51
|
Brunetti G, Di Rosa G, Scuto M, Leri M, Stefani M, Schmitz-Linneweber C, Calabrese V, Saul N. Healthspan Maintenance and Prevention of Parkinson's-like Phenotypes with Hydroxytyrosol and Oleuropein Aglycone in C. elegans. Int J Mol Sci 2020; 21:ijms21072588. [PMID: 32276415 PMCID: PMC7178172 DOI: 10.3390/ijms21072588] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 12/22/2022] Open
Abstract
Numerous studies highlighted the beneficial effects of the Mediterranean diet (MD) in maintaining health, especially during ageing. Even neurodegeneration, which is part of the natural ageing process, as well as the foundation of ageing-related neurodegenerative disorders like Alzheimer’s and Parkinson’s disease (PD), was successfully targeted by MD. In this regard, olive oil and its polyphenolic constituents have received increasing attention in the last years. Thus, this study focuses on two main olive oil polyphenols, hydroxytyrosol (HT) and oleuropein aglycone (OLE), and their effects on ageing symptoms with special attention to PD. In order to avoid long-lasting, expensive, and ethically controversial experiments, the established invertebrate model organism Caenorhabditis elegans was used to test HT and OLE treatments. Interestingly, both polyphenols were able to increase the survival after heat stress, but only HT could prolong the lifespan in unstressed conditions. Furthermore, in aged worms, HT and OLE caused improvements of locomotive behavior and the attenuation of autofluorescence as a marker for ageing. In addition, by using three different C. elegans PD models, HT and OLE were shown i) to enhance locomotion in worms suffering from α-synuclein-expression in muscles or rotenone exposure, ii) to reduce α-synuclein accumulation in muscles cells, and iii) to prevent neurodegeneration in α-synuclein-containing dopaminergic neurons. Hormesis, antioxidative capacities and an activity-boost of the proteasome & phase II detoxifying enzymes are discussed as potential underlying causes for these beneficial effects. Further biological and medical trials are indicated to assess the full potential of HT and OLE and to uncover their mode of action.
Collapse
Affiliation(s)
- Giovanni Brunetti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (G.B.); (G.D.R.); (M.S.)
| | - Gabriele Di Rosa
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (G.B.); (G.D.R.); (M.S.)
| | - Maria Scuto
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (G.B.); (G.D.R.); (M.S.)
| | - Manuela Leri
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.S.)
- Department of Neuroscience, Psychology, Area of Medicine and Health of the Child of the University of Florence, Viale Pieraccini, 6 - 50139 Florence, Italy
| | - Massimo Stefani
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.S.)
| | - Christian Schmitz-Linneweber
- Humboldt University of Berlin, Faculty of Life Sciences, Institute of Biology, Molecular Genetics Group, Philippstr. 13, House 22, 10115 Berlin, Germany;
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (G.B.); (G.D.R.); (M.S.)
- Correspondence: (V.C.); (N.S.)
| | - Nadine Saul
- Humboldt University of Berlin, Faculty of Life Sciences, Institute of Biology, Molecular Genetics Group, Philippstr. 13, House 22, 10115 Berlin, Germany;
- Correspondence: (V.C.); (N.S.)
| |
Collapse
|
52
|
Wong SQ, Kumar AV, Mills J, Lapierre LR. C. elegans to model autophagy-related human disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 172:325-373. [PMID: 32620247 DOI: 10.1016/bs.pmbts.2020.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Autophagy is a highly conserved degradation process that clears damaged intracellular macromolecules and organelles in order to maintain cellular health. Dysfunctional autophagy is fundamentally linked to the development of various human disorders and pathologies. The use of the nematode Caenorhabditis elegans as a model system to study autophagy has improved our understanding of its regulation and function in organismal physiology. Here, we review the genetic, functional, and regulatory conservation of the autophagy pathway in C. elegans and we describe tools to quantify and study the autophagy process in this incredibly useful model organism. We further discuss how these nematodes have been modified to model autophagy-related human diseases and underscore the important insights obtained from such models. Altogether, we highlight the strengths of C. elegans as an exceptional tool to understand the genetic and molecular foundations underlying autophagy-related human diseases.
Collapse
Affiliation(s)
- Shi Quan Wong
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Anita V Kumar
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Joslyn Mills
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Louis R Lapierre
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States.
| |
Collapse
|
53
|
de Araújo Boleti AP, de Oliveira Flores TM, Moreno SE, Anjos LD, Mortari MR, Migliolo L. Neuroinflammation: An overview of neurodegenerative and metabolic diseases and of biotechnological studies. Neurochem Int 2020; 136:104714. [PMID: 32165170 DOI: 10.1016/j.neuint.2020.104714] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/19/2020] [Accepted: 03/04/2020] [Indexed: 12/11/2022]
Abstract
Neuroinflammation is an important factor contributing to cognitive impairment and neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), ischemic injury, and multiple sclerosis (MS). These diseases are characterized by inexorable progressive injury of neuron cells, and loss of motor or cognitive functions. Microglia, which are the resident macrophages in the brain, play an important role in both physiological and pathological conditions. In this review, we provide an updated discussion on the role of ROS and metabolic disease in the pathological mechanisms of activation of the microglial cells and release of cytotoxins, leading to the neurodegenerative process. In addition, we also discuss in vivo models, such as zebrafish and Caenorhabditis elegans, and provide new insights into therapeutics bioinspired by neuropeptides from venomous animals, supporting high throughput drug screening in the near future, searching for a complementary approach to elucidating crucial mechanisms associated with neurodegenerative disorders.
Collapse
Affiliation(s)
- Ana Paula de Araújo Boleti
- S-InovaBiotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117-900, Campo Grande, MS, Brazil
| | - Taylla Michelle de Oliveira Flores
- S-InovaBiotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117-900, Campo Grande, MS, Brazil; Programa de Pós-graduação em Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Susana Elisa Moreno
- S-InovaBiotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117-900, Campo Grande, MS, Brazil
| | - Lilian Dos Anjos
- Laboratório de Neurofarmacologia, Departmento Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brazil
| | - Márcia Renata Mortari
- Laboratório de Neurofarmacologia, Departmento Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brazil
| | - Ludovico Migliolo
- S-InovaBiotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117-900, Campo Grande, MS, Brazil; Programa de Pós-graduação em Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, Brazil; Programa de Pós-graduação em Bioquímica, Universidade Federal do Rio Grande do Norte, Natal, Brazil.
| |
Collapse
|
54
|
Eckenhoff RG, Maze M, Xie Z, Culley DJ, Goodlin SJ, Zuo Z, Wei H, Whittington RA, Terrando N, Orser BA, Eckenhoff MF. Perioperative Neurocognitive Disorder: State of the Preclinical Science. Anesthesiology 2020; 132:55-68. [PMID: 31834869 PMCID: PMC6913778 DOI: 10.1097/aln.0000000000002956] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The purpose of this article is to provide a succinct summary of the different experimental approaches that have been used in preclinical postoperative cognitive dysfunction research, and an overview of the knowledge that has accrued. This is not intended to be a comprehensive review, but rather is intended to highlight how the many different approaches have contributed to our understanding of postoperative cognitive dysfunction, and to identify knowledge gaps to be filled by further research. The authors have organized this report by the level of experimental and systems complexity, starting with molecular and cellular approaches, then moving to intact invertebrates and vertebrate animal models. In addition, the authors' goal is to improve the quality and consistency of postoperative cognitive dysfunction and perioperative neurocognitive disorder research by promoting optimal study design, enhanced transparency, and "best practices" in experimental design and reporting to increase the likelihood of corroborating results. Thus, the authors conclude with general guidelines for designing, conducting and reporting perioperative neurocognitive disorder rodent research.
Collapse
Affiliation(s)
- Roderic G Eckenhoff
- From Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania (R.G.E., H.W., M.F.E.) Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California (M.M.) Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts (Z.X.) Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts (D.J.C.) Harvard Medical School, Boston, Massachusetts (Z.X., D.J.C.) Department of Medicine, Oregon Health and Science University and Veterans Administration Portland Health Care System, Portland, Oregon (S.J.G.) Department of Anesthesiology, University of Virginia School of Medicine, Charlottesville, Virginia (Z.Z.) Department of Anesthesiology, Columbia University Irving Medical Center, New York, New York (R.A.W.) Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina (N.T.) Department of Anesthesia, University of Toronto, Toronto, Canada (B.A.O.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Lin C, Su Z, Luo J, Jiang L, Shen S, Zheng W, Gu W, Cao Y, Chen Y. Polysaccharide extracted from the leaves of Cyclocarya paliurus (Batal.) Iljinskaja enhanced stress resistance in Caenorhabditis elegans via skn-1 and hsf-1. Int J Biol Macromol 2020; 143:243-254. [DOI: 10.1016/j.ijbiomac.2019.12.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 11/26/2019] [Accepted: 12/03/2019] [Indexed: 01/12/2023]
|
56
|
Huber RJ, Hughes SM, Liu W, Morgan A, Tuxworth RI, Russell C. The contribution of multicellular model organisms to neuronal ceroid lipofuscinosis research. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165614. [PMID: 31783156 DOI: 10.1016/j.bbadis.2019.165614] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/14/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023]
Abstract
The NCLs (neuronal ceroid lipofuscinosis) are forms of neurodegenerative disease that affect people of all ages and ethnicities but are most prevalent in children. Commonly known as Batten disease, this debilitating neurological disorder is comprised of 13 different subtypes that are categorized based on the particular gene that is mutated (CLN1-8, CLN10-14). The pathological mechanisms underlying the NCLs are not well understood due to our poor understanding of the functions of NCL proteins. Only one specific treatment (enzyme replacement therapy) is approved, which is for the treating the brain in CLN2 disease. Hence there remains a desperate need for further research into disease-modifying treatments. In this review, we present and evaluate the genes, proteins and studies performed in the social amoeba, nematode, fruit fly, zebrafish, mouse and large animals pertinent to NCL. In particular, we highlight the use of multicellular model organisms to study NCL protein function, pathology and pathomechanisms. Their use in testing novel therapeutic approaches is also presented. With this information, we highlight how future research in these systems may be able to provide new insight into NCL protein functions in human cells and aid in the development of new therapies.
Collapse
Affiliation(s)
- Robert J Huber
- Department of Biology, Trent University, Peterborough, Ontario K9L 0G2, Canada
| | - Stephanie M Hughes
- Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre and Genetics Otago, University of Otago, Dunedin, New Zealand
| | - Wenfei Liu
- School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Alan Morgan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown St., Liverpool L69 3BX, UK
| | - Richard I Tuxworth
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Claire Russell
- Dept. Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK.
| |
Collapse
|
57
|
Pohl F, Teixeira-Castro A, Costa MD, Lindsay V, Fiúza-Fernandes J, Goua M, Bermano G, Russell W, Maciel P, Kong Thoo Lin P. GST-4-Dependent Suppression of Neurodegeneration in C. elegans Models of Parkinson's and Machado-Joseph Disease by Rapeseed Pomace Extract Supplementation. Front Neurosci 2019; 13:1091. [PMID: 31680826 PMCID: PMC6811615 DOI: 10.3389/fnins.2019.01091] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022] Open
Abstract
Genetic mutations and aging-associated oxidative damage underlie the onset and progression of neurodegenerative diseases, like Parkinson’s disease (PD) and Machado-Joseph disease (MJD). Natural products derived from plants have been regarded as important sources of novel bioactive compounds to counteract neurodegeneration. Here, we tested the neuroprotective effect of an ethanolic extract of rapeseed pomace (RSP), a rapeseed (canola) oil production by-product, in C. elegans models of MJD and PD. The extract, containing sinapine and other phenolics, restored motor function of mutant ataxin-3 (ATXN3) animals (MJD) and prevented degeneration of dopaminergic neurons in one toxin-induced and two genetic models of PD. Whole-organism sensors of antioxidant and xenobiotic response activation revealed the induction of phase II detoxification enzymes, including glutathione S- transferase (GST-4) upon RSP extract supplementation. Furthermore in vivo pharmacogenetic studies confirmed gst-4 is required for the therapeutic effect of RSP extract in the two disease models. The results suggest that GST-4-mediated antioxidant pathways may constitute promising therapeutic co-targets for neurodegenerative diseases and confirm the utility of searching for bioactive compounds in novel sources, including food and agricultural waste/by-products, such as RSP.
Collapse
Affiliation(s)
- Franziska Pohl
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen, United Kingdom
| | - Andreia Teixeira-Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Marta Daniela Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Victoria Lindsay
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen, United Kingdom
| | - Juliana Fiúza-Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Marie Goua
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen, United Kingdom
| | - Giovanna Bermano
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen, United Kingdom
| | - Wendy Russell
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, United Kingdom
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Paul Kong Thoo Lin
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen, United Kingdom
| |
Collapse
|
58
|
Shanmuganathan B, Sathya S, Balasubramaniam B, Balamurugan K, Devi KP. Amyloid-β induced neuropathological actions are suppressed by Padina gymnospora (Phaeophyceae) and its active constituent α-bisabolol in Neuro2a cells and transgenic Caenorhabditis elegans Alzheimer's model. Nitric Oxide 2019; 91:52-66. [PMID: 31362072 DOI: 10.1016/j.niox.2019.07.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/11/2019] [Accepted: 07/26/2019] [Indexed: 12/23/2022]
Abstract
The inhibition of Aβ peptide development and aggregation is a hopeful curative approach for the discovery of disease modifying drugs for Alzheimer's disease (AD) treatment. Recent research mainly focuses on the discovery of drugs from marine setting due to their immense therapeutic potential. The present study aims to evaluate the brown macroalga Padina gymnospora and its active constituent α-bisabolol against Aβ25-35 induced neurotoxicity in Neuro2a cells and transgenic Caenorhabditis elegans (CL2006 and CL4176). The results of the in vitro study revealed that the acetone extract of P. gymnospora (ACTPG) and its active constituent α-bisabolol restores the Aβ25-35 induced alteration in the oxidation of intracellular protein and lipids. In addition, ACTPG and α-bisabolol inhibited cholinesterase and β-secretase activity in Neuro2a cells. Moreover, the intracellular reactive oxygen species (ROS) and reactive nitrogen species (RNS) production was reduced by ACTPG and α-bisabolol in Neuro2a cells. The decrease in the expression level of apoptotic proteins such as Bax and caspase-3 in ACTPG and α-bisabolol treated group indicates that the seaweed and its bioactive compound have anti-apoptotic property. Further, the in vivo study revealed that the ACTPG and α-bisabolol exerts neuroprotective effect against Aβ induced proteotoxicity in transgenic C. elegans strains of AD. Moreover it altered the Aβ mediated pathways, lifespan, macromolecular damage and down regulated the AD related gene expression of ace-1, hsp-4 and Aβ, thereby preventing Aβ synthesis. Overall, the outcome of the study signifies the neuroprotective effect of ACTPG and α-bisabolol against Aβ mediated AD pathology.
Collapse
Affiliation(s)
| | - Sethuraman Sathya
- Department of Biotechnology, Alagappa University, Karaikudi, 630003, Tamil Nadu, India
| | | | | | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University, Karaikudi, 630003, Tamil Nadu, India.
| |
Collapse
|
59
|
Fuellen G, Jansen L, Cohen AA, Luyten W, Gogol M, Simm A, Saul N, Cirulli F, Berry A, Antal P, Köhling R, Wouters B, Möller S. Health and Aging: Unifying Concepts, Scores, Biomarkers and Pathways. Aging Dis 2019; 10:883-900. [PMID: 31440392 PMCID: PMC6675520 DOI: 10.14336/ad.2018.1030] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/30/2018] [Indexed: 12/30/2022] Open
Abstract
Despite increasing research efforts, there is a lack of consensus on defining aging or health. To understand the underlying processes, and to foster the development of targeted interventions towards increasing one's health, there is an urgent need to find a broadly acceptable and useful definition of health, based on a list of (molecular) features; to operationalize features of health so that it can be measured; to identify predictive biomarkers and (molecular) pathways of health; and to suggest interventions, such as nutrition and exercise, targeted at putative causal pathways and processes. Based on a survey of the literature, we propose to define health as a state of an individual characterized by the core features of physiological, cognitive, physical and reproductive function, and a lack of disease. We further define aging as the aggregate of all processes in an individual that reduce its wellbeing, that is, its health or survival or both. We define biomarkers of health by their attribute of predicting future health better than chronological age. We define healthspan pathways as molecular features of health that relate to each other by belonging to the same molecular pathway. Our conceptual framework may integrate diverse operationalizations of health and guide precision prevention efforts.
Collapse
Affiliation(s)
- Georg Fuellen
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research (IBIMA), Rostock, Germany.
| | - Ludger Jansen
- Institute of Philosophy, University of Rostock, Germany.
| | - Alan A Cohen
- Department of Family Medicine, University of Sherbrooke, Sherbrooke, Canada.
| | - Walter Luyten
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium.
| | - Manfred Gogol
- Institute of Gerontology, University Heidelberg, Germany.
| | - Andreas Simm
- Department of Cardiac Surgery, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| | - Nadine Saul
- Humboldt-University of Berlin, Institute of Biology, Berlin, Germany.
| | - Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Italy.
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Italy.
| | - Peter Antal
- Budapest University of Technology and Economics, Budapest, Hungary.
- Abiomics Europe Ltd., Hungary.
| | - Rüdiger Köhling
- Rostock University Medical Center, Institute for Physiology, Rostock, Germany.
| | | | - Steffen Möller
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research (IBIMA), Rostock, Germany.
| |
Collapse
|
60
|
Brown DG, Wobst HJ. Opportunities and Challenges in Phenotypic Screening for Neurodegenerative Disease Research. J Med Chem 2019; 63:1823-1840. [PMID: 31268707 DOI: 10.1021/acs.jmedchem.9b00797] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Toxic misfolded proteins potentially underly many neurodegenerative diseases, but individual targets which regulate these proteins and their downstream detrimental effects are often unknown. Phenotypic screening is an unbiased method to screen for novel targets and therapeutic molecules and span the range from primitive model organisms such as Sacchaomyces cerevisiae, which allow for high-throughput screening to patient-derived cell-lines that have a close connection to the disease biology but are limited in screening capacity. This perspective will review current phenotypic models, as well as the chemical screening strategies most often employed. Advances in in 3D cell cultures, high-content screens, robotic microscopy, CRISPR screening, and use of machine learning methods to process the enormous amount of data generated by these screens are certain to change the paradigm for phenotypic screening and will be discussed.
Collapse
Affiliation(s)
- Dean G Brown
- Hit Discovery, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Heike J Wobst
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| |
Collapse
|
61
|
da Silveira TL, Machado ML, Arantes LP, Zamberlan DC, Cordeiro LM, Obetine FBB, da Silva AF, Tassi CL, Soares FAA. Guanosine Prevents against Glutamatergic Excitotoxicity in C. elegans. Neuroscience 2019; 414:265-272. [PMID: 31306683 DOI: 10.1016/j.neuroscience.2019.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 02/08/2023]
Abstract
Glutamatergic neurotransmission is present in most mammalian excitatory synapses and plays a key role in central nervous system homeostasis. When over-activated, it can induce excitotoxicity, which is present in several neuropathologies. The nucleoside guanosine (GUO) is a guanine-based purine known to have neuroprotective effects by modulating glutamatergic system during glutamate excitotoxicity in mammals. However, GUO action in Caenorhabditis elegans, as well as on C. elegans glutamatergic excitotoxicity model, is not known. The GUO effects on behavioral parameters in Wild Type (WT) and knockouts worms for glutamate transporters (GLT-3, GLT-1), glutamate vesicular transporter (EAT-4), and NMDA and non-NMDA receptors were used to evaluate the GUO modulatory effects. The GUO tested concentrations did not alter the animals' development, but GUO reduced pharyngeal pumps in WT animals in a dose-dependent manner. The same effect was observed in pharyngeal pumps, when the animals were treated with 4 mM of GUO in glr-1, nmr-1 and eat-4, but not in glt-3 and glt-3;glt-1 knockouts. The double mutant glt-3; glt-1 for GluTs had decreased body bends and an increased number of reversions. This effect was reverted after treatment with GUO. Furthermore, GUO did not alter the sensory response in worms with altered glutamatergic signaling. Thus, GUO seems to modulate the worm's glutamatergic system in situations of exacerbated glutamatergic signaling, which are represented by knockout strains to glutamate transporters. However, in WT animals, GUO appears to reinforce glutamatergic signaling in specific neurons. Our findings indicate that C. elegans strains are useful models to study new compounds that could be used in glutamate-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Tássia Limana da Silveira
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Marina Lopes Machado
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Leticia Priscilla Arantes
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Daniele Coradini Zamberlan
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Larissa Marafiga Cordeiro
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Fabiane Bicca Baptista Obetine
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Aline Franzen da Silva
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Cintia Letícia Tassi
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Felix Alexandre Antunes Soares
- Universidade Federal de Santa Maria, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Bioquímica Toxicológica, Camobi, 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
62
|
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
|
63
|
Zhou L, Huang PP, Chen LL, Wang P. Panax Notoginseng Saponins Ameliorate A β-Mediated Neurotoxicity in C. elegans through Antioxidant Activities. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:7621043. [PMID: 31275419 PMCID: PMC6582912 DOI: 10.1155/2019/7621043] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/21/2019] [Indexed: 11/25/2022]
Abstract
The deposition of amyloid beta (Aβ) is the main hallmark of Alzheimer's disease (AD) and there is no effective drug to cure the progressive cognitive loss or memory deficits caused by the aggregative toxicity of Aβ protein. Oxidative stress has been hypothesized to play a role in progressive neurodegenerative diseases like AD. Panax notoginseng saponin (PNS) from the rhizome of "pseudo-ginseng" exhibits potent antioxidant effects on aging process in neuron cells and animals. By using C. elegans as an ideal model organism, the present study shows that PNS (0.5-4 mg/mL) can significantly inhibit AD-like symptoms of worm paralysis and enhance resistance to oxidative stress induced by paraquat and aging conditions. Additionally, PNS extends lifespan and maintains healthspan of C. elegans by improving the swimming prowess and fertility at old age. It markedly activates the expression of SKN-1 mRNA, which further supports SKN-1 signaling pathway which is involved in the therapeutic effect of PNS on AD C. elegans. Our results provide direct evidence on PNS for treating AD on gene level and theoretical foundation for reshaping medicinal products of PNS in the future.
Collapse
Affiliation(s)
- Ling Zhou
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Pan-Pan Huang
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Lin-Lin Chen
- Key Laboratory of Traditional Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ping Wang
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
| |
Collapse
|
64
|
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
|
65
|
Nyarko JNK, Quartey MO, Baker GB, Mousseau DD. Can Animal Models Inform on the Relationship between Depression and Alzheimer Disease? CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2019; 64:18-29. [PMID: 29685068 PMCID: PMC6364140 DOI: 10.1177/0706743718772514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The focus on the β-amyloid (Aβ) peptide in clinical Alzheimer disease (AD) as well as in animal models of AD has perhaps biased our understanding of what contributes to the heterogeneity in disease onset and progression. Part of this heterogeneity could reflect the various neuropsychiatric risk factors that present with common symptomatology and can predispose the brain to AD-like changes. One such risk factor is depression. Animal models, particularly mouse models carrying variants of AD-related gene(s), many of which lead to an accumulation of Aβ, suggest that a fundamental shift in depression-related monoaminergic systems (including serotonin and noradrenaline) is a strong indicator of the altered cellular function associated with the earlier(est) stages of AD-related pathology. These changes in monoaminergic neurochemistry could provide for relevant targets for intervention in clinical AD and/or could support a polypharmacy strategy, which might include the targeting of Aβ, in vulnerable populations. Future studies must also include female mice as well as male mice in animal model studies on the relationship between depression and AD.
Collapse
Affiliation(s)
- Jennifer N K Nyarko
- 1 Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Maa O Quartey
- 1 Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Glen B Baker
- 2 Department of Psychiatry, Neuroscience and Mental Health Institute, Neurochemical Research Unit, University of Alberta, Edmonton, Alberta, Canada
| | - Darrell D Mousseau
- 1 Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
66
|
Worms on a Chip. Bioanalysis 2019. [DOI: 10.1007/978-981-13-6229-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
67
|
Moliner C, Barros L, Dias MI, López V, Langa E, Ferreira ICFR, Gómez-Rincón C. Edible Flowers of Tagetes erecta L. as Functional Ingredients: Phenolic Composition, Antioxidant and Protective Effects on Caenorhabditis elegans. Nutrients 2018; 10:nu10122002. [PMID: 30567311 PMCID: PMC6316237 DOI: 10.3390/nu10122002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/03/2018] [Accepted: 12/13/2018] [Indexed: 12/29/2022] Open
Abstract
Tagetes erecta L. has long been consumed for culinary and medicinal purposes in different countries. The aim of this study was to explore the potential benefits from two cultivars of T. erecta related to its polyphenolic profile as well as antioxidant and anti-aging properties. The phenolic composition was analyzed by LC-DAD-ESI/MSn. Folin-Ciocalteu, DPPH·, and FRAP assays were performed in order to evaluate reducing antiradical properties. The neuroprotective potential was evaluated using the enzymes acetylcholinesterase and monoamine oxidase. Caenorhabditis elegans was used as an in vivo model to assess extract toxicity, antioxidant activity, delayed aging, and reduced β-amyloid toxicity. Both extracts showed similar phenolic profiles and bioactivities. The main polyphenols found were laricitin and its glycosides. No acute toxicity was detected for extracts in the C. elegans model. T. erecta flower extracts showed promising antioxidant and neuroprotective properties in the different tested models. Hence, these results may add some information supporting the possibilities of using these plants as functional foods and/or as nutraceutical ingredients.
Collapse
Affiliation(s)
- Cristina Moliner
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, 50830 Villanueva de Gállego (Zaragoza), Spain.
| | - Lillian Barros
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal.
| | - Maria Inês Dias
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal.
| | - Víctor López
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, 50830 Villanueva de Gállego (Zaragoza), Spain.
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), 50013 Zaragoza, Spain.
| | - Elisa Langa
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, 50830 Villanueva de Gállego (Zaragoza), Spain.
| | - Isabel C F R Ferreira
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal.
| | - Carlota Gómez-Rincón
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, 50830 Villanueva de Gállego (Zaragoza), Spain.
| |
Collapse
|
68
|
Pohl F, Kong Thoo Lin P. The Potential Use of Plant Natural Products and Plant Extracts with Antioxidant Properties for the Prevention/Treatment of Neurodegenerative Diseases: In Vitro, In Vivo and Clinical Trials. Molecules 2018; 23:E3283. [PMID: 30544977 PMCID: PMC6321248 DOI: 10.3390/molecules23123283] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 01/08/2023] Open
Abstract
Neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease and Huntington's disease, present a major health issue and financial burden for health care systems around the world. The impact of these diseases will further increase over the next decades due to increasing life expectancies. No cure is currently available for the treatment of these conditions; only drugs, which merely alleviate the symptoms. Oxidative stress has long been associated with neurodegeneration, whether as a cause or as part of the downstream results caused by other factors. Thus, the use of antioxidants to counter cellular oxidative stress within the nervous system has been suggested as a potential treatment option for neurological disorders. Over the last decade, significant research has focused on the potential use of natural antioxidants to target oxidative stress. However, clinical trial results have lacked success for the treatment of patients with neurological disorders. The knowledge that natural extracts show other positive molecular activities in addition to antioxidant activity, however, has led to further research of natural extracts for their potential use as prevention or treatment/management of neurodegenerative diseases. This review will cover several in vitro and in vivo research studies, as well as clinical trials, and highlight the potential of natural antioxidants.
Collapse
Affiliation(s)
- Franziska Pohl
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen AB10 7GJ, UK.
| | - Paul Kong Thoo Lin
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen AB10 7GJ, UK.
| |
Collapse
|
69
|
Folch J, Busquets O, Ettcheto M, Sánchez-López E, Pallàs M, Beas-Zarate C, Marin M, Casadesus G, Olloquequi J, Auladell C, Camins A. Experimental Models for Aging and their Potential for Novel Drug Discovery. Curr Neuropharmacol 2018; 16:1466-1483. [PMID: 28685671 PMCID: PMC6295931 DOI: 10.2174/1570159x15666170707155345] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/22/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023] Open
Abstract
Background: An interesting area of scientific research is the development of potential antiaging drugs. In order to pursue this goal, it is necessary to gather the specific knowledge about the adequate preclinical models that are available to evaluate the beneficial effects of new potential drugs. This review is focused on invertebrate and vertebrate preclinical models used to evaluate the efficacy of antiaging compounds, with the objective to extend life span and health span. Methods: Research and online content related to aging, antiaging drugs, experimental aging models is reviewed. Moreover, in this review, the main experimental preclinical models of organisms that have contributed to the research in the pharmacol-ogy of lifespan extension and the understanding of the aging process are discussed. Results: Dietary restriction (DR) constitutes a common experimental process to extend life span in all organisms. Besides, classical antiaging drugs such as resveratrol, rapamycin and metformin denominated as DR mimetics are also discussed. Likewise, the main therapeutic targets of these drugs include sirtuins, IGF-1, and mTOR, all of them being modulated by DR. Conclusion: Advances in molecular biology have uncovered the potential molecular pathways involved in the aging process. Due to their characteristics, invertebrate models are mainly used for drug screening. The National Institute on Aging (NIA) developed the Interventions Testing Program (ITP). At the pre-clinical level, the ITP uses Heterogeneous mouse model (HET) which is probably the most suitable rodent model to study potential drugs against aging prevention. The accelerated-senescence mouse P8 is also a mammalian rodent model for aging research. However, when evaluating the effect of drugs on a preclinical level, the evaluation must be done in non-human primates since it is the mammalian specie closest to humans. Research is needed to investigate the impact of new potential drugs for the increase of human quality of
Collapse
Affiliation(s)
- Jaume Folch
- Unitat de Bioquimica i Biotecnologia, Facultat de Medicina i Ciencies de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Oriol Busquets
- Unitat de Bioquimica i Biotecnologia, Facultat de Medicina i Ciencies de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Departament Deaprtament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Miren Ettcheto
- Unitat de Bioquimica i Biotecnologia, Facultat de Medicina i Ciencies de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Departament Deaprtament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Elena Sánchez-López
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Unitat de Farmacia, Tecnologia Farmacèutica i Fisico-química, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Mercè Pallàs
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Departament Deaprtament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Carlos Beas-Zarate
- Departamento de Biologia Celulary Molecular, C.U.C.B.A., Universidad de Guadalajara and Division de Neurociencias, Sierra Mojada 800, Col. Independencia, Guadalajara, Jalisco 44340, Mexico
| | - Miguel Marin
- Centro de Biotecnologia. Universidad Nacional de Loja, Av. Pío Jaramillo Alvarado y Reinaldo Espinosa, La Argelia. Loja, Ecuador
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Jordi Olloquequi
- Instituto de Ciencias Biomedicas, Facultad de Ciencias de la Salud, Universidad Autonoma de Chile, Talca, Chile
| | - Carme Auladell
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Departamento de Biologia Celulary Molecular, C.U.C.B.A., Universidad de Guadalajara and Division de Neurociencias, Sierra Mojada 800, Col. Independencia, Guadalajara, Jalisco 44340, Mexico.,Departament de Biologia Cellular, Fisiologia i Inmunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Antoni Camins
- Departament Deaprtament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Centro de Biotecnologia. Universidad Nacional de Loja, Av. Pío Jaramillo Alvarado y Reinaldo Espinosa, La Argelia. Loja, Ecuador
| |
Collapse
|
70
|
Marshall LJ, Willett C. Parkinson's disease research: adopting a more human perspective to accelerate advances. Drug Discov Today 2018; 23:1950-1961. [PMID: 30240875 DOI: 10.1016/j.drudis.2018.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/20/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) affects 1% of the population over 60 years old and, with global increases in the aging population, presents huge economic and societal burdens. The etiology of PD remains unknown; most cases are idiopathic, presumed to result from genetic and environmental risk factors. Despite 200 years since the first description of PD, the mechanisms behind initiation and progression of the characteristic neurodegenerative processes are not known. Here, we review progress and limitations of the multiple PD animal models available and identify advances that could be implemented to better understand pathological processes, improve disease outcome, and reduce dependence on animal models. Lessons learned from reducing animal use in PD research could serve as guideposts for wider biomedical research.
Collapse
Affiliation(s)
- Lindsay J Marshall
- Humane Society International, The Humane Society of the United States, 700 Professional Drive, Gaithersburg, MD 20879, USA
| | - Catherine Willett
- Humane Society International, The Humane Society of the United States, 700 Professional Drive, Gaithersburg, MD 20879, USA.
| |
Collapse
|
71
|
Wong SQ, Jones A, Dodd S, Grimes D, Barclay JW, Marson AG, Cunliffe VT, Burgoyne RD, Sills GJ, Morgan A. A Caenorhabditis elegans assay of seizure-like activity optimised for identifying antiepileptic drugs and their mechanisms of action. J Neurosci Methods 2018; 309:132-142. [PMID: 30189284 PMCID: PMC6200019 DOI: 10.1016/j.jneumeth.2018.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/14/2018] [Accepted: 09/02/2018] [Indexed: 11/28/2022]
Abstract
Worms with mutant GABAA receptors exhibit convulsions upon exposure to pentylenetetrazol. Convulsions are prevented by the approved anti-epileptic drug, ethosuximide. C. elegans model is a higher throughput, ethical alternative to rodent seizure models.
Background Epilepsy affects around 1% of people, but existing antiepileptic drugs (AEDs) only offer symptomatic relief and are ineffective in approximately 30% of patients. Hence, new AEDs are sorely needed. However, a major bottleneck is the low-throughput nature of early-stage AED screens in conventional rodent models. This process could potentially be expedited by using simpler invertebrate systems, such as the nematode Caenorhabditis elegans. New method Head-bobbing convulsions were previously reported to be inducible by pentylenetetrazol (PTZ) in C. elegans with loss-of-function mutations in unc-49, which encodes a GABAA receptor. Given that epilepsy-linked mutations in human GABAA receptors are well documented, this could represent a clinically-relevant system for early-stage AED screens. However, the original agar plate-based assay is unsuited to large-scale screening and has not been validated for identifying AEDs. Therefore, we established an alternative streamlined, higher-throughput approach whereby mutants were treated with PTZ and AEDs via liquid-based incubation. Results Convulsions induced within minutes of PTZ exposure in unc-49 mutants were strongly inhibited by the established AED ethosuximide. This protective activity was independent of ethosuximide’s suggested target, the T-type calcium channel, as a null mutation in the worm cca-1 ortholog did not affect ethosuximide’s anticonvulsant action. Comparison with existing method Our streamlined assay is AED-validated, feasible for higher throughput compound screens, and can facilitate insights into AED mechanisms of action. Conclusions Based on an epilepsy-associated genetic background, this C. elegans unc-49 model of seizure-like activity presents an ethical, higher throughput alternative to conventional rodent seizure models for initial AED screens.
Collapse
Affiliation(s)
- Shi Quan Wong
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Alistair Jones
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Steven Dodd
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Douglas Grimes
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Jeff W Barclay
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Anthony G Marson
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Vincent T Cunliffe
- Department of Biomedical Science, University of Sheffield, Sheffield, UK.
| | - Robert D Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Graeme J Sills
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Alan Morgan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
72
|
Savelieff MG, Nam G, Kang J, Lee HJ, Lee M, Lim MH. Development of Multifunctional Molecules as Potential Therapeutic Candidates for Alzheimer’s Disease, Parkinson’s Disease, and Amyotrophic Lateral Sclerosis in the Last Decade. Chem Rev 2018; 119:1221-1322. [DOI: 10.1021/acs.chemrev.8b00138] [Citation(s) in RCA: 270] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Masha G. Savelieff
- SciGency Science Communications, Ann Arbor, Michigan 48104, United States
| | - Geewoo Nam
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Juhye Kang
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Hyuck Jin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Misun Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
73
|
Wong SQ, Pontifex MG, Phelan MM, Pidathala C, Kraemer BC, Barclay JW, Berry NG, O'Neill PM, Burgoyne RD, Morgan A. α-Methyl-α-phenylsuccinimide ameliorates neurodegeneration in a C. elegans model of TDP-43 proteinopathy. Neurobiol Dis 2018; 118:40-54. [PMID: 29940336 PMCID: PMC6097874 DOI: 10.1016/j.nbd.2018.06.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/06/2018] [Accepted: 06/15/2018] [Indexed: 12/12/2022] Open
Abstract
The antiepileptic drug ethosuximide has recently been shown to be neuroprotective in various Caenorhabditis elegans and rodent neurodegeneration models. It is therefore a promising repurposing candidate for the treatment of multiple neurodegenerative diseases. However, high concentrations of the drug are required for its protective effects in animal models, which may impact on its translational potential and impede the identification of its molecular mechanism of action. Therefore, we set out to develop more potent neuroprotective lead compounds based on ethosuximide as a starting scaffold. Chemoinformatic approaches were used to identify compounds with structural similarity to ethosuximide and to prioritise these based on good predicated blood-brain barrier permeability and C. elegans bioaccumulation properties. Selected compounds were initially screened for anti-convulsant activity in a C. elegans pentylenetetrazol-induced seizure assay, as a rapid primary readout of bioactivity; and then assessed for neuroprotective properties in a C. elegans TDP-43 proteinopathy model based on pan-neuronal expression of human A315T mutant TDP-43. The most potent compound screened, α-methyl-α-phenylsuccinimide (MPS), ameliorated the locomotion defects and extended the shortened lifespan of TDP-43 mutant worms. MPS also directly protected against neurodegeneration by reducing the number of neuronal breaks and cell body losses in GFP-labelled GABAergic motor neurons. Importantly, optimal neuroprotection was exhibited by external application of 50 μM MPS, compared to 8 mM for ethosuximide. This greater potency of MPS was not due to bioaccumulation to higher internal levels within the worm, based on 1H-nuclear magnetic resonance analysis. Like ethosuximide, the activity of MPS was abolished by mutation of the evolutionarily conserved FOXO transcription factor, daf-16, suggesting that both compounds act via the same neuroprotective pathway(s). In conclusion, we have revealed a novel neuroprotective activity of MPS that is >100-fold more potent than ethosuximide. This increased potency will facilitate future biochemical studies to identify the direct molecular target(s) of both compounds, as we have shown here that they share a common downstream DAF-16-dependent mechanism of action. Furthermore, MPS is the active metabolite of another approved antiepileptic drug, methsuximide. Therefore, methsuximide may have repurposing potential for treatment of TDP-43 proteinopathies and possibly other human neurodegenerative diseases.
Collapse
Affiliation(s)
- Shi Quan Wong
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Matthew G Pontifex
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Marie M Phelan
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK.
| | | | - Brian C Kraemer
- Geriatrics Research Education and Clinical Center, Seattle Veterans Affairs Puget Sound Health Care System, University of Washington Department of Medicine, Seattle, WA 98108, USA.
| | - Jeff W Barclay
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Neil G Berry
- Department of Chemistry, University of Liverpool, Liverpool, UK.
| | - Paul M O'Neill
- Department of Chemistry, University of Liverpool, Liverpool, UK.
| | - Robert D Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Alan Morgan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
74
|
Berté TE, Dalmagro AP, Zimath PL, Gonçalves AE, Meyre-Silva C, Bürger C, Weber CJ, Dos Santos DA, Cechinel-Filho V, de Souza MM. Taraxerol as a possible therapeutic agent on memory impairments and Alzheimer's disease: Effects against scopolamine and streptozotocin-induced cognitive dysfunctions. Steroids 2018; 132:5-11. [PMID: 29355563 DOI: 10.1016/j.steroids.2018.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 12/19/2017] [Accepted: 01/11/2018] [Indexed: 01/23/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with cognitive impairment and cholinergic neuronal death, characteristic of the effect of time on biochemical neuronal function. The use of medicinal plants as an alternative form of prevention, or even as a possible treatment of AD, is therefore interesting areas of research, since the standard drugs have many side effects. Taraxerol (TRX) is a triterpene that has been isolated from several plant species, and its various pharmacological properties have already been identified, such the acetylcholinesterase (AChE) inhibition activity in vitro. There is a lack of information in literature that confirms the effect of TRX in an animal AD-like model. Seeking to fill this gap in the literature, in the present work we assessed the effect of TRX on AChE activity in the animals' encephalon and hippocampus. We also investigated the effect of TRX (1.77 µM/side, 0.5 μL) isolated from leaves of Eugenia umbelliflora Berg. on aversive memory impairments induced by scopolamine (2 µg/side, 0.5 µL) infused into rat hippocampus, and the effect of TRX (0.89 and 1.77 µM/side, 0.5 μL) on aversive memory impairments induced by streptozotocin (STZ) (2.5 mg/mL, 2.0 µL) infused i.c.v. into mice, using the step-down inhibitory avoidance task. We found that TRX significantly inhibited AChE activity in the animal's hippocampus. Furthermore, TRX significantly improved scopolamine and STZ-induced memory impairment. Taking together, these results confirms its AChE activity inhibition in animals and indicate that TRX has anti-amnesic activity that may hold significant therapeutic value in alleviating certain memory impairments observed in AD.
Collapse
Affiliation(s)
- Talita Elisa Berté
- Centro de Ciências da Saúde, CCS - Núcleo de Investigações Químico Farmacêuticas NIQFAR/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil
| | - Ana Paula Dalmagro
- Centro de Ciências da Saúde, CCS - Núcleo de Investigações Químico Farmacêuticas NIQFAR/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil.
| | - Priscila Laiz Zimath
- Centro de Ciências da Saúde, CCS - Núcleo de Investigações Químico Farmacêuticas NIQFAR/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil
| | - Ana Elisa Gonçalves
- Centro de Ciências da Saúde, CCS - Núcleo de Investigações Químico Farmacêuticas NIQFAR/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil
| | - Christiane Meyre-Silva
- Centro de Ciências da Saúde, CCS - Núcleo de Investigações Químico Farmacêuticas NIQFAR/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil
| | - Cristiani Bürger
- Centro de Ciências da Saúde, CCS - Núcleo de Investigações Químico Farmacêuticas NIQFAR/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil
| | - Carla J Weber
- Centro de Ciências da Saúde, CCS - Núcleo de Investigações Químico Farmacêuticas NIQFAR/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil
| | - Diogo Adolfo Dos Santos
- Centro de Ciências da Saúde, CCS - Núcleo de Investigações Químico Farmacêuticas NIQFAR/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil
| | - Valdir Cechinel-Filho
- Centro de Ciências da Saúde, CCS - Núcleo de Investigações Químico Farmacêuticas NIQFAR/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil
| | - Márcia M de Souza
- Centro de Ciências da Saúde, CCS - Núcleo de Investigações Químico Farmacêuticas NIQFAR/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas/UNIVALI, Rua Uruguai 458, Centro, CEP: 88302-202 Itajaí, SC, Brazil
| |
Collapse
|
75
|
Tenreiro S, Franssens V, Winderickx J, Outeiro TF. Yeast models of Parkinson's disease-associated molecular pathologies. Curr Opin Genet Dev 2018; 44:74-83. [PMID: 28232272 DOI: 10.1016/j.gde.2017.01.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/30/2017] [Indexed: 12/15/2022]
Abstract
The aging of the human population is resulting in an increase in the number of people afflicted by neurodegenerative disorders such as Parkinson's disease (PD), creating tremendous socio-economic challenges. This requires the urgent for the development of effective therapies, and of tools for early diagnosis of the disease. However, our understanding of the molecular mechanisms underlying PD pathogenesis is still incomplete, hampering progress in those areas. In recent years, the progression made in genetics has considerably contributed to our knowledge, by identifying several novel PD genes. Furthermore, many cellular and animal models have proven their value to decipher pathways involved in PD development. In this review we highlight the value of the yeast Saccharomyces cerevisiae as a model for PD. This unicellular eukaryote has contributed to our understanding of the cellular mechanisms targeted by most important PD genes and offers an excellent tool for discovering novel players via powerful and informative high throughput screens that accelerate further validation in more complex models.
Collapse
Affiliation(s)
- Sandra Tenreiro
- CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Vanessa Franssens
- Department of Biology, Functional Biology, KU Leuven, 3001 Heverlee, Belgium
| | - Joris Winderickx
- Department of Biology, Functional Biology, KU Leuven, 3001 Heverlee, Belgium
| | - Tiago Fleming Outeiro
- CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal; Department of Neurodegeneration and Restorative Research, University Medical Center Goettingen, Goettingen, Germany.
| |
Collapse
|
76
|
Chongtham A, Barbaro B, Filip T, Syed A, Huang W, Smith MR, Marsh JL. Nonmammalian Models of Huntington's Disease. Methods Mol Biol 2018; 1780:75-96. [PMID: 29856015 DOI: 10.1007/978-1-4939-7825-0_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Flies, worms, yeast and more recently zebra fish have all been engineered to express expanded polyglutamine repeat versions of Huntingtin with various resulting pathologies including early death, neurodegeneration, and loss of motor function. Each of these models present particular features that make it useful in studying the mechanisms of polyglutamine pathology. However, one particular unbiased readout of mHTT pathology is functional loss of motor control. Loss of motor control is prominent in patients, but it remains unresolved whether pathogenic symptoms in patients result from overt degeneration and loss of neurons or from malfunctioning of surviving neurons as the pathogenic insult builds up. This is why a functional assay such as motor control can be uniquely powerful in revealing early as well as late neurological deficits and does not rely on assumptions such as that the level of inclusions or the degree of neuronal loss can be equated with the level of pathology. Drosophila is well suited for such assays because it contains a functioning nervous system with many parallels to the human condition. In addition, the ability to readily express mHTT transgenes in different tissues and subsets of neurons allows one the possibility of isolating a particular effect to a subset of neurons where one can correlate subcellular events in response to mHTT challenge with pathology at both the cellular and organismal levels. Here we describe methods to monitor the degree of motor function disruption in Drosophila models of HD and we include a brief summary of other nonmammalian models of HD and discussion of their unique strengths.
Collapse
Affiliation(s)
- Anjalika Chongtham
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, 92697, CA, USA
| | - Brett Barbaro
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, 92697, CA, USA.,The Scripps Research Institute, La Jolla, CA, USA
| | - Tomas Filip
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, 92697, CA, USA.,Biology Centre Czech Acad. Sci., Ceske Budejovice, Czech Republic
| | - Adeela Syed
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, 92697, CA, USA
| | - Weijian Huang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, 92697, CA, USA
| | - Marianne R Smith
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, 92697, CA, USA.,University Advancement, UC Irvine, Irvine, CA, USA
| | - J Lawrence Marsh
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, 92697, CA, USA.
| |
Collapse
|
77
|
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide. It is known that there are many factors, either genetic or environmental factors, involved in PD, but the mechanism of PD is still not fully understood. Several animal models have been established to study the mechanisms of PD. Among these models, Drosophila melanogaster has been utilized as a valuable model to get insight into important features of PD. Drosophila melanogaster possesses a well-developed dopaminergic (DA) neuron system which is known to play an important role in PD pathogenesis. The well understanding of DA neurons from early larval through adult stage makes Drosophila as a powerful model for investigating the progressive neurodegeneration in PD. Besides, the short life cycle of Drosophila melanogaster serves an advantage in studying epidemiological features of PD. Most of PD symptoms can be mimicked in Drosophila model such as progressive impairment in locomotion, DA neuron degeneration, and some other non-motor symptoms. The Drosophila models of PD, therefore, show a great potential in application for PD genetic and drug screening.
Collapse
Affiliation(s)
- Vuu My Dung
- University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | | |
Collapse
|
78
|
Brunquell J, Morris S, Snyder A, Westerheide SD. Coffee extract and caffeine enhance the heat shock response and promote proteostasis in an HSF-1-dependent manner in Caenorhabditis elegans. Cell Stress Chaperones 2018; 23:65-75. [PMID: 28674941 PMCID: PMC5741582 DOI: 10.1007/s12192-017-0824-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 06/12/2017] [Accepted: 06/14/2017] [Indexed: 01/05/2023] Open
Abstract
As the population ages, there is a critical need to uncover strategies to combat diseases of aging. Studies in the soil-dwelling nematode Caenorhabditis elegans have demonstrated the protective effects of coffee extract and caffeine in promoting the induction of conserved longevity pathways including the insulin-like signaling pathway and the oxidative stress response. We were interested in determining the effects of coffee and caffeine treatment on the regulation of the heat shock response. The heat shock response is a highly conserved cellular response that functions as a cytoprotective mechanism during stress, mediated by the heat shock transcription factor HSF-1. In the worm, HSF-1 not only promotes protection against stress but is also essential for development and longevity. Induction of the heat shock response has been suggested to be beneficial for diseases of protein conformation by preventing protein misfolding and aggregation, and as such has been proposed as a therapeutic target for age-associated neurodegenerative disorders. In this study, we demonstrate that coffee is a potent, dose-dependent, inducer of the heat shock response. Treatment with a moderate dose of pure caffeine was also able to induce the heat shock response, indicating caffeine as an important component within coffee for producing this response. The effects that we observe with both coffee and pure caffeine on the heat shock response are both dependent on HSF-1. In a C. elegans Huntington's disease model, worms treated with caffeine were protected from polyglutamine aggregates and toxicity, an effect that was also HSF-1-dependent. In conclusion, these results demonstrate caffeinated coffee, and pure caffeine, as protective substances that promote proteostasis through induction of the heat shock response.
Collapse
Affiliation(s)
- Jessica Brunquell
- Department of Cell Biology, Microbiology, and Molecular Biology, College of Arts and Sciences, University of South Florida, 4202 E. Fowler Ave, ISA 2015, Tampa, FL, 33620, USA
| | - Stephanie Morris
- Department of Cell Biology, Microbiology, and Molecular Biology, College of Arts and Sciences, University of South Florida, 4202 E. Fowler Ave, ISA 2015, Tampa, FL, 33620, USA
| | - Alana Snyder
- Department of Cell Biology, Microbiology, and Molecular Biology, College of Arts and Sciences, University of South Florida, 4202 E. Fowler Ave, ISA 2015, Tampa, FL, 33620, USA
| | - Sandy D Westerheide
- Department of Cell Biology, Microbiology, and Molecular Biology, College of Arts and Sciences, University of South Florida, 4202 E. Fowler Ave, ISA 2015, Tampa, FL, 33620, USA.
| |
Collapse
|
79
|
Strange K. Drug Discovery in Fish, Flies, and Worms. ILAR J 2017; 57:133-143. [PMID: 28053067 DOI: 10.1093/ilar/ilw034] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 10/21/2016] [Indexed: 12/22/2022] Open
Abstract
Nonmammalian model organisms such as the nematode Caenorhabditis elegans, the fruit fly Drosophila melanogaster, and the zebrafish Danio rerio provide numerous experimental advantages for drug discovery including genetic and molecular tractability, amenability to high-throughput screening methods and reduced experimental costs and increased experimental throughput compared to traditional mammalian models. An interdisciplinary approach that strategically combines the study of nonmammalian and mammalian animal models with diverse experimental tools has and will continue to provide deep molecular and genetic understanding of human disease and will significantly enhance the discovery and application of new therapies to treat those diseases. This review will provide an overview of C. elegans, Drosophila, and zebrafish biology and husbandry and will discuss how these models are being used for phenotype-based drug screening and for identification of drug targets and mechanisms of action. The review will also describe how these and other nonmammalian model organisms are uniquely suited for the discovery of drug-based regenerative medicine therapies.
Collapse
Affiliation(s)
- Kevin Strange
- Kevin Strange, Ph.D., is President and CEO of the MDI Biological Laboratory and CEO of Novo Biosciences, Inc
| |
Collapse
|
80
|
Ma L, Zhao Y, Chen Y, Cheng B, Peng A, Huang K. Caenorhabditis elegans as a model system for target identification and drug screening against neurodegenerative diseases. Eur J Pharmacol 2017; 819:169-180. [PMID: 29208474 DOI: 10.1016/j.ejphar.2017.11.051] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/30/2017] [Indexed: 12/12/2022]
Abstract
Over the past decades, Caenorhabditis elegans (C. elegans) has been widely used as a model system because of its small size, transparent body, short generation time and lifespan (~3 days and 3 weeks, respectively), completely sequenced genome and tractability to genetic manipulation. Protein misfolding and aggregation are key pathological features in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease and Amyotrophic lateral sclerosis. Animal models, including C. elegans, have been extensively used to discover and validate new drugs against neurodegenerative diseases. The well-defined and genetically tractable nervous system of C. elegans offers an effective model to explore basic mechanistic pathways of neurodegenerative diseases. Recent progress in high-throughput drug screening also provides a powerful approach for identifying chemical modulators of biological processes. Here, we summarize the latest progress of using C. elegans as a model system for target identification and drug screening in neurodegenerative diseases.
Collapse
Affiliation(s)
- Liang Ma
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yudan Zhao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuchen Chen
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Biao Cheng
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Anlin Peng
- Department of Pharmacy, The Third Hospital of Wuhan, Wuhan 430060, China
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China; Center for Biomedicine Research, Wuhan Institute of Biotechnology, Wuhan 430075, China.
| |
Collapse
|
81
|
Wei CC, Chang CH, Liao VHC. Anti-Parkinsonian effects of β-amyrin are regulated via LGG-1 involved autophagy pathway in Caenorhabditis elegans. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 36:118-125. [PMID: 29157804 DOI: 10.1016/j.phymed.2017.09.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/11/2017] [Accepted: 09/19/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disease that is associated with aging and is characterized as a movement disorder. Currently, there is still no complete therapy for PD. In recent years, the identification and characterization of medicinal plants to cure or treat PD has gained increasing scientific interest. PURPOSE In this study, we investigated a pentacyclic triterpenoid compound, β-amyrin, which is found in many medicinal plants for its anti-Parkinsonian effects, using Caenorhabditis elegans (C. elegans) disease models and their underlying mechanisms. METHODS C. elegans treated or untreated with β-amyrin were investigated for oxidative stress resistance, neurodegeneration, and α-synuclein aggregation assays. The C. elegans ortholog of Atg8/LC3, LGG-1 that is involved in the autophagy pathway was also evaluated by quantitative RT-PCR and transgenic strain experiments. RESULTS β-Amyrin exerted excellent antioxidant activity and reduced intracellular oxygen species in C. elegans. Using the transgenic strain BZ555, β-amyrin showed a protective effect on dopaminergic neurons reducing cell damage induced by 6-hydroxydopamine (6-OHDA). In addition, β-amyrin significantly reduced the α-synuclein aggregation in the transgenic strain NL5901. Moreover, β-amyrin up-regulated LGG-1 mRNA expression and increased the number of localized LGG-1 puncta in the transgenic strain DA2123. CONCLUSION The results from this study suggest that the anti-Parkinsonian effects of β-amyrin might be regulated via LGG-1 involved autophagy pathway in C. elegans. Therefore, β-amyrin may be useful for therapeutic applications or supplements to treat or slow the progression of PD.
Collapse
Affiliation(s)
- Chia-Cheng Wei
- Department of Bioenvironmental Systems Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 106, Taiwan
| | - Chun-Han Chang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 106, Taiwan
| | - Vivian Hsiu-Chuan Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 106, Taiwan.
| |
Collapse
|
82
|
Cornaglia M, Lehnert T, Gijs MAM. Microfluidic systems for high-throughput and high-content screening using the nematode Caenorhabditis elegans. LAB ON A CHIP 2017; 17:3736-3759. [PMID: 28840220 DOI: 10.1039/c7lc00509a] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In a typical high-throughput drug screening (HTS) process, up to millions of chemical compounds are applied to cells cultured in well plates, aiming to find molecules that exhibit a robust dose-response, as evidenced for example by a fluorescence signal. In high-content screening (HCS), one goes a step further by linking the tested compounds to phenotypic information, obtained, for instance, from microscopic cell images, thereby creating richer data sets that also require more advanced analysis methods. The nematode Caenorhabditis elegans came into the screening picture due to the wide availability of its mutants and human disease models, its relatively easy culture and short life cycle. Being a whole-organism model, it allows drug testing under physiological conditions at multi-tissue levels and provides additional observable phenotypes with respect to cell models, related, for instance, to development, aging, behavior or motility. Worm-based HTS studies in liquid environments on microwell plates have been demonstrated, while microfluidic devices allowed surpassing the performance of plates by enabling more versatile and accurate assays, precise and dynamic dosing of compounds, and readouts down to single-animal resolution. In this review, we discuss microfluidic devices for C. elegans analysis and related studies, published in the period from 2012 to 2017. After an introduction to the different screening approaches, we first focus on microfluidic systems with potential for screening applications. Various enabling technologies, e.g. electrophysiological on-chip recordings or laser axotomy, have been implemented, as well as techniques for reversible worm immobilization and high-resolution imaging, combined with algorithms for automated experimentation and analysis. Several devices for developmental or behavioral assays, and worm sorting based on different phenotypes, have been proposed too. In a subsequent section, we review the application of microfluidic-based systems for medium- and high-throughput screens, including neurobiology and neurodegeneration studies, aging and developmental assays, toxicity and pathogenesis screens, as well as behavioral and motility assays. A thorough analysis of this work reveals a trend towards microfluidic systems more and more capable of offering high-quality analyses of large worm populations, based on multi-phenotypic and/or longitudinal readouts, with clear potential for their application in larger HTS/HCS contexts.
Collapse
Affiliation(s)
- Matteo Cornaglia
- Laboratory of Microsystems, École Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland.
| | | | | |
Collapse
|
83
|
Sonane M, Moin N, Satish A. The role of antioxidants in attenuation of Caenorhabditis elegans lethality on exposure to TiO 2 and ZnO nanoparticles. CHEMOSPHERE 2017; 187:240-247. [PMID: 28854380 DOI: 10.1016/j.chemosphere.2017.08.080] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/05/2017] [Accepted: 08/16/2017] [Indexed: 05/26/2023]
Abstract
The exponential increase in the usage of engineered nanoparticles (ENPs) has raised global concerns due to their potential toxicity and environmental impacts. Nano-TiO2 and nano-ZnO have been extensively used in various applications. Thus, there is a need for determining the toxic potentials of ENPs as well as, to develop the possible attenuation method for ENPs toxicity. Both in the in vitro and in vivo systems, exposure to the majority of ENPs have shown Reactive Oxygen Species (ROS) generation, which leads to oxidative stress mediated inflammation, genotoxicity, and cytotoxicity. Hence, with the rationale of determining easy and economical protection against ENPs exposure, the amelioration effect of the antioxidants (curcumin and vitamin-C) against the nano-TiO2 and nano-ZnO induced ROS and lethality were investigated in Caenorhabditis elegans. We not only employed pre-treatment and along with treatment approach, but also determined the effect of antioxidants at different time points of treatment. Our study revealed that both the antioxidants efficiently ameliorate nanoparticles induced ROS as well as lethality in worms. Further, the pretreatment approach was more effective than the along with treatment. Therefore, our study indicates the possibility of evading the nanotoxicity by incorporating curcumin and vitamin-C in everyday diet.
Collapse
Affiliation(s)
- Madhavi Sonane
- Ecotoxicology Laboratory, Nanotherapeutics & Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, M.G. Marg, Post Box-80, Lucknow 226 001, Uttar Pradesh, India; Department of Biochemistry, Babu Banarasi Das University, Lucknow 227015, India
| | - Nida Moin
- Ecotoxicology Laboratory, Nanotherapeutics & Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, M.G. Marg, Post Box-80, Lucknow 226 001, Uttar Pradesh, India; Department of Biochemistry, Babu Banarasi Das University, Lucknow 227015, India
| | - Aruna Satish
- Ecotoxicology Laboratory, Nanotherapeutics & Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, M.G. Marg, Post Box-80, Lucknow 226 001, Uttar Pradesh, India.
| |
Collapse
|
84
|
Manalo RV, Silvestre MA, Barbosa ALA, Medina PM. Coconut (Cocos nucifera) Ethanolic Leaf Extract Reduces Amyloid-β (1-42) Aggregation and Paralysis Prevalence in Transgenic Caenorhabditis elegans Independently of Free Radical Scavenging and Acetylcholinesterase Inhibition. Biomedicines 2017; 5:biomedicines5020017. [PMID: 28536360 PMCID: PMC5489803 DOI: 10.3390/biomedicines5020017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 04/17/2017] [Accepted: 04/17/2017] [Indexed: 01/15/2023] Open
Abstract
Virgin coconut oil (VCO) has been the subject of several studies which have aimed to alleviate Alzheimer’s disease (AD) pathology, focusing on in vitro antioxidant and acetylcholinesterase (AChE) inhibitory activities. Here, we studied an underutilized and lesser-valued part of the coconut tree, specifically the leaves, using in vitro and in vivo approaches. Coconut leaf extract (CLE) was screened for antioxidant and AChE inhibitory properties in vitro and therapeutic effects in two strains of transgenic Caenorhabditis elegans expressing amyloid-β1–42 (Aβ1-42) in muscle cells. CLE demonstrated free radical scavenging activity with an EC50 that is 79-fold less compared to ascorbic acid, and an AChE inhibitory activity that is 131-fold less compared to Rivastigmine. Surprisingly, in spite of its low antioxidant activity and AChE inhibition, CLE reduced Aβ deposits by 30.31% in CL2006 in a dose-independent manner, and reduced the percentage of paralyzed nematodes at the lowest concentration of CLE (159.38 μg/mL), compared to dH2O/vehicle (control). Phytochemical analysis detected glycosides, anthocyanins, and hydrolyzable tannins in CLE, some of which are known to be anti-amyloidogenic. Taken together, these findings suggest that CLE metabolites alternatively decrease AB1–42 aggregation and paralysis prevalence independently of free radical scavenging and AChE inhibition, and this warrants further investigation on the bioactive compounds of CLE.
Collapse
Affiliation(s)
- Rafael Vincent Manalo
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Ermita, Manila 1000, Philippines.
| | | | | | - Paul Mark Medina
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Ermita, Manila 1000, Philippines.
| |
Collapse
|
85
|
Sunderhaus ER, Kretzschmar D. Mass Histology to Quantify Neurodegeneration in Drosophila. J Vis Exp 2016. [PMID: 28060320 DOI: 10.3791/54809] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Progressive neurodegenerative diseases like Alzheimer's disease (AD) or Parkinson's disease (PD) are an increasing threat to human health worldwide. Although mammalian models have provided important insights into the underlying mechanisms of pathogenicity, the complexity of mammalian systems together with their high costs are limiting their use. Therefore, the simple but well-established Drosophila model-system provides an alternative for investigating the molecular pathways that are affected in these diseases. Besides behavioral deficits, neurodegenerative diseases are characterized by histological phenotypes such as neuronal death and axonopathy. To quantify neuronal degeneration and to determine how it is affected by genetic and environmental factors, we use a histological approach that is based on measuring the vacuoles in adult fly brains. To minimize the effects of systematic error and to directly compare sections from control and experimental flies in one preparation, we use the 'collar' method for paraffin sections. Neurodegeneration is then assessed by measuring the size and/or number of vacuoles that have developed in the fly brain. This can either be done by focusing on a specific region of interest or by analyzing the entire brain by obtaining serial sections that span the complete head. Therefore, this method allows one to measure not only severe degeneration but also relatively mild phenotypes that are only detectable in a few sections, as occurs during normal aging.
Collapse
Affiliation(s)
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, Oregon Health & Sciences University;
| |
Collapse
|
86
|
Grüner BM, Schulze CJ, Yang D, Ogasawara D, Dix MM, Rogers ZN, Chuang CH, McFarland CD, Chiou SH, Brown JM, Cravatt BF, Bogyo M, Winslow MM. An in vivo multiplexed small-molecule screening platform. Nat Methods 2016; 13:883-889. [PMID: 27617390 DOI: 10.1038/nmeth.3992] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 08/01/2016] [Indexed: 01/04/2023]
Abstract
Phenotype-based small-molecule screening is a powerful method to identify molecules that regulate cellular functions. However, such screens are generally performed in vitro under conditions that do not necessarily model complex physiological conditions or disease states. Here, we use molecular cell barcoding to enable direct in vivo phenotypic screening of small-molecule libraries. The multiplexed nature of this approach allows rapid in vivo analysis of hundreds to thousands of compounds. Using this platform, we screened >700 covalent inhibitors directed toward hydrolases for their effect on pancreatic cancer metastatic seeding. We identified multiple hits and confirmed the relevant target of one compound as the lipase ABHD6. Pharmacological and genetic studies confirmed the role of this enzyme as a regulator of metastatic fitness. Our results highlight the applicability of this multiplexed screening platform for investigating complex processes in vivo.
Collapse
Affiliation(s)
- Barbara M Grüner
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Dian Yang
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Daisuke Ogasawara
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Melissa M Dix
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Zoë N Rogers
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Chen-Hua Chuang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Shin-Heng Chiou
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - J Mark Brown
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Benjamin F Cravatt
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.,Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
87
|
Link P, Roth K, Sporer F, Wink M. Carlina acaulis Exhibits Antioxidant Activity and Counteracts Aβ Toxicity in Caenorhabditis elegans. Molecules 2016; 21:molecules21070871. [PMID: 27384550 PMCID: PMC6273941 DOI: 10.3390/molecules21070871] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/18/2016] [Accepted: 06/28/2016] [Indexed: 01/03/2023] Open
Abstract
Carlina acaulis is a medicinal plant that has shown antioxidant activity in in vitro studies, but to date no corresponding in vivo data is available. Therefore, in the present study the antioxidant activity and its impact in counteracting Aβ toxicity were studied in the Caenorhabditis elegans model. A dichloromethane extract of the roots of C. acaulis was prepared and characterised via gas-liquid-chromatography/mass-spectrometry (GLC-MS). The in vitro antioxidant activity was confirmed via 2,2-diphenyl-1-picrylhydracyl assay. The extract was further separated by thin layer chromatography into two fractions, one of which was a fraction of the dichloromethane extract of C. acaulis containing mostly Carlina oxide (CarOx). Different strains of C. elegans were employed to study the expression of hsp-16.2p::GFP as a marker for oxidative stress, delocalisation of the transcription factor DAF-16 as a possible mechanism of antioxidant activity, the effect of the drug under lethal oxidative stress, and the effect against beta-amyloid (Aβ) toxicity in a paralysis assay. The C. acaulis extract and CarOx showed high antioxidant activity (stress reduction by 47% and 64%, respectively) in C. elegans and could activate the transcription factor DAF-16 which directs the expression of anti-stress genes. In paralysis assay, only the total extract was significantly active, delaying paralysis by 1.6 h. In conclusion, in vivo antioxidant activity was shown for C. acaulis for the first time in the C. elegans model. The active antioxidant compound is Carlina oxide. This activity, however, is not sufficient to counteract Aβ toxicity. Other mechanisms and possibly other active compounds are involved in this effect.
Collapse
Affiliation(s)
- Pille Link
- Institute of Pharmacy and Molecular Biology, Heidelberg University, Heidelberg 69120, Germany.
| | - Kevin Roth
- Institute of Pharmacy and Molecular Biology, Heidelberg University, Heidelberg 69120, Germany.
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, Scotland DD1 9SY, UK.
| | - Frank Sporer
- Institute of Pharmacy and Molecular Biology, Heidelberg University, Heidelberg 69120, Germany.
| | - Michael Wink
- Institute of Pharmacy and Molecular Biology, Heidelberg University, Heidelberg 69120, Germany.
| |
Collapse
|