1
|
Torres AK, Mira RG, Pinto C, Inestrosa NC. Studying the mechanisms of neurodegeneration: C. elegans advantages and opportunities. Front Cell Neurosci 2025; 19:1559151. [PMID: 40207239 PMCID: PMC11979225 DOI: 10.3389/fncel.2025.1559151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Caenorhabditis elegans has been widely used as a model organism in neurodevelopment for several decades due to its simplicity, rapid growth, short life cycle, transparency, and rather simple genetics. It has been useful in modeling neurodegenerative diseases by the heterologous expression of the major proteins that form neurodegenerative-linked aggregates such as amyloid-β peptide, tau protein, and α-synuclein, among others. Furthermore, chemical treatments as well as the existence of several interference RNA libraries, transgenic worm lines, and the possibility of generating new transgenic strains create a magnificent range of possible tools to study the signaling pathways that could confer protection against protein aggregates or, on the contrary, are playing a detrimental role. In this review, we summarize the different C. elegans models of neurodegenerative diseases with a focus on Alzheimer's and Parkinson's diseases and how genetic tools could be used to dissect the signaling pathways involved in their pathogenesis mentioning several examples. Finally, we discuss the use of pharmacological agents in C. elegans models that could help to study these disease-associated signaling pathways and the powerful combinations of experimental designs with genetic tools. This review highlights the advantages of C. elegans as a valuable intermediary between in vitro and mammalian in vivo models in the development of potential new therapies.
Collapse
Affiliation(s)
- Angie K. Torres
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo G. Mira
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | - Cristina Pinto
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | - Nibaldo C. Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
2
|
Ganguly U, Carroll T, Nehrke K, Johnson GVW. Mitochondrial Quality Control in Alzheimer's Disease: Insights from Caenorhabditis elegans Models. Antioxidants (Basel) 2024; 13:1343. [PMID: 39594485 PMCID: PMC11590956 DOI: 10.3390/antiox13111343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder that is classically defined by the extracellular deposition of senile plaques rich in amyloid-beta (Aβ) protein and the intracellular accumulation of neurofibrillary tangles (NFTs) that are rich in aberrantly modified tau protein. In addition to aggregative and proteostatic abnormalities, neurons affected by AD also frequently possess dysfunctional mitochondria and disrupted mitochondrial maintenance, such as the inability to eliminate damaged mitochondria via mitophagy. Decades have been spent interrogating the etiopathogenesis of AD, and contributions from model organism research have aided in developing a more fundamental understanding of molecular dysfunction caused by Aβ and toxic tau aggregates. The soil nematode C. elegans is a genetic model organism that has been widely used for interrogating neurodegenerative mechanisms including AD. In this review, we discuss the advantages and limitations of the many C. elegans AD models, with a special focus and discussion on how mitochondrial quality control pathways (namely mitophagy) may contribute to AD development. We also summarize evidence on how targeting mitophagy has been therapeutically beneficial in AD. Lastly, we delineate possible mechanisms that can work alone or in concert to ultimately lead to mitophagy impairment in neurons and may contribute to AD etiopathology.
Collapse
Affiliation(s)
- Upasana Ganguly
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Trae Carroll
- Department of Pathology, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Keith Nehrke
- Department of Medicine, Nephrology Division, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Gail V. W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| |
Collapse
|
3
|
Rani N, Alam MM, Jamal A, Bin Ghaffar U, Parvez S. Caenorhabditis elegans: A transgenic model for studying age-associated neurodegenerative diseases. Ageing Res Rev 2023; 91:102036. [PMID: 37598759 DOI: 10.1016/j.arr.2023.102036] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
Neurodegenerative diseases (NDs) are a heterogeneous group of aging-associated ailments characterized by interrupting cellular proteostasic machinery and the misfolding of distinct proteins to form toxic aggregates in neurons. Neurodegenerative diseases, which include Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and others, are becoming an increasing threat to human health worldwide. The degeneration and death of certain specific groups of neurons are the hallmarks of these diseases. Over the past decades, Caenorhabditis eleganshas beenwidely used as a transgenic model to investigate biological processes related to health and disease. The nematode Caenorhabditis elegans (C. elegans) has developed as a powerful tool for studying disease mechanisms due to its ease of genetic handling and instant cultivation while providing a whole-animal system amendable to several molecular and biochemical techniques. In this review, we elucidate the potential of C. elegans as a versatile platform for systematic dissection of the molecular basis of human disease, focusing on neurodegenerative disorders, and may help better our understanding of the disease mechanisms and search for new therapeutics for these devastating diseases.
Collapse
Affiliation(s)
- Nisha Rani
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Mumtaz Alam
- Drug Design and Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Azfar Jamal
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Usama Bin Ghaffar
- Department of Basic Science, College of Medicine, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
4
|
Witten G, DeMott E, Huang G, Zelasko F, de Jesus B, Mulchand C, Schuck L, Pullman S, Perez A, Mahableshwarkar P, Wu Z, Cardona EA, Pierce JT, Dickinson DJ, Doonan R. mScarlet and split fluorophore mScarlet resources for plasmid-based CRISPR/Cas9 knock-in in C. elegans. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000871. [PMID: 37396790 PMCID: PMC10308244 DOI: 10.17912/micropub.biology.000871] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 05/30/2023] [Accepted: 06/14/2023] [Indexed: 07/04/2023]
Abstract
Fluorescent proteins allow the expression of a gene and the behavior of its protein product to be observed in living animals. The ability to create endogenous fluorescent protein tags via CRISPR genome engineering has revolutionized the authenticity of this expression, and mScarlet is currently our first-choice red fluorescent protein (RFP) for visualizing gene expression in vivo . Here, we have cloned versions of mScarlet and split fluorophore mScarlet previously optimized for C. elegans into the SEC-based system of plasmids for CRISPR/Cas9 knock-in. Ideally, an endogenous tag will be easily visible while not interfering with the normal expression and function of the targeted protein. For low molecular weight proteins that are a fraction of the size of a fluorescent protein tag (e.g. GFP or mCherry) and/or proteins known to be non-functional when tagged in this way, split fluorophore tagging could be an alternative. Here, we used CRISPR/Cas9 knock-in to tag three such proteins with split-fluorophore wrmScarlet: HIS-72, EGL-1, and PTL-1. Although we find that split fluorophore tagging does not disrupt the function of any of these proteins, we were unfortunately unable to observe the expression of most of these tags with epifluorescence, suggesting that split fluorophore tags are often very limited as endogenous reporters. Nevertheless, our plasmid toolkit provides a new resource that enables straightforward knock-in of either mScarlet or split mScarlet in C. elegans.
Collapse
Affiliation(s)
- Gillian Witten
- Glow Worms Stream, Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, Texas, USA
| | - Ella DeMott
- Glow Worms Stream, Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, Texas, USA
| | - George Huang
- Glow Worms Stream, Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, Texas, USA
| | - Francis Zelasko
- Glow Worms Stream, Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, Texas, USA
| | - Bailey de Jesus
- Glow Worms Stream, Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, Texas, USA
| | - Chandi Mulchand
- Glow Worms Stream, Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, Texas, USA
| | - Liam Schuck
- Glow Worms Stream, Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, Texas, USA
| | - Stephen Pullman
- Glow Worms Stream, Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, Texas, USA
| | - Amelie Perez
- Glow Worms Stream, Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, Texas, USA
| | - Priya Mahableshwarkar
- Glow Worms Stream, Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, Texas, USA
| | - Zheng Wu
- Department of Neuroscience, The University of Texas at Austin, Austin, Texas, USA
| | - Eric Andrew Cardona
- Department of Neuroscience, The University of Texas at Austin, Austin, Texas, USA
| | - Jonathan T Pierce
- Department of Neuroscience, The University of Texas at Austin, Austin, Texas, USA
| | - Daniel J Dickinson
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Ryan Doonan
- Glow Worms Stream, Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
5
|
Aquino Nunez W, Combs B, Gamblin TC, Ackley BD. Age-dependent accumulation of tau aggregation in Caenorhabditis elegans. FRONTIERS IN AGING 2022; 3:928574. [PMID: 36062211 PMCID: PMC9437221 DOI: 10.3389/fragi.2022.928574] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
Aging is the primary risk factor for Alzheimer's disease (AD) and related disorders (ADRDs). Tau aggregation is a hallmark of AD and other tauopathies. Even in normal aging, tau aggregation is found in brains, but in disease states, significantly more aggregated tau is present in brain regions demonstrating synaptic degeneration and neuronal loss. It is unclear how tau aggregation and aging interact to give rise to the phenotypes observed in disease states. Most AD/ADRD animal models have focused on late stages, after significant tau aggregation has occurred. There are fewer where we can observe the early aggregation events and progression during aging. In an attempt to address this gap, we created C. elegans models expressing a GFP-tagged version of the human tau protein. Here we examined how tau-gfp behaved during aging, comparing wild-type tau (hTau40), a disease-associated mutation (P301S), and an aggregation-prone variant (3PO). We measured age-dependent changes in GFP intensity and correlated those changes to normal aging in the nematode. We found differences in tau stability and accumulation depending on the tau variant expressed. hTau40GFP and P301SGFP were localized to axons and cell bodies, while 3POGFP was more concentrated within cell bodies. Expression of 3POGFP resulted in decreased lifespan and variations in locomotor rate, consistent with a pathological effect. Finally, we found that the human tau interacted genetically with the C. elegans ortholog of human tau, ptl-1, where the loss of ptl-1 significantly accelerated the time to death in animals expressing 3PO.
Collapse
Affiliation(s)
- Wendy Aquino Nunez
- Laboratory Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, United States
| | - Benjamin Combs
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - T. Chris Gamblin
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas San Antonio, San Antonio, TX, United States
| | - Brian D. Ackley
- Laboratory Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, United States
| |
Collapse
|
6
|
Navarro-Hortal MD, Romero-Márquez JM, Osta S, Jiménez-Trigo V, Muñoz-Ollero P, Varela-López A. Natural Bioactive Products and Alzheimer’s Disease Pathology: Lessons from Caenorhabditis elegans Transgenic Models. Diseases 2022; 10:diseases10020028. [PMID: 35645249 PMCID: PMC9149938 DOI: 10.3390/diseases10020028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is an age-dependent, progressive disorder affecting millions of people. Currently, the therapeutics for AD only treat the symptoms. Although they have been used to discover new products of interest for this disease, mammalian models used to investigate the molecular determinants of this disease are often prohibitively expensive, time-consuming and very complex. On the other hand, cell cultures lack the organism complexity involved in AD. Given the highly conserved neurological pathways between mammals and invertebrates, Caenorhabditis elegans has emerged as a powerful tool for the investigation of the pathophysiology of human AD. Numerous models of both Tau- and Aβ-induced toxicity, the two prime components observed to correlate with AD pathology and the ease of performing RNA interference for any gene in the C. elegans genome, allow for the identification of multiple therapeutic targets. The effects of many natural products in main AD hallmarks using these models suggest promising health-promoting effects. However, the way in which they exert such effects is not entirely clear. One of the reasons is that various possible therapeutic targets have not been evaluated in many studies. The present review aims to explore shared therapeutical targets and the potential of each of them for AD treatment or prevention.
Collapse
|
7
|
Caenorhabditis elegans Models to Investigate the Mechanisms Underlying Tau Toxicity in Tauopathies. Brain Sci 2020; 10:brainsci10110838. [PMID: 33187241 PMCID: PMC7697895 DOI: 10.3390/brainsci10110838] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
The understanding of the genetic, biochemical, and structural determinants underlying tau aggregation is pivotal in the elucidation of the pathogenic process driving tauopathies and the design of effective therapies. Relevant information on the molecular basis of human neurodegeneration in vivo can be obtained using the nematode Caenorhabditis elegans (C. elegans). To this end, two main approaches can be applied: the overexpression of genes/proteins leading to neuronal dysfunction and death, and studies in which proteins prone to misfolding are exogenously administered to induce a neurotoxic phenotype. Thanks to the easy generation of transgenic strains expressing human disease genes, C. elegans allows the identification of genes and/or proteins specifically associated with pathology and the specific disruptions of cellular processes involved in disease. Several transgenic strains expressing human wild-type or mutated tau have been developed and offer significant information concerning whether transgene expression regulates protein production and aggregation in soluble or insoluble form, onset of the disease, and the degenerative process. C. elegans is able to specifically react to the toxic assemblies of tau, thus developing a neurodegenerative phenotype that, even when exogenously administered, opens up the use of this assay to investigate in vivo the relationship between the tau sequence, its folding, and its proteotoxicity. These approaches can be employed to screen drugs and small molecules that can interact with the biogenesis and dynamics of formation of tau aggregates and to analyze their interactions with other cellular proteins.
Collapse
|
8
|
Liang JJH, McKinnon IA, Rankin CH. The contribution of C. elegans neurogenetics to understanding neurodegenerative diseases. J Neurogenet 2020; 34:527-548. [DOI: 10.1080/01677063.2020.1803302] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Joseph J. H. Liang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Issa A. McKinnon
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Catharine H. Rankin
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Psychology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
9
|
Gamir-Morralla A, Sacristán S, Medina M, Iglesias T. Effects of Thioflavin T and GSK-3 Inhibition on Lifespan and Motility in a Caenorhabditis elegans Model of Tauopathy. J Alzheimers Dis Rep 2019; 3:47-57. [PMID: 30842997 PMCID: PMC6400111 DOI: 10.3233/adr-180087] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The nematode Caenorhabditis elegans (C. elegans) is a powerful model organism to study lifespan and aging, protein aggregation, and neurodegeneration, as well as to carry out drug screenings. The C. elegans strain aex-3/T337 expresses human pathogenic V337M mutant tau under a pan-neuronal promoter and presents uncoordinated locomotion, accumulation of phosphorylated insoluble tau, and shortened lifespan. Herein we have used this strain to assay two compounds that could affect tau aggregation and/or phosphorylation, and looked for phenotypic changes in their lifespan and motility. The first compound is Thioflavin T (ThT), a member of the tetracycline family with protein antiaggregant properties, yet to be tested in a tauopathy model. The second is a novel small molecule, NP103, a highly selective inhibitor of glycogen synthase kinase-3 (GSK-3), the main kinase contributing to pathogenic tau hyperphosphorylation. Importantly, we find that ThT extends lifespan of aex-3/T337 worms as it does with control N2 animals, showing both strains similar locomotion features under this treatment. By contrast, NP103 improves the paralysis phenotype of aex-3/T337 mutants but not their lifespan. Our results show that both treatments present beneficial effects for this model of tauopathy and encourage pursuing further investigations on their therapeutic potential for AD and other tauopathies.
Collapse
Affiliation(s)
- Andrea Gamir-Morralla
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Sacristán
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Miguel Medina
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
10
|
Experimental Models of Tauopathy - From Mechanisms to Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1184:381-391. [PMID: 32096051 DOI: 10.1007/978-981-32-9358-8_28] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Animal models have been instrumental in reproducing key aspects of human tauopathy. In pursuing these efforts, the mouse continues to have a prominent role. In this chapter, we focus on models that overexpress wild-type or mutant forms of tau, the latter being based on mutations found in familial cases of frontotemporal dementia. We review some of these models in more detail and discuss what they have revealed about the underlying pathomechanisms, as well as highlighting new developments that exploit gene editing tools such as TALEN and CRISPR. Interestingly, when investigating the role of tau in impairing cellular functions, common themes emerge. Because tau is a scaffolding protein that aggregates in the somatodendritic domain under pathological conditions, it traps proteins such as parkin and JIP1, preventing them from executing their normal function in mitophagy and axonal transport, respectively. Another aspect is the emerging role of tau in the translational machinery and the finding that the somatodendritic accumulation of tau in Alzheimer's disease may in part be due to the induction of the de novo synthesis of tau by amyloid-β via the Fyn/ERK/S6 pathway. We further discuss treatment strategies such as tau-based vaccinations and therapeutic ultrasound and conclude by discussing whether there is a future for animal models of tauopathies.
Collapse
|
11
|
V363I and V363A mutated tau affect aggregation and neuronal dysfunction differently in C. elegans. Neurobiol Dis 2018; 117:226-234. [PMID: 29936232 DOI: 10.1016/j.nbd.2018.06.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 11/24/2022] Open
Abstract
Mutations in the microtubule-associated protein tau (MAPT) gene have been linked to a heterogeneous group of progressive neurodegenerative disorders commonly called tauopathies. From patients with frontotemporal lobar degeneration with distinct atypical clinical phenotypes, we recently identified two new mutations on the same codon, in position 363 of the MAPT gene, which resulted in the production of Val-to-Ala (tauV363A) or Val-to-Ile (tauV363I) mutated tau. These substitutions specifically affected microtubule polymerization and propensity of tau to aggregate in vitro suggesting that single amino acid modification may dictate the fate of the neuropathology. To clarify whether tauV363A and tauV363I affect protein misfolding differently in vivo driving certain phenotypes, we generated new transgenic C. elegans strains. Human 2N4R tau carrying the mutation was expressed in all the neurons of worms. The behavioral defects, misfolding and proteotoxicity caused by the tauV363A and tauV363I mutated proteins were compared to that induced by the expression of wild-type tau (tauwt). Pan-neuronal expression of human 2N4R tauWT in worms resulted in a neuromuscular defect with characteristics of a neurodegenerative phenotype. This defect was worsened by the expression of mutated proteins which drive distinct neuronal dysfunctions and synaptic impairments involving, in transgenic worms expressing tauV363A (V363A) also a pharyngeal defect never linked before to other mutations. The two mutations differently affected the tau phosphorylation and misfolding propensities: tauV363I was highly phosphorylated on epitopes corresponding to Thr231 and Ser202/Thr205, and accumulated as insoluble tau assemblies whereas tauV363A showed a greater propensity to form soluble oligomeric assemblies. These findings uphold the role of a single amino acid substitution in specifically affecting the ability of tau to form soluble and insoluble assemblies, opening up new perspectives in the pathogenic mechanism underlying tauopathies.
Collapse
|
12
|
Choudhary B, Mandelkow E, Mandelkow EM, Pir GJ. Glutamatergic nervous system degeneration in a C. elegans Tau A152T tauopathy model involves pathways of excitotoxicity and Ca 2+ dysregulation. Neurobiol Dis 2018; 117:189-202. [PMID: 29894752 DOI: 10.1016/j.nbd.2018.06.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/28/2018] [Accepted: 06/07/2018] [Indexed: 10/14/2022] Open
Abstract
Mutations in the gene encoding Tau (MAPT-microtubule-associated protein tau) cause a group of neurodegenerative diseases called tauopathies. A recently identified Tau variant, p.A152T, has been reported as a risk factor for frontotemporal dementia-related disorders and Alzheimer disease. However, the mechanism for the pathologies still remain poorly understood. Transgenic Caenorhabditis elegans expressing mutant 2N4R-TauA152T (TauAT) panneuronally show locomotor defects, neurodegeneration and accelerated aging. Here we report that, in TauAT animals, the glutamatergic nervous system is at a high risk of progressive neuronal loss. We present genetic data that this loss occurs predominantly through necrosis. The neuronal loss is caused by several determinants, such as altered adenylyl cyclase (type AC9) pathway, prevalence of excitotoxicity-like conditions, aging-related factors and finally dyshomeostasis of intracellular calcium (Ca2+). The study provides novel insights into the mechanisms involved in selective loss of glutamatergic neurons in a TauAT tauopathy model which could point to new therapeutic targets.
Collapse
Affiliation(s)
- Bikash Choudhary
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud St. 27, 53127 Bonn, Germany; Max-Planck-Institute for Metabolism Research, Hamburg Outstation, c/o DESY, Notkestrsse 85, 22607 Hamburg, Germany.
| | - Eckhard Mandelkow
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud St. 27, 53127 Bonn, Germany; Caesar Research Center, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany; Max-Planck-Institute for Metabolism Research, Hamburg Outstation, c/o DESY, Notkestrsse 85, 22607 Hamburg, Germany.
| | - Eva-Maria Mandelkow
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud St. 27, 53127 Bonn, Germany; Caesar Research Center, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany; Max-Planck-Institute for Metabolism Research, Hamburg Outstation, c/o DESY, Notkestrsse 85, 22607 Hamburg, Germany.
| | - Ghulam Jeelani Pir
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud St. 27, 53127 Bonn, Germany; Max-Planck-Institute for Metabolism Research, Hamburg Outstation, c/o DESY, Notkestrsse 85, 22607 Hamburg, Germany.
| |
Collapse
|
13
|
Shen P, Yue Y, Zheng J, Park Y. Caenorhabditis elegans: A Convenient In Vivo Model for Assessing the Impact of Food Bioactive Compounds on Obesity, Aging, and Alzheimer's Disease. Annu Rev Food Sci Technol 2018; 9:1-22. [DOI: 10.1146/annurev-food-030117-012709] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Peiyi Shen
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Yiren Yue
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | | | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA
| |
Collapse
|
14
|
Pir GJ, Choudhary B, Mandelkow E. Caenorhabditis elegans models of tauopathy. FASEB J 2017; 31:5137-5148. [PMID: 29191965 DOI: 10.1096/fj.201701007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 12/14/2022]
Abstract
One of the hallmarks of the tauopathies, which include the neurodegenerative disorders, such as Alzheimer disease (AD), corticobasal degeneration, frontotemporal dementia, and progressive supranuclear palsy (PSP), is the abnormal accumulation of post-translationally modified, insoluble tau. The result is a loss of neurons, decreased mental function, and complete dependence of patients on others. Aggregation of tau, which under physiologic conditions is a highly soluble protein, is thought to be central to the pathogenesis of these diseases. Indeed one of the strongest lines of evidence is the MAPT gene polymorphisms that lead to the familial forms of tauopathy. Extensive research in animal models over the years has contributed some of the most important findings regarding the pathogenesis of these diseases. Despite this, the precise molecular mechanisms that lead to abnormal tau folding, accumulation, and spreading remain unknown. Owing to the fact that most of the biochemical pathways are conserved, Caenorhabditis elegans provides an alternative approach to identify cellular mechanisms and druggable genes that operate in such disorders. Many human genes implicated in neurodegenerative diseases have counterparts in C. elegans, making it an excellent model in which to study their pathogenesis. In this article, we review some of the important findings gained from C. elegans tauopathy models.-Pir, G. J., Choudhary, B., Mandelkow, E. Caenorhabditiselegans models of tauopathy.
Collapse
Affiliation(s)
- Ghulam Jeelani Pir
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; .,Max-Planck-Institute for Metabolism Research-Cologne, Hamburg, Germany
| | - Bikash Choudhary
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Max-Planck-Institute for Metabolism Research-Cologne, Hamburg, Germany
| | - Eckhard Mandelkow
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Max-Planck-Institute for Metabolism Research-Cologne, Hamburg, Germany.,Caesar Research Center, Bonn, Germany
| |
Collapse
|
15
|
Microtubule Organization Determines Axonal Transport Dynamics. Neuron 2017; 92:449-460. [PMID: 27764672 DOI: 10.1016/j.neuron.2016.09.036] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 08/02/2016] [Accepted: 09/15/2016] [Indexed: 12/31/2022]
Abstract
Axonal microtubule (MT) arrays are the major cytoskeleton substrate for cargo transport. How MT organization, i.e., polymer length, number, and minus-end spacing, is regulated and how it impinges on axonal transport are unclear. We describe a method for analyzing neuronal MT organization using light microscopy. This method circumvents the need for electron microscopy reconstructions and is compatible with live imaging of cargo transport and MT dynamics. Examination of a C. elegans motor neuron revealed how age, MT-associated proteins, and signaling pathways control MT length, minus-end spacing, and coverage. In turn, MT organization determines axonal transport progression: cargoes pause at polymer termini, suggesting that switching MT tracks is rate limiting for efficient transport. Cargo run length is set by MT length, and higher MT coverage correlates with shorter pauses. These results uncover the principles and mechanisms of neuronal MT organization and its regulation of axonal cargo transport.
Collapse
|
16
|
Farina F, Lambert E, Commeau L, Lejeune FX, Roudier N, Fonte C, Parker JA, Boddaert J, Verny M, Baulieu EE, Neri C. The stress response factor daf-16/FOXO is required for multiple compound families to prolong the function of neurons with Huntington's disease. Sci Rep 2017. [PMID: 28638078 PMCID: PMC5479833 DOI: 10.1038/s41598-017-04256-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Helping neurons to compensate for proteotoxic stress and maintain function over time (neuronal compensation) has therapeutic potential in aging and neurodegenerative disease. The stress response factor FOXO3 is neuroprotective in models of Huntington’s disease (HD), Parkinson’s disease and motor-neuron diseases. Neuroprotective compounds acting in a FOXO-dependent manner could thus constitute bona fide drugs for promoting neuronal compensation. However, whether FOXO-dependent neuroprotection is a common feature of several compound families remains unknown. Using drug screening in C. elegans nematodes with neuronal expression of human exon-1 huntingtin (128Q), we found that 3ß-Methoxy-Pregnenolone (MAP4343), 17ß-oestradiol (17ßE2) and 12 flavonoids including isoquercitrin promote neuronal function in 128Q nematodes. MAP4343, 17ßE2 and isoquercitrin also promote stress resistance in mutant Htt striatal cells derived from knock-in HD mice. Interestingly, daf-16/FOXO is required for MAP4343, 17ßE2 and isoquercitrin to sustain neuronal function in 128Q nematodes. This similarly applies to the GSK3 inhibitor lithium chloride (LiCl) and, as previously described, to resveratrol and the AMPK activator metformin. Daf-16/FOXO and the targets engaged by these compounds define a sub-network enriched for stress-response and neuronally-active pathways. Collectively, these data highlights the dependence on a daf-16/FOXO-interaction network as a common feature of several compound families for prolonging neuronal function in HD.
Collapse
Affiliation(s)
- Francesca Farina
- CNRS, Laboratory of Neuronal Cell Biology & Pathology and University Hospital Department Fight Aging and Stress (DHU FAST), UMR 8256, Paris, France.,Sorbonne Universités, University Pierre and Marie Curie (UPMC) Univ Paris 06, Paris, France
| | - Emmanuel Lambert
- CNRS, Laboratory of Neuronal Cell Biology & Pathology and University Hospital Department Fight Aging and Stress (DHU FAST), UMR 8256, Paris, France.,Sorbonne Universités, University Pierre and Marie Curie (UPMC) Univ Paris 06, Paris, France
| | - Lucie Commeau
- CNRS, Laboratory of Neuronal Cell Biology & Pathology and University Hospital Department Fight Aging and Stress (DHU FAST), UMR 8256, Paris, France.,Sorbonne Universités, University Pierre and Marie Curie (UPMC) Univ Paris 06, Paris, France
| | - François-Xavier Lejeune
- CNRS, Laboratory of Neuronal Cell Biology & Pathology and University Hospital Department Fight Aging and Stress (DHU FAST), UMR 8256, Paris, France.,Sorbonne Universités, University Pierre and Marie Curie (UPMC) Univ Paris 06, Paris, France
| | | | - Cosima Fonte
- Inserm, UMR 1195, 94276, Le Kremlin-Bicêtre, Cedex, France
| | - J Alex Parker
- CNRS, Laboratory of Neuronal Cell Biology & Pathology and University Hospital Department Fight Aging and Stress (DHU FAST), UMR 8256, Paris, France.,Sorbonne Universités, University Pierre and Marie Curie (UPMC) Univ Paris 06, Paris, France.,CRCHUM, Montréal, Canada and Department de Neurosciences, Faculté de médecine, Université de Montréal, Montréal, Canada
| | - Jacques Boddaert
- CNRS, Laboratory of Neuronal Cell Biology & Pathology and University Hospital Department Fight Aging and Stress (DHU FAST), UMR 8256, Paris, France.,Sorbonne Universités, University Pierre and Marie Curie (UPMC) Univ Paris 06, Paris, France.,Department of Geriatrics, Pitié-Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris (APHP), 75013, Paris, France
| | - Marc Verny
- CNRS, Laboratory of Neuronal Cell Biology & Pathology and University Hospital Department Fight Aging and Stress (DHU FAST), UMR 8256, Paris, France.,Sorbonne Universités, University Pierre and Marie Curie (UPMC) Univ Paris 06, Paris, France.,Department of Geriatrics, Pitié-Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris (APHP), 75013, Paris, France
| | - Etienne-Emile Baulieu
- Inserm, UMR 1195, 94276, Le Kremlin-Bicêtre, Cedex, France. .,MAPREG, 94276, Le Kremlin-Bicêtre, Cedex, France.
| | - Christian Neri
- CNRS, Laboratory of Neuronal Cell Biology & Pathology and University Hospital Department Fight Aging and Stress (DHU FAST), UMR 8256, Paris, France. .,Sorbonne Universités, University Pierre and Marie Curie (UPMC) Univ Paris 06, Paris, France.
| |
Collapse
|
17
|
Maulik M, Mitra S, Bult-Ito A, Taylor BE, Vayndorf EM. Behavioral Phenotyping and Pathological Indicators of Parkinson's Disease in C. elegans Models. Front Genet 2017; 8:77. [PMID: 28659967 PMCID: PMC5468440 DOI: 10.3389/fgene.2017.00077] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/22/2017] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder with symptoms that progressively worsen with age. Pathologically, PD is characterized by the aggregation of α-synuclein in cells of the substantia nigra in the brain and loss of dopaminergic neurons. This pathology is associated with impaired movement and reduced cognitive function. The etiology of PD can be attributed to a combination of environmental and genetic factors. A popular animal model, the nematode roundworm Caenorhabditis elegans, has been frequently used to study the role of genetic and environmental factors in the molecular pathology and behavioral phenotypes associated with PD. The current review summarizes cellular markers and behavioral phenotypes in transgenic and toxin-induced PD models of C. elegans.
Collapse
Affiliation(s)
- Malabika Maulik
- Department of Chemistry and Biochemistry, University of Alaska FairbanksFairbanks, AK, United States
| | - Swarup Mitra
- Department of Chemistry and Biochemistry, University of Alaska FairbanksFairbanks, AK, United States
| | - Abel Bult-Ito
- Department of Biology and Wildlife, University of Alaska FairbanksFairbanks, AK, United States
| | - Barbara E Taylor
- Department of Biological Sciences, California State University, Long BeachLong Beach, CA, United States
| | - Elena M Vayndorf
- Institute of Arctic Biology, University of Alaska FairbanksFairbanks, AK, United States
| |
Collapse
|
18
|
Krieg M, Stühmer J, Cueva JG, Fetter R, Spilker K, Cremers D, Shen K, Dunn AR, Goodman MB. Genetic defects in β-spectrin and tau sensitize C. elegans axons to movement-induced damage via torque-tension coupling. eLife 2017; 6. [PMID: 28098556 PMCID: PMC5298879 DOI: 10.7554/elife.20172] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 01/17/2017] [Indexed: 12/24/2022] Open
Abstract
Our bodies are in constant motion and so are the neurons that invade each tissue. Motion-induced neuron deformation and damage are associated with several neurodegenerative conditions. Here, we investigated the question of how the neuronal cytoskeleton protects axons and dendrites from mechanical stress, exploiting mutations in UNC-70 β-spectrin, PTL-1 tau/MAP2-like and MEC-7 β-tubulin proteins in Caenorhabditis elegans. We found that mechanical stress induces supercoils and plectonemes in the sensory axons of spectrin and tau double mutants. Biophysical measurements, super-resolution, and electron microscopy, as well as numerical simulations of neurons as discrete, elastic rods provide evidence that a balance of torque, tension, and elasticity stabilizes neurons against mechanical deformation. We conclude that the spectrin and microtubule cytoskeletons work in combination to protect axons and dendrites from mechanical stress and propose that defects in β-spectrin and tau may sensitize neurons to damage. DOI:http://dx.doi.org/10.7554/eLife.20172.001
Collapse
Affiliation(s)
- Michael Krieg
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States.,Department of Chemical Engineering, Stanford University, Stanford, United States
| | - Jan Stühmer
- Department of Informatics, Technical University of Munich, , Germany
| | - Juan G Cueva
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| | - Richard Fetter
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| | - Kerri Spilker
- Department of Biology, Stanford University, Stanford, United States
| | - Daniel Cremers
- Department of Informatics, Technical University of Munich, , Germany
| | - Kang Shen
- Department of Biology, Stanford University, Stanford, United States
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, United States
| | - Miriam B Goodman
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| |
Collapse
|
19
|
Bodea L, Eckert A, Ittner LM, Piguet O, Götz J. Tau physiology and pathomechanisms in frontotemporal lobar degeneration. J Neurochem 2016; 138 Suppl 1:71-94. [PMID: 27306859 PMCID: PMC5094566 DOI: 10.1111/jnc.13600] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/31/2016] [Accepted: 02/24/2016] [Indexed: 12/27/2022]
Abstract
Frontotemporal lobar degeneration (FTLD) has been associated with toxic intracellular aggregates of hyperphosphorylated tau (FTLD-tau). Moreover, genetic studies identified mutations in the MAPT gene encoding tau in familial cases of the disease. In this review, we cover a range of aspects of tau function, both in the healthy and diseased brain, discussing several in vitro and in vivo models. Tau structure and function in the healthy brain is presented, accentuating its distinct compartmentalization in neurons and its role in microtubule stabilization and axonal transport. Furthermore, tau-driven pathology is discussed, introducing current concepts and the underlying experimental evidence. Different aspects of pathological tau phosphorylation, the protein's genomic and domain organization as well as its spreading in disease, together with MAPT-associated mutations and their respective models are presented. Dysfunction related to other post-transcriptional modifications and their effect on normal neuronal functions such as cell cycle, epigenetics and synapse dynamics are also discussed, providing a mechanistic explanation for the observations made in FTLD-tau cases, with the possibility for therapeutic intervention. In this review, we cover aspects of tau function, both in the healthy and diseased brain, referring to different in vitro and in vivo models. In healthy neurons, tau is compartmentalized, with higher concentrations found in the distal part of the axon. Cargo molecules are sensitive to this gradient. A disturbed tau distribution, as found in frontotemporal lobar degeneration (FTLD-tau), has severe consequences for cellular physiology: tau accumulates in the neuronal soma and dendrites, leading among others to microtubule depolymerization and impaired axonal transport. Tau forms insoluble aggregates that sequester additional molecules stalling cellular physiology. Neuronal communication is gradually lost as toxic tau accumulates in dendritic spines with subsequent degeneration of synapses and synaptic loss. Thus, by providing a mechanistic explanation for the observations made in FTLD-tau cases, arises a possibility for therapeutic interventions. This article is part of the Frontotemporal Dementia special issue.
Collapse
Affiliation(s)
- Liviu‐Gabriel Bodea
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Anne Eckert
- Neurobiology LaboratoryPsychiatric University Clinics BaselUniversity of BaselBaselSwitzerland
| | - Lars Matthias Ittner
- Dementia Research UnitSchool of Medical SciencesFaculty of MedicineUniversity of New South WalesSydneyNSWAustralia
| | | | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
20
|
Abstract
The sense of touch informs us of the physical properties of our surroundings and is a critical aspect of communication. Before touches are perceived, mechanical signals are transmitted quickly and reliably from the skin's surface to mechano-electrical transduction channels embedded within specialized sensory neurons. We are just beginning to understand how soft tissues participate in force transmission and how they are deformed. Here, we review empirical and theoretical studies of single molecules and molecular ensembles thought to be involved in mechanotransmission and apply the concepts emerging from this work to the sense of touch. We focus on the nematode Caenorhabditis elegans as a well-studied model for touch sensation in which mechanics can be studied on the molecular, cellular, and systems level. Finally, we conclude that force transmission is an emergent property of macromolecular cellular structures that mutually stabilize one another.
Collapse
Affiliation(s)
- Michael Krieg
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alex Dunn
- Department of Chemical Engineering, Stanford University School of Engineering, Stanford, CA, USA
| | - Miriam B. Goodman
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
21
|
Alexander AG, Marfil V, Li C. Use of Caenorhabditis elegans as a model to study Alzheimer's disease and other neurodegenerative diseases. Front Genet 2014; 5:279. [PMID: 25250042 PMCID: PMC4155875 DOI: 10.3389/fgene.2014.00279] [Citation(s) in RCA: 207] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/31/2014] [Indexed: 12/12/2022] Open
Abstract
Advances in research and technology has increased our quality of life, allowed us to combat diseases, and achieve increased longevity. Unfortunately, increased longevity is accompanied by a rise in the incidences of age-related diseases such as Alzheimer’s disease (AD). AD is the sixth leading cause of death, and one of the leading causes of dementia amongst the aged population in the USA. It is a progressive neurodegenerative disorder, characterized by the prevalence of extracellular Aβ plaques and intracellular neurofibrillary tangles, derived from the proteolysis of the amyloid precursor protein (APP) and the hyperphosphorylation of microtubule-associated protein tau, respectively. Despite years of extensive research, the molecular mechanisms that underlie the pathology of AD remain unclear. Model organisms, such as the nematode, Caenorhabditis elegans, present a complementary approach to addressing these questions. C. elegans has many advantages as a model system to study AD and other neurodegenerative diseases. Like their mammalian counterparts, they have complex biochemical pathways, most of which are conserved. Genes in which mutations are correlated with AD have counterparts in C. elegans, including an APP-related gene, apl-1, a tau homolog, ptl-1, and presenilin homologs, such as sel-12 and hop-1. Since the neuronal connectivity in C. elegans has already been established, C. elegans is also advantageous in modeling learning and memory impairments seen during AD. This article addresses the insights C. elegans provide in studying AD and other neurodegenerative diseases. Additionally, we explore the advantages and drawbacks associated with using this model.
Collapse
Affiliation(s)
- Adanna G Alexander
- Department of Biology, City College of New York New York, NY, USA ; Department of Biology, The Graduate Center, City University of New York New York, NY, USA
| | - Vanessa Marfil
- Department of Biology, City College of New York New York, NY, USA
| | - Chris Li
- Department of Biology, City College of New York New York, NY, USA ; Department of Biology, The Graduate Center, City University of New York New York, NY, USA
| |
Collapse
|
22
|
Chew YL, Fan X, Götz J, Nicholas HR. Regulation of age-related structural integrity in neurons by protein with tau-like repeats (PTL-1) is cell autonomous. Sci Rep 2014; 4:5185. [PMID: 24898126 PMCID: PMC4046136 DOI: 10.1038/srep05185] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 05/15/2014] [Indexed: 11/09/2022] Open
Abstract
PTL-1 is the sole homolog of the MAP2/MAP4/tau family in Caenorhabditis elegans. Accumulation of tau is a pathological hallmark of neurodegenerative diseases such as Alzheimer's disease. Therefore, reducing tau levels has been suggested as a therapeutic strategy. We previously showed that PTL-1 maintains age-related structural integrity in neurons, implying that excessive reduction in the levels of a tau-like protein is detrimental. Here, we demonstrate that the regulation of neuronal ageing by PTL-1 occurs via a cell-autonomous mechanism. We re-expressed PTL-1 in a null mutant background using a pan-neuronal promoter to show that PTL-1 functions in neurons to maintain structural integrity. We next expressed PTL-1 only in touch neurons and showed rescue of the neuronal ageing phenotype of ptl-1 mutant animals in these neurons but not in another neuronal subset, the ventral nerve cord GABAergic neurons. Knockdown of PTL-1 in touch neurons also resulted in premature neuronal ageing in these neurons but not in GABAergic neurons. Additionally, expression of PTL-1 in touch neurons alone was unable to rescue the shortened lifespan observed in ptl-1 mutants, but pan-neuronal re-expression restored wild-type longevity, indicating that, at least for a specific group of mechanosensory neurons, premature neuronal ageing and organismal ageing can be decoupled.
Collapse
Affiliation(s)
- Yee Lian Chew
- 1] School of Molecular Bioscience, University of Sydney, Australia [2]
| | - Xiaochen Fan
- 1] School of Molecular Bioscience, University of Sydney, Australia [2]
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research (CJCADR) at the Queensland Brain Institute (QBI), University of Queensland, Australia
| | | |
Collapse
|
23
|
Chege PM, McColl G. Caenorhabditis elegans: a model to investigate oxidative stress and metal dyshomeostasis in Parkinson's disease. Front Aging Neurosci 2014; 6:89. [PMID: 24904406 PMCID: PMC4032941 DOI: 10.3389/fnagi.2014.00089] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 04/27/2014] [Indexed: 12/04/2022] Open
Abstract
Parkinson's disease (PD) is characterized by progressive motor impairment attributed to progressive loss of dopaminergic (DAergic) neurons in the substantia nigra pars compacta. Additional clinical manifestations include non-motor symptoms such as insomnia, depression, psychosis, and cognitive impairment. PD patients with mild cognitive impairment have an increased risk of developing dementia. The affected brain regions also show perturbed metal ion levels, primarily iron. These observations have led to speculation that metal ion dyshomeostasis plays a key role in the neuronal death of this disease. However, the mechanisms underlying this metal-associated neurodegeneration have yet to be completely elucidated. Mammalian models have traditionally been used to investigate PD pathogenesis. However, alternate animal models are also being adopted, bringing to bear their respective experimental advantage. The nematode, Caenorhabditis elegans, is one such system that has well-developed genetics, is amenable to transgenesis and has relatively low associated experimental costs. C. elegans has a well characterized neuronal network that includes a simple DAergic system. In this review we will discuss mechanisms thought to underlie PD and the use of C. elegans to investigate these processes.
Collapse
Affiliation(s)
| | - Gawain McColl
- The Florey Institute of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| |
Collapse
|
24
|
Frédéric MY, Lundin VF, Whiteside MD, Cueva JG, Tu DK, Kang SYC, Singh H, Baillie DL, Hutter H, Goodman MB, Brinkman FSL, Leroux MR. Identification of 526 conserved metazoan genetic innovations exposes a new role for cofactor E-like in neuronal microtubule homeostasis. PLoS Genet 2013; 9:e1003804. [PMID: 24098140 PMCID: PMC3789837 DOI: 10.1371/journal.pgen.1003804] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 08/03/2013] [Indexed: 11/30/2022] Open
Abstract
The evolution of metazoans from their choanoflagellate-like unicellular ancestor coincided with the acquisition of novel biological functions to support a multicellular lifestyle, and eventually, the unique cellular and physiological demands of differentiated cell types such as those forming the nervous, muscle and immune systems. In an effort to understand the molecular underpinnings of such metazoan innovations, we carried out a comparative genomics analysis for genes found exclusively in, and widely conserved across, metazoans. Using this approach, we identified a set of 526 core metazoan-specific genes (the ‘metazoanome’), approximately 10% of which are largely uncharacterized, 16% of which are associated with known human disease, and 66% of which are conserved in Trichoplax adhaerens, a basal metazoan lacking neurons and other specialized cell types. Global analyses of previously-characterized core metazoan genes suggest a prevalent property, namely that they act as partially redundant modifiers of ancient eukaryotic pathways. Our data also highlights the importance of exaptation of pre-existing genetic tools during metazoan evolution. Expression studies in C. elegans revealed that many metazoan-specific genes, including tubulin folding cofactor E-like (TBCEL/coel-1), are expressed in neurons. We used C. elegans COEL-1 as a representative to experimentally validate the metazoan-specific character of our dataset. We show that coel-1 disruption results in developmental hypersensitivity to the microtubule drug paclitaxel/taxol, and that overexpression of coel-1 has broad effects during embryonic development and perturbs specialized microtubules in the touch receptor neurons (TRNs). In addition, coel-1 influences the migration, neurite outgrowth and mechanosensory function of the TRNs, and functionally interacts with components of the tubulin acetylation/deacetylation pathway. Together, our findings unveil a conserved molecular toolbox fundamental to metazoan biology that contains a number of neuronally expressed and disease-related genes, and reveal a key role for TBCEL/coel-1 in regulating microtubule function during metazoan development and neuronal differentiation. The evolution of multicellular animals (metazoans) from their single-celled ancestor required new molecular tools to create and coordinate the various biological functions involved in a communal, or multicellular, lifestyle. This would eventually include the unique cellular and physiological demands of specialized tissues like the nervous system. To identify and understand the genetic bases of such unique metazoan traits, we used a comparative genomics approach to identify 526 metazoan-specific genes which have been evolutionarily conserved throughout the diversification of the animal kingdom. Interestingly, we found that some of those genes are still completely uncharacterized or poorly studied. We used the metazoan model organism C. elegans to examine the expression of some poorly characterized metazoan-specific genes and found that many, including one encoding tubulin folding cofactor E-like (TBCEL; C. elegans COEL-1), are expressed in cells of the nervous system. Using COEL-1 as an example to understand the metazoan-specific character of our dataset, our studies reveal a new role for this protein in regulating the stability of the microtubule cytoskeleton during development, and function of the touch receptor neurons. In summary, our findings help define a conserved molecular toolbox important for metazoan biology, and uncover an important role for COEL-1/TBCEL during development and in the nervous system of the metazoan C. elegans.
Collapse
Affiliation(s)
- Melissa Y. Frédéric
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Victor F. Lundin
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
| | - Matthew D. Whiteside
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Juan G. Cueva
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
| | - Domena K. Tu
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - S. Y. Catherine Kang
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Cancer Control Research, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Hansmeet Singh
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
| | - David L. Baillie
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Harald Hutter
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Miriam B. Goodman
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
| | - Fiona S. L. Brinkman
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Michel R. Leroux
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- * E-mail:
| |
Collapse
|
25
|
Eckert A, Nisbet R, Grimm A, Götz J. March separate, strike together--role of phosphorylated TAU in mitochondrial dysfunction in Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2013; 1842:1258-66. [PMID: 24051203 DOI: 10.1016/j.bbadis.2013.08.013] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/08/2013] [Accepted: 08/12/2013] [Indexed: 12/17/2022]
Abstract
The energy demand and calcium buffering requirements of the brain are met by the high number of mitochondria in neurons and in these, especially at the synapses. Mitochondria are the major producer of reactive oxygen species (ROS); at the same time, they are damaged by ROS that are induced by abnormal protein aggregates that characterize human neurodegenerative diseases such as Alzheimer's disease (AD). Because synaptic mitochondria are long-lived, any damage exerted by these aggregates impacts severely on neuronal function. Here we review how increased TAU, a defining feature of AD and related tauopathies, impairs mitochondrial function by following the principle: 'March separate, strike together!' In the presence of amyloid-β, TAU's toxicity is augmented suggesting synergistic pathomechanisms. In order to restore mitochondrial functions in neurodegeneration as a means of therapeutic intervention it will be important to integrate the various aspects of dysfunction and get a handle on targeting distinct cell types and subcellular compartments.
Collapse
Affiliation(s)
- Anne Eckert
- Neurobiology Laboratory, Psychiatric University Clinics Basel, University of Basel, Switzerland
| | - Rebecca Nisbet
- Centre for Ageing Dementia Research (CADR), Queensland Brain Institute (QBI), The University of Queensland, Australia
| | - Amandine Grimm
- Neurobiology Laboratory, Psychiatric University Clinics Basel, University of Basel, Switzerland
| | - Jürgen Götz
- Centre for Ageing Dementia Research (CADR), Queensland Brain Institute (QBI), The University of Queensland, Australia.
| |
Collapse
|
26
|
Chew YL, Fan X, Götz J, Nicholas HR. Aging in the nervous system of Caenorhabditis elegans. Commun Integr Biol 2013; 6:e25288. [PMID: 24255742 PMCID: PMC3829903 DOI: 10.4161/cib.25288] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 06/04/2013] [Accepted: 06/05/2013] [Indexed: 11/28/2022] Open
Abstract
It has recently been described that aging in C. elegans is accompanied by the progressive development of morphological changes in the nervous system. These include novel outgrowths from the cell body or axonal process, as well as blebbing and beading along the length of the axon. The formation of these structures is regulated by numerous molecular players including members of the well-conserved insulin/insulin growth factor-like (IGF)-1 signaling and mitogen-activated protein (MAP) kinase pathways. This review summarizes the recent literature on neuronal aging in C. elegans, including our own findings, which indicate a role for protein with tau-like repeats (PTL-1), the homolog of mammalian tau and MAP2/4, in maintaining neuronal integrity during aging.
Collapse
Affiliation(s)
- Yee Lian Chew
- School of Molecular Bioscience; University of Sydney; Sydney, NSW Australia
| | | | | | | |
Collapse
|
27
|
Götz J, Xia D, Leinenga G, Chew YL, Nicholas HR. What Renders TAU Toxic. Front Neurol 2013; 4:72. [PMID: 23772223 PMCID: PMC3677143 DOI: 10.3389/fneur.2013.00072] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 05/28/2013] [Indexed: 12/21/2022] Open
Abstract
TAU is a microtubule-associated protein that under pathological conditions such as Alzheimer's disease (AD) forms insoluble, filamentous aggregates. When 20 years after TAU's discovery the first TAU transgenic mouse models were established, one declared goal that was achieved was the modeling of authentic TAU aggregate formation in the form of neurofibrillary tangles. However, as we review here, it has become increasingly clear that TAU causes damage much before these filamentous aggregates develop. In fact, because TAU is a scaffolding protein, increased levels and an altered subcellular localization (due to an increased insolubility and impaired clearance) result in the interaction of TAU with cellular proteins with which it would otherwise either not interact or do so to a lesser degree, thereby impairing their physiological functions. We specifically discuss the non-axonal localization of TAU, the role phosphorylation has in TAU toxicity and how TAU impairs mitochondrial functions. A major emphasis is on what we have learned from the four available TAU knock-out models in mice, and the knock-out of the TAU/MAP2 homolog PTL-1 in worms. It has been proposed that in human pathological conditions such as AD, a rare toxic TAU species exists which needs to be specifically removed to abrogate TAU's toxicity and restore neuronal functions. However, what is toxic in one context may not be in another, and simply reducing, but not fully abolishing TAU levels may be sufficient to abrogate TAU toxicity.
Collapse
Affiliation(s)
- Jürgen Götz
- Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Sydney Medical School, Brain and Mind Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Di Xia
- Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Gerhard Leinenga
- Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Yee Lian Chew
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, Australia
| | - Hannah R. Nicholas
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
28
|
Raslan AA, Kee Y. Tackling neurodegenerative diseases: animal models of Alzheimer’s disease and Parkinson’s disease. Genes Genomics 2013. [DOI: 10.1007/s13258-013-0116-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
29
|
Chew YL, Fan X, Götz J, Nicholas HR. PTL-1 regulates neuronal integrity and lifespan in C. elegans. J Cell Sci 2013; 126:2079-91. [PMID: 23525010 DOI: 10.1242/jcs.jcs124404] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Protein with tau-like repeats (PTL-1) is the sole Caenorhabditis elegans homolog of tau and MAP2, which are members of the mammalian family of microtubule-associated proteins (MAPs). In mammalian neurons, tau and MAP2 are segregated, with tau being mainly localised to the axon and MAP2 mainly to the dendrite. In particular, tau plays a crucial role in pathology, as elevated levels lead to the formation of tau aggregates in many neurodegenerative conditions including Alzheimer's disease. We used PTL-1 in C. elegans to model the biological functions of a tau-like protein without the complication of functional redundancy that is observed among the mammalian MAPs. Our findings indicate that PTL-1 is important for the maintenance of neuronal health as animals age, as well as in the regulation of whole organism lifespan. In addition, gene dosage of PTL-1 is crucial because variations from wild-type levels are detrimental. We also observed that human tau is unable to robustly compensate for loss of PTL-1, although phenotypes observed in tau transgenic worms are dependent on the presence of endogenous PTL-1. Our data suggest that some of the effects of tau pathology result from the loss of physiological tau function and not solely from a toxic gain-of-function due to accumulation of tau.
Collapse
Affiliation(s)
- Yee Lian Chew
- School of Molecular Bioscience, University of Sydney, New South Wales, 2006, Australia
| | | | | | | |
Collapse
|
30
|
Mancozeb-induced behavioral deficits precede structural neural degeneration. Neurotoxicology 2013; 34:74-81. [DOI: 10.1016/j.neuro.2012.10.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 09/10/2012] [Accepted: 10/11/2012] [Indexed: 01/03/2023]
|
31
|
Bellanger JM, Cueva JG, Baran R, Tang G, Goodman MB, Debant A. The doublecortin-related gene zyg-8 is a microtubule organizer in Caenorhabditis elegans neurons. J Cell Sci 2012; 125:5417-27. [PMID: 22956537 DOI: 10.1242/jcs.108381] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Doublecortin-domain containing (DCDC) genes play key roles in the normal and pathological development of the human brain cortex. The origin of the cellular specialisation and the functional redundancy of these microtubule (MT)-associated proteins (MAPs), especially those of Doublecortin (DCX) and Doublecortin-like kinase (DCLKs) genes, is still unclear. The DCX domain has the ability to control MT architecture and bundling. However, the physiological significance of such properties is not fully understood. To address these issues, we sought post-mitotic roles for zyg-8, the sole representative of the DCX-DCLK subfamily of genes in C. elegans. Previously, zyg-8 has been shown to control anaphase-spindle positioning in one-cell stage embryos, but functions of the gene later in development have not been investigated. Here we show that wild-type zyg-8 is required beyond early embryonic divisions for proper development, spontaneous locomotion and touch sensitivity of adult worms. Consistently, we find zyg-8 expression in the six touch receptor neurons (TRNs), as well as in a subset of other neuronal and non-neuronal cells. In TRNs and motoneurons, zyg-8 controls cell body shape/polarity and process outgrowth and morphology. Ultrastructural analysis of mutant animals reveals that zyg-8 promotes structural integrity, length and number of individual MTs, as well as their bundled organisation in TRNs, with no impact on MT architecture.
Collapse
Affiliation(s)
- Jean-Michel Bellanger
- CRBM-CNRS, Université Montpellier 2, 1919, route de Mende, 34293 Montpellier, France.
| | | | | | | | | | | |
Collapse
|
32
|
Fatouros C, Pir GJ, Biernat J, Koushika SP, Mandelkow E, Mandelkow EM, Schmidt E, Baumeister R. Inhibition of tau aggregation in a novel Caenorhabditis elegans model of tauopathy mitigates proteotoxicity. Hum Mol Genet 2012; 21:3587-603. [PMID: 22611162 DOI: 10.1093/hmg/dds190] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Increased Tau protein amyloidogenicity has been causatively implicated in several neurodegenerative diseases, collectively called tauopathies. In pathological conditions, Tau becomes hyperphosphorylated and forms intracellular aggregates. The deletion of K280, which is a mutation that commonly appears in patients with frontotemporal dementia with Parkinsonism linked to chromosome 17, enhances Tau aggregation propensity (pro-aggregation). In contrast, introduction of the I277P and I308P mutations prevents β-sheet formation and subsequent aggregation (anti-aggregation). In this study, we created a tauopathy model by expressing pro- or anti-aggregant Tau species in the nervous system of Caenorhabditis elegans. Animals expressing the highly amyloidogenic Tau species showed accelerated Tau aggregation and pathology manifested by severely impaired motility and evident neuronal dysfunction. In addition, we observed that the axonal transport of mitochondria was perturbed in these animals. Control animals expressing the anti-aggregant combination had rather mild phenotype. We subsequently tested several Tau aggregation inhibitor compounds and observed a mitigation of Tau proteotoxicity. In particular, a novel compound that crosses the blood-brain barrier of mammals proved effective in ameliorating the motility as well as delaying the accumulation of neuronal defects. Our study establishes a new C. elegans model of Tau aggregation-mediated toxicity and supports the emerging notion that inhibiting the nucleation of Tau aggregation can be neuroprotective.
Collapse
Affiliation(s)
- Chronis Fatouros
- Institute of Biology III, University of Freiburg, 79104 Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Pandey UB, Nichols CD. Human disease models in Drosophila melanogaster and the role of the fly in therapeutic drug discovery. Pharmacol Rev 2011; 63:411-36. [PMID: 21415126 PMCID: PMC3082451 DOI: 10.1124/pr.110.003293] [Citation(s) in RCA: 706] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The common fruit fly, Drosophila melanogaster, is a well studied and highly tractable genetic model organism for understanding molecular mechanisms of human diseases. Many basic biological, physiological, and neurological properties are conserved between mammals and D. melanogaster, and nearly 75% of human disease-causing genes are believed to have a functional homolog in the fly. In the discovery process for therapeutics, traditional approaches employ high-throughput screening for small molecules that is based primarily on in vitro cell culture, enzymatic assays, or receptor binding assays. The majority of positive hits identified through these types of in vitro screens, unfortunately, are found to be ineffective and/or toxic in subsequent validation experiments in whole-animal models. New tools and platforms are needed in the discovery arena to overcome these limitations. The incorporation of D. melanogaster into the therapeutic discovery process holds tremendous promise for an enhanced rate of discovery of higher quality leads. D. melanogaster models of human diseases provide several unique features such as powerful genetics, highly conserved disease pathways, and very low comparative costs. The fly can effectively be used for low- to high-throughput drug screens as well as in target discovery. Here, we review the basic biology of the fly and discuss models of human diseases and opportunities for therapeutic discovery for central nervous system disorders, inflammatory disorders, cardiovascular disease, cancer, and diabetes. We also provide information and resources for those interested in pursuing fly models of human disease, as well as those interested in using D. melanogaster in the drug discovery process.
Collapse
Affiliation(s)
- Udai Bhan Pandey
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, 1901 Perdido St., New Orleans, LA 70112, USA
| | | |
Collapse
|
34
|
Tien NW, Wu GH, Hsu CC, Chang CY, Wagner OI. Tau/PTL-1 associates with kinesin-3 KIF1A/UNC-104 and affects the motor's motility characteristics in C. elegans neurons. Neurobiol Dis 2011; 43:495-506. [PMID: 21569846 DOI: 10.1016/j.nbd.2011.04.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 04/06/2011] [Accepted: 04/25/2011] [Indexed: 11/29/2022] Open
Abstract
Tauopathies are neurodegenerative diseases based on pathological tau-aggregation including Alzheimer's disease, frontotemporal dementia (FTD) and Pick's disease. In general, cargo (e.g., β-amyloid precursor protein, tau, neurofilaments) accumulation is a commonly observed phenomenon in degenerated neurons. Therefore, it is crucial to investigate the interaction between cargo, microtubule-binding proteins and molecular motors. We report the effect of tau/PTL-1 (protein with tau-like repeats) on the transport characteristics of the major axonal transporter kinesin-3 KIF1A/UNC-104 in the nervous system of Caenorhabditis elegans. Using confocal spinning disk time-lapse imaging we analyzed the motility of UNC-104::mRFP in ptl-1 knockout worms and found that predominantly retrograde moving characteristics are affected (rather than the motor's anterograde displacements). A similar motility pattern was observed for synaptobrevin-1-containing vesicles, a major cargo of UNC-104. Moreover, UNC-104 and PTL-1 colocalize and occasionally co-migrate. We further confirmed physical interactions between PTL-1 and UNC-104 in living animals using the bimolecular fluorescence complementation assay (BiFC) as well as in co-immunoprecipitation experiments. Though this study focuses on PTL-1/UNC-104 interactions, we extended our research on monitoring conventional kinesin-1 (UNC-116) as well as dynein motility pattern and found that in ptl-1 mutants retrograde displacements were also affected for UNC-116, while for dynein, interestingly, its anterograde movements were affected.
Collapse
Affiliation(s)
- Nai-Wen Tien
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, 30013 Hsinchu, Taiwan, ROC
| | | | | | | | | |
Collapse
|
35
|
Dimitriadi M, Hart AC. Neurodegenerative disorders: insights from the nematode Caenorhabditis elegans. Neurobiol Dis 2010; 40:4-11. [PMID: 20493260 DOI: 10.1016/j.nbd.2010.05.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 05/06/2010] [Accepted: 05/11/2010] [Indexed: 10/19/2022] Open
Abstract
Neurodegenerative diseases impose a burden on society, yet for the most part, the mechanisms underlying neuronal dysfunction and death in these disorders remain unclear despite the identification of relevant disease genes. Given the molecular conservation in neuronal signaling pathways across vertebrate and invertebrate species, many researchers have turned to the nematode Caenorhabditis elegans to identify the mechanisms underlying neurodegenerative disease pathology. C. elegans can be engineered to express human proteins associated with neurodegeneration; additionally, the function of C. elegans orthologs of human neurodegenerative disease genes can be dissected. Herein, we examine major C. elegans neurodegeneration models that recapitulate many aspects of human neurodegenerative disease and we survey the screens that have identified modifier genes. This review highlights how the C. elegans community has used this versatile organism to model several aspects of human neurodegeneration and how these studies have contributed to our understanding of human disease.
Collapse
Affiliation(s)
- Maria Dimitriadi
- Department of Neuroscience, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| | | |
Collapse
|
36
|
Ewald CY, Li C. Understanding the molecular basis of Alzheimer's disease using a Caenorhabditis elegans model system. Brain Struct Funct 2010; 214:263-83. [PMID: 20012092 PMCID: PMC3902020 DOI: 10.1007/s00429-009-0235-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Accepted: 11/17/2009] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is the major cause of dementia in the United States. At the cellular level, the brains of AD patients are characterized by extracellular dense plaques and intracellular neurofibrillary tangles whose major components are the beta-amyloid peptide and tau, respectively. The beta-amyloid peptide is a cleavage product of the amyloid precursor protein (APP); mutations in APP have been correlated with a small number of cases of familial Alzheimer's disease. APP is the canonical member of the APP family, whose functions remain unclear. The nematode Caenorhabditis elegans, one of the premier genetic workhorses, is being used in a variety of ways to address the functions of APP and determine how the beta-amyloid peptide and tau can induce toxicity. First, the function of the C. elegans APP-related gene, apl-1, is being examined. Although different organisms may use APP and related proteins, such as APL-1, in different functional contexts, the pathways in which they function and the molecules with which they interact are usually conserved. Second, components of the gamma-secretase complex and their respective functions are being revealed through genetic analyses in C. elegans. Third, to address questions of toxicity, onset of degeneration, and protective mechanisms, different human beta-amyloid peptide and tau variants are being introduced into C. elegans and the resultant transgenic lines examined. Here, we summarize how a simple system such as C. elegans can be used as a model to understand APP function and suppression of beta-amyloid peptide and tau toxicity in higher organisms.
Collapse
Affiliation(s)
- Collin Y. Ewald
- Graduate Center and Department of Biology, City College of the City University of New York, MR526, 160 Convent Avenue, New York, NY 10031, USA
| | - Chris Li
- Graduate Center and Department of Biology, City College of the City University of New York, MR526, 160 Convent Avenue, New York, NY 10031, USA
| |
Collapse
|
37
|
The multipurpose 15-protofilament microtubules in C. elegans have specific roles in mechanosensation. Curr Biol 2009; 19:1362-7. [PMID: 19615905 DOI: 10.1016/j.cub.2009.06.036] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 06/14/2009] [Accepted: 06/15/2009] [Indexed: 02/04/2023]
Abstract
Because microtubules perform many essential functions in neurons, delineating unique roles attributable to these organelles presents a formidable challenge. Microtubules endow neurons with shape and structure and are required for developmental processes including neurite outgrowth, intracellular transport, and synapse formation and plasticity; microtubules in sensory neurons may be required for the above processes in addition to a specific sensory function. In Caenorhabditis elegans, six touch receptor neurons (TRNs) sense gentle touch and uniquely contain 15-protofilament microtubules. Disruption of these microtubules by loss of either the MEC-7 beta-tubulin or MEC-12 alpha-tubulin or by growth in 1 mM colchicine causes touch insensitivity, altered distribution of the touch transduction channel, and a general reduction in protein levels. We show that the effect on touch sensitivity can be separated from the others; microtubule depolymerization in mature TRNs causes touch insensitivity but does not result in protein distribution and production defects. In addition, the mec-12(e1605) mutation selectively causes touch insensitivity without affecting microtubule formation and other cellular processes. Touching e1605 animals produces a reduced mechanoreceptor current that inactivates more rapidly than in wild-type, suggesting a specific role of the microtubules in mechanotransduction.
Collapse
|