1
|
Van Pelt KM, Truttmann MC. Loss of FIC-1-mediated AMPylation activates the UPR ER and upregulates cytosolic HSP70 chaperones to suppress polyglutamine toxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625751. [PMID: 39651313 PMCID: PMC11623694 DOI: 10.1101/2024.11.27.625751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Targeted regulation of cellular proteostasis machinery represents a promising strategy for the attenuation of pathological protein aggregation. Recent work suggests that the unfolded protein response in the endoplasmic reticulum (UPR ER ) directly regulates the aggregation and toxicity of expanded polyglutamine (polyQ) proteins. However, the mechanisms underlying this phenomenon remain poorly understood. In this study, we report that perturbing ER homeostasis in Caenorhabditis elegans through the depletion of either BiP ortholog, hsp-3 or hsp-4, causes developmental arrest in worms expressing aggregation-prone polyQ proteins. This phenotype is rescued by the genetic deletion of the conserved UPR ER regulator, FIC-1. We demonstrate that the beneficial effects of fic-1 knock-out (KO) extend into adulthood, where the loss of FIC-1-mediated protein AMPylation in polyQ-expressing animals is sufficient to prevent declines in fitness and lifespan. We further show that loss of hsp-3 and hsp-4 leads to distinct, but complementary transcriptomic responses to ER stress involving all three UPR ER stress sensors (IRE-1, PEK-1, and ATF-6). We identify the cytosolic HSP70 family chaperone F44E5.4 , whose expression is increased in fic-1 -deficient animals upon ER dysregulation, as a key effector suppressing polyQ toxicity. Over-expression of F44E5.4 , but not other HSP70 family chaperones, is sufficient to rescue developmental arrest in polyQ-expressing embryos upon hsp-3 knock-down. Finally, we show that knock-down of ire-1 , pek-1 , or atf-6 blocks the upregulation of F44E5.4 in fic-1 -deficient worms. Taken together, our findings support a model in which the loss of FIC-1-mediated AMPylation engages UPR ER signaling to upregulate cytosolic chaperone activity in response to polyQ toxicity.
Collapse
|
2
|
Bej E, Cesare P, d'Angelo M, Volpe AR, Castelli V. Neuronal Cell Rearrangement During Aging: Antioxidant Compounds as a Potential Therapeutic Approach. Cells 2024; 13:1945. [PMID: 39682694 DOI: 10.3390/cells13231945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/02/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Aging is a natural process that leads to time-related changes and a decrease in cognitive abilities, executive functions, and attention. In neuronal aging, brain cells struggle to respond to oxidative stress. The structure, function, and survival of neurons can be mediated by different pathways that are sensitive to oxidative stress and age-related low-energy states. Mitochondrial impairment is one of the most noticeable signs of brain aging. Damaged mitochondria are thought to be one of the main causes that feed the inflammation related to aging. Also, protein turnover is involved in age-related impairments. The brain, due to its high oxygen usage, is particularly susceptible to oxidative damage. This review explores the mechanisms underlying neuronal cell rearrangement during aging, focusing on morphological changes that contribute to cognitive decline and increased susceptibility to neurodegenerative diseases. Potential therapeutic approaches are discussed, including the use of antioxidants (e.g., Vitamin C, Vitamin E, glutathione, carotenoids, quercetin, resveratrol, and curcumin) to mitigate oxidative damage, enhance mitochondrial function, and maintain protein homeostasis. This comprehensive overview aims to provide insights into the cellular and molecular processes of neuronal aging and highlight promising therapeutic avenues to counteract age-related neuronal deterioration.
Collapse
Affiliation(s)
- Erjola Bej
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
- Department of the Chemical-Toxicological and Pharmacological Evaluation of Drugs, Faculty of Pharmacy, Catholic University Our Lady of Good Counsel, 1001 Tirana, Albania
| | - Patrizia Cesare
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Anna Rita Volpe
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
- Department of the Chemical-Toxicological and Pharmacological Evaluation of Drugs, Faculty of Pharmacy, Catholic University Our Lady of Good Counsel, 1001 Tirana, Albania
| |
Collapse
|
3
|
Baron KR, Oviedo S, Krasny S, Zaman M, Aldakhlallah R, Bora P, Mathur P, Pfeffer G, Bollong MJ, Shutt TE, Grotjahn DA, Wiseman RL. Pharmacologic Activation of Integrated Stress Response Kinases Inhibits Pathologic Mitochondrial Fragmentation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598126. [PMID: 38915623 PMCID: PMC11195119 DOI: 10.1101/2024.06.10.598126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Excessive mitochondrial fragmentation is associated with the pathologic mitochondrial dysfunction implicated in the pathogenesis of etiologically-diverse diseases, including many neurodegenerative disorders. The integrated stress response (ISR) - comprising the four eIF2α kinases PERK, GCN2, PKR, and HRI - is a prominent stress-responsive signaling pathway that regulates mitochondrial morphology and function in response to diverse types of pathologic insult. This suggests that pharmacologic activation of the ISR represents a potential strategy to mitigate pathologic mitochondrial fragmentation associated with human disease. Here, we show that pharmacologic activation of the ISR kinases HRI or GCN2 promotes adaptive mitochondrial elongation and prevents mitochondrial fragmentation induced by the calcium ionophore ionomycin. Further, we show that pharmacologic activation of the ISR reduces mitochondrial fragmentation and restores basal mitochondrial morphology in patient fibroblasts expressing the pathogenic D414V variant of the pro-fusion mitochondrial GTPase MFN2 associated with neurological dysfunctions including ataxia, optic atrophy, and sensorineural hearing loss. These results identify pharmacologic activation of ISR kinases as a potential strategy to prevent pathologic mitochondrial fragmentation induced by disease-relevant chemical and genetic insults, further motivating the pursuit of highly selective ISR kinase-activating compounds as a therapeutic strategy to mitigate mitochondrial dysfunction implicated in diverse human diseases.
Collapse
Affiliation(s)
- Kelsey R. Baron
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
- These authors contributed equally
| | - Samantha Oviedo
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
- Department of Integrative Structural and Computation Biology, The Scripps Research Institute, La Jolla, CA 92037
- These authors contributed equally
| | - Sophia Krasny
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Mashiat Zaman
- Department of Biochemistry and Molecular Biology, Cummings School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rama Aldakhlallah
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Prerona Bora
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Prakhyat Mathur
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Gerald Pfeffer
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary; Alberta Child Health Research Institute, Department of Medical Genetics, Cumming School of Medicine, University of Calgary
| | - Michael J. Bollong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Timothy E. Shutt
- Departments of Medical Genetics and Biochemistry & Molecular Biology, Cumming School of Medicine, Hotchkiss Brain Institute, Snyder Institute for Chronic Diseases, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Danielle A. Grotjahn
- Department of Integrative Structural and Computation Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - R. Luke Wiseman
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
4
|
Williams D, Glasstetter LM, Jong TT, Chen T, Kapoor A, Zhu S, Zhu Y, Calvo R, Gehrlein A, Wong K, Hogan AN, Vocadlo DJ, Jagasia R, Marugan JJ, Sidransky E, Henderson MJ, Chen Y. High-throughput screening for small-molecule stabilizers of misfolded glucocerebrosidase in Gaucher disease and Parkinson's disease. Proc Natl Acad Sci U S A 2024; 121:e2406009121. [PMID: 39388267 PMCID: PMC11494340 DOI: 10.1073/pnas.2406009121] [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: 03/25/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
Glucocerebrosidase (GCase) is implicated in both a rare, monogenic disorder (Gaucher disease, GD) and a common, multifactorial condition (Parkinson's disease, PD); hence, it is an urgent therapeutic target. To identify correctors of severe protein misfolding and trafficking obstruction manifested by the pathogenic L444P-variant of GCase, we developed a suite of quantitative, high-throughput, cell-based assays. First, we labeled GCase with a small proluminescent HiBiT peptide reporter tag, enabling quantitation of protein stabilization in cells while faithfully maintaining target biology. TALEN-based gene editing allowed for stable integration of a single HiBiT-GBA1 transgene into an intragenic safe-harbor locus in GBA1-knockout H4 (neuroglioma) cells. This GD cell model was amenable to lead discovery via titration-based quantitative high-throughput screening and lead optimization via structure-activity relationships. A primary screen of 10,779 compounds from the NCATS bioactive collections identified 140 stabilizers of HiBiT-GCase-L444P, including both pharmacological chaperones (ambroxol and noninhibitory chaperone NCGC326) and proteostasis regulators (panobinostat, trans-ISRIB, and pladienolide B). Two complementary high-content imaging-based assays were deployed to triage hits: The fluorescence-quenched substrate LysoFix-GBA captured functional lysosomal GCase activity, while an immunofluorescence assay featuring antibody hGCase-1/23 directly visualized GCase lysosomal translocation. NCGC326 was active in both secondary assays and completely reversed pathological glucosylsphingosine accumulation. Finally, we tested the concept of combination therapy by demonstrating synergistic actions of NCGC326 with proteostasis regulators in enhancing GCase-L444P levels. Looking forward, these physiologically relevant assays can facilitate the identification, pharmacological validation, and medicinal chemistry optimization of small molecules targeting GCase, ultimately leading to a viable therapeutic for GD and PD.
Collapse
Affiliation(s)
- Darian Williams
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD20850
| | - Logan M. Glasstetter
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Tiffany T. Jong
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Tiffany Chen
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Abhijeet Kapoor
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD20850
| | - Sha Zhu
- Department of Chemistry, Simon Fraser University, Burnaby, BCV5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BCV5A 1S6, Canada
| | - Yanping Zhu
- Department of Chemistry, Simon Fraser University, Burnaby, BCV5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BCV5A 1S6, Canada
| | - Raul Calvo
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD20850
| | - Alexandra Gehrlein
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, 4070Basel, Switzerland
| | - Kimberly Wong
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Andrew N. Hogan
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - David J. Vocadlo
- Department of Chemistry, Simon Fraser University, Burnaby, BCV5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BCV5A 1S6, Canada
| | - Ravi Jagasia
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, 4070Basel, Switzerland
| | - Juan J. Marugan
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD20850
| | - Ellen Sidransky
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Mark J. Henderson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD20850
| | - Yu Chen
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD20892
| |
Collapse
|
5
|
Yang X, Zhao X, Zhao Z, Du J. Genome-wide analysis reveals transcriptional and translational changes during diapause of the Asian corn borer (Ostrinia furnacalis). BMC Biol 2024; 22:206. [PMID: 39272107 PMCID: PMC11401443 DOI: 10.1186/s12915-024-02000-1] [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: 12/14/2023] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Diapause, a pivotal phase in the insect life cycle, enables survival during harsh environmental conditions. Unraveling the gene expression profiles of the diapause process helps uncover the molecular mechanisms that underlying diapause, which is crucial for understanding physiological adaptations. In this study, we utilize RNA-seq and Ribo-seq data to examine differentially expressed genes (DEGs) and translational efficiency during diapause of Asian corn borer (Ostrinia furnacalis, ACB). RESULTS Our results unveil genes classified as "forwarded", "exclusive", "intensified", or "buffered" during diapause, shedding light on their transcription and translation regulation patterns. Furthermore, we explore the landscape of lncRNAs (long non-coding RNAs) during diapause and identify differentially expressed lncRNAs, suggesting their roles in diapause regulation. Comparative analysis of different types of diapause in insects uncovers shared and unique KEGG pathways. While shared pathways highlight energy balance, exclusive pathways in the ACB larvae indicate insect-specific adaptations related to nutrient utilization and stress response. Interestingly, our study also reveals dynamic changes in the HSP70 gene family and proteasome pathway during diapause. Manipulating HSP protein levels and proteasome pathway by HSP activator or inhibitor and proteasome inhibitor affects diapause, indicating their vital role in the process. CONCLUSIONS In summary, these findings enhance our knowledge of how insects navigate challenging conditions through intricate molecular mechanisms.
Collapse
Affiliation(s)
- Xingzhuo Yang
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Xianguo Zhao
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Zhangwu Zhao
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Juan Du
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
6
|
Chang KH, Chen CM. The Role of NRF2 in Trinucleotide Repeat Expansion Disorders. Antioxidants (Basel) 2024; 13:649. [PMID: 38929088 PMCID: PMC11200942 DOI: 10.3390/antiox13060649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Trinucleotide repeat expansion disorders, a diverse group of neurodegenerative diseases, are caused by abnormal expansions within specific genes. These expansions trigger a cascade of cellular damage, including protein aggregation and abnormal RNA binding. A key contributor to this damage is oxidative stress, an imbalance of reactive oxygen species that harms cellular components. This review explores the interplay between oxidative stress and the NRF2 pathway in these disorders. NRF2 acts as the master regulator of the cellular antioxidant response, orchestrating the expression of enzymes that combat oxidative stress. Trinucleotide repeat expansion disorders often exhibit impaired NRF2 signaling, resulting in inadequate responses to excessive ROS production. NRF2 activation has been shown to upregulate antioxidative gene expression, effectively alleviating oxidative stress damage. NRF2 activators, such as omaveloxolone, vatiquinone, curcumin, sulforaphane, dimethyl fumarate, and resveratrol, demonstrate neuroprotective effects by reducing oxidative stress in experimental cell and animal models of these diseases. However, translating these findings into successful clinical applications requires further research. In this article, we review the literature supporting the role of NRF2 in the pathogenesis of these diseases and the potential therapeutics of NRF2 activators.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Kueishan, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Kueishan, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
7
|
Akdag M, van Schijndel V, Sinnige T. Islet amyloid polypeptide tagged with green fluorescent protein localises to mitochondria and forms filamentous aggregates in Caenorhabditis elegans. Biophys Chem 2024; 307:107180. [PMID: 38241827 DOI: 10.1016/j.bpc.2024.107180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/22/2023] [Accepted: 01/12/2024] [Indexed: 01/21/2024]
Abstract
Type 2 diabetes (T2D) is the most common form of diabetes and represents a growing health concern. A characteristic feature of T2D is the aggregation of islet amyloid polypeptide (IAPP), which is thought to be associated with the death of pancreatic β-cells. Inhibiting IAPP aggregation is a promising therapeutic avenue to treat T2D, but the mechanisms of aggregation and toxicity are not yet fully understood. Caenorhabditis elegans is a well-characterised multicellular model organism that has been extensively used to study protein aggregation diseases. In this study, we aimed to develop a simple in vivo model to investigate IAPP aggregation and toxicity based on expression in the C. elegans body wall muscle cells. We show that IAPP tagged with green fluorescent protein (GFP) localises to mitochondria not only in muscle cells but also when expressed in the intestine, in line with previous observations in mouse and human pancreatic β-cells. The IAPP-GFP fusion protein forms solid aggregates, which have a filamentous appearance as seen by electron microscopy. However, the animals expressing IAPP-GFP in the body wall muscle cells do not display a strong motility phenotype, suggesting that the IAPP-GFP aggregates are not considerably toxic. Nevertheless, the mitochondrial localisation and aggregate formation may be useful read-outs to screen for IAPP-solubilizing compounds as a therapeutic strategy for T2D.
Collapse
Affiliation(s)
- Mehmet Akdag
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Vera van Schijndel
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Tessa Sinnige
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands.
| |
Collapse
|
8
|
Williams D, Glasstetter LM, Jong TT, Kapoor A, Zhu S, Zhu Y, Gehrlein A, Vocadlo DJ, Jagasia R, Marugan JJ, Sidransky E, Henderson MJ, Chen Y. Development of quantitative high-throughput screening assays to identify, validate, and optimize small-molecule stabilizers of misfolded β-glucocerebrosidase with therapeutic potential for Gaucher disease and Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586364. [PMID: 38712038 PMCID: PMC11071283 DOI: 10.1101/2024.03.22.586364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Glucocerebrosidase (GCase) is implicated in both a rare, monogenic disorder (Gaucher disease, GD) and a common, multifactorial condition (Parkinson's disease); hence, it is an urgent therapeutic target. To identify correctors of severe protein misfolding and trafficking obstruction manifested by the pathogenic L444P-variant of GCase, we developed a suite of quantitative, high-throughput, cell-based assays. First, we labeled GCase with a small pro-luminescent HiBiT peptide reporter tag, enabling quantitation of protein stabilization in cells while faithfully maintaining target biology. TALEN-based gene editing allowed for stable integration of a single HiBiT-GBA1 transgene into an intragenic safe-harbor locus in GBA1-knockout H4 (neuroglioma) cells. This GD cell model was amenable to lead discovery via titration-based quantitative high-throughput screening and lead optimization via structure-activity relationships. A primary screen of 10,779 compounds from the NCATS bioactive collections identified 140 stabilizers of HiBiT-GCase-L444P, including both pharmacological chaperones (ambroxol and non-inhibitory chaperone NCGC326) and proteostasis regulators (panobinostat, trans-ISRIB, and pladienolide B). Two complementary high-content imaging-based assays were deployed to triage hits: the fluorescence-quenched substrate LysoFix-GBA captured functional lysosomal GCase activity, while an immunofluorescence assay featuring antibody hGCase-1/23 provided direct visualization of GCase lysosomal translocation. NCGC326 was active in both secondary assays and completely reversed pathological glucosylsphingosine accumulation. Finally, we tested the concept of combination therapy, by demonstrating synergistic actions of NCGC326 with proteostasis regulators in enhancing GCase-L444P levels. Looking forward, these physiologically-relevant assays can facilitate the identification, pharmacological validation, and medicinal chemistry optimization of new chemical matter targeting GCase, ultimately leading to a viable therapeutic for two protein-misfolding diseases.
Collapse
Affiliation(s)
- Darian Williams
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Logan M. Glasstetter
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Tiffany T. Jong
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Abhijeet Kapoor
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Sha Zhu
- Department of Chemistry and Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Yanping Zhu
- Department of Chemistry and Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Alexandra Gehrlein
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - David J. Vocadlo
- Department of Chemistry and Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Ravi Jagasia
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Juan J. Marugan
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Ellen Sidransky
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Mark J. Henderson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Yu Chen
- Molecular Neurogenetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
9
|
Claeyssen C, Bulangalire N, Bastide B, Agbulut O, Cieniewski-Bernard C. Desmin and its molecular chaperone, the αB-crystallin: How post-translational modifications modulate their functions in heart and skeletal muscles? Biochimie 2024; 216:137-159. [PMID: 37827485 DOI: 10.1016/j.biochi.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/04/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023]
Abstract
Maintenance of the highly organized striated muscle tissue requires a cell-wide dynamic network through protein-protein interactions providing an effective mechanochemical integrator of morphology and function. Through a continuous and complex trans-cytoplasmic network, desmin intermediate filaments ensure this essential role in heart and in skeletal muscle. Besides their role in the maintenance of cell shape and architecture (permitting contractile activity efficiency and conferring resistance towards mechanical stress), desmin intermediate filaments are also key actors of cell and tissue homeostasis. Desmin participates to several cellular processes such as differentiation, apoptosis, intracellular signalisation, mechanotransduction, vesicle trafficking, organelle biogenesis and/or positioning, calcium homeostasis, protein homeostasis, cell adhesion, metabolism and gene expression. Desmin intermediate filaments assembly requires αB-crystallin, a small heat shock protein. Over its chaperone activity, αB-crystallin is involved in several cellular functions such as cell integrity, cytoskeleton stabilization, apoptosis, autophagy, differentiation, mitochondria function or aggresome formation. Importantly, both proteins are known to be strongly associated to the aetiology of several cardiac and skeletal muscles pathologies related to desmin filaments disorganization and a strong disturbance of desmin interactome. Note that these key proteins of cytoskeleton architecture are extensively modified by post-translational modifications that could affect their functional properties. Therefore, we reviewed in the herein paper the impact of post-translational modifications on the modulation of cellular functions of desmin and its molecular chaperone, the αB-crystallin.
Collapse
Affiliation(s)
- Charlotte Claeyssen
- University of Lille, University of Artois, University of Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France
| | - Nathan Bulangalire
- University of Lille, University of Artois, University of Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France; Université de Lille, CHU Lille, F-59000 Lille, France
| | - Bruno Bastide
- University of Lille, University of Artois, University of Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 75005, Paris, France
| | - Caroline Cieniewski-Bernard
- University of Lille, University of Artois, University of Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France.
| |
Collapse
|
10
|
Naus E, Derweduwe M, Lampi Y, Claeys A, Pauwels J, Langenberg T, Claes F, Xu J, Haemels V, Atak ZK, van der Kant R, Van Durme J, De Baets G, Ligon KL, Fiers M, Gevaert K, Aerts S, Rousseau F, Schymkowitz J, De Smet F. Reduced Levels of Misfolded and Aggregated Mutant p53 by Proteostatic Activation. Cells 2023; 12:cells12060960. [PMID: 36980299 PMCID: PMC10047295 DOI: 10.3390/cells12060960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 12/21/2022] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
In malignant cancer, excessive amounts of mutant p53 often lead to its aggregation, a feature that was recently identified as druggable. Here, we describe that induction of a heat shock-related stress response mediated by Foldlin, a small-molecule tool compound, reduces the protein levels of misfolded/aggregated mutant p53, while contact mutants or wild-type p53 remain largely unaffected. Foldlin also prevented the formation of stress-induced p53 nuclear inclusion bodies. Despite our inability to identify a specific molecular target, Foldlin also reduced protein levels of aggregating SOD1 variants. Finally, by screening a library of 778 FDA-approved compounds for their ability to reduce misfolded mutant p53, we identified the proteasome inhibitor Bortezomib with similar cellular effects as Foldlin. Overall, the induction of a cellular heat shock response seems to be an effective strategy to deal with pathological protein aggregation. It remains to be seen however, how this strategy can be translated to a clinical setting.
Collapse
Affiliation(s)
- Evelyne Naus
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Switch Laboratory, Department for Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Marleen Derweduwe
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium (V.H.); (K.G.)
| | - Youlia Lampi
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Switch Laboratory, Department for Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Annelies Claeys
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium (V.H.); (K.G.)
| | - Jarne Pauwels
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium;
- Department of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Tobias Langenberg
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Switch Laboratory, Department for Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Filip Claes
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Switch Laboratory, Department for Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Jie Xu
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Switch Laboratory, Department for Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Veerle Haemels
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium (V.H.); (K.G.)
| | - Zeynep Kalender Atak
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Laboratory of Computational Biology, Center for Human Genetics, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Rob van der Kant
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Switch Laboratory, Department for Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Joost Van Durme
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Switch Laboratory, Department for Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Greet De Baets
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Switch Laboratory, Department for Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Keith L. Ligon
- Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
- The Broad Institute, Cambridge, MA 02142, USA
- Department of Pathology, Division of Neuropathology, Brigham and Women’s Hospital and Children’s Hospital Boston, Boston, MA 02215, USA
- Department of Pathology, Harvard Medical School, Boston, MA 02215, USA
| | - Mark Fiers
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
| | - Kris Gevaert
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium (V.H.); (K.G.)
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium;
| | - Stein Aerts
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Laboratory of Computational Biology, Center for Human Genetics, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Frederic Rousseau
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Switch Laboratory, Department for Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Joost Schymkowitz
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; (E.N.); (Y.L.); (T.L.); (F.C.); (J.X.); (Z.K.A.); (R.v.d.K.); (J.V.D.); (G.D.B.); (M.F.); (S.A.); (F.R.); (J.S.)
- Switch Laboratory, Department for Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Frederik De Smet
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, Katholieke Universiteit Leuven, 3000 Leuven, Belgium (V.H.); (K.G.)
- Correspondence:
| |
Collapse
|
11
|
Lucaciu SA, Figliuzzi R, Neumann R, Nazarali S, Del Sordo L, Leighton SE, Hauser A, Shao Q, Johnston D, Bai D, Laird DW. GJB4 variants linked to skin disease exhibit a trafficking deficiency en route to gap junction formation that can be restored by co-expression of select connexins. Front Cell Dev Biol 2023; 11:1073805. [PMID: 36861039 PMCID: PMC9968944 DOI: 10.3389/fcell.2023.1073805] [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: 10/18/2022] [Accepted: 02/01/2023] [Indexed: 02/15/2023] Open
Abstract
Epidermal keratinocytes are enriched with at least nine connexins that are key regulators of epidermal homeostasis. The role of Cx30.3 in keratinocytes and epidermal health became evident when fourteen autosomal dominant mutations in the Cx30.3-encoding GJB4 gene were linked to a rare and incurable skin disorder called erythrokeratodermia variabilis et progressiva (EKVP). While these variants are linked to EKVP, they remain largely uncharacterized hindering therapeutic options. In this study, we characterize the expression and functional status of three EKVP-linked Cx30.3 mutants (G12D, T85P, and F189Y) in tissue-relevant and differentiation-competent rat epidermal keratinocytes. We found that GFP-tagged Cx30.3 mutants were non-functional likely due to their impaired trafficking and primary entrapment within the endoplasmic reticulum (ER). However, all mutants failed to increase BiP/GRP78 levels suggesting they were not inducing an unfolded protein response. FLAG-tagged Cx30.3 mutants were also trafficking impaired yet occasionally exhibited some capacity to assemble into gap junctions. The pathological impact of these mutants may extend beyond their trafficking deficiencies as keratinocytes expressing FLAG-tagged Cx30.3 mutants exhibited increased propidium iodide uptake in the absence of divalent cations. Attempts to rescue the delivery of trafficking impaired GFP-tagged Cx30.3 mutants into gap junctions by chemical chaperone treatment were ineffective. However, co-expression of wild type Cx30.3 greatly enhanced the assembly of Cx30.3 mutants into gap junctions, although endogenous levels of Cx30.3 do not appear to prevent the skin pathology found in patients harboring these autosomal dominant mutations. In addition, a spectrum of connexin isoforms (Cx26, Cx30, and Cx43) exhibited the differential ability to trans-dominantly rescue the assembly of GFP-tagged Cx30.3 mutants into gap junctions suggesting a broad range of connexins found in keratinocytes may favourably interact with Cx30.3 mutants. We conclude that selective upregulation of compatible wild type connexins in keratinocytes may have potential therapeutic value in rescuing epidermal defects invoked by Cx30.3 EKVP-linked mutants.
Collapse
Affiliation(s)
- Sergiu A. Lucaciu
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada,Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Rhett Figliuzzi
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Ruth Neumann
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Samina Nazarali
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Luigi Del Sordo
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada,Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Stephanie E. Leighton
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Alexandra Hauser
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Qing Shao
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Danielle Johnston
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Donglin Bai
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Dale W. Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada,Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada,*Correspondence: Dale W. Laird,
| |
Collapse
|
12
|
Kim H, Gomez-Pastor R. HSF1 and Its Role in Huntington's Disease Pathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1410:35-95. [PMID: 36396925 DOI: 10.1007/5584_2022_742] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
PURPOSE OF REVIEW Heat shock factor 1 (HSF1) is the master transcriptional regulator of the heat shock response (HSR) in mammalian cells and is a critical element in maintaining protein homeostasis. HSF1 functions at the center of many physiological processes like embryogenesis, metabolism, immune response, aging, cancer, and neurodegeneration. However, the mechanisms that allow HSF1 to control these different biological and pathophysiological processes are not fully understood. This review focuses on Huntington's disease (HD), a neurodegenerative disease characterized by severe protein aggregation of the huntingtin (HTT) protein. The aggregation of HTT, in turn, leads to a halt in the function of HSF1. Understanding the pathways that regulate HSF1 in different contexts like HD may hold the key to understanding the pathomechanisms underlying other proteinopathies. We provide the most current information on HSF1 structure, function, and regulation, emphasizing HD, and discussing its potential as a biological target for therapy. DATA SOURCES We performed PubMed search to find established and recent reports in HSF1, heat shock proteins (Hsp), HD, Hsp inhibitors, HSF1 activators, and HSF1 in aging, inflammation, cancer, brain development, mitochondria, synaptic plasticity, polyglutamine (polyQ) diseases, and HD. STUDY SELECTIONS Research and review articles that described the mechanisms of action of HSF1 were selected based on terms used in PubMed search. RESULTS HSF1 plays a crucial role in the progression of HD and other protein-misfolding related neurodegenerative diseases. Different animal models of HD, as well as postmortem brains of patients with HD, reveal a connection between the levels of HSF1 and HSF1 dysfunction to mutant HTT (mHTT)-induced toxicity and protein aggregation, dysregulation of the ubiquitin-proteasome system (UPS), oxidative stress, mitochondrial dysfunction, and disruption of the structural and functional integrity of synaptic connections, which eventually leads to neuronal loss. These features are shared with other neurodegenerative diseases (NDs). Currently, several inhibitors against negative regulators of HSF1, as well as HSF1 activators, are developed and hold promise to prevent neurodegeneration in HD and other NDs. CONCLUSION Understanding the role of HSF1 during protein aggregation and neurodegeneration in HD may help to develop therapeutic strategies that could be effective across different NDs.
Collapse
Affiliation(s)
- Hyuck Kim
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rocio Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
13
|
Rapid differentiation of hiPSCs into functional oligodendrocytes using an OLIG2 synthetic modified messenger RNA. Commun Biol 2022; 5:1095. [PMID: 36241911 PMCID: PMC9568531 DOI: 10.1038/s42003-022-04043-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/27/2022] [Indexed: 11/28/2022] Open
Abstract
Transcription factors (TFs) have been introduced to drive the highly efficient differentiation of human-induced pluripotent stem cells (hiPSCs) into lineage-specific oligodendrocytes (OLs). However, effective strategies currently rely mainly on genome-integrating viruses. Here we show that a synthetic modified messenger RNA (smRNA)-based reprogramming method that leads to the generation of transgene-free OLs has been developed. An smRNA encoding a modified form of OLIG2, in which the serine 147 phosphorylation site is replaced with alanine, OLIG2S147A, is designed to reprogram hiPSCs into OLs. We demonstrate that repeated administration of the smRNA encoding OLIG2S147A lead to higher and more stable protein expression. Using the single-mutant OLIG2 smRNA morphogen, we establish a 6-day smRNA transfection protocol, and glial induction lead to rapid NG2+ OL progenitor cell (OPC) generation (>70% purity) from hiPSC. The smRNA-induced NG2+ OPCs can mature into functional OLs in vitro and promote remyelination in vivo. Taken together, we present a safe and efficient smRNA-driven strategy for hiPSC differentiation into OLs, which may be utilized for therapeutic OPC/OL transplantation in patients with neurodegenerative disease. The use of synthetic modified messenger RNA (smRNA) allows for the differentiation of human-induced pluripotent stem cells (hiPSCs) into lineage-specific oligodendrocytes.
Collapse
|
14
|
Pariollaud M, Ibrahim LH, Irizarry E, Mello RM, Chan AB, Altman BJ, Shaw RJ, Bollong MJ, Wiseman RL, Lamia KA. Circadian disruption enhances HSF1 signaling and tumorigenesis in Kras-driven lung cancer. SCIENCE ADVANCES 2022; 8:eabo1123. [PMID: 36170373 PMCID: PMC9519049 DOI: 10.1126/sciadv.abo1123] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/12/2022] [Indexed: 05/04/2023]
Abstract
Disrupted circadian rhythmicity is a prominent feature of modern society and has been designated as a probable carcinogen by the World Health Organization. However, the biological mechanisms that connect circadian disruption and cancer risk remain largely undefined. We demonstrate that exposure to chronic circadian disruption [chronic jetlag (CJL)] increases tumor burden in a mouse model of KRAS-driven lung cancer. Molecular characterization of tumors and tumor-bearing lung tissues revealed that CJL enhances the expression of heat shock factor 1 (HSF1) target genes. Consistently, exposure to CJL disrupted the highly rhythmic nuclear trafficking of HSF1 in the lung, resulting in an enhanced accumulation of HSF1 in the nucleus. HSF1 has been shown to promote tumorigenesis in other systems, and we find that pharmacological or genetic inhibition of HSF1 reduces the growth of KRAS-mutant human lung cancer cells. These findings implicate HSF1 as a molecular link between circadian disruption and enhanced tumorigenesis.
Collapse
Affiliation(s)
- Marie Pariollaud
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Lara H. Ibrahim
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Chemistry, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Emanuel Irizarry
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Rebecca M. Mello
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Alanna B. Chan
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Brian J. Altman
- Department of Biomedical Genetics and Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Reuben J. Shaw
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Michael J. Bollong
- Department of Chemistry, Scripps Research Institute, La Jolla, CA 92037, USA
| | - R. Luke Wiseman
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Katja A. Lamia
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
15
|
Mossiah I, Perez SM, Stanley TR, Foley MK, Kim Guisbert KS, Guisbert E. Geranylgeranylacetone Ameliorates Beta-Amyloid Toxicity and Extends Lifespan via the Heat Shock Response in Caenorhabditis elegans. FRONTIERS IN AGING 2022; 3:846977. [PMID: 35821801 PMCID: PMC9261441 DOI: 10.3389/fragi.2022.846977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 04/01/2022] [Indexed: 11/26/2022]
Abstract
Activation of a cytoprotective cellular pathway known as the heat shock response (HSR) is a promising strategy for the treatment of Alzheimer’s disease and other neurodegenerative diseases. Geranylgeranylacetone (GGA) is a commonly used anti-ulcer drug in Japan that has been shown to activate the HSR. Here, we establish C. elegans as a model system to investigate the effects of GGA. First, we show that GGA-mediated activation of the HSR is conserved in worms. Then, we show that GGA can ameliorate beta-amyloid toxicity in both muscle and neuronal worm Alzheimer’s disease models. Finally, we find that exposure to GGA is sufficient to extend the lifespan of wild-type worms. Significantly, the beneficial effects of GGA on both beta-amyloid toxicity and lifespan are dependent on HSR activation. Taken together, this research supports further development of GGA as a therapeutic for Alzheimer’s disease, provides evidence that HSR activation is a relevant therapeutic mechanism, and indicates that the beneficial effects of GGA are not limited to disease.
Collapse
|
16
|
Mesgarzadeh JS, Buxbaum JN, Wiseman RL. Stress-responsive regulation of extracellular proteostasis. J Cell Biol 2022; 221:213026. [PMID: 35191945 PMCID: PMC8868021 DOI: 10.1083/jcb.202112104] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 12/18/2022] Open
Abstract
Genetic, environmental, and aging-related insults can promote the misfolding and subsequent aggregation of secreted proteins implicated in the pathogenesis of numerous diseases. This has led to considerable interest in understanding the molecular mechanisms responsible for regulating proteostasis in extracellular environments such as the blood and cerebrospinal fluid (CSF). Extracellular proteostasis is largely dictated by biological pathways comprising chaperones, folding enzymes, and degradation factors localized to the ER and extracellular space. These pathways limit the accumulation of nonnative, potentially aggregation-prone proteins in extracellular environments. Many reviews discuss the molecular mechanisms by which these pathways impact the conformational integrity of the secreted proteome. Here, we instead focus on describing the stress-responsive mechanisms responsible for adapting ER and extracellular proteostasis pathways to protect the secreted proteome from pathologic insults that challenge these environments. Further, we highlight new strategies to identify stress-responsive pathways involved in regulating extracellular proteostasis and describe the pathologic and therapeutic implications for these pathways in human disease.
Collapse
Affiliation(s)
| | - Joel N Buxbaum
- Department of Molecular Medicine, Scripps Research, La Jolla, CA
| | - R Luke Wiseman
- Department of Molecular Medicine, Scripps Research, La Jolla, CA
| |
Collapse
|
17
|
Guan XN, Zhang T, Yang T, Dong Z, Yang S, Lan L, Gan J, Yang CG. Covalent sortase A inhibitor ML346 prevents Staphylococcus aureus infection of Galleria mellonella. RSC Med Chem 2022; 13:138-149. [PMID: 35308030 PMCID: PMC8864484 DOI: 10.1039/d1md00316j] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/31/2021] [Indexed: 12/16/2023] Open
Abstract
The housekeeping sortase A (SrtA), a membrane-associated cysteine transpeptidase, is responsible for anchoring surface proteins to the cell wall peptidoglycan in Gram-positive bacteria. This process is essential for the regulation of bacterial virulence and pathogenicity. Therefore, SrtA is considered to be an ideal target for antivirulence therapy. In this study, we report that ML346, a compound with a barbituric acid and cinnamaldehyde scaffold, functions as an irreversible inhibitor of Staphylococcus aureus SrtA (SaSrtA) and Streptococcus pyogenes SrtA (SpSrtA) in vitro at low micromolar concentrations. According to our X-ray crystal structure of the SpSrtAΔN81/ML346 complex (Protein Data Bank ID: 7V6K), ML346 covalently modifies the thiol group of Cys208 in the active site of SpSrtA. Importantly, ML346 significantly attenuated the virulence phenotypes of S. aureus and exhibited inhibitory effects on Galleria mellonella larva infection caused by S. aureus. Collectively, our results indicate that ML346 has potential for development as a covalent antivirulence agent for treating S. aureus infections, including methicillin-resistant S. aureus.
Collapse
Affiliation(s)
- Xiang-Na Guan
- Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of the Chinese Academy of Sciences Beijing 100049 China
| | - Tao Zhang
- Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Teng Yang
- Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals, Guizhou University Guiyang 550025 China
| | - Ze Dong
- Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Song Yang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals, Guizhou University Guiyang 550025 China
| | - Lefu Lan
- Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of the Chinese Academy of Sciences Beijing 100049 China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences Hangzhou 310024 China
| | - Jianhua Gan
- School of Life Sciences, Fudan University Shanghai 200433 China
| | - Cai-Guang Yang
- Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of the Chinese Academy of Sciences Beijing 100049 China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences Hangzhou 310024 China
| |
Collapse
|
18
|
Owyong TC, Hong Y. Emerging fluorescence tools for the study of proteostasis in cells. Curr Opin Chem Biol 2022; 67:102116. [PMID: 35176555 DOI: 10.1016/j.cbpa.2022.102116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/22/2021] [Accepted: 01/12/2022] [Indexed: 11/25/2022]
Abstract
Understanding how cells maintain the functional proteome and respond to stress conditions is critical for deciphering molecular pathogenesis and developing treatments for conditions such as neurodegenerative diseases. Efforts towards finer quantification of cellular proteostasis machinery efficiency, phase transitions and local environment changes remain a priority. Herein, we describe recent developments in fluorescence-based strategy and methodology, building on the experimental toolkit, for the study of proteostasis (protein homeostasis) in cells. We hope this review can assist in bridging gaps between a multitude of research disciplines and promote interdisciplinary collaboration to address the crucial topic of proteostasis.
Collapse
Affiliation(s)
- Tze Cin Owyong
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia; ARC Centre of Excellence in Exciton Science, School of Chemistry, Bio21 Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Yuning Hong
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
19
|
Giovannucci TA, Salomons FA, Haraldsson M, Elfman LHM, Wickström M, Young P, Lundbäck T, Eirich J, Altun M, Jafari R, Gustavsson AL, Johnsen JI, Dantuma NP. Inhibition of the ubiquitin-proteasome system by an NQO1-activatable compound. Cell Death Dis 2021; 12:914. [PMID: 34615851 PMCID: PMC8494907 DOI: 10.1038/s41419-021-04191-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/03/2021] [Accepted: 09/16/2021] [Indexed: 11/10/2022]
Abstract
Malignant cells display an increased sensitivity towards drugs that reduce the function of the ubiquitin-proteasome system (UPS), which is the primary proteolytic system for destruction of aberrant proteins. Here, we report on the discovery of the bioactivatable compound CBK77, which causes an irreversible collapse of the UPS, accompanied by a general accumulation of ubiquitylated proteins and caspase-dependent cell death. CBK77 caused accumulation of ubiquitin-dependent, but not ubiquitin-independent, reporter substrates of the UPS, suggesting a selective effect on ubiquitin-dependent proteolysis. In a genome-wide CRISPR interference screen, we identified the redox enzyme NAD(P)H:quinone oxidoreductase 1 (NQO1) as a critical mediator of CBK77 activity, and further demonstrated its role as the compound bioactivator. Through affinity-based proteomics, we found that CBK77 covalently interacts with ubiquitin. In vitro experiments showed that CBK77-treated ubiquitin conjugates were less susceptible to disassembly by deubiquitylating enzymes. In vivo efficacy of CBK77 was validated by reduced growth of NQO1-proficient human adenocarcinoma cells in nude mice treated with CBK77. This first-in-class NQO1-activatable UPS inhibitor suggests that it may be possible to exploit the intracellular environment in malignant cells for leveraging the impact of compounds that impair the UPS.
Collapse
Affiliation(s)
- Tatiana A Giovannucci
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, Sweden
| | - Florian A Salomons
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, Sweden
| | - Martin Haraldsson
- Chemical Biology Consortium Sweden (CBCS), Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Lotta H M Elfman
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Malin Wickström
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Patrick Young
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, Sweden
| | - Thomas Lundbäck
- Chemical Biology Consortium Sweden (CBCS), Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Stockholm, Sweden
- Mechanistic & Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Jürgen Eirich
- Science for Life Laboratory, Department of Oncology-Pathology, Clinical Proteomics Mass Spectrometry, Karolinska Institutet, Solna, Stockholm, Sweden
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics (MBB), Karolinska Institutet, Solna, Stockholm, Sweden
- Institute of Plant Biology and Biotechnology, University of Muenster, 48143, Muenster, Germany
| | - Mikael Altun
- Science for Life Laboratory, Department of Laboratory Medicine, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Rozbeh Jafari
- Science for Life Laboratory, Department of Oncology-Pathology, Clinical Proteomics Mass Spectrometry, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Anna-Lena Gustavsson
- Chemical Biology Consortium Sweden (CBCS), Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Stockholm, Sweden
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Nico P Dantuma
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
20
|
Powers ET, Gierasch LM. The Proteome Folding Problem and Cellular Proteostasis. J Mol Biol 2021; 433:167197. [PMID: 34391802 DOI: 10.1016/j.jmb.2021.167197] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 12/16/2022]
Abstract
Stunning advances have been achieved in addressing the protein folding problem, providing deeper understanding of the mechanisms by which proteins navigate energy landscapes to reach their native states and enabling powerful algorithms to connect sequence to structure. However, the realities of the in vivo protein folding problem remain a challenge to reckon with. Here, we discuss the concept of the "proteome folding problem"-the problem of how organisms build and maintain a functional proteome-by admitting that folding energy landscapes are characterized by many misfolded states and that cells must deploy a network of chaperones and degradation enzymes to minimize deleterious impacts of these off-pathway species. The resulting proteostasis network is an inextricable part of in vivo protein folding and must be understood in detail if we are to solve the proteome folding problem. We discuss how the development of computational models for the proteostasis network's actions and the relationship to the biophysical properties of the proteome has begun to offer new insights and capabilities.
Collapse
Affiliation(s)
- Evan T Powers
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Lila M Gierasch
- Departments of Biochemistry & Molecular Biology and Chemistry, University of Massachusetts-Amherst, Amherst, MA 01003, USA.
| |
Collapse
|
21
|
Joshi P, Perni M, Limbocker R, Mannini B, Casford S, Chia S, Habchi J, Labbadia J, Dobson CM, Vendruscolo M. Two human metabolites rescue a C. elegans model of Alzheimer's disease via a cytosolic unfolded protein response. Commun Biol 2021; 4:843. [PMID: 34234268 PMCID: PMC8263720 DOI: 10.1038/s42003-021-02218-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
Age-related changes in cellular metabolism can affect brain homeostasis, creating conditions that are permissive to the onset and progression of neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Although the roles of metabolites have been extensively studied with regard to cellular signaling pathways, their effects on protein aggregation remain relatively unexplored. By computationally analysing the Human Metabolome Database, we identified two endogenous metabolites, carnosine and kynurenic acid, that inhibit the aggregation of the amyloid beta peptide (Aβ) and rescue a C. elegans model of Alzheimer's disease. We found that these metabolites act by triggering a cytosolic unfolded protein response through the transcription factor HSF-1 and downstream chaperones HSP40/J-proteins DNJ-12 and DNJ-19. These results help rationalise previous observations regarding the possible anti-ageing benefits of these metabolites by providing a mechanism for their action. Taken together, our findings provide a link between metabolite homeostasis and protein homeostasis, which could inspire preventative interventions against neurodegenerative disorders.
Collapse
Affiliation(s)
- Priyanka Joshi
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK ,grid.47840.3f0000 0001 2181 7878Present Address: The California Institute for Quantitative Biosciences (QB3-Berkeley), University of California, Berkeley, CA USA
| | - Michele Perni
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Ryan Limbocker
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK ,grid.419884.80000 0001 2287 2270Present Address: Department of Chemistry and Life Science, United States Military Academy, West Point, NY USA
| | - Benedetta Mannini
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Sam Casford
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Sean Chia
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Johnny Habchi
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Johnathan Labbadia
- grid.83440.3b0000000121901201Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK
| | - Christopher M. Dobson
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Michele Vendruscolo
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| |
Collapse
|
22
|
Vertti-Quintero N, Berger S, Casadevall I Solvas X, Statzer C, Annis J, Ruppen P, Stavrakis S, Ewald CY, Gunawan R, deMello AJ. Stochastic and Age-Dependent Proteostasis Decline Underlies Heterogeneity in Heat-Shock Response Dynamics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2102145. [PMID: 34196492 DOI: 10.1002/smll.202102145] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/18/2021] [Indexed: 06/13/2023]
Abstract
Significant non-genetic stochastic factors affect aging, causing lifespan differences among individuals, even those sharing the same genetic and environmental background. In Caenorhabditis elegans, differences in heat-shock response (HSR) are predictive of lifespan. However, factors contributing to the heterogeneity of HSR are still not fully elucidated. Here, the authors characterized HSR dynamics in isogenic C. elegans expressing GFP reporter for hsp-16.2 for identifying the key contributors of HSR heterogeneity. Specifically, microfluidic devices that enable cross-sectional and longitudinal measurements of HSR dynamics in C. elegans at different scales are developed: in populations, within individuals, and in embryos. The authors adapted a mathematical model of HSR to single C. elegans and identified model parameters associated with proteostasis-maintenance of protein homeostasis-more specifically, protein turnover, as the major drivers of heterogeneity in HSR dynamics. It is verified that individuals with enhanced proteostasis fidelity in early adulthood live longer. The model-based comparative analysis of protein turnover in day-1 and day-2 adult C. elegans revealed a stochastic-onset of age-related proteostasis decline that increases the heterogeneity of HSR capacity. Finally, the analysis of C. elegans embryos showed higher HSR and proteostasis capacity than young adults and established transgenerational contribution to HSR heterogeneity that depends on maternal age.
Collapse
Affiliation(s)
| | - Simon Berger
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, 8093, Switzerland
| | - Xavier Casadevall I Solvas
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, 8093, Switzerland
- Department of Biosystems, KU Leuven, Leuven, B-3001, Belgium
| | - Cyril Statzer
- Institute of Translational Medicine, ETH Zurich, Schwerzenbach, 8603, Switzerland
| | - Jillian Annis
- Department of Chemical and Biological Engineering, University at Buffalo - SUNY, Buffalo, NY, 14260, USA
| | - Peter Ruppen
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, 8093, Switzerland
| | - Stavros Stavrakis
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, 8093, Switzerland
| | - Collin Y Ewald
- Institute of Translational Medicine, ETH Zurich, Schwerzenbach, 8603, Switzerland
| | - Rudiyanto Gunawan
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, 8093, Switzerland
- Department of Chemical and Biological Engineering, University at Buffalo - SUNY, Buffalo, NY, 14260, USA
| | - Andrew J deMello
- Institute of Chemical and Bioengineering, ETH Zurich, Zurich, 8093, Switzerland
| |
Collapse
|
23
|
Modrego A, Amaranto M, Godino A, Mendoza R, Barra JL, Corchero JL. Human α-Galactosidase A Mutants: Priceless Tools to Develop Novel Therapies for Fabry Disease. Int J Mol Sci 2021; 22:6518. [PMID: 34204583 PMCID: PMC8234732 DOI: 10.3390/ijms22126518] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 12/25/2022] Open
Abstract
Fabry disease (FD) is a lysosomal storage disease caused by mutations in the gene for the α-galactosidase A (GLA) enzyme. The absence of the enzyme or its activity results in the accumulation of glycosphingolipids, mainly globotriaosylceramide (Gb3), in different tissues, leading to a wide range of clinical manifestations. More than 1000 natural variants have been described in the GLA gene, most of them affecting proper protein folding and enzymatic activity. Currently, FD is treated by enzyme replacement therapy (ERT) or pharmacological chaperone therapy (PCT). However, as both approaches show specific drawbacks, new strategies (such as new forms of ERT, organ/cell transplant, substrate reduction therapy, or gene therapy) are under extensive study. In this review, we summarize GLA mutants described so far and discuss their putative application for the development of novel drugs for the treatment of FD. Unfavorable mutants with lower activities and stabilities than wild-type enzymes could serve as tools for the development of new pharmacological chaperones. On the other hand, GLA mutants showing improved enzymatic activity have been identified and produced in vitro. Such mutants could overcome several complications associated with current ERT, as lower-dose infusions of these mutants could achieve a therapeutic effect equivalent to that of the wild-type enzyme.
Collapse
Affiliation(s)
- Andrea Modrego
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (A.M.); (R.M.)
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain
| | - Marilla Amaranto
- Departamento de Química Biológica Ranwel Caputto, Centro de Investigaciones en Química Biológica de Córdoba, CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5016, Argentina; (M.A.); (A.G.); (J.L.B.)
| | - Agustina Godino
- Departamento de Química Biológica Ranwel Caputto, Centro de Investigaciones en Química Biológica de Córdoba, CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5016, Argentina; (M.A.); (A.G.); (J.L.B.)
| | - Rosa Mendoza
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (A.M.); (R.M.)
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), c/Monforte de Lemos 3–5, 28029 Madrid, Spain
| | - José Luis Barra
- Departamento de Química Biológica Ranwel Caputto, Centro de Investigaciones en Química Biológica de Córdoba, CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5016, Argentina; (M.A.); (A.G.); (J.L.B.)
| | - José Luis Corchero
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; (A.M.); (R.M.)
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), c/Monforte de Lemos 3–5, 28029 Madrid, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| |
Collapse
|
24
|
Ilyinsky NS, Nesterov SV, Shestoperova EI, Fonin AV, Uversky VN, Gordeliy VI. On the Role of Normal Aging Processes in the Onset and Pathogenesis of Diseases Associated with the Abnormal Accumulation of Protein Aggregates. BIOCHEMISTRY (MOSCOW) 2021; 86:275-289. [PMID: 33838629 DOI: 10.1134/s0006297921030056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Aging is a prime systemic cause of various age-related diseases, in particular, proteinopathies. In fact, most diseases associated with protein misfolding are sporadic, and their incidence increases with aging. This review examines the process of protein aggregate formation, the toxicity of such aggregates, the organization of cellular systems involved in proteostasis, and the impact of protein aggregates on important cellular processes leading to proteinopathies. We also analyze how manifestations of aging (mitochondrial dysfunction, dysfunction of signaling systems, changes in the genome and epigenome) facilitate pathogenesis of various proteinopathies either directly, by increasing the propensity of key proteins for aggregation, or indirectly, through dysregulation of stress responses. Such analysis might help in outlining approaches for treating proteinopathies and extending healthy longevity.
Collapse
Affiliation(s)
- Nikolay S Ilyinsky
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia.
| | - Semen V Nesterov
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia.,Institute of Cytochemistry and Molecular Pharmacology, Moscow, 115404, Russia
| | - Elizaveta I Shestoperova
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia
| | - Alexander V Fonin
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia.,Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, 194064, Russia
| | - Vladimir N Uversky
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia.,Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Valentin I Gordeliy
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia.,Forschungszentrum Juelich, Juelich, 52428, Germany.,Institut de Biologie Structurale, Grenoble, 38000, France
| |
Collapse
|
25
|
Sharma R, Srivastava T, Pandey AR, Mishra T, Gupta B, Reddy SS, Singh SP, Narender T, Tripathi A, Chandramouli B, Sashidhara KV, Priya S, Kumar N. Identification of Natural Products as Potential Pharmacological Chaperones for Protein Misfolding Diseases. ChemMedChem 2021; 16:2146-2156. [PMID: 33760394 DOI: 10.1002/cmdc.202100147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Indexed: 01/12/2023]
Abstract
Defective protein folding and accumulation of misfolded proteins is associated with neurodegenerative, cardiovascular, secretory, and metabolic disorders. Efforts are being made to identify small-molecule modulators or structural-correctors for conformationally destabilized proteins implicated in various protein aggregation diseases. Using a metastable-reporter-based primary screen, we evaluated pharmacological chaperone activity of a diverse class of natural products. We found that a flavonoid glycoside (C-10, chrysoeriol-7-O-β-D-glucopyranoside) stabilizes metastable proteins, prevents its aggregation, and remodels the oligomers into protease-sensitive species. Data was corroborated with additional secondary screen with disease-specific pathogenic protein. In vitro and cell-based experiments showed that C-10 inhibits α-synuclein aggregation which is implicated in synucleinopathies-related neurodegeneration. C-10 interferes in its structural transition into β-sheeted fibrils and mitigates α-synuclein aggregation-associated cytotoxic effects. Computational modeling suggests that C-10 binds to unique sites in α-synuclein which may interfere in its aggregation amplification. These findings open an avenue for comprehensive SAR development for flavonoid glycosides as pharmacological chaperones for metastable and aggregation-prone proteins implicated in protein conformational diseases.
Collapse
Affiliation(s)
- Richa Sharma
- CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Tulika Srivastava
- CSIR-Indian Institute of Toxicology Research, Lucknow, 226 001, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India
| | - Alka Raj Pandey
- CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India
| | - Tripti Mishra
- CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Bhagyashri Gupta
- CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | | | - Suriya P Singh
- CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Tadigoppula Narender
- CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India
| | - Aradhya Tripathi
- CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | | | - Koneni V Sashidhara
- CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India
| | - Smriti Priya
- CSIR-Indian Institute of Toxicology Research, Lucknow, 226 001, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India
| | - Niti Kumar
- CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India
| |
Collapse
|
26
|
Lu M, Mishra A, Boschetti C, Lin J, Liu Y, Huang H, Kaminski CF, Huang Z, Tunnacliffe A, Kaminski Schierle GS. Sea Cucumber-Derived Peptides Alleviate Oxidative Stress in Neuroblastoma Cells and Improve Survival in C. elegans Exposed to Neurotoxic Paraquat. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8842926. [PMID: 33959216 PMCID: PMC8075690 DOI: 10.1155/2021/8842926] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/16/2021] [Accepted: 03/30/2021] [Indexed: 01/22/2023]
Abstract
Oxidative stress results when the production of oxidants outweighs the capacity of the antioxidant defence mechanisms. This can lead to pathological conditions including cancer and neurodegeneration. Consequently, there is considerable interest in compounds with antioxidant activity, including those from natural sources. Here, we characterise the antioxidant activity of three novel peptides identified in protein hydrolysates from the sea cucumber Apostichopus japonicus. Under oxidative stress conditions, synthetic versions of the sea cucumber peptides significantly compensate for glutathione depletion, decrease mitochondrial superoxide levels, and alleviate mitophagy in human neuroblastoma cells. Moreover, orally supplied peptides improve survival of the Caenorhabditis elegans after treatment with paraquat, the latter of which leads to the production of excessive oxidative stress. Thus, the sea cucumber peptides exhibit antioxidant activity at both the cellular and organism levels and might prove attractive as nutritional supplements for healthy ageing.
Collapse
Affiliation(s)
- Meng Lu
- Cambridge Infinitus Research Centre, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge CB3 0AS, UK
| | - Ajay Mishra
- Cambridge Infinitus Research Centre, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge CB3 0AS, UK
| | - Chiara Boschetti
- Cambridge Infinitus Research Centre, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge CB3 0AS, UK
| | - Jing Lin
- Research Institute for Food Nutrition and Human Health, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yushuang Liu
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hongliang Huang
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Clemens F. Kaminski
- Cambridge Infinitus Research Centre, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge CB3 0AS, UK
| | - Zebo Huang
- Research Institute for Food Nutrition and Human Health, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Alan Tunnacliffe
- Cambridge Infinitus Research Centre, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge CB3 0AS, UK
| | - Gabriele S. Kaminski Schierle
- Cambridge Infinitus Research Centre, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge CB3 0AS, UK
| |
Collapse
|
27
|
Dutta N, Ghosh S, Nelson VK, Sareng HR, Majumder C, Mandal SC, Pal M. Andrographolide upregulates protein quality control mechanisms in cell and mouse through upregulation of mTORC1 function. Biochim Biophys Acta Gen Subj 2021; 1865:129885. [PMID: 33639218 DOI: 10.1016/j.bbagen.2021.129885] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Heat shock response (HSR), a component of cellular protein quality control mechanisms, is defective in different neurodegenerative conditions such as Parkinson's disease (PD). Forced upregulation of heat shock factor 1 (HSF1), an HSR master regulator, showed therapeutic promise in PD models. Many of the reported small-molecule HSF1 activators have limited functions. Therefore, identification and understanding the molecular bases of action of new HSF1 activating molecules is necessary. METHOD We used a cell-based reporter system to screen Andrographis paniculata leaf extract to isolate andrographolide as an inducer of HSF1 activity. The andrographolide activity was characterized by analyzing its role in different protein quality control mechanisms. RESULT We find that besides ameliorating the PD in MPTP-treated mice, andrographolide upregulated different machineries controlled by HSF1 and NRF2 in both cell and mouse brain. Andrographolide achieves these functions through mTORC1 activated via p38 MAPK and ERK pathways. NRF2 activation is reflected in the upregulation of proteasome as well as autophagy pathways. We further show that NRF2 activation is mediated through mTORC1 driven phosphorylation of p62/sequestosome 1. Studies with different cell types suggested that andrographolide-mediated induction of ROS level underlies all these activities in agreement with the upregulation of mTORC1 and NRF2-antioxidant pathway in mice. CONCLUSION Andrographolide through upregulating HSF1 activity ameliorates protein aggregation induced cellular toxicity. GENERAL SIGNIFICANCE Our results provide a reasonable basis for use of andrographolide in the therapy regimen for the treatment of PD.
Collapse
Affiliation(s)
- Naibedya Dutta
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Suvranil Ghosh
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Vinod K Nelson
- Pharmacognosy and Phytotherapy Research Laboratory, Division of Pharmacognosy, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Hossainoor R Sareng
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Chirantan Majumder
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Subhash C Mandal
- Pharmacognosy and Phytotherapy Research Laboratory, Division of Pharmacognosy, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Mahadeb Pal
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India.
| |
Collapse
|
28
|
Silva MC, Haggarty SJ. Tauopathies: Deciphering Disease Mechanisms to Develop Effective Therapies. Int J Mol Sci 2020; 21:ijms21238948. [PMID: 33255694 PMCID: PMC7728099 DOI: 10.3390/ijms21238948] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/20/2020] [Accepted: 11/22/2020] [Indexed: 12/13/2022] Open
Abstract
Tauopathies are neurodegenerative diseases characterized by the pathological accumulation of microtubule-associated protein tau (MAPT) in the form of neurofibrillary tangles and paired helical filaments in neurons and glia, leading to brain cell death. These diseases include frontotemporal dementia (FTD) and Alzheimer's disease (AD) and can be sporadic or inherited when caused by mutations in the MAPT gene. Despite an incredibly high socio-economic burden worldwide, there are still no effective disease-modifying therapies, and few tau-focused experimental drugs have reached clinical trials. One major hindrance for therapeutic development is the knowledge gap in molecular mechanisms of tau-mediated neuronal toxicity and death. For the promise of precision medicine for brain disorders to be fulfilled, it is necessary to integrate known genetic causes of disease, i.e., MAPT mutations, with an understanding of the dysregulated molecular pathways that constitute potential therapeutic targets. Here, the growing understanding of known and proposed mechanisms of disease etiology will be reviewed, together with promising experimental tau-directed therapeutics, such as recently developed tau degraders. Current challenges faced by the fields of tau research and drug discovery will also be addressed.
Collapse
|
29
|
Caenorhabditis elegans Models to Investigate the Mechanisms Underlying Tau Toxicity in Tauopathies. Brain Sci 2020; 10:brainsci10110838. [PMID: 33187241 PMCID: PMC7697895 DOI: 10.3390/brainsci10110838] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
The understanding of the genetic, biochemical, and structural determinants underlying tau aggregation is pivotal in the elucidation of the pathogenic process driving tauopathies and the design of effective therapies. Relevant information on the molecular basis of human neurodegeneration in vivo can be obtained using the nematode Caenorhabditis elegans (C. elegans). To this end, two main approaches can be applied: the overexpression of genes/proteins leading to neuronal dysfunction and death, and studies in which proteins prone to misfolding are exogenously administered to induce a neurotoxic phenotype. Thanks to the easy generation of transgenic strains expressing human disease genes, C. elegans allows the identification of genes and/or proteins specifically associated with pathology and the specific disruptions of cellular processes involved in disease. Several transgenic strains expressing human wild-type or mutated tau have been developed and offer significant information concerning whether transgene expression regulates protein production and aggregation in soluble or insoluble form, onset of the disease, and the degenerative process. C. elegans is able to specifically react to the toxic assemblies of tau, thus developing a neurodegenerative phenotype that, even when exogenously administered, opens up the use of this assay to investigate in vivo the relationship between the tau sequence, its folding, and its proteotoxicity. These approaches can be employed to screen drugs and small molecules that can interact with the biogenesis and dynamics of formation of tau aggregates and to analyze their interactions with other cellular proteins.
Collapse
|
30
|
Grandjean JMD, Madhavan A, Cech L, Seguinot BO, Paxman RJ, Smith E, Scampavia L, Powers ET, Cooley CB, Plate L, Spicer TP, Kelly JW, Wiseman RL. Pharmacologic IRE1/XBP1s activation confers targeted ER proteostasis reprogramming. Nat Chem Biol 2020; 16:1052-1061. [PMID: 32690944 PMCID: PMC7502540 DOI: 10.1038/s41589-020-0584-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
Abstract
Activation of the IRE1/XBP1s signaling arm of the unfolded protein response (UPR) is a promising strategy to correct defects in endoplasmic reticulum (ER) proteostasis implicated in diverse diseases. However, no pharmacologic activators of this pathway identified to date are suitable for ER proteostasis remodeling through selective activation of IRE1/XBP1s signaling. Here, we use high-throughput screening to identify non-toxic compounds that induce ER proteostasis remodeling through IRE1/XBP1s activation. We employ transcriptional profiling to stringently confirm that our prioritized compounds selectively activate IRE1/XBP1s signaling without activating other cellular stress-responsive signaling pathways. Furthermore, we demonstrate that our compounds improve ER proteostasis of destabilized variants of amyloid precursor protein (APP) through an IRE1-dependent mechanism and reduce APP-associated mitochondrial toxicity in cellular models. These results establish highly selective IRE1/XBP1s activating compounds that can be widely employed to define the functional importance of IRE1/XBP1s activity for ER proteostasis regulation in the context of health and disease.
Collapse
Affiliation(s)
- Julia M D Grandjean
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Aparajita Madhavan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Lauren Cech
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Bryan O Seguinot
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Ryan J Paxman
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Emery Smith
- Scripps Research Molecular Screening Center, The Scripps Research Institute, Jupiter, FL, USA
| | - Louis Scampavia
- Scripps Research Molecular Screening Center, The Scripps Research Institute, Jupiter, FL, USA
| | - Evan T Powers
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Lars Plate
- Departments of Chemistry and Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Timothy P Spicer
- Scripps Research Molecular Screening Center, The Scripps Research Institute, Jupiter, FL, USA
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
31
|
Abstract
Sustaining a healthy proteome is a lifelong challenge for each individual cell of an organism. However, protein homeostasis or proteostasis is constantly jeopardized since damaged proteins accumulate under proteotoxic stress that originates from ever-changing metabolic, environmental, and pathological conditions. Proteostasis is achieved via a conserved network of quality control pathways that orchestrate the biogenesis of correctly folded proteins, prevent proteins from misfolding, and remove potentially harmful proteins by selective degradation. Nevertheless, the proteostasis network has a limited capacity and its collapse deteriorates cellular functionality and organismal viability, causing metabolic, oncological, or neurodegenerative disorders. While cell-autonomous quality control mechanisms have been described intensely, recent work on Caenorhabditis elegans has demonstrated the systemic coordination of proteostasis between distinct tissues of an organism. These findings indicate the existence of intricately balanced proteostasis networks important for integration and maintenance of the organismal proteome, opening a new door to define novel therapeutic targets for protein aggregation diseases. Here, we provide an overview of individual protein quality control pathways and the systemic coordination between central proteostatic nodes. We further provide insights into the dynamic regulation of cellular and organismal proteostasis mechanisms that integrate environmental and metabolic changes. The use of C. elegans as a model has pioneered our understanding of conserved quality control mechanisms important to safeguard the organismal proteome in health and disease.
Collapse
Affiliation(s)
- Thorsten Hoppe
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne 50931, Germany and
| | - Ehud Cohen
- Department of Biochemistry and Molecular Biology, the Institute for Medical Research Israel-Canada (IMRIC), the Hebrew University School of Medicine, Jerusalem 91120, Israel
| |
Collapse
|
32
|
Proteostasis regulators modulate proteasomal activity and gene expression to attenuate multiple phenotypes in Fabry disease. Biochem J 2020; 477:359-380. [PMID: 31899485 PMCID: PMC6993862 DOI: 10.1042/bcj20190513] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/17/2019] [Accepted: 01/02/2020] [Indexed: 12/22/2022]
Abstract
The lysosomal storage disorder Fabry disease is characterized by a deficiency of the lysosomal enzyme α-Galactosidase A. The observation that missense variants in the encoding GLA gene often lead to structural destabilization, endoplasmic reticulum retention and proteasomal degradation of the misfolded, but otherwise catalytically functional enzyme has resulted in the exploration of alternative therapeutic approaches. In this context, we have investigated proteostasis regulators (PRs) for their potential to increase cellular enzyme activity, and to reduce the disease-specific accumulation of the biomarker globotriaosylsphingosine in patient-derived cell culture. The PRs also acted synergistically with the clinically approved 1-deoxygalactonojirimycine, demonstrating the potential of combination treatment in a therapeutic application. Extensive characterization of the effective PRs revealed inhibition of the proteasome and elevation of GLA gene expression as paramount effects. Further analysis of transcriptional patterns of the PRs exposed a variety of genes involved in proteostasis as potential modulators. We propose that addressing proteostasis is an effective approach to discover new therapeutic targets for diseases involving folding and trafficking-deficient protein mutants.
Collapse
|
33
|
Lakshmi PK, Kumar S, Pawar S, Kuriakose BB, Sudheesh MS, Pawar RS. Targeting metabolic syndrome with phytochemicals: Focus on the role of molecular chaperones and hormesis in drug discovery. Pharmacol Res 2020; 159:104925. [PMID: 32492491 DOI: 10.1016/j.phrs.2020.104925] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 05/10/2020] [Accepted: 05/10/2020] [Indexed: 12/21/2022]
Abstract
Adaptive cellular stress response confers stress tolerance against inflammatory and metabolic disorders. In response to metabolic stress, the key mediator of cellular adaptation and tolerance is a class of molecules called the molecular chaperones (MCs). MCs are highly conserved molecules that play critical role in maintaining protein stability and functionality. Hormesis in this context is a unique adaptation mechanism where a low dose of a stressor (which is toxic at high dose) confers a stress-resistant adaptive cellular phenotype. Hormesis can be observed at different level of biological organization at various measurable endpoints. The MCs are believed to play a key role in adaptation during hormesis. Several phytochemicals are known for their hormetic response and are called phytochemical hormetins. The role of phytochemical-mediated hormesis on the adaptive cellular processes is proposed as a potential therapeutic approach to target inflammation associated with metabolic syndrome. However, the screening of phytochemical hormetins would require a paradigm shift in the methods currently used in drug discovery.
Collapse
Affiliation(s)
- P K Lakshmi
- Pharmacognosy and Phytochemistry Laboratory, Faculty of Pharmacy, VNS Group of Institutions, VNS Campus, Vidya Vihar, Neelbad-462044, Bhopal, MP, India
| | - Shweta Kumar
- Pharmacognosy and Phytochemistry Laboratory, Faculty of Pharmacy, VNS Group of Institutions, VNS Campus, Vidya Vihar, Neelbad-462044, Bhopal, MP, India
| | - Sulakshhna Pawar
- Ravi Shankar College of Pharmacy, Bypass Road, Bhanpur Square, Bhopal, MP 462010, India
| | - Beena Briget Kuriakose
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King Khalid University, Khamis, Mushayt, Saudi Arabia
| | - M S Sudheesh
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India
| | - Rajesh Singh Pawar
- Truba Institute of Pharmacy, Karond-Gandhi Nagar, By Pass Road, Bhopal, 462038, India.
| |
Collapse
|
34
|
La Rosa P, Petrillo S, Bertini ES, Piemonte F. Oxidative Stress in DNA Repeat Expansion Disorders: A Focus on NRF2 Signaling Involvement. Biomolecules 2020; 10:biom10050702. [PMID: 32369911 PMCID: PMC7277112 DOI: 10.3390/biom10050702] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
DNA repeat expansion disorders are a group of neuromuscular and neurodegenerative diseases that arise from the inheritance of long tracts of nucleotide repetitions, located in the regulatory region, introns, or inside the coding sequence of a gene. Although loss of protein expression and/or the gain of function of its transcribed mRNA or translated product represent the major pathogenic effect of these pathologies, mitochondrial dysfunction and imbalance in redox homeostasis are reported as common features in these disorders, deeply affecting their severity and progression. In this review, we examine the role that the redox imbalance plays in the pathological mechanisms of DNA expansion disorders and the recent advances on antioxidant treatments, particularly focusing on the expression and the activity of the transcription factor NRF2, the main cellular regulator of the antioxidant response.
Collapse
|
35
|
Luh LM, Bertolotti A. Potential benefit of manipulating protein quality control systems in neurodegenerative diseases. Curr Opin Neurobiol 2020; 61:125-132. [PMID: 32199101 DOI: 10.1016/j.conb.2020.02.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 12/19/2022]
Abstract
The deposition of proteins of abnormal conformation is one of the major hallmarks of the common neurodegenerative diseases including Alzheimer's, Parkinson's, amyotrophic lateral sclerosis, frontotemporal dementia, and prion diseases. Protein quality control systems have evolved to protect cells and organisms against the harmful consequences of abnormally folded proteins that are constantly produced in small amounts. Mutations in rare inherited forms of neurodegenerative diseases have provided compelling evidence that failure of protein quality control systems can drive neurodegeneration. With extensive knowledge of these systems, and the notion that protein quality control may decline with age, many laboratories are now focussing on manipulating these evolutionarily optimized defence mechanisms to reduce the protein misfolding burden for therapeutic benefit.
Collapse
Affiliation(s)
- Laura M Luh
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, United Kingdom
| | - Anne Bertolotti
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, United Kingdom.
| |
Collapse
|
36
|
Abstract
Ageing is a major risk factor for the development of many diseases, prominently including neurodegenerative disorders such as Alzheimer disease and Parkinson disease. A hallmark of many age-related diseases is the dysfunction in protein homeostasis (proteostasis), leading to the accumulation of protein aggregates. In healthy cells, a complex proteostasis network, comprising molecular chaperones and proteolytic machineries and their regulators, operates to ensure the maintenance of proteostasis. These factors coordinate protein synthesis with polypeptide folding, the conservation of protein conformation and protein degradation. However, sustaining proteome balance is a challenging task in the face of various external and endogenous stresses that accumulate during ageing. These stresses lead to the decline of proteostasis network capacity and proteome integrity. The resulting accumulation of misfolded and aggregated proteins affects, in particular, postmitotic cell types such as neurons, manifesting in disease. Recent analyses of proteome-wide changes that occur during ageing inform strategies to improve proteostasis. The possibilities of pharmacological augmentation of the capacity of proteostasis networks hold great promise for delaying the onset of age-related pathologies associated with proteome deterioration and for extending healthspan.
Collapse
|
37
|
Abstract
Protein folding in the cell is mediated by an extensive network of >1,000 chaperones, quality control factors, and trafficking mechanisms collectively termed the proteostasis network. While the components and organization of this network are generally well established, our understanding of how protein-folding problems are identified, how the network components integrate to successfully address challenges, and what types of biophysical issues each proteostasis network component is capable of addressing remains immature. We describe a chemical biology-informed framework for studying cellular proteostasis that relies on selection of interesting protein-folding problems and precise researcher control of proteostasis network composition and activities. By combining these methods with multifaceted strategies to monitor protein folding, degradation, trafficking, and aggregation in cells, researchers continue to rapidly generate new insights into cellular proteostasis.
Collapse
Affiliation(s)
- Rebecca M Sebastian
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| | - Matthew D Shoulders
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| |
Collapse
|
38
|
Wille H, Dorosh L, Amidian S, Schmitt-Ulms G, Stepanova M. Combining molecular dynamics simulations and experimental analyses in protein misfolding. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 118:33-110. [PMID: 31928730 DOI: 10.1016/bs.apcsb.2019.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The fold of a protein determines its function and its misfolding can result in loss-of-function defects. In addition, for certain proteins their misfolding can lead to gain-of-function toxicities resulting in protein misfolding diseases such as Alzheimer's, Parkinson's, or the prion diseases. In all of these diseases one or more proteins misfold and aggregate into disease-specific assemblies, often in the form of fibrillar amyloid deposits. Most, if not all, protein misfolding diseases share a fundamental molecular mechanism that governs the misfolding and subsequent aggregation. A wide variety of experimental methods have contributed to our knowledge about misfolded protein aggregates, some of which are briefly described in this review. The misfolding mechanism itself is difficult to investigate, as the necessary timescale and resolution of the misfolding events often lie outside of the observable parameter space. Molecular dynamics simulations fill this gap by virtue of their intrinsic, molecular perspective and the step-by-step iterative process that forms the basis of the simulations. This review focuses on molecular dynamics simulations and how they combine with experimental analyses to provide detailed insights into protein misfolding and the ensuing diseases.
Collapse
Affiliation(s)
- Holger Wille
- Department of Biochemistry, University of Alberta, Edmonton, Canada; Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Lyudmyla Dorosh
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, Canada
| | - Sara Amidian
- Department of Biochemistry, University of Alberta, Edmonton, Canada; Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Canada
| | - Gerold Schmitt-Ulms
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Maria Stepanova
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, Canada
| |
Collapse
|
39
|
Wang C, Scott SM, Sun S, Zhao P, Hutt DM, Shao H, Gestwicki JE, Balch WE. Individualized management of genetic diversity in Niemann-Pick C1 through modulation of the Hsp70 chaperone system. Hum Mol Genet 2020; 29:1-19. [PMID: 31509197 PMCID: PMC7001602 DOI: 10.1093/hmg/ddz215] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 08/05/2019] [Accepted: 09/02/2019] [Indexed: 12/21/2022] Open
Abstract
Genetic diversity provides a rich repository for understanding the role of proteostasis in the management of the protein fold in human biology. Failure in proteostasis can trigger multiple disease states, affecting both human health and lifespan. Niemann-Pick C1 (NPC1) disease is a rare genetic disorder triggered by mutations in NPC1, a multi-spanning transmembrane protein that is trafficked through the exocytic pathway to late endosomes (LE) and lysosomes (Ly) (LE/Ly) to globally manage cholesterol homeostasis. Defects triggered by >300 NPC1 variants found in the human population inhibit export of NPC1 protein from the endoplasmic reticulum (ER) and/or function in downstream LE/Ly, leading to cholesterol accumulation and onset of neurodegeneration in childhood. We now show that the allosteric inhibitor JG98, that targets the cytosolic Hsp70 chaperone/co-chaperone complex, can significantly improve the trafficking and post-ER protein level of diverse NPC1 variants. Using a new approach to model genetic diversity in human disease, referred to as variation spatial profiling, we show quantitatively how JG98 alters the Hsp70 chaperone/co-chaperone system to adjust the spatial covariance (SCV) tolerance and set-points on an amino acid residue-by-residue basis in NPC1 to differentially regulate variant trafficking, stability, and cholesterol homeostasis, results consistent with the role of BCL2-associated athanogene family co-chaperones in managing the folding status of NPC1 variants. We propose that targeting the cytosolic Hsp70 system by allosteric regulation of its chaperone/co-chaperone based client relationships can be used to adjust the SCV tolerance of proteostasis buffering capacity to provide an approach to mitigate systemic and neurological disease in the NPC1 population.
Collapse
Affiliation(s)
- Chao Wang
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Samantha M Scott
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Shuhong Sun
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Pei Zhao
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Darren M Hutt
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Hao Shao
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, CA 94158, USA
| | - William E Balch
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
- The Skaggs Institute for Chemical Biology, Scripps Research, La Jolla, CA 92037, USA
| |
Collapse
|
40
|
Liu Y, He G, Zhang Z, Yin H, Liu H, Chen J, Zhang S, Yang B, Xu LP, Zhang X. Size-effect of gold nanorods on modulating the kinetic process of amyloid-β aggregation. Chem Phys Lett 2019. [DOI: 10.1016/j.cplett.2019.136702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
41
|
Sándor S, Kubinyi E. Genetic Pathways of Aging and Their Relevance in the Dog as a Natural Model of Human Aging. Front Genet 2019; 10:948. [PMID: 31681409 PMCID: PMC6813227 DOI: 10.3389/fgene.2019.00948] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 09/05/2019] [Indexed: 12/15/2022] Open
Abstract
Aging research has experienced a burst of scientific efforts in the last decades as the growing ratio of elderly people has begun to pose an increased burden on the healthcare and pension systems of developed countries. Although many breakthroughs have been reported in understanding the cellular mechanisms of aging, the intrinsic and extrinsic factors that contribute to senescence on higher biological levels are still barely understood. The dog, Canis familiaris, has already served as a valuable model of human physiology and disease. The possible role the dog could play in aging research is still an open question, although utilization of dogs may hold great promises as they naturally develop age-related cognitive decline, with behavioral and histological characteristics very similar to those of humans. In this regard, family dogs may possess unmatched potentials as models for investigations on the complex interactions between environmental, behavioral, and genetic factors that determine the course of aging. In this review, we summarize the known genetic pathways in aging and their relevance in dogs, putting emphasis on the yet barely described nature of certain aging pathways in canines. Reasons for highlighting the dog as a future aging and gerontology model are also discussed, ranging from its unique evolutionary path shared with humans, its social skills, and the fact that family dogs live together with their owners, and are being exposed to the same environmental effects.
Collapse
Affiliation(s)
- Sára Sándor
- Department of Ethology, Eötvös Loránd University, Budapest, Hungary
| | | |
Collapse
|
42
|
Gandhi J, Antonelli AC, Afridi A, Vatsia S, Joshi G, Romanov V, Murray IVJ, Khan SA. Protein misfolding and aggregation in neurodegenerative diseases: a review of pathogeneses, novel detection strategies, and potential therapeutics. Rev Neurosci 2019; 30:339-358. [PMID: 30742586 DOI: 10.1515/revneuro-2016-0035] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022]
Abstract
Protein folding is a complex, multisystem process characterized by heavy molecular and cellular footprints. Chaperone machinery enables proper protein folding and stable conformation. Other pathways concomitant with the protein folding process include transcription, translation, post-translational modifications, degradation through the ubiquitin-proteasome system, and autophagy. As such, the folding process can go awry in several different ways. The pathogenic basis behind most neurodegenerative diseases is that the disruption of protein homeostasis (i.e. proteostasis) at any level will eventually lead to protein misfolding. Misfolded proteins often aggregate and accumulate to trigger neurotoxicity through cellular stress pathways and consequently cause neurodegenerative diseases. The manifestation of a disease is usually dependent on the specific brain region that the neurotoxicity affects. Neurodegenerative diseases are age-associated, and their incidence is expected to rise as humans continue to live longer and pursue a greater life expectancy. We presently review the sequelae of protein misfolding and aggregation, as well as the role of these phenomena in several neurodegenerative diseases including Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, Parkinson's disease, transmissible spongiform encephalopathies, and spinocerebellar ataxia. Strategies for treatment and therapy are also conferred with respect to impairing, inhibiting, or reversing protein misfolding.
Collapse
Affiliation(s)
- Jason Gandhi
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, 101 Nicolls Road, Health Sciences Center, Stony Brook, NY 11794-8434, USA.,Medical Student Research Institute, St. George's University School of Medicine, Grenada, West Indies
| | - Anthony C Antonelli
- Department of Pathology, Stony Brook University School of Medicine, 101 Nicolls Road, Health Sciences Center, Stony Brook, NY 11794-8434, USA
| | - Adil Afridi
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, 101 Nicolls Road, Health Sciences Center, Stony Brook, NY 11794-8434, USA
| | - Sohrab Vatsia
- Department of Cardiothoracic Surgery, Lenox Hill Hospital, 130 East 77th Street, New York, NY 10075, USA
| | - Gunjan Joshi
- Department of Internal Medicine, Stony Brook Southampton Hospital, 240 Meeting House Lane, Southampton, NY 11968, USA
| | - Victor Romanov
- Department of Urology, Health Sciences Center T9-040, Stony Brook University School of Medicine, 101 Nicolls Road, Stony Brook, NY 11794-8093, USA
| | - Ian V J Murray
- Department of Physiology and Neuroscience, St. George's University School of Medicine, Grenada, West Indies
| | - Sardar Ali Khan
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, 101 Nicolls Road, Health Sciences Center, Stony Brook, NY 11794-8434, USA.,Department of Urology, Health Sciences Center T9-040, Stony Brook University School of Medicine, 101 Nicolls Road, Stony Brook, NY 11794-8093, USA
| |
Collapse
|
43
|
Beaver SK, Mesa-Torres N, Pey AL, Timson DJ. NQO1: A target for the treatment of cancer and neurological diseases, and a model to understand loss of function disease mechanisms. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2019; 1867:663-676. [PMID: 31091472 DOI: 10.1016/j.bbapap.2019.05.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 01/08/2023]
Abstract
NAD(P)H quinone oxidoreductase 1 (NQO1) is a multi-functional protein that catalyses the reduction of quinones (and other molecules), thus playing roles in xenobiotic detoxification and redox balance, and also has roles in stabilising apoptosis regulators such as p53. The structure and enzymology of NQO1 is well-characterised, showing a substituted enzyme mechanism in which NAD(P)H binds first and reduces an FAD cofactor in the active site, assisted by a charge relay system involving Tyr-155 and His-161. Protein dynamics play important role in physio-pathological aspects of this protein. NQO1 is a good target to treat cancer due to its overexpression in cancer cells. A polymorphic form of NQO1 (p.P187S) is associated with increased cancer risk and certain neurological disorders (such as multiple sclerosis and Alzheimer´s disease), possibly due to its roles in the antioxidant defence. p.P187S has greatly reduced FAD affinity and stability, due to destabilization of the flavin binding site and the C-terminal domain, which leading to reduced activity and enhanced degradation. Suppressor mutations partially restore the activity of p.P187S by local stabilization of these regions, and showing long-range allosteric communication within the protein. Consequently, the correction of NQO1 misfolding by pharmacological chaperones is a viable strategy, which may be useful to treat cancer and some neurological conditions, targeting structural spots linked to specific disease-mechanisms. Thus, NQO1 emerges as a good model to investigate loss of function mechanisms in genetic diseases as well as to improve strategies to discriminate between neutral and pathogenic variants in genome-wide sequencing studies.
Collapse
Affiliation(s)
- Sarah K Beaver
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Huxley Building, Lewes Road, Brighton BN2 4GJ, UK
| | - Noel Mesa-Torres
- Department of Physical Chemistry, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, 18071, Spain
| | - Angel L Pey
- Department of Physical Chemistry, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, 18071, Spain.
| | - David J Timson
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Huxley Building, Lewes Road, Brighton BN2 4GJ, UK.
| |
Collapse
|
44
|
Marinko J, Huang H, Penn WD, Capra JA, Schlebach JP, Sanders CR. Folding and Misfolding of Human Membrane Proteins in Health and Disease: From Single Molecules to Cellular Proteostasis. Chem Rev 2019; 119:5537-5606. [PMID: 30608666 PMCID: PMC6506414 DOI: 10.1021/acs.chemrev.8b00532] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Indexed: 12/13/2022]
Abstract
Advances over the past 25 years have revealed much about how the structural properties of membranes and associated proteins are linked to the thermodynamics and kinetics of membrane protein (MP) folding. At the same time biochemical progress has outlined how cellular proteostasis networks mediate MP folding and manage misfolding in the cell. When combined with results from genomic sequencing, these studies have established paradigms for how MP folding and misfolding are linked to the molecular etiologies of a variety of diseases. This emerging framework has paved the way for the development of a new class of small molecule "pharmacological chaperones" that bind to and stabilize misfolded MP variants, some of which are now in clinical use. In this review, we comprehensively outline current perspectives on the folding and misfolding of integral MPs as well as the mechanisms of cellular MP quality control. Based on these perspectives, we highlight new opportunities for innovations that bridge our molecular understanding of the energetics of MP folding with the nuanced complexity of biological systems. Given the many linkages between MP misfolding and human disease, we also examine some of the exciting opportunities to leverage these advances to address emerging challenges in the development of therapeutics and precision medicine.
Collapse
Affiliation(s)
- Justin
T. Marinko
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
- Center
for Structural Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Hui Huang
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
- Center
for Structural Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Wesley D. Penn
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - John A. Capra
- Center
for Structural Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
- Department
of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37245, United States
| | - Jonathan P. Schlebach
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Charles R. Sanders
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
| |
Collapse
|
45
|
Grandjean JM, Plate L, Morimoto RI, Bollong MJ, Powers ET, Wiseman RL. Deconvoluting Stress-Responsive Proteostasis Signaling Pathways for Pharmacologic Activation Using Targeted RNA Sequencing. ACS Chem Biol 2019; 14:784-795. [PMID: 30821953 PMCID: PMC6474822 DOI: 10.1021/acschembio.9b00134] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cellular proteostasis is maintained by stress-responsive signaling pathways such as the heat shock response (HSR), the oxidative stress response (OSR), and the unfolded protein response (UPR). Activation of these pathways results in the transcriptional upregulation of select subsets of stress-responsive genes that restore proteostasis and adapt cellular physiology to promote recovery following various types of acute insult. The capacity for these pathways to regulate cellular proteostasis makes them attractive therapeutic targets for correcting proteostasis defects associated with diverse diseases. High-throughput screening (HTS) using cell-based reporter assays is highly effective for identifying putative activators of stress-responsive signaling pathways. However, the development of these compounds is hampered by the lack of medium-throughput assays to define compound potency and selectivity for a given pathway. Here, we describe a targeted RNA sequencing (RNAseq) assay that allows cost-effective, medium-throughput screening of stress-responsive signaling pathway activation. We demonstrate that this assay allows deconvolution of stress-responsive signaling activated by chemical genetic or pharmacologic agents. Furthermore, we use this assay to define the selectivity of putative OSR and HSR activating compounds previously identified by HTS. Our results demonstrate the potential for integrating this adaptable targeted RNAseq assay into screening programs focused on developing pharmacologic activators of stress-responsive signaling pathways.
Collapse
Affiliation(s)
- Julia M.D. Grandjean
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Lars Plate
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Richard I. Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL, USA
| | - Michael J. Bollong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Evan T. Powers
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - R. Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
46
|
Yu A, Fox SG, Cavallini A, Kerridge C, O'Neill MJ, Wolak J, Bose S, Morimoto RI. Tau protein aggregates inhibit the protein-folding and vesicular trafficking arms of the cellular proteostasis network. J Biol Chem 2019; 294:7917-7930. [PMID: 30936201 DOI: 10.1074/jbc.ra119.007527] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/29/2019] [Indexed: 11/06/2022] Open
Abstract
Tauopathies are a diverse class of neurodegenerative diseases characterized by the formation of insoluble tau aggregates and the loss of cellular function and neuronal death. Tau inclusions have been shown to contain a number of proteins, including molecular chaperones, but the consequences of these entrapments are not well established. Here, using a human cell system for seeding-dependent tau aggregation, we demonstrate that the molecular chaperones heat-shock cognate 71-kDa protein (HSC70)/heat-shock protein 70 (HSP70), HSP90, and J-domain co-chaperones are sequestered by tau aggregates. By employing single-cell analysis of protein-folding and clathrin-mediated endocytosis, we show that both chaperone-dependent cellular activities are significantly impaired by tau aggregation and can be reversed by treatment with small-molecule regulators of heat-shock transcription factor 1 (HSF1) proteostasis that induce the expression of cytosolic chaperones. These results reveal that the sequestration of cytoplasmic molecular chaperones by tau aggregates interferes with two arms of the proteostasis network, likely having profound negative consequences for cellular function.
Collapse
Affiliation(s)
- Anan Yu
- From the Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208 and
| | - Susan G Fox
- From the Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208 and
| | - Annalisa Cavallini
- the Lilly Research Centre, Eli Lilly and Co. Ltd., Erl Wood Manor, Sunninghill Road, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Caroline Kerridge
- the Lilly Research Centre, Eli Lilly and Co. Ltd., Erl Wood Manor, Sunninghill Road, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Michael J O'Neill
- the Lilly Research Centre, Eli Lilly and Co. Ltd., Erl Wood Manor, Sunninghill Road, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Joanna Wolak
- the Lilly Research Centre, Eli Lilly and Co. Ltd., Erl Wood Manor, Sunninghill Road, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Suchira Bose
- the Lilly Research Centre, Eli Lilly and Co. Ltd., Erl Wood Manor, Sunninghill Road, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Richard I Morimoto
- From the Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208 and
| |
Collapse
|
47
|
Modulation of proteostasis and protein trafficking: a therapeutic avenue for misfolded G protein-coupled receptors causing disease in humans. Emerg Top Life Sci 2019; 3:39-52. [PMID: 33523195 DOI: 10.1042/etls20180055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 11/17/2022]
Abstract
Proteostasis refers to the process whereby the cell maintains in equilibrium the protein content of different compartments. This system consists of a highly interconnected network intended to efficiently regulate the synthesis, folding, trafficking, and degradation of newly synthesized proteins. Molecular chaperones are key players of the proteostasis network. These proteins assist in the assembly and folding processes of newly synthesized proteins in a concerted manner to achieve a three-dimensional structure compatible with export from the endoplasmic reticulum to other cell compartments. Pharmacologic interventions intended to modulate the proteostasis network and tackle the devastating effects of conformational diseases caused by protein misfolding are under development. These include small molecules called pharmacoperones, which are highly specific toward the target protein serving as a molecular framework to cause misfolded mutant proteins to fold and adopt a stable conformation suitable for passing the scrutiny of the quality control system and reach its correct location within the cell. Here, we review the main components of the proteostasis network and how pharmacoperones may be employed to correct misfolding of two G protein-coupled receptors, the vasopressin 2 receptor and the gonadotropin-releasing hormone receptor, whose mutations lead to X-linked nephrogenic diabetes insipidus and congenital hypogonadotropic hypogonadism in humans respectively.
Collapse
|
48
|
Ben-Yakar A. High-Content and High-Throughput In Vivo Drug Screening Platforms Using Microfluidics. Assay Drug Dev Technol 2019; 17:8-13. [DOI: 10.1089/adt.2018.908] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Adela Ben-Yakar
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, Texas
- Adela Ben-Yakar from the Department of Mechanical Engineering, The University of Texas at Austin was awarded The President's Innovation award at the annual Society of Biomolecular Imaging and Informatics (SBI2) meeting held in Boston, September 2018
| |
Collapse
|
49
|
Voisine C, Brehme M. HSP90 et al.: Chaperome and Proteostasis Deregulation in Human Disease. HEAT SHOCK PROTEINS 2019. [DOI: 10.1007/978-3-030-23158-3_27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
50
|
Veit G, Xu H, Dreano E, Avramescu RG, Bagdany M, Beitel LK, Roldan A, Hancock MA, Lay C, Li W, Morin K, Gao S, Mak PA, Ainscow E, Orth AP, McNamara P, Edelman A, Frenkiel S, Matouk E, Sermet-Gaudelus I, Barnes WG, Lukacs GL. Structure-guided combination therapy to potently improve the function of mutant CFTRs. Nat Med 2018; 24:1732-1742. [PMID: 30297908 PMCID: PMC6301090 DOI: 10.1038/s41591-018-0200-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 08/08/2018] [Indexed: 12/17/2022]
Abstract
Available corrector drugs are unable to effectively rescue the folding defects of CFTR-ΔF508 (or CFTR-F508del), the most common disease-causing mutation of the cystic fibrosis transmembrane conductance regulator, a plasma membrane (PM) anion channel, and thus to substantially ameliorate clinical phenotypes of cystic fibrosis (CF). To overcome the corrector efficacy ceiling, here we show that compounds targeting distinct structural defects of CFTR can synergistically rescue mutant expression and function at the PM. High-throughput cell-based screens and mechanistic analysis identified three small-molecule series that target defects at nucleotide-binding domain (NBD1), NBD2 and their membrane-spanning domain (MSD) interfaces. Although individually these compounds marginally improve ΔF508-CFTR folding efficiency, function and stability, their combinations lead to ~50-100% of wild-type-level correction in immortalized and primary human airway epithelia and in mouse nasal epithelia. Likewise, corrector combinations were effective against rare missense mutations in various CFTR domains, probably acting via structural allostery, suggesting a mechanistic framework for their broad application.
Collapse
Affiliation(s)
- Guido Veit
- Department of Physiology, McGill University, Montréal, Quebec, Canada.
| | - Haijin Xu
- Department of Physiology, McGill University, Montréal, Quebec, Canada
| | - Elise Dreano
- Institut Necker-Enfants Malades (INEM)-INSERM U1151, Paris, France
| | - Radu G Avramescu
- Department of Physiology, McGill University, Montréal, Quebec, Canada
| | - Miklos Bagdany
- Department of Physiology, McGill University, Montréal, Quebec, Canada
| | - Lenore K Beitel
- Department of Physiology, McGill University, Montréal, Quebec, Canada
| | - Ariel Roldan
- Department of Physiology, McGill University, Montréal, Quebec, Canada
| | - Mark A Hancock
- SPR-MS Facility, McGill University, Montréal, Quebec, Canada
| | - Cecilia Lay
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Wei Li
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Katelin Morin
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Sandra Gao
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Puiying A Mak
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Edward Ainscow
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Anthony P Orth
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Peter McNamara
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | | | - Saul Frenkiel
- Department of Otolaryngology - Head and Neck Surgery, McGill University, Montréal, Quebec, Canada
| | - Elias Matouk
- Adult Cystic Fibrosis Clinic, Montreal Chest Institute, McGill University, Montréal, Quebec, Canada
| | | | - William G Barnes
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Gergely L Lukacs
- Department of Physiology, McGill University, Montréal, Quebec, Canada. .,Department of Biochemistry, McGill University, Montréal, Quebec, Canada. .,Groupe de Recherche Axé sur la Structure des Protéines (GRASP), McGill University, Montréal, Quebec, Canada.
| |
Collapse
|