1
|
Xu R, Kang Q, Yang X, Yi P, Zhang R. Unraveling Molecular Targets for Neurodegenerative Diseases Through Caenorhabditis elegans Models. Int J Mol Sci 2025; 26:3030. [PMID: 40243699 PMCID: PMC11988803 DOI: 10.3390/ijms26073030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Neurodegenerative diseases (NDDs), including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and prion disease, represent a group of age-related disorders that pose a growing and formidable challenge to global health. Despite decades of extensive research that has uncovered key genetic factors and biochemical pathways, the precise molecular mechanisms underlying these diseases and effective therapeutic strategies remain elusive. Caenorhabditis elegans (C. elegans) has emerged as a powerful model organism for studying NDDs due to its unique biological features such as genetic tractability, conserved molecular pathways, and ease of high-throughput screening. This model provides an exceptional platform for identifying molecular targets associated with NDDs and developing novel therapeutic interventions. This review highlights the critical role of C. elegans in elucidating the complex molecular mechanisms of human NDDs, with a particular focus on recent advancements and its indispensable contributions to the discovery of molecular targets and therapeutic strategies for these NDDs.
Collapse
Affiliation(s)
- Rongmei Xu
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei 230002, China; (R.X.); (X.Y.)
| | - Qiaoju Kang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; (Q.K.); (P.Y.)
| | - Xuefei Yang
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei 230002, China; (R.X.); (X.Y.)
| | - Ping Yi
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; (Q.K.); (P.Y.)
| | - Rongying Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; (Q.K.); (P.Y.)
| |
Collapse
|
2
|
Higashitani A, Teranishi M, Nakagawa Y, Itoh Y, Sudevan S, Szewczyk NJ, Kubota Y, Abe T, Kobayashi T. Increased mitochondrial Ca 2+ contributes to health decline with age and Duchene muscular dystrophy in C. elegans. FASEB J 2023; 37:e22851. [PMID: 36935171 PMCID: PMC10946577 DOI: 10.1096/fj.202201489rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 03/21/2023]
Abstract
Sarcopenia is a geriatric syndrome characterized by an age-related decline in skeletal muscle mass and strength. Here, we show that suppression of mitochondrial calcium uniporter (MCU)-mediated Ca2+ influx into mitochondria in the body wall muscles of the nematode Caenorhabditis elegans improved the sarcopenic phenotypes, blunting movement and mitochondrial structural and functional decline with age. We found that normally aged muscle cells exhibited elevated resting mitochondrial Ca2+ levels and increased mitophagy to eliminate damaged mitochondria. Similar to aging muscle, we found that suppressing MCU function in muscular dystrophy improved movement via reducing elevated resting mitochondrial Ca2+ levels. Taken together, our results reveal that elevated resting mitochondrial Ca2+ levels contribute to muscle decline with age and muscular dystrophy. Further, modulation of MCU activity may act as a potential pharmacological target in various conditions involving muscle loss.
Collapse
Affiliation(s)
| | - Mika Teranishi
- Graduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Yui Nakagawa
- Graduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Yukou Itoh
- Graduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Surabhi Sudevan
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby HospitalUniversity of NottinghamDerbyUK
| | - Nathaniel J. Szewczyk
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby HospitalUniversity of NottinghamDerbyUK
- Ohio Musculoskeletal and Neurologic Institute, Heritage College of Osteopathic MedicineOhio UniversityAthensOhioUSA
| | | | - Takaaki Abe
- Division of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Department of Clinical Biology and Hormonal RegulationTohoku University Graduate School of MedicineSendaiJapan
| | | |
Collapse
|
3
|
Shrivastava A, Sandhof CA, Reinle K, Jawed A, Ruger-Herreros C, Schwarz D, Creamer D, Nussbaum-Krammer C, Mogk A, Bukau B. The cytoprotective sequestration activity of small heat shock proteins is evolutionarily conserved. J Cell Biol 2022; 221:213447. [PMID: 36069810 PMCID: PMC9458469 DOI: 10.1083/jcb.202202149] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/21/2022] [Accepted: 08/16/2022] [Indexed: 11/22/2022] Open
Abstract
The chaperone-mediated sequestration of misfolded proteins into inclusions is a pivotal cellular strategy to maintain proteostasis in Saccharomyces cerevisiae, executed by small heat shock proteins (sHsps) Hsp42 and Btn2. Direct homologs of Hsp42 and Btn2 are absent in other organisms, questioning whether sequestration represents a conserved proteostasis strategy and, if so, which factors are involved. We examined sHsps from Escherchia coli, Caenorhabditis elegans, and humans for their ability to complement the defects of yeast sequestrase mutants. We show that sequestration of misfolded proteins is an original and widespread activity among sHsps executed by specific family members. Sequestrase positive C. elegans' sHsps harbor specific sequence features, including a high content of aromatic and methionine residues in disordered N-terminal extensions. Those sHsps buffer limitations in Hsp70 capacity in C. elegans WT animals and are upregulated in long-lived daf-2 mutants, contributing to lifespan extension. Cellular protection by sequestration of misfolded proteins is, therefore, an evolutionarily conserved activity of the sHsp family.
Collapse
Affiliation(s)
- Aseem Shrivastava
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carl Alexander Sandhof
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kevin Reinle
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Areeb Jawed
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carmen Ruger-Herreros
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominic Schwarz
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Declan Creamer
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Carmen Nussbaum-Krammer
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Axel Mogk
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernd Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
4
|
Bizat N, Parrales V, Laoues S, Normant S, Levavasseur E, Roussel J, Privat N, Gougerot A, Ravassard P, Beaudry P, Brandel JP, Laplanche JL, Haïk S. An in vivo Caenorhabditis elegans model for therapeutic research in human prion diseases. Brain 2021; 144:2745-2758. [PMID: 34687213 DOI: 10.1093/brain/awab152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/11/2021] [Accepted: 02/27/2021] [Indexed: 11/12/2022] Open
Abstract
Human prion diseases are fatal neurodegenerative disorders that include sporadic, infectious and genetic forms. Inherited Creutzfeldt-Jakob disease due to the E200K mutation of the prion protein-coding gene is the most common form of genetic prion disease. The phenotype resembles that of sporadic Creutzfeldt-Jakob disease at both the clinical and pathological levels, with a median disease duration of 4 months. To date, there is no available treatment for delaying the occurrence or slowing the progression of human prion diseases. Existing in vivo models do not allow high-throughput approaches that may facilitate the discovery of compounds targeting pathological assemblies of human prion protein or their effects on neuronal survival. Here, we generated a genetic model in the nematode Caenorhabditis elegans, which is devoid of any homologue of the prion protein, by expressing human prion protein with the E200K mutation in the mechanosensitive neuronal system. Expression of E200K prion protein induced a specific behavioural pattern and neurodegeneration of green fluorescent protein-expressing mechanosensitive neurons, in addition to the formation of intraneuronal inclusions associated with the accumulation of a protease-resistant form of the prion protein. We demonstrated that this experimental system is a powerful tool for investigating the efficacy of anti-prion compounds on both prion-induced neurodegeneration and prion protein misfolding, as well as in the context of human prion protein. Within a library of 320 compounds that have been approved for human use and cross the blood-brain barrier, we identified five molecules that were active against the aggregation of the E200K prion protein and the neurodegeneration it induced in transgenic animals. This model breaks a technological limitation in prion therapeutic research and provides a key tool to study the deleterious effects of misfolded prion protein in a well-described neuronal system.
Collapse
Affiliation(s)
- Nicolas Bizat
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne University, Hospital Pitié-Salpêtrière, F-75013 Paris, France.,Faculté de Pharmacie de Paris, Paris University, Paris F-75006, France
| | - Valeria Parrales
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne University, Hospital Pitié-Salpêtrière, F-75013 Paris, France
| | - Sofian Laoues
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne University, Hospital Pitié-Salpêtrière, F-75013 Paris, France
| | - Sébastien Normant
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne University, Hospital Pitié-Salpêtrière, F-75013 Paris, France
| | - Etienne Levavasseur
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne University, Hospital Pitié-Salpêtrière, F-75013 Paris, France
| | - Julian Roussel
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne University, Hospital Pitié-Salpêtrière, F-75013 Paris, France
| | - Nicolas Privat
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne University, Hospital Pitié-Salpêtrière, F-75013 Paris, France
| | - Alexianne Gougerot
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne University, Hospital Pitié-Salpêtrière, F-75013 Paris, France
| | - Philippe Ravassard
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne University, Hospital Pitié-Salpêtrière, F-75013 Paris, France
| | - Patrice Beaudry
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne University, Hospital Pitié-Salpêtrière, F-75013 Paris, France
| | - Jean-Philippe Brandel
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne University, Hospital Pitié-Salpêtrière, F-75013 Paris, France.,AP-HP, Cellule Nationale de Référence des Maladies de Creutzfeldt-Jakob, University Hospital Pitié-Salpêtrière, Paris F-75013, France
| | - Jean-Louis Laplanche
- Faculté de Pharmacie de Paris, Paris University, Paris F-75006, France.,Inserm, UMR-S 1144, Paris F-75006, France
| | - Stéphane Haïk
- Paris Brain Institute, Inserm U 1127, CNRS UMR 7225, Sorbonne University, Hospital Pitié-Salpêtrière, F-75013 Paris, France.,AP-HP, Cellule Nationale de Référence des Maladies de Creutzfeldt-Jakob, University Hospital Pitié-Salpêtrière, Paris F-75013, France
| |
Collapse
|
5
|
Cooper JF, Guasp RJ, Arnold ML, Grant BD, Driscoll M. Stress increases in exopher-mediated neuronal extrusion require lipid biosynthesis, FGF, and EGF RAS/MAPK signaling. Proc Natl Acad Sci U S A 2021; 118:e2101410118. [PMID: 34475208 PMCID: PMC8433523 DOI: 10.1073/pnas.2101410118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 06/23/2021] [Indexed: 01/08/2023] Open
Abstract
In human neurodegenerative diseases, neurons can transfer toxic protein aggregates to surrounding cells, promoting pathology via poorly understood mechanisms. In Caenorhabditis elegans, proteostressed neurons can expel neurotoxic proteins in large, membrane-bound vesicles called exophers. We investigated how specific stresses impact neuronal trash expulsion to show that neuronal exopher production can be markedly elevated by oxidative and osmotic stress. Unexpectedly, we also found that fasting dramatically increases exophergenesis. Mechanistic dissection focused on identifying nonautonomous factors that sense and activate the fasting-induced exopher response revealed that DAF16/FOXO-dependent and -independent processes are engaged. Fasting-induced exopher elevation requires the intestinal peptide transporter PEPT-1, lipid synthesis transcription factors Mediator complex MDT-15 and SBP-1/SREPB1, and fatty acid synthase FASN-1, implicating remotely initiated lipid signaling in neuronal trash elimination. A conserved fibroblast growth factor (FGF)/RAS/MAPK signaling pathway that acts downstream of, or in parallel to, lipid signaling also promotes fasting-induced neuronal exopher elevation. A germline-based epidermal growth factor (EGF) signal that acts through neurons is also required for exopher production. Our data define a nonautonomous network that links food availability changes to remote, and extreme, neuronal homeostasis responses relevant to aggregate transfer biology.
Collapse
Affiliation(s)
- Jason F Cooper
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| | - Ryan J Guasp
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| | - Meghan Lee Arnold
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| | - Barth D Grant
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
- Rutgers Center for Lipid Research, Rutgers University, New Brunswick, NJ 08901
| | - Monica Driscoll
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854;
| |
Collapse
|
6
|
Easy ultrastructural insight into the internal morphology of biological specimens by Atomic Force Microscopy. Sci Rep 2021; 11:10214. [PMID: 33986350 PMCID: PMC8119718 DOI: 10.1038/s41598-021-89633-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/27/2021] [Indexed: 02/08/2023] Open
Abstract
As a topographical technique, Atomic Force Microscopy (AFM) needs to establish direct interactions between a given sample and the measurement probe in order to create imaging information. The elucidation of internal features of organisms, tissues and cells by AFM has therefore been a challenging process in the past. To overcome this hindrance, simple and fast embedding, sectioning and dehydration techniques are presented, allowing the easy access to the internal morphology of virtually any organism, tissue or cell by AFM. The study at hand shows the applicability of the proposed protocol to exemplary biological samples, the resolution currently allowed by the approach as well as advantages and shortcomings compared to classical ultrastructural microscopic techniques like electron microscopy. The presented cheap, facile, fast and non-toxic experimental protocol might introduce AFM as a universal tool for the elucidation of internal ultrastructural detail of virtually any given organism, tissue or cell.
Collapse
|
7
|
Kinetic analysis reveals that independent nucleation events determine the progression of polyglutamine aggregation in C. elegans. Proc Natl Acad Sci U S A 2021; 118:2021888118. [PMID: 33836595 PMCID: PMC7980373 DOI: 10.1073/pnas.2021888118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Protein aggregation is associated with a wide range of degenerative human diseases with devastating consequences, as exemplified by Alzheimer's, Parkinson's, and Huntington's diseases. In vitro kinetic studies have provided a mechanistic understanding of the aggregation process at the molecular level. However, it has so far remained largely unclear to what extent the biophysical principles of amyloid formation learned in vitro translate to the complex environment of living organisms. Here, we take advantage of the unique properties of a Caenorhabditis elegans model expressing a fluorescently tagged polyglutamine (polyQ) protein, which aggregates into discrete micrometer-sized inclusions that can be directly visualized in real time. We provide a quantitative analysis of protein aggregation in this system and show that the data are described by a molecular model where stochastic nucleation occurs independently in each cell, followed by rapid aggregate growth. Global fitting of the image-based aggregation kinetics reveals a nucleation rate corresponding to 0.01 h-1 per cell at 1 mM intracellular protein concentration, and shows that the intrinsic molecular stochasticity of nucleation accounts for a significant fraction of the observed animal-to-animal variation. Our results highlight how independent, stochastic nucleation events in individual cells control the overall progression of polyQ aggregation in a living animal. The key finding that the biophysical principles associated with protein aggregation in small volumes remain the governing factors, even in the complex environment of a living organism, will be critical for the interpretation of in vivo data from a wide range of protein aggregation diseases.
Collapse
|
8
|
Arnold ML, Cooper J, Grant BD, Driscoll M. Quantitative Approaches for Scoring in vivo Neuronal Aggregate and Organelle Extrusion in Large Exopher Vesicles in C. elegans. J Vis Exp 2020:10.3791/61368. [PMID: 33016946 PMCID: PMC7805482 DOI: 10.3791/61368] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Toxicity of misfolded proteins and mitochondrial dysfunction are pivotal factors that promote age-associated functional neuronal decline and neurodegenerative disease across species. Although these neurotoxic challenges have long been considered to be cell-intrinsic, considerable evidence now supports that misfolded human disease proteins originating in one neuron can appear in neighboring cells, a phenomenon proposed to promote pathology spread in human neurodegenerative disease. C. elegans adult neurons that express aggregating proteins can extrude large (~4 µm) membrane-surrounded vesicles that can include the aggregated protein, mitochondria, and lysosomes. These large vesicles are called "exophers" and are distinct from exosomes (which are about 100x smaller and have different biogenesis). Throwing out cellular debris in exophers may occur by a conserved mechanism that constitutes a fundamental, but formerly unrecognized, branch of neuronal proteostasis and mitochondrial quality control, relevant to processes by which aggregates spread in human neurodegenerative diseases. While exophers have been mostly studied in animals that express high copy transgenic mCherry within touch neurons, these protocols are equally useful in the study of exophergenesis using fluorescently tagged organelles or other proteins of interest in various classes of neurons. Described here are the physical features of C. elegans exophers, strategies for their detection, identification criteria, optimal timing for quantitation, and animal growth protocols that control for stresses that can modulate exopher production levels. Together, details of protocols outlined here should serve to establish a standard for quantitative analysis of exophers across laboratories. This document seeks to serve as a resource in the field for laboratories seeking to elaborate molecular mechanisms by which exophers are produced and by which exophers are reacted to by neighboring and distant cells.
Collapse
Affiliation(s)
- Meghan Lee Arnold
- Department of Molecular Biology and Biochemistry, Rutgers University
| | - Jason Cooper
- Department of Molecular Biology and Biochemistry, Rutgers University
| | - Barth D Grant
- Department of Molecular Biology and Biochemistry, Rutgers University
| | - Monica Driscoll
- Department of Molecular Biology and Biochemistry, Rutgers University;
| |
Collapse
|
9
|
C. elegans Models to Study the Propagation of Prions and Prion-Like Proteins. Biomolecules 2020; 10:biom10081188. [PMID: 32824215 PMCID: PMC7464663 DOI: 10.3390/biom10081188] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022] Open
Abstract
A hallmark common to many age-related neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS), is that patients develop proteinaceous deposits in their central nervous system (CNS). The progressive spreading of these inclusions from initially affected sites to interconnected brain areas is reminiscent of the behavior of bona fide prions in transmissible spongiform encephalopathies (TSEs), hence the term prion-like proteins has been coined. Despite intensive research, the exact mechanisms that facilitate the spreading of protein aggregation between cells, and the associated loss of neurons, remain poorly understood. As population demographics in many countries continue to shift to higher life expectancy, the incidence of neurodegenerative diseases is also rising. This represents a major challenge for healthcare systems and patients’ families, since patients require extensive support over several years and there is still no therapy to cure or stop these diseases. The model organism Caenorhabditis elegans offers unique opportunities to accelerate research and drug development due to its genetic amenability, its transparency, and the high degree of conservation of molecular pathways. Here, we will review how recent studies that utilize this soil dwelling nematode have proceeded to investigate the propagation and intercellular transmission of prions and prion-like proteins and discuss their relevance by comparing their findings to observations in other model systems and patients.
Collapse
|
10
|
McCombe PA, Garton FC, Katz M, Wray NR, Henderson RD. What do we know about the variability in survival of patients with amyotrophic lateral sclerosis? Expert Rev Neurother 2020; 20:921-941. [PMID: 32569484 DOI: 10.1080/14737175.2020.1785873] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION ALS is a fatal neurodegenerative disease. However, patients show variability in the length of survival after symptom onset. Understanding the mechanisms of long survival could lead to possible avenues for therapy. AREAS COVERED This review surveys the reported length of survival in ALS, the clinical features that predict survival in individual patients, and possible factors, particularly genetic factors, that could cause short or long survival. The authors also speculate on possible mechanisms. EXPERT OPINION a small number of known factors can explain some variability in ALS survival. However, other disease-modifying factors likely exist. Factors that alter motor neurone vulnerability and immune, metabolic, and muscle function could affect survival by modulating the disease process. Knowing these factors could lead to interventions to change the course of the disease. The authors suggest a broad approach is needed to quantify the proportion of variation survival attributable to genetic and non-genetic factors and to identify and estimate the effect size of specific factors. Studies of this nature could not only identify novel avenues for therapeutic research but also play an important role in clinical trial design and personalized medicine.
Collapse
Affiliation(s)
- Pamela A McCombe
- Centre for Clinical Research, The University of Queensland , Brisbane, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital , Brisbane, Australia
| | - Fleur C Garton
- Institute for Molecular Biosciences, The University of Queensland , Brisbane, Australia
| | - Matthew Katz
- Department of Neurology, Royal Brisbane and Women's Hospital , Brisbane, Australia
| | - Naomi R Wray
- Institute for Molecular Biosciences, The University of Queensland , Brisbane, Australia.,Queensland Brain Institute, The University of Queensland , Brisbane, Australia
| | - Robert D Henderson
- Centre for Clinical Research, The University of Queensland , Brisbane, Australia
| |
Collapse
|
11
|
Van Pelt KM, Truttmann MC. Caenorhabditis elegans as a model system for studying aging-associated neurodegenerative diseases. TRANSLATIONAL MEDICINE OF AGING 2020; 4:60-72. [PMID: 34327290 PMCID: PMC8317484 DOI: 10.1016/j.tma.2020.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases (NDs) are a heterogeneous group of aging-associated disorders characterized by the disruption of cellular proteostasis machinery and the misfolding of distinct protein species to form toxic aggregates in neurons. The increasing prevalence of NDs represents a growing healthcare burden worldwide, a concern compounded by the fact that few, if any, treatments exist to target the underlying cause of these diseases. Consequently, the application of a high-throughput, physiologically relevant model system to studies dissecting the molecular mechanisms governing ND pathology is crucial for identifying novel avenues for the development of targeted therapeutics. The nematode Caenorhabditis elegans (C. elegans) has emerged as a powerful tool for the study of disease mechanisms due to its ease of genetic manipulation and swift cultivation, while providing a whole-animal system amendable to numerous molecular and biochemical techniques. To date, numerous C. elegans models have been generated for a variety of NDs, allowing for the large-scale in vivo study of protein-conformation disorders. Furthermore, the comparatively low barriers to entry in the development of transgenic worm models have facilitated the modeling of rare or "orphan" NDs, thereby providing unparalleled insight into the shared mechanisms underlying these pathologies. In this review, we summarize findings from a comprehensive collection of C. elegans neurodegenerative disease models of varying prevalence to emphasize shared mechanisms of proteotoxicity, and highlight the utility of these models in elucidating the molecular basis of ND pathologies.
Collapse
Affiliation(s)
- Kate M. Van Pelt
- Cellular & Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Matthias C. Truttmann
- Cellular & Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Geriatrics Center, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
12
|
Tittelmeier J, Sandhof CA, Ries HM, Druffel-Augustin S, Mogk A, Bukau B, Nussbaum-Krammer C. The HSP110/HSP70 disaggregation system generates spreading-competent toxic α-synuclein species. EMBO J 2020; 39:e103954. [PMID: 32449565 PMCID: PMC7327497 DOI: 10.15252/embj.2019103954] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 04/19/2020] [Accepted: 04/23/2020] [Indexed: 12/21/2022] Open
Abstract
The accumulation and prion-like propagation of α-synuclein and other amyloidogenic proteins are associated with devastating neurodegenerative diseases. Metazoan heat shock protein HSP70 and its co-chaperones DNAJB1 and HSP110 constitute a disaggregation machinery that is able to disassemble α-synuclein fibrils in vitro, but its physiological effects on α-synuclein toxicity are unknown. Here, we depleted Caenorhabditis elegans HSP-110 and monitored the consequences on α-synuclein-related pathological phenotypes such as misfolding, intercellular spreading, and toxicity in C. elegans in vivo models. Depletion of HSP-110 impaired HSP70 disaggregation activity, prevented resolubilization of amorphous aggregates, and compromised the overall cellular folding capacity. At the same time, HSP-110 depletion reduced α-synuclein foci formation, cell-to-cell transmission, and toxicity. These data demonstrate that the HSP70 disaggregation activity constitutes a double-edged sword, as it is essential for maintaining cellular proteostasis but also involved in the generation of toxic amyloid-type protein species.
Collapse
Affiliation(s)
- Jessica Tittelmeier
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Carl Alexander Sandhof
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Heidrun Maja Ries
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Silke Druffel-Augustin
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Axel Mogk
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Bernd Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Carmen Nussbaum-Krammer
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| |
Collapse
|
13
|
Sandhof CA, Hoppe SO, Druffel-Augustin S, Gallrein C, Kirstein J, Voisine C, Nussbaum-Krammer C. Reducing INS-IGF1 signaling protects against non-cell autonomous vesicle rupture caused by SNCA spreading. Autophagy 2020; 16:878-899. [PMID: 31354022 PMCID: PMC7144869 DOI: 10.1080/15548627.2019.1643657] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 07/02/2019] [Accepted: 07/11/2019] [Indexed: 01/28/2023] Open
Abstract
Aging is associated with a gradual decline of cellular proteostasis, giving rise to devastating protein misfolding diseases, such as Alzheimer disease (AD) or Parkinson disease (PD). These diseases often exhibit a complex pathology involving non-cell autonomous proteotoxic effects, which are still poorly understood. Using Caenorhabditis elegans we investigated how local protein misfolding is affecting neighboring cells and tissues showing that misfolded PD-associated SNCA/α-synuclein is accumulating in highly dynamic endo-lysosomal vesicles. Irrespective of whether being expressed in muscle cells or dopaminergic neurons, accumulated proteins were transmitted into the hypodermis with increasing age, indicating that epithelial cells might play a role in remote degradation when the local endo-lysosomal degradation capacity is overloaded. Cell biological and genetic approaches revealed that inter-tissue dissemination of SNCA was regulated by endo- and exocytosis (neuron/muscle to hypodermis) and basement membrane remodeling (muscle to hypodermis). Transferred SNCA conformers were, however, inefficiently cleared and induced endo-lysosomal membrane permeabilization. Remarkably, reducing INS (insulin)-IGF1 (insulin-like growth factor 1) signaling provided protection by maintaining endo-lysosomal integrity. This study suggests that the degradation of lysosomal substrates is coordinated across different tissues in metazoan organisms. Because the chronic dissemination of poorly degradable disease proteins into neighboring tissues exerts a non-cell autonomous toxicity, this implies that restoring endo-lysosomal function not only in cells with pathological inclusions, but also in apparently unaffected cell types might help to halt disease progression.Abbreviations: AD: Alzheimer disease; BM: basement membrane; BWM: body wall muscle; CEP: cephalic sensilla; CLEM: correlative light and electron microscopy; CTNS-1: cystinosin (lysosomal protein) homolog; DA: dopaminergic; DAF-2: abnormal dauer formation; ECM: extracellular matrix; FLIM: fluorescence lifetime imaging microscopy; fps: frames per second; GFP: green fluorescent protein; HPF: high pressure freezing; IGF1: insulin-like growth factor 1; INS: insulin; KD: knockdown; LMP: lysosomal membrane permeabilization; MVB: multivesicular body; NOC: nocodazole; PD: Parkinson disease; RFP: red fluorescent protein; RNAi: RNA interference; sfGFP: superfolder GFP; SNCA: synuclein alpha; TEM: transmission electron microscopy; TNTs: tunneling nanotubes; TCSPC: time correlated single photon counting; YFP: yellow fluorescent protein.
Collapse
Affiliation(s)
- Carl Alexander Sandhof
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Simon Oliver Hoppe
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Silke Druffel-Augustin
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Christian Gallrein
- Department of Molecular Physiology and Cell Biology, Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin e.V, Berlin, Germany
| | - Janine Kirstein
- Department of Molecular Physiology and Cell Biology, Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin e.V, Berlin, Germany
| | - Cindy Voisine
- Department of Biology, Northeastern Illinois University, Chicago, IL, USA
| | - Carmen Nussbaum-Krammer
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| |
Collapse
|
14
|
Youssef K, Tandon A, Rezai P. Studying Parkinson’s disease using Caenorhabditis elegans models in microfluidic devices. Integr Biol (Camb) 2019; 11:186-207. [DOI: 10.1093/intbio/zyz017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/30/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022]
Abstract
Abstract
Parkinson’s disease (PD) is a progressive neurological disorder associated with the loss of dopaminergic neurons (DNs) in the substantia nigra and the widespread accumulation of α-synuclein (α-syn) protein, leading to motor impairments and eventual cognitive dysfunction. In-vitro cell cultures and in-vivo animal models have provided the opportunity to investigate the PD pathological hallmarks and identify different therapeutic compounds. However, PD pathogenesis and causes are still not well understood, and effective inhibitory drugs for PD are yet to be discovered. Biologically simple but pathologically relevant disease models and advanced screening technologies are needed to reveal the mechanisms underpinning protein aggregation and PD progression. For instance, Caenorhabditis elegans (C. elegans) offers many advantages for fundamental PD neurobehavioral studies including a simple, well-mapped, and accessible neuronal system, genetic homology to humans, body transparency and amenability to genetic manipulation. Several transgenic worm strains that exhibit multiple PD-related phenotypes have been developed to perform neuronal and behavioral assays and drug screening. However, in conventional worm-based assays, the commonly used techniques are equipment-intensive, slow and low in throughput. Over the past two decades, microfluidics technology has contributed significantly to automation and control of C. elegans assays. In this review, we focus on C. elegans PD models and the recent advancements in microfluidic platforms used for manipulation, handling and neurobehavioral screening of these models. Moreover, we highlight the potential of C. elegans to elucidate the in-vivo mechanisms of neuron-to-neuron protein transfer that may underlie spreading Lewy pathology in PD, and its suitability for in-vitro studies. Given the advantages of C. elegans and microfluidics technology, their integration has the potential to facilitate the investigation of disease pathology and discovery of potential chemical leads for PD.
Collapse
Affiliation(s)
- Khaled Youssef
- Department of Mechanical Engineering, York University, Toronto, ON, Canada
| | - Anurag Tandon
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Pouya Rezai
- Department of Mechanical Engineering, York University, Toronto, ON, Canada
| |
Collapse
|
15
|
Bistaffa E, Rossi M, De Luca CMG, Cazzaniga F, Carletta O, Campagnani I, Tagliavini F, Legname G, Giaccone G, Moda F. Prion Efficiently Replicates in α-Synuclein Knockout Mice. Mol Neurobiol 2019; 56:7448-7457. [PMID: 31041657 DOI: 10.1007/s12035-019-1602-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/09/2019] [Indexed: 10/26/2022]
Abstract
Prion diseases are a group of neurodegenerative disorders associated with the conformational conversion of the cellular prion protein (PrPC) into an abnormal misfolded form named PrPSc. Other than accumulating in the brain, PrPSc can bind PrPC and force it to change conformation to PrPSc. The exact mechanism which underlies the process of PrPC/PrPSc conversion still needs to be defined and many molecules or cofactors might be involved. Several studies have documented an important role of PrPC to act as receptor for abnormally folded forms of α-synuclein which are responsible of a group of diseases known as synucleinopathies. The presence of PrPC was required to promote efficient internalization and spreading of abnormal α-synuclein between cells. In this work, we have assessed whether α-synuclein exerts any role in PrPSc conversion and propagation either in vitro or in vivo. Indeed, understanding the mechanism of PrPC/PrPSc conversion and the identification of cofactors involved in this process is crucial for developing new therapeutic strategies. Our results showed that PrPSc was able to efficiently propagate in the brain of animals even in the absence of α-synuclein thus suggesting that this protein did not act as key modulator of prion propagation. Thus, α-synuclein might take part in this process but is not specifically required for sustaining prion conversion and propagation.
Collapse
Affiliation(s)
- Edoardo Bistaffa
- Unit of Neuropathology and Neurology 5, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Martina Rossi
- Department of Neuroscience, Laboratory of Prion Biology, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Chiara Maria Giulia De Luca
- Unit of Neuropathology and Neurology 5, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Department of Neuroscience, Laboratory of Prion Biology, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Federico Cazzaniga
- Unit of Neuropathology and Neurology 5, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Olga Carletta
- Unit of Neuropathology and Neurology 5, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ilaria Campagnani
- Unit of Neuropathology and Neurology 5, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fabrizio Tagliavini
- Unit of Neuropathology and Neurology 5, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Legname
- Department of Neuroscience, Laboratory of Prion Biology, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Giorgio Giaccone
- Unit of Neuropathology and Neurology 5, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fabio Moda
- Unit of Neuropathology and Neurology 5, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| |
Collapse
|
16
|
Sil TB, Sahoo B, Bera SC, Garai K. Quantitative Characterization of Metastability and Heterogeneity of Amyloid Aggregates. Biophys J 2019; 114:800-811. [PMID: 29490242 DOI: 10.1016/j.bpj.2017.12.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/23/2017] [Accepted: 12/11/2017] [Indexed: 01/13/2023] Open
Abstract
Amyloids are heterogeneous assemblies of extremely stable fibrillar aggregates of proteins. Although biological activities of the amyloids are dependent on its conformation, quantitative evaluation of heterogeneity of amyloids has been difficult. Here we use disaggregation of the amyloids of tetramethylrhodamine-labeled Aβ (TMR-Aβ) to characterize its stability and heterogeneity. Disaggregation of TMR-Aβ amyloids, monitored by fluorescence recovery of TMR, was negligible in native buffer even at low nanomolar concentrations but the kinetics increased exponentially with addition of denaturants such as urea or GdnCl. However, dissolution of TMR-Aβ amyloids is different from what is expected in the case of thermodynamic solubility. For example, the fraction of soluble amyloids is found to be independent of total concentration of the peptide at all concentrations of the denaturants. Additionally, soluble fraction is dependent on growth conditions such as temperature, pH, and aging of the amyloids. Furthermore, amyloids undissolved in a certain concentration of the denaturant do not show any further dissolution after dilution in the same solvent; instead, these require higher concentrations of the denaturant. Taken together, our results indicate that amyloids are a heterogeneous ensemble of metastable states. Furthermore, dissolution of each structurally homogeneous member requires a unique threshold concentration of denaturant. Fraction of soluble amyloids as a function of concentration of denaturants is found to be sigmoidal. The sigmoidal curve becomes progressively steeper with progressive seeding of the amyloids, although the midpoint remains unchanged. Therefore, heterogeneity of the amyloids is a major determinant of the steepness of the sigmoidal curve. The sigmoidal curve can be fit assuming a normal distribution for the population of the amyloids of various kinetic stabilities. We propose that the mean and the standard deviation of the normal distribution provide quantitative estimates of mean kinetic stability and heterogeneity, respectively, of the amyloids in a certain preparation.
Collapse
Affiliation(s)
- Timir Baran Sil
- Tata Institute of Fundamental Research, Serilingampally, Hyderabad, India
| | - Bankanidhi Sahoo
- Tata Institute of Fundamental Research, Serilingampally, Hyderabad, India
| | | | - Kanchan Garai
- Tata Institute of Fundamental Research, Serilingampally, Hyderabad, India.
| |
Collapse
|
17
|
Madhivanan K, Greiner ER, Alves-Ferreira M, Soriano-Castell D, Rouzbeh N, Aguirre CA, Paulsson JF, Chapman J, Jiang X, Ooi FK, Lemos C, Dillin A, Prahlad V, Kelly JW, Encalada SE. Cellular clearance of circulating transthyretin decreases cell-nonautonomous proteotoxicity in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2018; 115:E7710-E7719. [PMID: 30061394 PMCID: PMC6099907 DOI: 10.1073/pnas.1801117115] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cell-autonomous and cell-nonautonomous mechanisms of neurodegeneration appear to occur in the proteinopathies, including Alzheimer's and Parkinson's diseases. However, how neuronal toxicity is generated from misfolding-prone proteins secreted by nonneuronal tissues and whether modulating protein aggregate levels at distal locales affects the degeneration of postmitotic neurons remains unknown. We generated and characterized animal models of the transthyretin (TTR) amyloidoses that faithfully recapitulate cell-nonautonomous neuronal proteotoxicity by expressing human TTR in the Caenorhabditis elegans muscle. We identified sensory neurons with affected morphological and behavioral nociception-sensing impairments. Nonnative TTR oligomer load and neurotoxicity increased following inhibition of TTR degradation in distal macrophage-like nonaffected cells. Moreover, reducing TTR levels by RNAi or by kinetically stabilizing natively folded TTR pharmacologically decreased TTR aggregate load and attenuated neuronal dysfunction. These findings reveal a critical role for in trans modulation of aggregation-prone degradation that directly affects postmitotic tissue degeneration observed in the proteinopathies.
Collapse
Affiliation(s)
- Kayalvizhi Madhivanan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| | - Erin R Greiner
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| | - Miguel Alves-Ferreira
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
- Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-171 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4150-171 Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, 4150-171 Porto, Portugal
| | - David Soriano-Castell
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| | - Nirvan Rouzbeh
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| | - Carlos A Aguirre
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| | - Johan F Paulsson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | | | - Xin Jiang
- Misfolding Diagnostics, San Diego, CA 92121
| | - Felicia K Ooi
- Department of Biology, Aging Mind and Brain Initiative, University of Iowa, Iowa City, IA 52242
| | - Carolina Lemos
- Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-171 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4150-171 Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, 4150-171 Porto, Portugal
| | - Andrew Dillin
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- Howard Hughes Medical Institute, University of California, Berkeley, CA 94720
| | - Veena Prahlad
- Department of Biology, Aging Mind and Brain Initiative, University of Iowa, Iowa City, IA 52242
| | - Jeffery W Kelly
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Sandra E Encalada
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037;
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
18
|
Arnold ML, Melentijevic I, Smart AJ, Driscoll M. Q&A: Trash talk: disposal and remote degradation of neuronal garbage. BMC Biol 2018; 16:17. [PMID: 29382333 PMCID: PMC5789552 DOI: 10.1186/s12915-018-0487-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Caenorhabditis elegans neurons have recently been found to throw out cellular debris for remote degradation and/or storage, adding an “extracellular garbage elimination” option to known intracellular protein and organelle degradation pathways. This Q&A describes initial insights into the biology of seemingly selective protein and organelle elimination by challenged neurons, highlighting mysteries of how garbage is distinguished and sorted in the sending neuron, how the garbage-filled “exophers” appear to elicit degradative responses as they transit neighboring tissue, and how non-digestible materials get thrown out of cells again via processes that may be highly relevant to human neurodegenerative disease mechanisms.
Collapse
Affiliation(s)
- Meghan Lee Arnold
- Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, A232 Nelson Biological Laboratories, 604 Allison Road, Piscataway, NJ, 08855, USA
| | - Ilija Melentijevic
- Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, A232 Nelson Biological Laboratories, 604 Allison Road, Piscataway, NJ, 08855, USA
| | - Anna Joelle Smart
- Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, A232 Nelson Biological Laboratories, 604 Allison Road, Piscataway, NJ, 08855, USA
| | - Monica Driscoll
- Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, A232 Nelson Biological Laboratories, 604 Allison Road, Piscataway, NJ, 08855, USA.
| |
Collapse
|
19
|
Griffin EF, Caldwell KA, Caldwell GA. Genetic and Pharmacological Discovery for Alzheimer's Disease Using Caenorhabditis elegans. ACS Chem Neurosci 2017; 8:2596-2606. [PMID: 29022701 DOI: 10.1021/acschemneuro.7b00361] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The societal burden presented by Alzheimer's disease warrants both innovative and expedient means by which its underlying molecular causes can be both identified and mechanistically exploited to discern novel therapeutic targets and strategies. The conserved characteristics, defined neuroanatomy, and advanced technological application of Caenorhabditis elegans render this metazoan an unmatched tool for probing neurotoxic factors. In addition, its short lifespan and importance in the field of aging make it an ideal organism for modeling age-related neurodegenerative disease. As such, this nematode system has demonstrated its value in predicting functional modifiers of human neurodegenerative disorders. Here, we review how C. elegans has been utilized to model Alzheimer's disease. Specifically, we present how the causative neurotoxic peptides, amyloid-β and tau, contribute to disease-like neurodegeneration in C. elegans and how they translate to human disease. Furthermore, we describe how a variety of transgenic animal strains, each with distinct utility, have been used to identify both genetic and pharmacological modifiers of toxicity in C. elegans. As technological advances improve the prospects for intervention, the rapidity, unparalleled accuracy, and scale that C. elegans offers researchers for defining functional modifiers of neurodegeneration should speed the discovery of improved therapies for Alzheimer's disease.
Collapse
Affiliation(s)
- Edward F. Griffin
- Department
of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Kim A. Caldwell
- Department
of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Guy A. Caldwell
- Department
of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama 35487, United States
- Departments
of Neurology and Neurobiology, Center for Neurodegeneration and Experimental
Therapeutics, The University of Alabama School of Medicine at Birmingham, Birmingham, Alabama 35294, United States
| |
Collapse
|
20
|
Kumar DKV, Choi SH, Washicosky KJ, Eimer WA, Tucker S, Ghofrani J, Lefkowitz A, McColl G, Goldstein LE, Tanzi RE, Moir RD. Amyloid-β peptide protects against microbial infection in mouse and worm models of Alzheimer's disease. Sci Transl Med 2017; 8:340ra72. [PMID: 27225182 DOI: 10.1126/scitranslmed.aaf1059] [Citation(s) in RCA: 722] [Impact Index Per Article: 90.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 05/10/2016] [Indexed: 12/19/2022]
Abstract
The amyloid-β peptide (Aβ) is a key protein in Alzheimer's disease (AD) pathology. We previously reported in vitro evidence suggesting that Aβ is an antimicrobial peptide. We present in vivo data showing that Aβ expression protects against fungal and bacterial infections in mouse, nematode, and cell culture models of AD. We show that Aβ oligomerization, a behavior traditionally viewed as intrinsically pathological, may be necessary for the antimicrobial activities of the peptide. Collectively, our data are consistent with a model in which soluble Aβ oligomers first bind to microbial cell wall carbohydrates via a heparin-binding domain. Developing protofibrils inhibited pathogen adhesion to host cells. Propagating β-amyloid fibrils mediate agglutination and eventual entrapment of unatttached microbes. Consistent with our model, Salmonella Typhimurium bacterial infection of the brains of transgenic 5XFAD mice resulted in rapid seeding and accelerated β-amyloid deposition, which closely colocalized with the invading bacteria. Our findings raise the intriguing possibility that β-amyloid may play a protective role in innate immunity and infectious or sterile inflammatory stimuli may drive amyloidosis. These data suggest a dual protective/damaging role for Aβ, as has been described for other antimicrobial peptides.
Collapse
Affiliation(s)
- Deepak Kumar Vijaya Kumar
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Kevin J Washicosky
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - William A Eimer
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Stephanie Tucker
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Jessica Ghofrani
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Aaron Lefkowitz
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Gawain McColl
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Lee E Goldstein
- Department of Psychiatry, Boston University, Boston, MA 02215, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| | - Robert D Moir
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
21
|
Shape matters: the complex relationship between aggregation and toxicity in protein-misfolding diseases. Essays Biochem 2017; 60:181-190. [PMID: 27744334 DOI: 10.1042/ebc20160008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/29/2016] [Indexed: 01/07/2023]
Abstract
A particular subgroup of protein-misfolding diseases, comprising Alzheimer's and Parkinson's disease, involves amyloidogenic proteins that can form alternative pathogenic conformations with a high tendency to self-assemble into oligomeric and fibrillar species. Although misfolded proteins have been clearly linked to disease, the exact nature of the toxic species remains highly controversial. Increasing evidence suggests that there is little correlation between the occurrence of macroscopic protein deposits and toxic phenotypes in affected cells and tissues. In this article, we recap amyloid aggregation pathways, describe prion-like propagation, elaborate on detrimental interactions of protein aggregates with the cellular protein quality control system and discuss why some aggregates are toxic, whereas others seem to be beneficial. On the basis of recent studies on prion strains, we reason that the specific aggregate conformation and the resulting individual interaction with the cellular environment might be the major determinant of toxicity.
Collapse
|
22
|
Regulation of cell-non-autonomous proteostasis in metazoans. Essays Biochem 2017; 60:133-142. [PMID: 27744329 PMCID: PMC5065704 DOI: 10.1042/ebc20160006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/28/2016] [Indexed: 12/24/2022]
Abstract
Cells have developed robust adaptation mechanisms to survive environmental conditions that challenge the integrity of their proteome and ensure cellular viability. These are stress signalling pathways that integrate extracellular signals with the ability to detect and efficiently respond to protein-folding perturbations within the cell. Within the context of an organism, the cell-autonomous effects of these signalling mechanisms are superimposed by cell-non-autonomous stress signalling pathways that allow co-ordination of stress responses across tissues. These transcellular stress signalling pathways orchestrate and maintain the cellular proteome at an organismal level. This article focuses on mechanisms in both invertebrate and vertebrate organisms that activate stress responses in a cell-non-autonomous manner. We discuss emerging insights and provide specific examples on how components of the cell-non-autonomous proteostasis network are used in cancer and protein-folding diseases to drive disease progression across tissues.
Collapse
|
23
|
Effects of Mutations on the Aggregation Propensity of the Human Prion-Like Protein hnRNPA2B1. Mol Cell Biol 2017; 37:MCB.00652-16. [PMID: 28137911 DOI: 10.1128/mcb.00652-16] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 01/19/2017] [Indexed: 12/31/2022] Open
Abstract
Hundreds of human proteins contain prion-like domains, which are a subset of low-complexity domains with high amino acid compositional similarity to yeast prion domains. A recently characterized mutation in the prion-like domain of the human heterogeneous nuclear ribonucleoprotein hnRNPA2B1 increases the aggregation propensity of the protein and causes multisystem proteinopathy. The mutant protein forms cytoplasmic inclusions when expressed in Drosophila, the mutation accelerates aggregation in vitro, and the mutant prion-like domain can substitute for a portion of a yeast prion domain in supporting prion activity. To examine the relationship between amino acid sequence and aggregation propensity, we made a diverse set of point mutations in the hnRNPA2B1 prion-like domain. We found that the effects on prion formation in Saccharomyces cerevisiae and aggregation in vitro could be predicted entirely based on amino acid composition. However, composition was an imperfect predictor of inclusion formation in Drosophila; while most mutations showed similar behaviors in yeast, in vitro, and in Drosophila, a few showed anomalous behavior. Collectively, these results demonstrate the significant progress that has been made in predicting the effects of mutations on intrinsic aggregation propensity while also highlighting the challenges of predicting the effects of mutations in more complex organisms.
Collapse
|
24
|
C. elegans neurons jettison protein aggregates and mitochondria under neurotoxic stress. Nature 2017; 542:367-371. [PMID: 28178240 PMCID: PMC5336134 DOI: 10.1038/nature21362] [Citation(s) in RCA: 300] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 01/03/2017] [Indexed: 12/12/2022]
|
25
|
Kikis EA. The struggle by Caenorhabditis elegans to maintain proteostasis during aging and disease. Biol Direct 2016; 11:58. [PMID: 27809888 PMCID: PMC5093949 DOI: 10.1186/s13062-016-0161-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 10/24/2016] [Indexed: 01/07/2023] Open
Abstract
The presence of only small amounts of misfolded protein is an indication of a healthy proteome. Maintaining proteome health, or more specifically, “proteostasis,” is the purview of the “proteostasis network.” This network must respond to constant fluctuations in the amount of destabilized proteins caused by errors in protein synthesis and exposure to acute proteotoxic conditions. Aging is associated with a gradual increase in damaged and misfolded protein, which places additional stress on the machinery of the proteostasis network. In fact, despite the ability of the proteostasis machinery to readjust its stoichiometry in an attempt to maintain homeostasis, the capacity of cells to buffer against misfolding is strikingly limited. Therefore, subtle changes in the folding environment that occur during aging can significantly impact the health of the proteome. This decline and eventual collapse in proteostasis is most pronounced in individuals with neurodegenerative disorders such as Alzheimer’s Disease, Parkinson’s Disease, and Huntington’s Disease that are caused by the misfolding, aggregation, and toxicity of certain proteins. This review discusses how C. elegans models of protein misfolding have contributed to our current understanding of the proteostasis network, its buffering capacity, and its regulation. Reviewers: This article was reviewed by Luigi Bubacco, Patrick Lewis and Xavier Roucou.
Collapse
Affiliation(s)
- Elise A Kikis
- Biology Department, The University of the South, 735 University Avenue, Sewanee, TN, 37383, USA.
| |
Collapse
|
26
|
Abstract
UNLABELLED Prions are infectious protein particles that replicate by templating their aggregated state onto soluble protein of the same type. Originally identified as the causative agent of transmissible spongiform encephalopathies, prions in yeast (Saccharomyces cerevisiae) are epigenetic elements of inheritance that induce phenotypic changes of their host cells. The prototype yeast prion is the translation termination factor Sup35. Prions composed of Sup35 or its modular prion domain NM are heritable and are transmitted vertically to progeny or horizontally during mating. Interestingly, in mammalian cells, protein aggregates derived from yeast Sup35 NM behave as true infectious entities that employ dissemination strategies similar to those of mammalian prions. While transmission is most efficient when cells are in direct contact, we demonstrate here that cytosolic Sup35 NM prions are also released into the extracellular space in association with nanometer-sized membrane vesicles. Importantly, extracellular vesicles are biologically active and are taken up by recipient cells, where they induce self-sustained Sup35 NM protein aggregation. Thus, in mammalian cells, extracellular vesicles can serve as dissemination vehicles for protein-based epigenetic information transfer. IMPORTANCE Prions are proteinaceous infectious particles that propagate by templating their quaternary structure onto nascent proteins of the same kind. Prions in yeast act as heritable epigenetic elements that can alter the phenotype when transmitted to daughter cells or during mating. Prion activity is conferred by so-called prion domains often enriched in glutamine and asparagine residues. Interestingly, many mammalian proteins also contain domains with compositional similarity to yeast prion domains. We have recently provided a proof-of-principle demonstration that a yeast prion domain also retains its prion activity in mammalian cells. We demonstrate here that cytosolic prions composed of a yeast prion domain are also packaged into extracellular vesicles that transmit the prion phenotype to bystander cells. Thus, proteins with prion-like domains can behave as proteinaceous information molecules that exploit the cellular vesicle trafficking machinery for intercellular long-distance dissemination.
Collapse
|
27
|
Feleciano DR, Kirstein J. Collapse of redox homeostasis during aging and stress. Mol Cell Oncol 2016; 3:e1091060. [PMID: 27308612 PMCID: PMC4905398 DOI: 10.1080/23723556.2015.1091060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 08/31/2015] [Accepted: 08/31/2015] [Indexed: 01/08/2023]
Abstract
Coordination of the protein homeostasis network and redox states in eukaryotic cells is crucial for cellular and organismal fitness. By employing endogenous in vivo redox sensors we demonstrate that the redox state of the ER and cytosol is subject to profound changes upon proteotoxic challenges and during aging.
Collapse
Affiliation(s)
- Diogo R. Feleciano
- Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund, Berlin, Germany
| | - Janine Kirstein
- Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund, Berlin, Germany
| |
Collapse
|
28
|
Chen X, Barclay JW, Burgoyne RD, Morgan A. Using C. elegans to discover therapeutic compounds for ageing-associated neurodegenerative diseases. Chem Cent J 2015; 9:65. [PMID: 26617668 PMCID: PMC4661952 DOI: 10.1186/s13065-015-0143-y] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/15/2015] [Indexed: 12/24/2022] Open
Abstract
Age-associated neurodegenerative disorders such as Alzheimer's disease are a major public health challenge, due to the demographic increase in the proportion of older individuals in society. However, the relatively few currently approved drugs for these conditions provide only symptomatic relief. A major goal of neurodegeneration research is therefore to identify potential new therapeutic compounds that can slow or even reverse disease progression, either by impacting directly on the neurodegenerative process or by activating endogenous physiological neuroprotective mechanisms that decline with ageing. This requires model systems that can recapitulate key features of human neurodegenerative diseases that are also amenable to compound screening approaches. Mammalian models are very powerful, but are prohibitively expensive for high-throughput drug screens. Given the highly conserved neurological pathways between mammals and invertebrates, Caenorhabditis elegans has emerged as a powerful tool for neuroprotective compound screening. Here we describe how C. elegans has been used to model various human ageing-associated neurodegenerative diseases and provide an extensive list of compounds that have therapeutic activity in these worm models and so may have translational potential.
Collapse
Affiliation(s)
- Xi Chen
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown St, Liverpool, L69 3BX UK ; Centre for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Avenue NE, Grand Rapids, Michigan, MI 49503 USA
| | - Jeff W Barclay
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown St, Liverpool, L69 3BX UK
| | - Robert D Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown St, Liverpool, L69 3BX UK
| | - Alan Morgan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown St, Liverpool, L69 3BX UK
| |
Collapse
|
29
|
Tipping KW, van Oosten-Hawle P, Hewitt EW, Radford SE. Amyloid Fibres: Inert End-Stage Aggregates or Key Players in Disease? Trends Biochem Sci 2015; 40:719-727. [PMID: 26541462 DOI: 10.1016/j.tibs.2015.10.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/30/2015] [Accepted: 10/05/2015] [Indexed: 01/08/2023]
Abstract
The formation of amyloid fibres is a hallmark of amyloid disorders. Nevertheless, the lack of correlation between fibre load and disease as observed, for example, in Alzheimer's disease, means that fibres are considered secondary contributors to the onset of cellular dysfunction. Instead, soluble intermediates of amyloid assembly are often described as the agents of toxicity. Here, we discuss recent experimental discoveries which suggest that amyloid fibres should be considered as disease-relevant species that can mediate a range of pathological processes. These include disruption of biological membranes, secondary nucleation, amyloid aggregate transmission, and the disruption of protein homeostasis (proteostasis). Thus, a greater understanding of amyloid fibre biology could enhance prospects of developing therapeutic interventions against this devastating class of protein-misfolding disorders.
Collapse
Affiliation(s)
- Kevin W Tipping
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, The University of Leeds, Leeds, LS2 9JT, UK
| | - Patricija van Oosten-Hawle
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, The University of Leeds, Leeds, LS2 9JT, UK
| | - Eric W Hewitt
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, The University of Leeds, Leeds, LS2 9JT, UK
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, The University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
30
|
Clavaguera F, Hench J, Goedert M, Tolnay M. Invited review: Prion-like transmission and spreading of tau pathology. Neuropathol Appl Neurobiol 2015; 41:47-58. [PMID: 25399729 DOI: 10.1111/nan.12197] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 11/13/2014] [Indexed: 12/20/2022]
Abstract
Filaments made of hyperphosphorylated tau protein are encountered in a number of neurodegenerative diseases referred to as 'tauopathies'. In the most prevalent tauopathy, Alzheimer's disease, tau pathology progresses in a stereotypical manner with the first lesions appearing in the locus coeruleus and the entorhinal cortex from where they appear to spread to the hippocampus and neocortex. Propagation of tau pathology is also characteristic of argyrophilic grain disease, where the tau lesions appear to spread throughout distinct regions of the limbic system. These findings strongly implicate neurone-to-neurone propagation of tau aggregates. Isoform composition and morphology of tau filaments can differ between tauopathies suggesting the existence of conformationally diverse tau strains. Altogether, this points to prion-like mechanisms in the pathogenesis of tauopathies.
Collapse
Affiliation(s)
- F Clavaguera
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | | | | | | |
Collapse
|
31
|
Teixeira-Castro A, Jalles A, Esteves S, Kang S, da Silva Santos L, Silva-Fernandes A, Neto MF, Brielmann RM, Bessa C, Duarte-Silva S, Miranda A, Oliveira S, Neves-Carvalho A, Bessa J, Summavielle T, Silverman RB, Oliveira P, Morimoto RI, Maciel P. Serotonergic signalling suppresses ataxin 3 aggregation and neurotoxicity in animal models of Machado-Joseph disease. Brain 2015; 138:3221-37. [PMID: 26373603 DOI: 10.1093/brain/awv262] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/13/2015] [Indexed: 11/13/2022] Open
Abstract
Polyglutamine diseases are a class of dominantly inherited neurodegenerative disorders for which there is no effective treatment. Here we provide evidence that activation of serotonergic signalling is beneficial in animal models of Machado-Joseph disease. We identified citalopram, a selective serotonin reuptake inhibitor, in a small molecule screen of FDA-approved drugs that rescued neuronal dysfunction and reduced aggregation using a Caenorhabditis elegans model of mutant ataxin 3-induced neurotoxicity. MOD-5, the C. elegans orthologue of the serotonin transporter and cellular target of citalopram, and the serotonin receptors SER-1 and SER-4 were strong genetic modifiers of ataxin 3 neurotoxicity and necessary for therapeutic efficacy. Moreover, chronic treatment of CMVMJD135 mice with citalopram significantly reduced ataxin 3 neuronal inclusions and astrogliosis, rescued diminished body weight and strikingly ameliorated motor symptoms. These results suggest that small molecule modulation of serotonergic signalling represents a promising therapeutic target for Machado-Joseph disease.
Collapse
Affiliation(s)
- Andreia Teixeira-Castro
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal 2 ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal 3 Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA 4 Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208, USA
| | - Ana Jalles
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal 2 ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sofia Esteves
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal 2 ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Soosung Kang
- 3 Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA 5 Department of Chemistry, Northwestern University, Evanston, Illinois 60208, USA 6 Chemistry of Life Processes Institute and Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, USA
| | - Liliana da Silva Santos
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal 2 ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Anabela Silva-Fernandes
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal 2 ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Mário F Neto
- 3 Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA 4 Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208, USA
| | - Renée M Brielmann
- 3 Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA 4 Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208, USA
| | - Carlos Bessa
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal 2 ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sara Duarte-Silva
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal 2 ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Adriana Miranda
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal 2 ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Stéphanie Oliveira
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal 2 ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Andreia Neves-Carvalho
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal 2 ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João Bessa
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal 2 ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Teresa Summavielle
- 7 IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal
| | - Richard B Silverman
- 3 Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA 5 Department of Chemistry, Northwestern University, Evanston, Illinois 60208, USA 6 Chemistry of Life Processes Institute and Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, USA
| | - Pedro Oliveira
- 8 ICBAS-Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
| | - Richard I Morimoto
- 3 Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA 4 Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208, USA
| | - Patrícia Maciel
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal 2 ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
32
|
Kirstein J, Morito D, Kakihana T, Sugihara M, Minnen A, Hipp MS, Nussbaum-Krammer C, Kasturi P, Hartl FU, Nagata K, Morimoto RI. Proteotoxic stress and ageing triggers the loss of redox homeostasis across cellular compartments. EMBO J 2015; 34:2334-49. [PMID: 26228940 DOI: 10.15252/embj.201591711] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 07/02/2015] [Indexed: 12/17/2022] Open
Abstract
The cellular proteostasis network integrates the protein folding and clearance machineries in multiple sub-cellular compartments of the eukaryotic cell. The endoplasmic reticulum (ER) is the site of synthesis and folding of membrane and secretory proteins. A distinctive feature of the ER is its tightly controlled redox homeostasis necessary for the formation of inter- and intra-molecular disulphide bonds. Employing genetically encoded in vivo sensors reporting on the redox state in an organelle-specific manner, we show in the nematode Caenorhabditis elegans that the redox state of the ER is subject to profound changes during worm lifetime. In young animals, the ER is oxidizing and this shifts towards reducing conditions during ageing, whereas in the cytosol the redox state becomes more oxidizing with age. Likewise, the redox state in the cytosol and the ER change in an opposing manner in response to proteotoxic challenges in C. elegans and in HeLa cells revealing conservation of redox homeostasis. Moreover, we show that organelle redox homeostasis is regulated across tissues within C. elegans providing a new measure for organismal fitness.
Collapse
Affiliation(s)
- Janine Kirstein
- Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin, Berlin, Germany
| | - Daisuke Morito
- Laboratory of Molecular and Cellular Biology, Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto, Japan
| | - Taichi Kakihana
- Laboratory of Molecular and Cellular Biology, Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto, Japan
| | - Munechika Sugihara
- Laboratory of Molecular and Cellular Biology, Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto, Japan
| | - Anita Minnen
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL, USA
| | - Mark S Hipp
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Carmen Nussbaum-Krammer
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL, USA
| | - Prasad Kasturi
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - F Ulrich Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Kazuhiro Nagata
- Laboratory of Molecular and Cellular Biology, Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto, Japan
| | - Richard I Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL, USA
| |
Collapse
|
33
|
Dictyostelium discoideum has a highly Q/N-rich proteome and shows an unusual resilience to protein aggregation. Proc Natl Acad Sci U S A 2015; 112:E2620-9. [PMID: 25941378 DOI: 10.1073/pnas.1504459112] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Many protein-misfolding diseases are caused by proteins carrying prion-like domains. These proteins show sequence similarity to yeast prion proteins, which can interconvert between an intrinsically disordered and an aggregated prion state. The natural presence of prions in yeast has provided important insight into disease mechanisms and cellular proteostasis. However, little is known about prions in other organisms, and it is not yet clear whether the findings in yeast can be generalized. Using bioinformatics tools, we show that Dictyostelium discoideum has the highest content of prion-like proteins of all organisms investigated to date, suggesting that its proteome has a high overall aggregation propensity. To study mechanisms regulating these proteins, we analyze the behavior of several well-characterized prion-like proteins, such as an expanded version of human huntingtin exon 1 (Q103) and the prion domain of the yeast prion protein Sup35 (NM), in D. discoideum. We find that these proteins remain soluble and are innocuous to D. discoideum, in contrast to other organisms, where they form cytotoxic cytosolic aggregates. However, when exposed to conditions that compromise molecular chaperones, these proteins aggregate and become cytotoxic. We show that the disaggregase Hsp101, a molecular chaperone of the Hsp100 family, dissolves heat-induced aggregates and promotes thermotolerance. Furthermore, prion-like proteins accumulate in the nucleus, where they are targeted by the ubiquitin-proteasome system. Our data suggest that D. discoideum has undergone specific adaptations that increase the proteostatic capacity of this organism and allow for an efficient regulation of its prion-like proteome.
Collapse
|
34
|
Abstract
Loss of protein homeostasis (proteostasis) is a common feature of aging and disease that is characterized by the appearance of nonnative protein aggregates in various tissues. Protein aggregation is routinely suppressed by the proteostasis network (PN), a collection of macromolecular machines that operate in diverse ways to maintain proteome integrity across subcellular compartments and between tissues to ensure a healthy life span. Here, we review the composition, function, and organizational properties of the PN in the context of individual cells and entire organisms and discuss the mechanisms by which disruption of the PN, and related stress response pathways, contributes to the initiation and progression of disease. We explore emerging evidence that disease susceptibility arises from early changes in the composition and activity of the PN and propose that a more complete understanding of the temporal and spatial properties of the PN will enhance our ability to develop effective treatments for protein conformational diseases.
Collapse
Affiliation(s)
- Johnathan Labbadia
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208;
| | | |
Collapse
|
35
|
Nussbaum-Krammer CI, Neto MF, Brielmann RM, Pedersen JS, Morimoto RI. Investigating the spreading and toxicity of prion-like proteins using the metazoan model organism C. elegans. J Vis Exp 2015:52321. [PMID: 25591151 DOI: 10.3791/52321] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Prions are unconventional self-propagating proteinaceous particles, devoid of any coding nucleic acid. These proteinaceous seeds serve as templates for the conversion and replication of their benign cellular isoform. Accumulating evidence suggests that many protein aggregates can act as self-propagating templates and corrupt the folding of cognate proteins. Although aggregates can be functional under certain circumstances, this process often leads to the disruption of the cellular protein homeostasis (proteostasis), eventually leading to devastating diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), or transmissible spongiform encephalopathies (TSEs). The exact mechanisms of prion propagation and cell-to-cell spreading of protein aggregates are still subjects of intense investigation. To further this knowledge, recently a new metazoan model in Caenorhabditis elegans, for expression of the prion domain of the cytosolic yeast prion protein Sup35 has been established. This prion model offers several advantages, as it allows direct monitoring of the fluorescently tagged prion domain in living animals and ease of genetic approaches. Described here are methods to study prion-like behavior of protein aggregates and to identify modifiers of prion-induced toxicity using C. elegans.
Collapse
Affiliation(s)
- Carmen I Nussbaum-Krammer
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University;
| | - Mário F Neto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University
| | - Renée M Brielmann
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University
| | - Jesper S Pedersen
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University
| | - Richard I Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University
| |
Collapse
|
36
|
Jenzer C, Simionato E, Legouis R. Tools and methods to analyze autophagy in C. elegans. Methods 2014; 75:162-71. [PMID: 25484340 DOI: 10.1016/j.ymeth.2014.11.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 11/27/2022] Open
Abstract
For a long time, autophagy has been mainly studied in yeast or mammalian cell lines, and assays for analyzing autophagy in these models have been well described. More recently, the involvement of autophagy in various physiological functions has been investigated in multicellular organisms. Modification of autophagy flux is involved in developmental processes, resistance to stress conditions, aging, cell death and multiple pathologies. So, the use of animal models is essential to understand these processes in the context of different cell types and during the whole life. For ten years, the nematode Caenorhabditis elegans has emerged as a powerful model to analyze autophagy in physiological or pathological contexts. In this article, we present some of the established approaches and the emerging tools available to monitor and manipulate autophagy in C. elegans, and discuss their advantages and limitations.
Collapse
Affiliation(s)
- Céline Jenzer
- Centre de Génétique Moléculaire, CNRS UPR3404, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France; Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris Sud, 1 Avenue de la Terrasse, 91198 Gif sur Yvette, France
| | - Elena Simionato
- Centre de Génétique Moléculaire, CNRS UPR3404, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France; Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris Sud, 1 Avenue de la Terrasse, 91198 Gif sur Yvette, France
| | - Renaud Legouis
- Centre de Génétique Moléculaire, CNRS UPR3404, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France; Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris Sud, 1 Avenue de la Terrasse, 91198 Gif sur Yvette, France.
| |
Collapse
|
37
|
Roth DM, Hutt DM, Tong J, Bouchecareilh M, Wang N, Seeley T, Dekkers JF, Beekman JM, Garza D, Drew L, Masliah E, Morimoto RI, Balch WE. Modulation of the maladaptive stress response to manage diseases of protein folding. PLoS Biol 2014; 12:e1001998. [PMID: 25406061 PMCID: PMC4236052 DOI: 10.1371/journal.pbio.1001998] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 10/07/2014] [Indexed: 12/31/2022] Open
Abstract
Diseases of protein folding arise because of the inability of an altered peptide sequence to properly engage protein homeostasis components that direct protein folding and function. To identify global principles of misfolding disease pathology we examined the impact of the local folding environment in alpha-1-antitrypsin deficiency (AATD), Niemann-Pick type C1 disease (NPC1), Alzheimer's disease (AD), and cystic fibrosis (CF). Using distinct models, including patient-derived cell lines and primary epithelium, mouse brain tissue, and Caenorhabditis elegans, we found that chronic expression of misfolded proteins not only triggers the sustained activation of the heat shock response (HSR) pathway, but that this sustained activation is maladaptive. In diseased cells, maladaptation alters protein structure-function relationships, impacts protein folding in the cytosol, and further exacerbates the disease state. We show that down-regulation of this maladaptive stress response (MSR), through silencing of HSF1, the master regulator of the HSR, restores cellular protein folding and improves the disease phenotype. We propose that restoration of a more physiological proteostatic environment will strongly impact the management and progression of loss-of-function and gain-of-toxic-function phenotypes common in human disease.
Collapse
Affiliation(s)
- Daniela Martino Roth
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Darren M. Hutt
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jiansong Tong
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Marion Bouchecareilh
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Ning Wang
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois, United States of America
| | - Theo Seeley
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Johanna F. Dekkers
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
- Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
| | - Jeffrey M. Beekman
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
- Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
| | - Dan Garza
- Proteostasis Therapeutics Inc., Cambridge, Massachusetts, United States of America
| | - Lawrence Drew
- Proteostasis Therapeutics Inc., Cambridge, Massachusetts, United States of America
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, California, United States of America
| | - Richard I. Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois, United States of America
| | - William E. Balch
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
- The Institute for Childhood and Neglected Diseases, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
38
|
Nussbaum-Krammer CI, Morimoto RI. Caenorhabditis elegans as a model system for studying non-cell-autonomous mechanisms in protein-misfolding diseases. Dis Model Mech 2014; 7:31-9. [PMID: 24396152 PMCID: PMC3882046 DOI: 10.1242/dmm.013011] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Caenorhabditis elegans has a number of distinct advantages that are useful for understanding the basis for cellular and organismal dysfunction underlying age-associated diseases of protein misfolding. Although protein aggregation, a key feature of human neurodegenerative diseases, has been typically explored in vivo at the single-cell level using cells in culture, there is now increasing evidence that proteotoxicity has a non-cell-autonomous component and is communicated between cells and tissues in a multicellular organism. These discoveries have opened up new avenues for the use of C. elegans as an ideal animal model system to study non-cell-autonomous proteotoxicity, prion-like propagation of aggregation-prone proteins, and the organismal regulation of stress responses and proteostasis. This Review focuses on recent evidence that C. elegans has mechanisms to transmit certain classes of toxic proteins between tissues and a complex stress response that integrates and coordinates signals from single cells and tissues across the organism. These findings emphasize the potential of C. elegans to provide insights into non-cell-autonomous proteotoxic mechanisms underlying age-related protein-misfolding diseases.
Collapse
Affiliation(s)
- Carmen I Nussbaum-Krammer
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, 2205 Tech Drive, Evanston, IL 60208, USA
| | | |
Collapse
|
39
|
van Oosten-Hawle P, Morimoto RI. Transcellular chaperone signaling: an organismal strategy for integrated cell stress responses. ACTA ACUST UNITED AC 2014; 217:129-36. [PMID: 24353212 DOI: 10.1242/jeb.091249] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ability of each cell within a metazoan to adapt to and survive environmental and physiological stress requires cellular stress-response mechanisms, such as the heat shock response (HSR). Recent advances reveal that cellular proteostasis and stress responses in metazoans are regulated by multiple layers of intercellular communication. This ensures that an imbalance of proteostasis that occurs within any single tissue 'at risk' is protected by a compensatory activation of a stress response in adjacent tissues that confers a community protective response. While each cell expresses the machinery for heat shock (HS) gene expression, the HSR is regulated cell non-autonomously in multicellular organisms, by neuronal signaling to the somatic tissues, and by transcellular chaperone signaling between somatic tissues and from somatic tissues to neurons. These cell non-autonomous processes ensure that the organismal HSR is orchestrated across multiple tissues and that transmission of stress signals between tissues can also override the neuronal control to reset cell- and tissue-specific proteostasis. Here, we discuss emerging concepts and insights into the complex cell non-autonomous mechanisms that control stress responses in metazoans and highlight the importance of intercellular communication for proteostasis maintenance in multicellular organisms.
Collapse
Affiliation(s)
- Patricija van Oosten-Hawle
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208, USA
| | | |
Collapse
|
40
|
Therrien M, Parker JA. Worming forward: amyotrophic lateral sclerosis toxicity mechanisms and genetic interactions in Caenorhabditis elegans. Front Genet 2014; 5:85. [PMID: 24860590 PMCID: PMC4029022 DOI: 10.3389/fgene.2014.00085] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/30/2014] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases share pathogenic mechanisms at the cellular level including protein misfolding, excitotoxicity and altered RNA homeostasis among others. Recent advances have shown that the genetic causes underlying these pathologies overlap, hinting at the existence of a genetic network for neurodegeneration. This is perhaps best illustrated by the recent discoveries of causative mutations for amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration (FTD). Once thought to be distinct entities, it is now recognized that these diseases exist along a genetic spectrum. With this wealth of discoveries comes the need to develop new genetic models of ALS and FTD to investigate not only pathogenic mechanisms linked to causative mutations, but to uncover potential genetic interactions that may point to new therapeutic targets. Given the conservation of many disease genes across evolution, Caenorhabditis elegans is an ideal system to investigate genetic interactions amongst these genes. Here we review the use of C. elegans to model ALS and investigate a putative genetic network for ALS/FTD that may extend to other neurological disorders.
Collapse
Affiliation(s)
- Martine Therrien
- Départment de Pathologie et Biologie Cellulaire, CRCHUM-Centre Hospitalier de l'Université de Montréal, Université de Montréal Montréal, QC, Canada
| | - J Alex Parker
- Départment de Pathologie et Biologie Cellulaire, Départment de Neurosciences, CRCHUM-Centre Hospitalier de l'Université de Montréal, Université de Montréal Montréal, QC, Canada
| |
Collapse
|
41
|
Abstract
Multiple prion elements, which are transmitted as heritable protein conformations and often linked to distinct phenotypes, have been identified in the budding yeast, Saccharomyces cerevisiae. It has been shown that overproduction of a prion protein Swi1 can promote the de novo conversion of another yeast prion [PSI(+)] when Sup35 is co-overproduced. However, the mechanism underlying this Pin(+) ([PSI(+)] inducible) activity is not clear. Moreover, how the Swi1 prion ([SWI(+)]) interacts with other yeast prions is unknown. Here, we demonstrate that the Pin(+) activity associated with Swi1 overproduction is independent of Rnq1 expression or [PIN(+)] conversion. We also show that [SWI(+)] enhances the appearance of [PSI(+)] and [PIN(+)]. However, [SWI(+)] significantly compromises the Pin(+) activity of [PIN(+)] when they coexist. We further demonstrate that a single yeast cell can harbor three prions, [PSI(+)], [PIN(+)], and [SWI(+)], simultaneously. However, under this condition, [SWI(+)] is significantly destabilized. While the propensity to aggregate underlies prionogenesis, Swi1 and Rnq1 aggregates resulting from overproduction are usually nonheritable. Conversely, prion protein aggregates formed in nonoverexpressing conditions or induced by preexisting prion(s) are more prionogenic. For [PSI(+)] and [PIN(+)] de novo formation, heterologous "facilitators," such as preexisting [SWI(+)] aggregates, colocalize only with the newly formed ring-/rod-shaped Sup35 or Rnq1 aggregates, but not with the dot-shaped mature prion aggregates. Their colocalization frequency is coordinated with their prion inducibility, indicating that prion-prion interactions mainly occur at the early initiation stage. Our results provide supportive evidence for the cross-seeding model of prionogenesis and highlight a complex interaction network among prions in yeast.
Collapse
|
42
|
Abstract
Prions are self-templating protein aggregates that were originally identified as the causative agent of prion diseases in mammals, but have since been discovered in other kingdoms. Mammalian prions represent a unique class of infectious agents that are composed of misfolded prion protein. Prion proteins usually exist as soluble proteins but can refold and assemble into highly ordered, self-propagating prion polymers. The prion concept is also applicable to a growing number of non-Mendelian elements of inheritance in lower eukaryotes. While prions identified in mammals are clearly pathogens, prions in lower eukaryotes can be either detrimental or beneficial to the host. Prion phenotypes in fungi are transmitted vertically from mother to daughter cells during cell division and horizontally during mating or abortive mating, but extracellular phases have not been reported. Recent findings now demonstrate that in a mammalian cell environment, protein aggregates derived from yeast prion domains exhibit a prion life cycle similar to mammalian prions propagated ex vivo. This life cycle includes a soluble state of the protein, an induction phase by exogenous prion fibrils, stable replication of prion entities, vertical transmission to progeny and natural horizontal transmission to neighboring cells. Our data reveal that mammalian cells contain all co-factors required for cytosolic prion propagation and dissemination. This has important implications for understanding prion-like properties of disease-related protein aggregates. In light of the growing number of identified functional amyloids, cell-to-cell propagation of cytosolic protein conformers might not only be relevant for the spreading of disease-associated proteins, but might also be of more general relevance under non-disease conditions.
Collapse
Affiliation(s)
- Julia Hofmann
- German Center for Neurodegenerative Diseases (DZNE e.V.); Bonn, Germany
| | - Ina Vorberg
- German Center for Neurodegenerative Diseases (DZNE e.V.); Bonn, Germany; Rheinische Friedrich-Wilhelms-Universität; Bonn, Germany
| |
Collapse
|
43
|
Shorter J, Taylor JP. Disease mutations in the prion-like domains of hnRNPA1 and hnRNPA2/B1 introduce potent steric zippers that drive excess RNP granule assembly. Rare Dis 2013; 1:e25200. [PMID: 25002999 PMCID: PMC3932941 DOI: 10.4161/rdis.25200] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 05/14/2013] [Accepted: 05/28/2013] [Indexed: 12/12/2022] Open
Abstract
Approximately 1% of human proteins harbor a prion-like domain (PrLD) of similar low complexity sequence and amino acid composition to domains that drive prionogenesis of yeast proteins like Sup35. PrLDs are over-represented in human RNA-binding proteins and mediate phase transitions underpinning RNP granule assembly. This modality renders PrLDs prone to misfold into conformers that accrue in pathological inclusions that characterize various fatal neurodegenerative diseases. For example, TDP-43 and FUS form cytoplasmic inclusions in amyotrophic lateral sclerosis (ALS) and mutations in TDP-43 and FUS can cause ALS. Here, we review our recent discovery of discrete missense mutations that alter a conserved gatekeeper aspartate residue in the PrLDs of hnRNPA2/B1 and hnRNPA1 and cause multisystem proteinopathy and ALS. The missense mutations generate potent steric zippers in the PrLDs, which enhance a natural propensity to form self-templating fibrils, promote recruitment to stress granules and drive cytoplasmic inclusion formation. PrLDs occur in ~250 human proteins and could contribute directly to the etiology of various degenerative disorders.
Collapse
Affiliation(s)
- James Shorter
- Department of Biochemistry and Biophysics; Perelman School of Medicine at The University of Pennsylvania; Philadelphia, PA USA
| | - J Paul Taylor
- Department of Developmental Neurobiology; St Jude Children's Research Hospital; Memphis, TN USA
| |
Collapse
|