1
|
Dos Santos E, Cochemé HM. Pharmacology of Aging: Drosophila as a Tool to Validate Drug Targets for Healthy Lifespan. AGING BIOLOGY 2024; 2:20240034. [PMID: 39346601 PMCID: PMC7616647 DOI: 10.59368/agingbio.20240034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Finding effective therapies to manage age-related conditions is an emerging public health challenge. Although disease-targeted treatments are important, a preventive approach focused on aging can be more efficient. Pharmacological targeting of aging-related processes can extend lifespan and improve health in animal models. However, drug development and translation are particularly challenging in geroscience. Preclinical studies have survival as a major endpoint for drug screening, which requires years of research in mammalian models. Shorter-lived invertebrates can be exploited to accelerate this process. In particular, the fruit fly Drosophila melanogaster allows the validation of new drug targets using precise genetic tools and proof-of-concept experiments on drugs impacting conserved aging processes. Screening for clinically approved drugs that act on aging-related targets may further accelerate translation and create new tools for aging research. To date, 31 drugs used in clinical practice have been shown to extend the lifespan of flies. Here, we describe recent advances in the pharmacology of aging, focusing on Drosophila as a tool to repurpose these drugs and study age-related processes.
Collapse
Affiliation(s)
- Eliano Dos Santos
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Helena M Cochemé
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
2
|
He S, Ru Q, Chen L, Xu G, Wu Y. Advances in animal models of Parkinson's disease. Brain Res Bull 2024; 215:111024. [PMID: 38969066 DOI: 10.1016/j.brainresbull.2024.111024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
Parkinson's disease is a complex neurodegenerative disease characterized by progressive movement impairments. Predominant symptoms encompass resting tremor, bradykinesia, limb rigidity, and postural instability. In addition, it also includes a series of non-motor symptoms such as sleep disorders, hyposmia, gastrointestinal dysfunction, autonomic dysfunction and cognitive impairment. Pathologically, the disease manifests through dopaminergic neuronal loss and the presence of Lewy bodies. At present, no significant breakthrough has been achieved in clinical Parkinson's disease treatment. Exploring treatment modalities necessitate the establishment of scientifically sound animal models. In recent years, researchers have focused on replicating the symptoms of human Parkinson's disease, resulting in the establishment of various experimental animal models primarily through drugs and transgenic methods to mimic relevant pathologies and identify more effective treatments. This review examines traditional neurotoxin and transgenic animal models as well as α-synuclein pre-formed fibrils models, non-human primate models and non-mammalian specie models. Additionally, it introduces emerging models, including models based on optogenetics, induced pluripotent stem cells, and gene editing, aiming to provide a reference for the utilization of experimental animal models and clinical research for researchers in this field.
Collapse
Affiliation(s)
- Sui He
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
3
|
Nainu F, Sartini S, Bahar MA, Asbah A, Rosa RA, Mudjahid M, As'ad MF, Latada NP. Anti-aging and immunomodulatory role of caffeine in Drosophila larvae. NARRA J 2024; 4:e818. [PMID: 39280322 PMCID: PMC11391967 DOI: 10.52225/narra.v4i2.818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 06/26/2024] [Indexed: 09/18/2024]
Abstract
Drug repurposing is a promising approach to identify new pharmacological indications for drugs that have already been established. However, there is still a limitation in the availability of a high-throughput in vivo preclinical system that is suitable for screening and investigating new pharmacological indications. The aim of this study was to introduce the application of Drosophila larvae as an in vivo platform to screen drug candidates with anti-aging and immunomodulatory activities. To determine whether Drosophila larvae can be utilized for assessing anti-aging and immunomodulatory activities, phenotypical and molecular assays were conducted using wildtype and mutant lines of Drosophila. The utilization of mutant lines (PGRP-LBΔ and Psh[1];;ModSP[KO]) mimics the autoinflammatory and immunodeficient conditions in humans, thereby enabling a thorough investigation of the effects of various compounds. The phenotypical assay was carried out using survival and locomotor observation in Drosophila larvae and adult flies. Meanwhile, the molecular assay was conducted using the RT-qPCR method. In vivo survival analysis revealed that caffeine was relatively safe for Drosophila larvae and exhibited the ability to extend Drosophila lifespan compared to the untreated controls, suggesting its anti-aging properties. Further analysis using the RT-qPCR method demonstrated that caffeine treatment induced transcriptional changes in the Drosophila larvae, particularly in the downstream of NF-κB and JAK-STAT pathways, two distinct immune-related pathways homologue to humans. In addition, caffeine enhanced the survival of Drosophila autoinflammatory model, further implying its immunosuppressive activity. Nevertheless, this compound had minimal to no effect on the survival of Staphylococcus aureus-infected wildtype and immunodeficient Drosophila, refuting its antibacterial and immunostimulant activities. Overall, our results suggest that the anti-aging and immunosuppressive activities of caffeine observed in Drosophila larvae align with those reported in mammalian model systems, emphasizing the suitability of Drosophila larvae as a model organism in drug repurposing endeavors, particularly for the screening of newly discovered chemical entities to assess their immunomodulatory activities before proceedings to investigations in mammalian animal models.
Collapse
Affiliation(s)
- Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Sartini Sartini
- Department of Pharmaceutical Science and Technology, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Muhammad A Bahar
- Department of Pharmacy, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Asbah Asbah
- Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Reski A Rosa
- Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Mukarram Mudjahid
- Department of Pharmacy, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| | - Muhammad F As'ad
- Postgraduate Program in Pharmacy, Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
- Pelamonia Health Sciences Institute, Makassar, Indonesia
| | - Nadila P Latada
- Faculty of Pharmacy, Universitas Hasanuddin, Makassar, Indonesia
| |
Collapse
|
4
|
Petitgas C, Seugnet L, Dulac A, Matassi G, Mteyrek A, Fima R, Strehaiano M, Dagorret J, Chérif-Zahar B, Marie S, Ceballos-Picot I, Birman S. Metabolic and neurobehavioral disturbances induced by purine recycling deficiency in Drosophila. eLife 2024; 12:RP88510. [PMID: 38700995 PMCID: PMC11068357 DOI: 10.7554/elife.88510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024] Open
Abstract
Adenine phosphoribosyltransferase (APRT) and hypoxanthine-guanine phosphoribosyltransferase (HGPRT) are two structurally related enzymes involved in purine recycling in humans. Inherited mutations that suppress HGPRT activity are associated with Lesch-Nyhan disease (LND), a rare X-linked metabolic and neurological disorder in children, characterized by hyperuricemia, dystonia, and compulsive self-injury. To date, no treatment is available for these neurological defects and no animal model recapitulates all symptoms of LND patients. Here, we studied LND-related mechanisms in the fruit fly. By combining enzymatic assays and phylogenetic analysis, we confirm that no HGPRT activity is expressed in Drosophila melanogaster, making the APRT homolog (Aprt) the only purine-recycling enzyme in this organism. Whereas APRT deficiency does not trigger neurological defects in humans, we observed that Drosophila Aprt mutants show both metabolic and neurobehavioral disturbances, including increased uric acid levels, locomotor impairments, sleep alterations, seizure-like behavior, reduced lifespan, and reduction of adenosine signaling and content. Locomotor defects could be rescued by Aprt re-expression in neurons and reproduced by knocking down Aprt selectively in the protocerebral anterior medial (PAM) dopaminergic neurons, the mushroom bodies, or glia subsets. Ingestion of allopurinol rescued uric acid levels in Aprt-deficient mutants but not neurological defects, as is the case in LND patients, while feeding adenosine or N6-methyladenosine (m6A) during development fully rescued the epileptic behavior. Intriguingly, pan-neuronal expression of an LND-associated mutant form of human HGPRT (I42T), but not the wild-type enzyme, resulted in early locomotor defects and seizure in flies, similar to Aprt deficiency. Overall, our results suggest that Drosophila could be used in different ways to better understand LND and seek a cure for this dramatic disease.
Collapse
Affiliation(s)
- Céline Petitgas
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
- Metabolomic and Proteomic Biochemistry Laboratory, Necker-Enfants Malades Hospital and Paris Cité UniversityParisFrance
| | - Laurent Seugnet
- Integrated Physiology of the Brain Arousal Systems (WAKING), Lyon Neuroscience Research Centre, INSERM/CNRS/UCBL1BronFrance
| | - Amina Dulac
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Giorgio Matassi
- Dipartimento di Scienze Agroalimentari, Ambientali e Animali, University of UdineUdineItaly
- UMR “Ecology and Dynamics of Anthropogenic Systems” (EDYSAN), CNRS, Université de Picardie Jules VerneAmiensFrance
| | - Ali Mteyrek
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Rebecca Fima
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Marion Strehaiano
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Joana Dagorret
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Baya Chérif-Zahar
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Sandrine Marie
- Laboratory of Metabolic Diseases, Cliniques Universitaires Saint-Luc, Université catholique de LouvainBrusselsBelgium
| | - Irène Ceballos-Picot
- Metabolomic and Proteomic Biochemistry Laboratory, Necker-Enfants Malades Hospital and Paris Cité UniversityParisFrance
| | - Serge Birman
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| |
Collapse
|
5
|
Shan L, Heusinkveld HJ, Paul KC, Hughes S, Darweesh SKL, Bloem BR, Homberg JR. Towards improved screening of toxins for Parkinson's risk. NPJ Parkinsons Dis 2023; 9:169. [PMID: 38114496 PMCID: PMC10730534 DOI: 10.1038/s41531-023-00615-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023] Open
Abstract
Parkinson's disease (PD) is a chronic, progressive and disabling neurodegenerative disorder. The prevalence of PD has risen considerably over the past decades. A growing body of evidence suggest that exposure to environmental toxins, including pesticides, solvents and heavy metals (collectively called toxins), is at least in part responsible for this rapid growth. It is worrying that the current screening procedures being applied internationally to test for possible neurotoxicity of specific compounds offer inadequate insights into the risk of developing PD in humans. Improved screening procedures are therefore urgently needed. Our review first substantiates current evidence on the relation between exposure to environmental toxins and the risk of developing PD. We subsequently propose to replace the current standard toxin screening by a well-controlled multi-tier toxin screening involving the following steps: in silico studies (tier 1) followed by in vitro tests (tier 2), aiming to prioritize agents with human relevant routes of exposure. More in depth studies can be undertaken in tier 3, with whole-organism (in)vertebrate models. Tier 4 has a dedicated focus on cell loss in the substantia nigra and on the presumed mechanisms of neurotoxicity in rodent models, which are required to confirm or refute the possible neurotoxicity of any individual compound. This improved screening procedure should not only evaluate new pesticides that seek access to the market, but also critically assess all pesticides that are being used today, acknowledging that none of these has ever been proven to be safe from a perspective of PD. Importantly, the improved screening procedures should not just assess the neurotoxic risk of isolated compounds, but should also specifically look at the cumulative risk conveyed by exposure to commonly used combinations of pesticides (cocktails). The worldwide implementation of such an improved screening procedure, would be an essential step for policy makers and governments to recognize PD-related environmental risk factors.
Collapse
Affiliation(s)
- Ling Shan
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands.
| | - Harm J Heusinkveld
- Centre for Health Protection, National Institute for Public Health and Environment (RIVM), Bilthoven, The Netherlands
| | - Kimberly C Paul
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Samantha Hughes
- A-LIFE Amsterdam Institute for Life and Environment, Section Environmental Health and Toxicology, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| | - Sirwan K L Darweesh
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Bastiaan R Bloem
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
6
|
Bertrand M, Szeremeta F, Hervouet-Coste N, Sarou-Kanian V, Landon C, Morisset-Lopez S, Decoville M. An adult Drosophila glioma model to highlight metabolic dysfunctions and evaluate the role of the serotonin 5-HT 7 receptor as a potential therapeutic target. FASEB J 2023; 37:e23230. [PMID: 37781977 DOI: 10.1096/fj.202300783rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 08/31/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023]
Abstract
Gliomas account for 50% of brain cancers and are therefore the most common brain tumors. Molecular alterations involved in adult gliomas have been identified and mainly affect tyrosine kinase receptors with amplification and/or mutation of the epidermal growth factor receptor (EGFR) and its associated signaling pathways. Several targeted therapies have been developed, but current treatments remain ineffective for glioblastomas, the most severe forms. Thus, it is a priority to identify new pharmacological targets. Drosophila glioma models established in larvae and adults are useful to identify new genes and signaling pathways involved in glioma progression. Here, we used a Drosophila glioma model in adults, to characterize metabolic disturbances associated with glioma and assess the consequences of 5-HT7 R expression on glioma development. First, by using in vivo magnetic resonance imaging, we have shown that expression of the constitutively active forms of EGFR and PI3K in adult glial cells induces brain enlargement. Then, we explored altered cellular metabolism by using high-resolution magic angle spinning NMR and 1 H-13 C heteronuclear single quantum coherence solution states. Discriminant metabolites identified highlight the rewiring of metabolic pathways in glioma and associated cachexia phenotypes. Finally, the expression of 5-HT7 R in this adult model attenuates phenotypes associated with glioma development. Collectively, this whole-animal approach in Drosophila allowed us to provide several rapid and robust phenotype readouts, such as enlarged brain volume and glioma-associated cachexia, as well as to determine the metabolic pathways involved in glioma genesis and finally to confirm the interest of the 5-HT7 R in the treatment of glioma.
Collapse
Affiliation(s)
- Marylène Bertrand
- Centre de Biophysique Moléculaire-CBM, UPR 4301, CNRS, Orléans, France
| | | | | | - Vincent Sarou-Kanian
- Conditions Extrêmes et Matériaux: Haute Température et Irradiation-CEMHTI-CNRS UPR 3079, Orléans, France
| | - Céline Landon
- Centre de Biophysique Moléculaire-CBM, UPR 4301, CNRS, Orléans, France
| | | | - Martine Decoville
- Centre de Biophysique Moléculaire-CBM, UPR 4301, CNRS, Orléans, France
- UFR Sciences et Techniques, Université d'Orléans, Orléans, France
| |
Collapse
|
7
|
Iwashita M, Tran A, Garcia M, Cashon J, Burbano D, Salgado V, Hasegawa M, Balmilero-Unciano R, Politan K, Wong M, Lee RWY, Yoshizawa M. Metabolic shift toward ketosis in asocial cavefish increases social-like affinity. BMC Biol 2023; 21:219. [PMID: 37840141 PMCID: PMC10577988 DOI: 10.1186/s12915-023-01725-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/04/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Social affinity and collective behavior are nearly ubiquitous in the animal kingdom, but many lineages feature evolutionarily asocial species. These solitary species may have evolved to conserve energy in food-sparse environments. However, the mechanism by which metabolic shifts regulate social affinity is not well investigated. RESULTS In this study, we used the Mexican tetra (Astyanax mexicanus), which features riverine sighted surface (surface fish) and cave-dwelling populations (cavefish), to address the impact of metabolic shifts on asociality and other cave-associated behaviors in cavefish, including repetitive turning, sleeplessness, swimming longer distances, and enhanced foraging behavior. After 1 month of ketosis-inducing ketogenic diet feeding, asocial cavefish exhibited significantly higher social affinity, whereas social affinity regressed in cavefish fed the standard diet. The ketogenic diet also reduced repetitive turning and swimming in cavefish. No major behavioral shifts were found regarding sleeplessness and foraging behavior, suggesting that other evolved behaviors are not largely regulated by ketosis. We further examined the effects of the ketogenic diet via supplementation with exogenous ketone bodies, revealing that ketone bodies are pivotal molecules positively associated with social affinity. CONCLUSIONS Our study indicated that fish that evolved to be asocial remain capable of exhibiting social affinity under ketosis, possibly linking the seasonal food availability and sociality.
Collapse
Affiliation(s)
- Motoko Iwashita
- School of Life Sciences, University of Hawai'I at Mānoa, Honolulu, HI, 96822, USA
| | - Amity Tran
- School of Life Sciences, University of Hawai'I at Mānoa, Honolulu, HI, 96822, USA
| | - Marianne Garcia
- School of Life Sciences, University of Hawai'I at Mānoa, Honolulu, HI, 96822, USA
| | - Jia Cashon
- Hawai'i Institute of Marine Biology, University of Hawai'i at Mānoa, Kāne'ohe, HI, 96744, USA
| | - Devanne Burbano
- School of Life Sciences, University of Hawai'I at Mānoa, Honolulu, HI, 96822, USA
| | - Vanessa Salgado
- School of Life Sciences, University of Hawai'I at Mānoa, Honolulu, HI, 96822, USA
| | - Malia Hasegawa
- School of Life Sciences, University of Hawai'I at Mānoa, Honolulu, HI, 96822, USA
| | | | - Kaylah Politan
- School of Life Sciences, University of Hawai'I at Mānoa, Honolulu, HI, 96822, USA
| | - Miki Wong
- Nā Pu'uwai Native Hawaiian Healthcare System, Kaunakakai, HI, 96748, USA
- Nutrition Services Department, Shriners Hospitals for Children, Honolulu, HI, 96826, USA
| | - Ryan W Y Lee
- Medical Staff Department, Shriners Hospitals for Children, Honolulu, HI, 96826, USA
| | - Masato Yoshizawa
- School of Life Sciences, University of Hawai'I at Mānoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
8
|
Fruit fly for anticancer drug discovery and repurposing. Ann Med Surg (Lond) 2023; 85:337-342. [PMID: 36845805 PMCID: PMC9949803 DOI: 10.1097/ms9.0000000000000222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/01/2023] [Indexed: 02/28/2023] Open
|
9
|
Teal LB, Ingram SM, Bubser M, McClure E, Jones CK. The Evolving Role of Animal Models in the Discovery and Development of Novel Treatments for Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2023; 30:37-99. [PMID: 36928846 DOI: 10.1007/978-3-031-21054-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Historically, animal models have been routinely used in the characterization of novel chemical entities (NCEs) for various psychiatric disorders. Animal models have been essential in the in vivo validation of novel drug targets, establishment of lead compound pharmacokinetic to pharmacodynamic relationships, optimization of lead compounds through preclinical candidate selection, and development of translational measures of target occupancy and functional target engagement. Yet, with decades of multiple NCE failures in Phase II and III efficacy trials for different psychiatric disorders, the utility and value of animal models in the drug discovery process have come under intense scrutiny along with the widespread withdrawal of the pharmaceutical industry from psychiatric drug discovery. More recently, the development and utilization of animal models for the discovery of psychiatric NCEs has undergone a dynamic evolution with the application of the Research Domain Criteria (RDoC) framework for better design of preclinical to clinical translational studies combined with innovative genetic, neural circuitry-based, and automated testing technologies. In this chapter, the authors will discuss this evolving role of animal models for improving the different stages of the discovery and development in the identification of next generation treatments for psychiatric disorders.
Collapse
Affiliation(s)
- Laura B Teal
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Shalonda M Ingram
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Michael Bubser
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Elliott McClure
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
10
|
Tello JA, Williams HE, Eppler RM, Steinhilb ML, Khanna M. Animal Models of Neurodegenerative Disease: Recent Advances in Fly Highlight Innovative Approaches to Drug Discovery. Front Mol Neurosci 2022; 15:883358. [PMID: 35514431 PMCID: PMC9063566 DOI: 10.3389/fnmol.2022.883358] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
Neurodegenerative diseases represent a formidable challenge to global health. As advances in other areas of medicine grant healthy living into later decades of life, aging diseases such as Alzheimer's disease (AD) and other neurodegenerative disorders can diminish the quality of these additional years, owed largely to the lack of efficacious treatments and the absence of durable cures. Alzheimer's disease prevalence is predicted to more than double in the next 30 years, affecting nearly 15 million Americans, with AD-associated costs exceeding $1 billion by 2050. Delaying onset of AD and other neurodegenerative diseases is critical to improving the quality of life for patients and reducing the burden of disease on caregivers and healthcare systems. Significant progress has been made to model disease pathogenesis and identify points of therapeutic intervention. While some researchers have contributed to our understanding of the proteins and pathways that drive biological dysfunction in disease using in vitro and in vivo models, others have provided mathematical, biophysical, and computational technologies to identify potential therapeutic compounds using in silico modeling. The most exciting phase of the drug discovery process is now: by applying a target-directed approach that leverages the strengths of multiple techniques and validates lead hits using Drosophila as an animal model of disease, we are on the fast-track to identifying novel therapeutics to restore health to those impacted by neurodegenerative disease.
Collapse
Affiliation(s)
- Judith A. Tello
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center of Innovation in Brain Science, Tucson, AZ, United States
| | - Haley E. Williams
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center of Innovation in Brain Science, Tucson, AZ, United States
| | - Robert M. Eppler
- Department of Biology, Central Michigan University, Mount Pleasant, MI, United States
| | - Michelle L. Steinhilb
- Department of Biology, Central Michigan University, Mount Pleasant, MI, United States
| | - May Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center of Innovation in Brain Science, Tucson, AZ, United States
- Department of Molecular Pathobiology, New York University, New York, NY, United States
| |
Collapse
|
11
|
Qiang SJ, Shi YQ, Wu TY, Wang JQ, Chen XL, Su J, Chen XP, Li JZ, Chen ZS. The Discovery of Novel PGK1 Activators as Apoptotic Inhibiting and Neuroprotective Agents. Front Pharmacol 2022; 13:877706. [PMID: 35387336 PMCID: PMC8978560 DOI: 10.3389/fphar.2022.877706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke is the second leading cause of death worldwide and the leading cause of long-term disability that seriously endangers health and quality of human life. Tissue-type fibrinogen activator is currently the only drug approved by FDA for the treatment of ischemic stroke. Neuroprotection is theoretically a common strategy for the treatment of both ischemic and hemorrhagic stroke; therefore, the development of neuroprotective agent has been the focus of research. However, no ideal neuroprotective drug is clinically available. Phosphoglycerate kinase-1 (PGK1) activator has the effect of inhibiting apoptosis and protecting tissue damage, and therefore could be a potential neuroprotective agent. To obtain effective PGK1 activators, we virtually screened a large chemical database and their evaluated the efficacy by the Drosophila oxidative stress model, PGK1 enzymatic activity assay, and oxygen-glucose stripping reperfusion (OGD/R) model. The results showed that compounds 7979989, Z112553128 and AK-693/21087020 are potential PGK1 activators with protective effects against PQ-induced oxidative stress in the Drosophila model and could effectively ameliorate apoptosis induced by OGD/R-induced neuronal cell injury. Additionally, compounds 7979989 and Z112553128 are effective in alleviating LPS-induced cellular inflammation. This study indicated that these compounds are promising lead compounds that provide theoretical and material basis to the neuroprotective drug discovery.
Collapse
Affiliation(s)
| | - Yu-Qi Shi
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Tong-Yu Wu
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Xue-Lian Chen
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Jie Su
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Xin-Ping Chen
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Jia-Zhong Li
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| |
Collapse
|
12
|
Maitra U, Stephen C, Ciesla LM. Drug discovery from natural products - Old problems and novel solutions for the treatment of neurodegenerative diseases. J Pharm Biomed Anal 2022; 210:114553. [PMID: 34968995 PMCID: PMC8792363 DOI: 10.1016/j.jpba.2021.114553] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/19/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022]
Abstract
The use of natural products has been shown to be a fruitful approach in the discovery of novel pharmaceuticals. In fact, many currently approved drugs originated from compounds that were first identified in nature. Chemical diversity of natural compounds cannot be matched by man-made libraries of chemically synthesized molecules. Many natural compounds interact with and modulate regulatory protein targets and can be considered evolutionarily-optimized drug-like molecules. Despite this, many pharmaceutical companies have reduced or eliminated their natural product discovery programs in the last two decades. Screening natural products for pharmacologically active compounds is a challenging task that requires high resource commitment. Novel approaches at the early stage of the drug discovery pipeline are needed to allow for rapid screening and identification of the most promising molecules. Here, we review the possible evolutionary roots for drug-like characteristics of numerous natural compounds. Since many of these compounds target evolutionarily conserved cellular signaling pathways, we propose novel, early-stage drug discovery approaches to identify drug candidates that can be used for the potential prevention and treatment of neurodegenerative diseases. Invertebrate in vivo animal models of neurodegenerative diseases and innovative tools used within these models are proposed here as a screening funnel to identify new drug candidates and to shuttle these hits into further stages of the drug discovery pipeline.
Collapse
Affiliation(s)
- Urmila Maitra
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Cayman Stephen
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Lukasz M Ciesla
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA.
| |
Collapse
|
13
|
Proof-of-Concept Preclinical Use of Drosophila melanogaster in the Initial Screening of Immunomodulators. Sci Pharm 2022. [DOI: 10.3390/scipharm90010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Drug discovery is a complex process, and the use of a comprehensive approach is deemed necessary to discover new chemical entities with novel mechanisms of action. This research was carried out to determine whether Drosophila melanogaster can serve as an appropriate model organism in the initial screening of drug candidates with immunomodulatory activities. To test this, we performed phenotypic assay and molecular analysis to investigate the immunomodulatory activities of aspirin, dexamethasone, curcumin, and epigallocatechin gallate (EGCG), that have been reported to yield such effects in the mammalian model system. In vivo survival analysis demonstrated that all drugs/compounds were relatively safe at the tested concentrations. In the infection assay, curcumin and EGCG showed a protective signature to bacterial infection in flies lacking Toll-mediated immune responses. Furthermore, dexamethasone and aspirin, drugs with immunosuppressive activity, could improve the survival of PGRP-LBΔ mutant flies with hyperactivated immune system. These phenotypes were supported by RT-qPCR-based molecular analysis, revealing that drugs/compounds used in this study could modulate the expression level of genes related to the immune system. In conclusion, while curcumin and EGCG could promote the improvement of fly survival against infection, aspirin and dexamethasone were able to suppress overactivation of immune responses in D. melanogaster. These results are in line with the ones observed in the mammalian model system, further emphasizing the notion that flies would serve as a prospective model organism in the initial screening of drug candidates for their immunomodulatory activities prior to further checking in the mammalian animal models. In the end, this will reduce the use of mammalian animal models for preliminary experiments in an effort to discover/repurpose drugs with immunomodulatory activity.
Collapse
|
14
|
Moraes KCM, Montagne J. Drosophila melanogaster: A Powerful Tiny Animal Model for the Study of Metabolic Hepatic Diseases. Front Physiol 2021; 12:728407. [PMID: 34603083 PMCID: PMC8481879 DOI: 10.3389/fphys.2021.728407] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/27/2021] [Indexed: 12/25/2022] Open
Abstract
Animal experimentation is limited by unethical procedures, time-consuming protocols, and high cost. Thus, the development of innovative approaches for disease treatment based on alternative models in a fast, safe, and economic manner is an important, yet challenging goal. In this paradigm, the fruit-fly Drosophila melanogaster has become a powerful model for biomedical research, considering its short life cycle and low-cost maintenance. In addition, biological processes are conserved and homologs of ∼75% of human disease-related genes are found in the fruit-fly. Therefore, this model has been used in innovative approaches to evaluate and validate the functional activities of candidate molecules identified via in vitro large-scale analyses, as putative agents to treat or reverse pathological conditions. In this context, Drosophila offers a powerful alternative to investigate the molecular aspects of liver diseases, since no effective therapies are available for those pathologies. Non-alcoholic fatty liver disease is the most common form of chronic hepatic dysfunctions, which may progress to the development of chronic hepatitis and ultimately to cirrhosis, thereby increasing the risk for hepatocellular carcinoma (HCC). This deleterious situation reinforces the use of the Drosophila model to accelerate functional research aimed at deciphering the mechanisms that sustain the disease. In this short review, we illustrate the relevance of using the fruit-fly to address aspects of liver pathologies to contribute to the biomedical area.
Collapse
Affiliation(s)
- Karen C M Moraes
- Laboratório de Sinalização Celular e Expressão Gênica, Departamento de Biologia Geral e Aplicada, Instituto de Biociências, UNESP, Rio Claro, Brazil
| | - Jacques Montagne
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
15
|
Liu Y, Weaver CM, Sen Y, Eitzen G, Simmonds AJ, Linchieh L, Lurette O, Hebert-Chatelain E, Rachubinski RA, Di Cara F. The Nitric Oxide Donor, S-Nitrosoglutathione, Rescues Peroxisome Number and Activity Defects in PEX1G843D Mild Zellweger Syndrome Fibroblasts. Front Cell Dev Biol 2021; 9:714710. [PMID: 34434934 PMCID: PMC8382563 DOI: 10.3389/fcell.2021.714710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/20/2021] [Indexed: 02/04/2023] Open
Abstract
Peroxisome biogenesis disorders (PBDs) are a group of metabolic developmental diseases caused by mutations in one or more genes encoding peroxisomal proteins. Zellweger syndrome spectrum (PBD-ZSS) results from metabolic dysfunction caused by damaged or non-functional peroxisomes and manifests as a multi-organ syndrome with significant morbidity and mortality for which there is no current drug therapy. Mild PBD-ZSS patients can exhibit a more progressive disease course and could benefit from the identification of drugs to improve the quality of life and extend the lifespan of affected individuals. Our study used a high-throughput screen of FDA-approved compounds to identify compounds that improve peroxisome function and biogenesis in human fibroblast cells carrying the mild PBD-ZSS variant, PEX1G843D. Our screen identified the nitrogen oxide donor, S-nitrosoglutathione (GSNO), as a potential therapeutic for this mild form of PBD-ZSS. Further biochemical characterization showed that GSNO enhances both peroxisome number and function in PEX1G843D mutant fibroblasts and leads to increased survival and longer lifespan in an in vivo humanized Drosophila model carrying the PEX1G843D mutation. GSNO is therefore a strong candidate to be translated to clinical trials as a potential therapeutic for mild PBD-ZSS.
Collapse
Affiliation(s)
- Yidi Liu
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | - Ceileigh M Weaver
- Department of Microbiology and Immunology, IWK Research Centre, Dalhousie University, Halifax, NS, Canada
| | - Yarina Sen
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | - Gary Eitzen
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | - Andrew J Simmonds
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | - Lilliana Linchieh
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | - Olivier Lurette
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | | | | | - Francesca Di Cara
- Department of Microbiology and Immunology, IWK Research Centre, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
16
|
Kukhtar D, Rubio-Peña K, Serrat X, Cerón J. Mimicking of splicing-related retinitis pigmentosa mutations in C. elegans allow drug screens and identification of disease modifiers. Hum Mol Genet 2021; 29:756-765. [PMID: 31919495 DOI: 10.1093/hmg/ddz315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/06/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
CRISPR/Cas and the high conservation of the spliceosome components facilitate the mimicking of human pathological mutations in splicing factors of model organisms. The degenerative retinal disease retinitis pigmentosa (RP) is caused by mutations in distinct types of genes, including missense mutations in splicing factors that provoke RP in an autosomal dominant form (s-adRP). Using CRISPR in Caenorhabditis elegans, we generated mutant strains to mimic s-adRP mutations reported in PRPF8 and SNRNP200. Whereas these inherited mutations are present in heterozygosis in patients, C. elegans allows the maintenance of these mutations as homozygotes, which is advantageous for genetic and drug screens. We found that snrp-200(cer23[V676L]) and prp-8(cer14[H2302del]) display pleiotropic phenotypes, including reduced fertility. However, snrp-200(cer24[S1080L]) and prp-8(cer22[R2303G]) are weak alleles suitable for RNAi screens for identifying genetic interactions, which could uncover potential disease modifiers. We screened a collection of RNAi clones for splicing-related genes and identified three splicing factors: isy-1/ISY1, cyn-15/PPWD1 and mog-2/SNRPA1, whose partial inactivation may modify the course of the disease. Interestingly, these three genes act as modifiers of prp-8(cer22) but not of snrp-200(cer24). Finally, a screen of the strong allele prp-8(cer14) with FDA-approved drugs did not identify molecules capable of alleviating the temperature-sensitive sterility. Instead, we detected drugs, such as dequalinium chloride, which exacerbated the phenotype, and therefore, are potentially harmful to s-adRP patients since they may accelerate the progression of the disease.
Collapse
Affiliation(s)
- Dmytro Kukhtar
- Modeling human diseases in C. elegans Group. Genes, Disease and Therapy Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, 08908 Barcelona, Spain
| | - Karinna Rubio-Peña
- Modeling human diseases in C. elegans Group. Genes, Disease and Therapy Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, 08908 Barcelona, Spain
| | - Xènia Serrat
- Modeling human diseases in C. elegans Group. Genes, Disease and Therapy Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, 08908 Barcelona, Spain
| | - Julián Cerón
- Modeling human diseases in C. elegans Group. Genes, Disease and Therapy Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, 08908 Barcelona, Spain
| |
Collapse
|
17
|
Zarini-Gakiye E, Sanadgol N, Parivar K, Vaezi G. Alpha-lipoic acid ameliorates tauopathy-induced oxidative stress, apoptosis, and behavioral deficits through the balance of DIAP1/DrICE ratio and redox homeostasis: Age is a determinant factor. Metab Brain Dis 2021; 36:669-683. [PMID: 33547995 DOI: 10.1007/s11011-021-00679-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/26/2021] [Indexed: 12/20/2022]
Abstract
Tauopathies belong to a heterogeneous class of neuronal diseases resulting in the metabolic disturbance. A disulfide natural compound of Alpha-Lipoic acid (ALA) has shown numerous pharmacologic, antioxidant, and neuroprotective activities under neuropathological conditions. The aim of this study was to investigate the neuroprotective effects of ALA on the tauopathy-induced oxidative disturbance and behavioral deficits. The transgenic Drosophila model of tauopathy induced by human tauR406W using GAL4/UAS system and effects of ALA (0.001, 0.005, and 0.025 % w/w of diet) on the neuropathology of tau in younger (20 days) and older (30 days) adults were investigated via biochemical, molecular, behavioral and in-situ tissue analyses. Expression of apoptosis-related proteins involving Drosophila Cyt-c-d (trigger of intrinsic apoptosis) and DrICE (effector caspase) were upregulated in both ages (20 and 30 days) and DIAP1 (caspase inhibitor) has reduced only in older model flies compared to the controls. Remarkably, all doses of ALA increased DIAP1 and glutathione (GSH) as well as reducing Cyt-c-d and lipid peroxidation (LPO) in the younger flies compared to the model flies. Moreover, the higher doses of ALA were able to decrease thiol concentrations, to increase total antioxidant capacity, and to improve the behavioral deficits (locomotor function, olfactory memory, and ethanol sensitivity) in the younger flies. On the other hand, only a higher dose of ALA was able to decrease DrICE, Cyt-c-d, LPO, and thiol as well as increasing antioxidant capacity and decreasing ethanol sensitivity (ST50, RT50) in the older flies. TUNEL assay showed that all doses of ALA could potentially increase the DIAP1/DrICE ratio and exert anti-apoptotic effects on younger, but not on the older adults. Furthermore, data obtained from the in-situ ROS assay confirmed that only a higher dose of ALA significantly decreased the ROS level at both ages. Our data showed that an effective neuroprotective dose of ALA and its mechanism of action on this model of tauopathy could potentially be influenced by longevity. Moreover, it was shown that ALA prevents apoptosis and decreases the redox homeostasis, and this partially explains the mechanism by which ALA diminishes behavioral deficits.
Collapse
Affiliation(s)
- Elahe Zarini-Gakiye
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nima Sanadgol
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran.
| | - Kazem Parivar
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Gholamhassan Vaezi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
18
|
Fernández-Hernández I, Marsh EB, Bonaguidi MA. Mechanosensory neuron regeneration in adult Drosophila. Development 2021; 148:dev.187534. [PMID: 33597190 DOI: 10.1242/dev.187534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/09/2021] [Indexed: 01/01/2023]
Abstract
Auditory and vestibular mechanosensory hair cells do not regenerate following injury or aging in the adult mammalian inner ear, inducing irreversible hearing loss and balance disorders for millions of people. Research on model systems showing replacement of mechanosensory cells can provide mechanistic insights into developing new regenerative therapies. Here, we developed lineage tracing systems to reveal the generation of mechanosensory neurons in the Johnston's organ (JO) of intact adult Drosophila, which are the functional counterparts to hair cells in vertebrates. New JO neurons develop cilia and target central brain circuitry. Unexpectedly, mitotic recombination clones point to JO neuron self-replication as a likely source of neuronal plasticity. This mechanism is further enhanced upon treatment with experimental and ototoxic compounds. Our findings introduce a new platform to expedite research on mechanisms and compounds mediating mechanosensory cell regeneration, with nascent implications for hearing and balance restoration.
Collapse
Affiliation(s)
- Ismael Fernández-Hernández
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Evan B Marsh
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Michael A Bonaguidi
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA .,Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Department of Gerontology, Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| |
Collapse
|
19
|
Rigon L, De Filippis C, Napoli B, Tomanin R, Orso G. Exploiting the Potential of Drosophila Models in Lysosomal Storage Disorders: Pathological Mechanisms and Drug Discovery. Biomedicines 2021; 9:biomedicines9030268. [PMID: 33800050 PMCID: PMC8000850 DOI: 10.3390/biomedicines9030268] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/18/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Lysosomal storage disorders (LSDs) represent a complex and heterogeneous group of rare genetic diseases due to mutations in genes coding for lysosomal enzymes, membrane proteins or transporters. This leads to the accumulation of undegraded materials within lysosomes and a broad range of severe clinical features, often including the impairment of central nervous system (CNS). When available, enzyme replacement therapy slows the disease progression although it is not curative; also, most recombinant enzymes cannot cross the blood-brain barrier, leaving the CNS untreated. The inefficient degradative capability of the lysosomes has a negative impact on the flux through the endolysosomal and autophagic pathways; therefore, dysregulation of these pathways is increasingly emerging as a relevant disease mechanism in LSDs. In the last twenty years, different LSD Drosophila models have been generated, mainly for diseases presenting with neurological involvement. The fruit fly provides a large selection of tools to investigate lysosomes, autophagy and endocytic pathways in vivo, as well as to analyse neuronal and glial cells. The possibility to use Drosophila in drug repurposing and discovery makes it an attractive model for LSDs lacking effective therapies. Here, ee describe the major cellular pathways implicated in LSDs pathogenesis, the approaches available for their study and the Drosophila models developed for these diseases. Finally, we highlight a possible use of LSDs Drosophila models for drug screening studies.
Collapse
Affiliation(s)
- Laura Rigon
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy; (C.D.F.); (R.T.)
- Correspondence:
| | - Concetta De Filippis
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy; (C.D.F.); (R.T.)
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children’s Health, University of Padova, Via Giustiniani 3, 35128 Padova, Italy
| | - Barbara Napoli
- Laboratory of Molecular Biology, Scientific Institute, IRCCS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, 23842 Lecco, Italy;
| | - Rosella Tomanin
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy; (C.D.F.); (R.T.)
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children’s Health, University of Padova, Via Giustiniani 3, 35128 Padova, Italy
| | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy;
| |
Collapse
|
20
|
Zimmermann M, Patil KR, Typas A, Maier L. Towards a mechanistic understanding of reciprocal drug-microbiome interactions. Mol Syst Biol 2021; 17:e10116. [PMID: 33734582 PMCID: PMC7970330 DOI: 10.15252/msb.202010116] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/10/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
Broad-spectrum antibiotics target multiple gram-positive and gram-negative bacteria, and can collaterally damage the gut microbiota. Yet, our knowledge of the extent of damage, the antibiotic activity spectra, and the resistance mechanisms of gut microbes is sparse. This limits our ability to mitigate microbiome-facilitated spread of antibiotic resistance. In addition to antibiotics, non-antibiotic drugs affect the human microbiome, as shown by metagenomics as well as in vitro studies. Microbiome-drug interactions are bidirectional, as microbes can also modulate drugs. Chemical modifications of antibiotics mostly function as antimicrobial resistance mechanisms, while metabolism of non-antibiotics can also change the drugs' pharmacodynamic, pharmacokinetic, and toxic properties. Recent studies have started to unravel the extensive capacity of gut microbes to metabolize drugs, the mechanisms, and the relevance of such events for drug treatment. These findings raise the question whether and to which degree these reciprocal drug-microbiome interactions will differ across individuals, and how to take them into account in drug discovery and precision medicine. This review describes recent developments in the field and discusses future study areas that will benefit from systems biology approaches to better understand the mechanistic role of the human gut microbiota in drug actions.
Collapse
Affiliation(s)
- Michael Zimmermann
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Kiran Raosaheb Patil
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- The Medical Research Council Toxicology UnitUniversity of CambridgeCambridgeUK
| | - Athanasios Typas
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- Genome Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Lisa Maier
- Interfaculty Institute of Microbiology and Infection MedicineUniversity of TübingenTübingenGermany
- Cluster of Excellence ‘Controlling Microbes to Fight Infections’University of TübingenTübingenGermany
| |
Collapse
|
21
|
Liu W, Wang Y, He H. CoFly: A gene coexpression database for the fruit fly Drosophila melanogaster. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2020; 105:e21693. [PMID: 32436316 DOI: 10.1002/arch.21693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 05/05/2020] [Accepted: 05/09/2020] [Indexed: 06/11/2023]
Abstract
The fruit fly Drosophila melanogaster can be used as a model organism for studying various problems in biomedicine and pest management. A large number of fruit fly transcriptomes have been profiled in various cell types, tissues, development stages, toxicological exposures, and other conditions by microarray. Until now, there are still no database developed for exploring those precious data. Microarray data for 4,367 samples from National Center for Biotechnology Information Gene Expression Omnibus was collected, and analyzed by weighted gene coexpression network analysis algorithm. Fifty one gene coexpression modules that are related to cell types, tissues, development stages, and other experimental conditions were identified. The high dimensional gene expression was reduced to tens of modules that were associated with experiments/traits, representing signatures for phenotypes. Six modules were enriched with genomic regions of clustered genes. Hub genes could also be screened by intramodule connectivity. By analyzing higher order module networks, we found that cell signaling modules are more connected than other modules. Module-based gene function identification may help to discover novel gene function. An easy-to-use database was developed, which provides a new source for gene function study in the fruit fly (http://bioinformatics.fafu.edu.cn/fly/).
Collapse
Affiliation(s)
- Wei Liu
- Department of Bioinformatics, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yanan Wang
- Department of Bioinformatics, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Huaqin He
- Department of Bioinformatics, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
22
|
Phenotypic Suppression of ALS/FTD-Associated Neurodegeneration Highlights Mechanisms of Dysfunction. J Neurosci 2020; 39:8217-8224. [PMID: 31619490 DOI: 10.1523/jneurosci.1159-19.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 12/14/2022] Open
Abstract
A fundamental question regarding the etiology of amyotrophic lateral sclerosis (ALS) is whether the various gene mutations associated with the disease converge on a single molecular pathway or act through multiple pathways to trigger neurodegeneration. Notably, several of the genes and cellular processes implicated in ALS have also been linked to frontotemporal dementia (FTD), suggesting these two diseases share common origins with varied clinical presentations. Scientists are rapidly identifying ALS/FTD suppressors that act on conserved pathways from invertebrates to vertebrates to alleviate degeneration. The elucidation of such genetic modifiers provides insight into the molecular pathways underlying this rapidly progressing neurodegenerative disease, while also revealing new targets for therapeutic development.
Collapse
|
23
|
Drosophila as a model for studying cystic fibrosis pathophysiology of the gastrointestinal system. Proc Natl Acad Sci U S A 2020; 117:10357-10367. [PMID: 32345720 DOI: 10.1073/pnas.1913127117] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cystic fibrosis (CF) is a recessive disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. The most common symptoms include progressive lung disease and chronic digestive conditions. CF is the first human genetic disease to benefit from having five different species of animal models. Despite the phenotypic differences among the animal models and human CF, these models have provided invaluable insight into understanding disease mechanisms at the organ-system level. Here, we identify a member of the ABCC4 family, CG5789, that has the structural and functional properties expected for encoding the Drosophila equivalent of human CFTR, and thus refer to it as Drosophila CFTR (Dmel\CFTR). We show that knockdown of Dmel\CFTR in the adult intestine disrupts osmotic homeostasis and displays CF-like phenotypes that lead to intestinal stem cell hyperplasia. We also show that expression of wild-type human CFTR, but not mutant variants of CFTR that prevent plasma membrane expression, rescues the mutant phenotypes of Dmel\CFTR Furthermore, we performed RNA sequencing (RNA-Seq)-based transcriptomic analysis using Dmel\CFTR fly intestine and identified a mucin gene, Muc68D, which is required for proper intestinal barrier protection. Altogether, our findings suggest that Drosophila can be a powerful model organism for studying CF pathophysiology.
Collapse
|
24
|
Large-Scale Transgenic Drosophila Resource Collections for Loss- and Gain-of-Function Studies. Genetics 2020; 214:755-767. [PMID: 32071193 DOI: 10.1534/genetics.119.302964] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/11/2020] [Indexed: 01/20/2023] Open
Abstract
The Transgenic RNAi Project (TRiP), a Drosophila melanogaster functional genomics platform at Harvard Medical School, was initiated in 2008 to generate and distribute a genome-scale collection of RNA interference (RNAi) fly stocks. To date, it has generated >15,000 RNAi fly stocks. As this covers most Drosophila genes, we have largely transitioned to development of new resources based on CRISPR technology. Here, we present an update on our libraries of publicly available RNAi and CRISPR fly stocks, and focus on the TRiP-CRISPR overexpression (TRiP-OE) and TRiP-CRISPR knockout (TRiP-KO) collections. TRiP-OE stocks express single guide RNAs targeting upstream of a gene transcription start site. Gene activation is triggered by coexpression of catalytically dead Cas9 fused to an activator domain, either VP64-p65-Rta or Synergistic Activation Mediator. TRiP-KO stocks express one or two single guide RNAs targeting the coding sequence of a gene or genes. Cutting is triggered by coexpression of Cas9, allowing for generation of indels in both germline and somatic tissue. To date, we have generated >5000 TRiP-OE or TRiP-KO stocks for the community. These resources provide versatile, transformative tools for gene activation, gene repression, and genome engineering.
Collapse
|
25
|
Abstract
Drosophila is an excellent system to investigate how the presence and composition of the gut microbiome influences the efficacy of therapeutic drugs and downstream consequences for host health. These opportunities derive from two key attributes of Drosophila. First, Drosophila is amenable for microbiome research, with simple, standardized methods to produce large numbers of microbiologically-sterile flies and flies with a standardized gut microbiome, thereby facilitating experimental reproducibility. Second, Drosophila is a well-established genetic model that is increasingly used to elucidate the molecular and physiological basis of lesions associated with human disease alleles; this provides the opportunity to link microbiome/drug interactions to previously-described processes shaping health and disease. In this way, Drosophila can fast-track understanding of fundamental biology to generate precise hypotheses for testing in mammalian systems. Drosophila is particularly well-suited to investigate the incidence of microbiome/drug interactions mediated by different mechanisms, including microbial drug metabolism (to active, inactive or toxic derivatives), microbial production of compounds that inhibit drug efficacy, and off-target effects of the drug on the microbiome, resulting in dysbiosis and host ill-health. Drosophila can also be used to investigate how interactions between the microbiome and host genotype may shape responses to therapeutic drugs, informing the reliability of precision medicine based exclusively on human genomic markers.
Collapse
Affiliation(s)
- Angela E Douglas
- Department of Entomology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
26
|
Ekowati H, Arai J, Damana Putri AS, Nainu F, Shiratsuchi A, Nakanishi Y. Protective effects of Phaseolus vulgaris lectin against viral infection in Drosophila. Drug Discov Ther 2019; 11:329-335. [PMID: 29332891 DOI: 10.5582/ddt.2017.01071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Phytohemagglutinin (PHA) isolated from the family of Phaseolus vulgaris beans is a promising agent against viral infection; however, it has not yet been demonstrated in vivo. We herein investigated this issue using Drosophila as a host. Adult flies were fed lectin approximately 12 h before they were subjected to a systemic viral infection. After a fatal infection with Drosophila C virus, death was delayed and survival was longer in flies fed PHA-P, a mixture of L4, L3E1, and L2E2, than in control unfed flies. We then examined PHA-L4, anticipating subunit L as the active form, and confirmed the protective effects of this lectin at markedly lower concentrations than PHA-P. In both experiments, lectin feeding reduced the viral load prior to the onset of fly death. Furthermore, we found a dramatic increase in the levels of the mRNAs of phagocytosis receptors in flies after feeding with PHA-L4 while a change in the levels of the mRNAs of antimicrobial peptides was marginal. We concluded that P. vulgaris PHA protects Drosophila against viral infection by augmenting the level of host immunity.
Collapse
Affiliation(s)
- Heny Ekowati
- Graduate School of Medical Sciences, Kanazawa University.,Faculty of Health Science, Jenderal Soedirman University
| | - Junko Arai
- Product Development Laboratory, J-Oil Mills, Inc
| | | | | | | | | |
Collapse
|
27
|
Maitra U, Ciesla L. Using Drosophila as a platform for drug discovery from natural products in Parkinson's disease. MEDCHEMCOMM 2019; 10:867-879. [PMID: 31303984 PMCID: PMC6596131 DOI: 10.1039/c9md00099b] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative movement disorder with no cure. Despite intensive research, most of the currently available therapies are only effective in alleviating symptoms with no effect on disease progression. There is an urgent need for new therapeutics to impede disease progression. Natural products are valuable sources of bioactive compounds that can be exploited for novel therapeutic potential in PD pathogenesis. However, rapid screening of plant-derived natural products and characterization of bioactive compounds is costly and challenging. Drosophila melanogaster, commonly known as the fruit fly, has recently emerged as an excellent model for human neurodegenerative diseases, including PD. The high degree of conserved molecular pathways with mammalian models make Drosophila PD models an inexpensive solution to preliminary phases of target validation in the drug discovery pipeline. The present review provides an overview of drug discovery from natural extracts using Drosophila as a screening platform to evaluate the therapeutic potential of phytochemicals against PD.
Collapse
Affiliation(s)
- Urmila Maitra
- Department of Biological Sciences , University of Alabama , Science and Engineering Complex 2320, 300 Hackberry Lane , Tuscaloosa , Alabama 35487-0344 , USA . ; Tel: +205 348 7599
| | - Lukasz Ciesla
- Department of Biological Sciences , University of Alabama , Science and Engineering Complex 2329, 300 Hackberry Lane , Tuscaloosa , Alabama 35487-0344 , USA . ; Tel: +205 348 1828
| |
Collapse
|
28
|
Su TT. Drug screening in Drosophila; why, when, and when not? WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 8:e346. [PMID: 31056843 DOI: 10.1002/wdev.346] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/08/2019] [Accepted: 04/10/2019] [Indexed: 12/17/2022]
Abstract
The best global seller among oncology drugs in 2018 is lenalidomide, an analog of thalidomide. It took 53 years and a circuitous route from the discovery of thalidomide to approval of an analog for use in treatment of cancer. We understand now a lot more about the genetic and molecular basis of diseases than we did in 1953 when thalidomide was discovered. We have also no shortage of chemical libraries with hundreds of thousands of compounds, both synthetic and natural. What we need are better ways to search among these rich resources for compounds with the potential to do what we want them to do. This review summarizes examples from the literature that make Drosophila melanogaster a good model to screen for drugs, and discusses knowledge gaps and technical challenges that make Drosophila models not as widely used as they could or should be. This article is categorized under: Technologies > Analysis of Cell, Tissue, and Animal Phenotypes.
Collapse
Affiliation(s)
- Tin Tin Su
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado.,Molecular, Cellular and Developmental Biology, University of Colorado Comprehensive Cancer Center, Aurora, Colorado
| |
Collapse
|
29
|
Wu Q, Kumar N, Velagala V, Zartman JJ. Tools to reverse-engineer multicellular systems: case studies using the fruit fly. J Biol Eng 2019; 13:33. [PMID: 31049075 PMCID: PMC6480878 DOI: 10.1186/s13036-019-0161-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/07/2019] [Indexed: 01/08/2023] Open
Abstract
Reverse-engineering how complex multicellular systems develop and function is a grand challenge for systems bioengineers. This challenge has motivated the creation of a suite of bioengineering tools to develop increasingly quantitative descriptions of multicellular systems. Here, we survey a selection of these tools including microfluidic devices, imaging and computer vision techniques. We provide a selected overview of the emerging cross-talk between engineering methods and quantitative investigations within developmental biology. In particular, the review highlights selected recent examples from the Drosophila system, an excellent platform for understanding the interplay between genetics and biophysics. In sum, the integrative approaches that combine multiple advances in these fields are increasingly necessary to enable a deeper understanding of how to analyze both natural and synthetic multicellular systems.
Collapse
Affiliation(s)
- Qinfeng Wu
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Nilay Kumar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Vijay Velagala
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Jeremiah J. Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
| |
Collapse
|
30
|
Papanikolopoulou K, Mudher A, Skoulakis E. An assessment of the translational relevance of Drosophila in drug discovery. Expert Opin Drug Discov 2019; 14:303-313. [PMID: 30664368 DOI: 10.1080/17460441.2019.1569624] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Drosophila melanogaster offers a powerful expedient and economical system with facile genetics. Because of the high sequence and functional conservation with human disease-associated genes, it has been cardinal in deciphering disease mechanisms at the genetic and molecular level. Drosophila are amenable to and respond well to pharmaceutical treatment which coupled to their genetic tractability has led to discovery, repositioning, and validation of a number of compounds. Areas covered: This review summarizes the generation of fly models of human diseases, their advantages and use in elucidation of human disease mechanisms. Representative studies provide examples of the utility of this system in modeling diseases and the discovery, repositioning and testing on pharmaceuticals to ameliorate them. Expert opinion: Drosophila offers a facile and economical whole animal system with many homologous organs to humans, high functional conservation and established methods of generating and validating human disease models. Nevertheless, it remains relatively underused as a drug discovery tool probably because its relevance to mammalian systems remains under question. However, recent exciting success stories using Drosophila disease models for drug screening, repositioning and validation strongly suggest that fly models should figure prominently in the drug discovery pipeline from bench to bedside.
Collapse
Affiliation(s)
- Katerina Papanikolopoulou
- a Division of Neuroscience , Biomedical Sciences Research Centre "Alexander Fleming" , Vari , Greece
| | - Amrit Mudher
- b Centre for Biological Sciences , University of Southampton , Southampton , UK
| | - Efthimios Skoulakis
- a Division of Neuroscience , Biomedical Sciences Research Centre "Alexander Fleming" , Vari , Greece
| |
Collapse
|
31
|
Aquilina B, Cauchi RJ. Modelling motor neuron disease in fruit flies: Lessons from spinal muscular atrophy. J Neurosci Methods 2018; 310:3-11. [DOI: 10.1016/j.jneumeth.2018.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 04/06/2018] [Accepted: 04/07/2018] [Indexed: 12/25/2022]
|
32
|
Bar S, Prasad M, Datta R. Neuromuscular degeneration and locomotor deficit in a Drosophila model of mucopolysaccharidosis VII is attenuated by treatment with resveratrol. Dis Model Mech 2018; 11:dmm036954. [PMID: 30459155 PMCID: PMC6262814 DOI: 10.1242/dmm.036954] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/05/2018] [Indexed: 12/12/2022] Open
Abstract
Mucopolysaccharidosis VII (MPS VII) is a recessively inherited lysosomal storage disorder caused by β-glucuronidase enzyme deficiency. The disease is characterized by widespread accumulation of non-degraded or partially degraded glycosaminoglycans, leading to cellular and multiple tissue dysfunctions. The patients exhibit diverse clinical symptoms, and eventually succumb to premature death. The only possible remedy is the recently approved enzyme replacement therapy, which is an expensive, invasive and lifelong treatment procedure. Small-molecule therapeutics for MPS VII have so far remained elusive primarily due to lack of molecular insights into the disease pathogenesis and unavailability of a suitable animal model that can be used for rapid drug screening. To address these issues, we developed a Drosophila model of MPS VII by knocking out the CG2135 gene, the fly β-glucuronidase orthologue. The CG2135-/- fly recapitulated cardinal features of MPS VII, such as reduced lifespan, progressive motor impairment and neuropathological abnormalities. Loss of dopaminergic neurons and muscle degeneration due to extensive apoptosis was implicated as the basis of locomotor deficit in this fly. Such hitherto unknown mechanistic links have considerably advanced our understanding of the MPS VII pathophysiology and warrant leveraging this genetically tractable model for deeper enquiry about the disease progression. We were also prompted to test whether phenotypic abnormalities in the CG2135-/- fly can be attenuated by resveratrol, a natural polyphenol with potential health benefits. Indeed, resveratrol treatment significantly ameliorated neuromuscular pathology and restored normal motor function in the CG2135-/- fly. This intriguing finding merits further preclinical studies for developing an alternative therapy for MPS VII.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sudipta Bar
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur 741246, West Bengal, India
| | - Mohit Prasad
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur 741246, West Bengal, India
| | - Rupak Datta
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur 741246, West Bengal, India
| |
Collapse
|
33
|
Drosophila Models of Sporadic Parkinson's Disease. Int J Mol Sci 2018; 19:ijms19113343. [PMID: 30373150 PMCID: PMC6275057 DOI: 10.3390/ijms19113343] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/17/2022] Open
Abstract
Parkinson’s disease (PD) is the most common cause of movement disorders and is characterized by the progressive loss of dopaminergic neurons in the substantia nigra. It is increasingly recognized as a complex group of disorders presenting widely heterogeneous symptoms and pathology. With the exception of the rare monogenic forms, the majority of PD cases result from an interaction between multiple genetic and environmental risk factors. The search for these risk factors and the development of preclinical animal models are in progress, aiming to provide mechanistic insights into the pathogenesis of PD. This review summarizes the studies that capitalize on modeling sporadic (i.e., nonfamilial) PD using Drosophilamelanogaster and discusses their methodologies, new findings, and future perspectives.
Collapse
|
34
|
Xu DC, Arthurton L, Baena-Lopez LA. Learning on the Fly: The Interplay between Caspases and Cancer. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5473180. [PMID: 29854765 PMCID: PMC5949197 DOI: 10.1155/2018/5473180] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 03/14/2018] [Indexed: 02/07/2023]
Abstract
The ease of genetic manipulation, as well as the evolutionary conservation of gene function, has placed Drosophila melanogaster as one of the leading model organisms used to understand the implication of many proteins with disease development, including caspases and their relation to cancer. The family of proteases referred to as caspases have been studied over the years as the major regulators of apoptosis: the most common cellular mechanism involved in eliminating unwanted or defective cells, such as cancerous cells. Indeed, the evasion of the apoptotic programme resulting from caspase downregulation is considered one of the hallmarks of cancer. Recent investigations have also shown an instrumental role for caspases in non-lethal biological processes, such as cell proliferation, cell differentiation, intercellular communication, and cell migration. Importantly, malfunction of these essential biological tasks can deeply impact the initiation and progression of cancer. Here, we provide an extensive review of the literature surrounding caspase biology and its interplay with many aspects of cancer, emphasising some of the key findings obtained from Drosophila studies. We also briefly describe the therapeutic potential of caspase modulation in relation to cancer, highlighting shortcomings and hopeful promises.
Collapse
Affiliation(s)
- Derek Cui Xu
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX13RE, UK
- Cell Biology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Lewis Arthurton
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX13RE, UK
| | | |
Collapse
|
35
|
Epigallocatechin-3-Gallate Protects and Prevents Paraquat-Induced Oxidative Stress and Neurodegeneration in Knockdown dj-1-β Drosophila melanogaster. Neurotox Res 2018; 34:401-416. [PMID: 29667128 DOI: 10.1007/s12640-018-9899-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 04/02/2018] [Accepted: 04/05/2018] [Indexed: 12/28/2022]
Abstract
Epigallocatechin-3-gallate (EGCG) is a polyhydroxyphenol constituent of green tea (e.g., Camellia sinensis) with known antioxidant properties. Due to these properties, others have proposed it as a potential therapeutic agent for the treatment of Parkinson's disease (PD). Previously, we demonstrated that EGCG prolonged the lifespan and locomotor activity in wild-type Canton-S flies exposed to the neurotoxicant paraquat (PQ), suggesting neuroprotective properties. Both gene mutations and environmental neurotoxicants (e.g., PQ) are factors involved in the development of PD. Thus, the first aim of this study was to create a suitable animal model of PD, which encompasses both of these factors. To create the model, we knocked down dj-1-β function specifically in the dopaminergic neurons to generate TH > dj-1-β-RNAi/+ Drosophila melanogaster flies. Next, we induced neurotoxicity in the transgenic flies with PQ. The second aim of this study was to validate the model by comparing the effects of vehicle, EGCG, and chemicals with known antioxidant and neuroprotective properties in vivo (e.g., propyl gallate and minocycline) on life-span, locomotor activity, lipid peroxidation, and neurodegeneration. The EGCG treatment provided protection and prevention from the PQ-induced reduction in the life-span and locomotor activity and from the PQ-induced increase in lipid peroxidation and neurodegeneration. These effects were augmented in the EGCG-treated flies when compared to the flies treated with either PG or MC. Altogether, these results suggest that the transgenic TH > dj-1-β-RNAi/+ flies treated with PQ serve as a suitable PD model for screening of potential therapeutic agents.
Collapse
|
36
|
Clatworthy AE, Romano KP, Hung DT. Whole-organism phenotypic screening for anti-infectives promoting host health. Nat Chem Biol 2018; 14:331-341. [PMID: 29556098 PMCID: PMC9843822 DOI: 10.1038/s41589-018-0018-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 11/20/2017] [Indexed: 01/19/2023]
Abstract
To date, antibiotics have been identified on the basis of their ability to kill bacteria or inhibit their growth rather than directly for their capacity to improve clinical outcomes of infected patients. Although historically successful, this approach has led to the development of an antibiotic armamentarium that suffers from a number of shortcomings, including the inevitable emergence of resistance and, in certain infections, suboptimal efficacy leading to long treatment durations, infection recurrence, or high mortality and morbidity rates despite apparent bacterial sterilization. Conventional antibiotics fail to address the complexities of in vivo bacterial physiology and virulence, as well as the role of the host underlying the complex, dynamic interactions that cause disease. New interventions are needed, aimed at host outcome rather than microbiological cure. Here we review the role of screening models for cellular and whole-organism infection, including worms, flies, zebrafish, and mice, to identify novel therapeutic strategies and discuss their future implications.
Collapse
Affiliation(s)
- Anne E. Clatworthy
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Keith P. Romano
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA,Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Deborah T. Hung
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA,Department of Genetics, Harvard Medical School, Boston, MA, USA,Correspondence and requests for materials should be addressed to D.T.H.
| |
Collapse
|
37
|
Modelling Cooperative Tumorigenesis in Drosophila. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4258387. [PMID: 29693007 PMCID: PMC5859872 DOI: 10.1155/2018/4258387] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/21/2018] [Indexed: 12/13/2022]
Abstract
The development of human metastatic cancer is a multistep process, involving the acquisition of several genetic mutations, tumour heterogeneity, and interactions with the surrounding microenvironment. Due to the complexity of cancer development in mammals, simpler model organisms, such as the vinegar fly, Drosophila melanogaster, are being utilized to provide novel insights into the molecular mechanisms involved. In this review, we highlight recent advances in modelling tumorigenesis using the Drosophila model, focusing on the cooperation of oncogenes or tumour suppressors, and the interaction of mutant cells with the surrounding tissue in epithelial tumour initiation and progression.
Collapse
|