basis of the interaction between these exported plasmodial chaperones, which could inform future antimalarial drug discovery studies.
Collapse
Affiliation(s)
- Tanima Dutta
- The Vice Chancellery, The University of Notre Dame Australia, Fremantle, WA, Australia
- The Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
| | - Harpreet Singh
- Department of Bioinformatics, Hans Raj Mahila Maha Vidyalaya, Jalandhar, Punjab, India
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Gregory L Blatch
- The Vice Chancellery, The University of Notre Dame Australia, Fremantle, WA, Australia.
- The Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia.
| |
Collapse
|
50
|
Serapian SA, Triveri A, Marchetti F, Castelli M, Colombo G. Exploiting Folding and Degradation Machineries To Target Undruggable Proteins: What Can a Computational Approach Tell Us? ChemMedChem 2021; 16:1593-1599. [PMID: 33443306 DOI: 10.1002/cmdc.202000960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Indexed: 01/03/2023]
Abstract
Advances in genomics and proteomics have unveiled an ever-growing number of key proteins and provided mechanistic insights into the genesis of pathologies. This wealth of data showed that changes in expression levels of specific proteins, mutations, and post-translational modifications can result in (often subtle) perturbations of functional protein-protein interaction networks, which ultimately determine disease phenotypes. Although many such validated pathogenic proteins have emerged as ideal drug targets, there are also several that escape traditional pharmacological regulation; these proteins have thus been labeled "undruggable". The challenges posed by undruggable targets call for new sorts of molecular intervention. One fascinating solution is to perturb a pathogenic protein's expression levels, rather than blocking its activities. In this Concept paper, we shall discuss chemical interventions aimed at recruiting undruggable proteins to the ubiquitin proteasome system, or aimed at disrupting protein-protein interactions in the chaperone-mediated cellular folding machinery: both kinds of intervention lead to a decrease in the amount of active pathogenic protein expressed. Specifically, we shall discuss the role of computational strategies in understanding the molecular determinants characterizing the function of synthetic molecules typically designed for either type of intervention. Finally, we shall provide our perspectives and views on the current limitations and possibilities to expand the scope of rational approaches to the design of chemical regulators of protein levels.
Collapse
Affiliation(s)
- Stefano A Serapian
- Department of Chemistry, University of Pavia, Via Taramelli 12, 27100, Pavia, Italy
| | - Alice Triveri
- Department of Chemistry, University of Pavia, Via Taramelli 12, 27100, Pavia, Italy
| | - Filippo Marchetti
- Department of Chemistry, University of Pavia, Via Taramelli 12, 27100, Pavia, Italy
| | - Matteo Castelli
- Department of Chemistry, University of Pavia, Via Taramelli 12, 27100, Pavia, Italy
| | - Giorgio Colombo
- Department of Chemistry, University of Pavia, Via Taramelli 12, 27100, Pavia, Italy
| |
Collapse
|