1
|
Chi JT, Lin CC, Lin YT, Chen SY, Setayeshpour Y, Chen Y, Dunn D, Soderblom E, Zhang GF, Filonenko V, Jeong SY, Floyd S, Hayflick S, Gout I. Coenzyme A protects against ferroptosis via CoAlation of thioredoxin reductase 2. RESEARCH SQUARE 2024:rs.3.rs-4522617. [PMID: 38947036 PMCID: PMC11213209 DOI: 10.21203/rs.3.rs-4522617/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The Cystine-xCT transporter-Glutathione (GSH)-GPX4 axis is the canonical pathway to protect against ferroptosis. While not required for ferroptosis-inducing compounds (FINs) targeting GPX4, FINs targeting the xCT transporter require mitochondria and its lipid peroxidation to trigger ferroptosis. However, the mechanism underlying the difference between these FINs is still unknown. Given that cysteine is also required for coenzyme A (CoA) biosynthesis, here we show that CoA supplementation specifically prevents ferroptosis induced by xCT inhibitors but not GPX4 inhibitors. We find that, auranofin, a thioredoxin reductase inhibitor, abolishes the protective effect of CoA. We also find that CoA availability determines the enzymatic activity of thioredoxin reductase, but not thioredoxin. Importantly, the mitochondrial thioredoxin system, but not the cytosolic thioredoxin system, determines CoA-mediated ferroptosis inhibition. Our data show that the CoA regulates the in vitro enzymatic activity of mitochondrial thioredoxin reductase (TXNRD2) by covalently modifying the thiol group of cysteine (CoAlation) on Cys-483. Replacing Cys-483 with alanine on TXNRD2 abolishes its in vitro enzymatic activity and ability to protect cells from ferroptosis. Targeting xCT to limit cysteine import and, therefore, CoA biosynthesis reduced CoAlation on TXNRD2, an effect that was rescued by CoA supplementation. Furthermore, the fibroblasts from patients with disrupted CoA metabolism demonstrate increased mitochondrial lipid peroxidation. In organotypic brain slice cultures, inhibition of CoA biosynthesis leads to an oxidized thioredoxin system, mitochondrial lipid peroxidation, and loss in cell viability, which were all rescued by ferrostatin-1. These findings identify CoA-mediated post-translation modification to regulate the thioredoxin system as an alternative ferroptosis protection pathway with potential clinical relevance for patients with disrupted CoA metabolism.
Collapse
|
2
|
Singh A, Ansari VA, Mahmood T, Ahsan F, Maheshwari S. Repercussion of Primary Nucleation Pathway: Dementia and Cognitive Impairment. Curr Aging Sci 2024; 17:196-204. [PMID: 38083895 DOI: 10.2174/0118746098243327231117113748] [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: 01/01/2023] [Revised: 07/05/2023] [Accepted: 09/08/2023] [Indexed: 09/10/2024]
Abstract
Neurodegenerative diseases, such as Alzheimer's, Parkinson's, and prion disease, are characterized by the conversion of normally soluble proteins or peptides into aggregated amyloidal fibrils. These diseases result in the permanent loss of specific types of neurons, making them incurable and devastating. Research on animal models of memory problems mentioned in this article contributes to our knowledge of brain health and functionality. Neurodegenerative disorders, which often lead to cognitive impairment and dementia, are becoming more prevalent as global life expectancy increases. These diseases cause severe neurological impairment and neuronal death, making them highly debilitating. Exploring and understanding these complex diseases offer significant insights into the fundamental processes essential for maintaining brain health. Exploring the intricate mechanisms underlying neurodegenerative diseases not only holds promise for potential treatments but also enhances our understanding of fundamental brain health and functionality. By unraveling the complexities of these disorders, researchers can pave the way for advancements in diagnosis, treatment, and ultimately, improving the lives of individuals affected by neurodegenerative diseases.
Collapse
Affiliation(s)
- Aditya Singh
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Vaseem A Ansari
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Tarique Mahmood
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Farogh Ahsan
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | | |
Collapse
|
3
|
Singh A, Ansari VA, Ansari TM, Hasan SM, Ahsan F, Singh K, Wasim R, Maheshwari S, Ahmad A. Consequence of Dementia and Cognitive Impairment by Primary Nucleation Pathway. Horm Metab Res 2023; 55:304-314. [PMID: 37130536 DOI: 10.1055/a-2052-8462] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
An acquired loss of cognition in several cognitive domains that is severe enough to interfere with social or professional functioning is called dementia. As well as a moderately in-depth mental status examination by a clinician to identify impairments in memory, language, attention, visuospatial cognition, such as spatial orientation, executive function, and mood, the diagnosis of dementia requires a history evaluating for cognitive decline and impairment in daily activities, with confirmation from a close friend or family member. The start and organization of the cognitive assessment can be helped by short screening tests for cognitive impairment. Clinical presentations show that neurodegenerative diseases are often incurable because patients permanently lose some types of neurons. It has been determined through an assessment that, at best, our understanding of the underlying processes is still rudimentary, which presents exciting new targets for further study as well as the development of diagnostics and drugs. A growing body of research suggests that they also advance our knowledge of the processes that are probably crucial for maintaining the health and functionality of the brain. We concentrate on a number of the animal models of memory problems that have been mentioned in this review article because dementia has numerous etiologies. Serious neurological impairment and neuronal death are the main features of neurodegenerative illnesses, which are also extremely crippling ailments. The most prevalent neurodegenerative disorders are followed by those primary nucleation pathways responsible for cognitive impairment and dementia.
Collapse
Affiliation(s)
- Aditya Singh
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | | | | | | - Farogh Ahsan
- Faculty of Pharmacy, Integral University, Lucknow, India
| | - Kuldeep Singh
- Faculty of Pharmacy, Integral University, Lucknow, India
| | - Rufaida Wasim
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | | - Asad Ahmad
- Faculty of Pharmacy, Integral University, Lucknow, India
| |
Collapse
|
4
|
Ahamad S, Bhat SA. The Emerging Landscape of Small-Molecule Therapeutics for the Treatment of Huntington's Disease. J Med Chem 2022; 65:15993-16032. [PMID: 36490325 DOI: 10.1021/acs.jmedchem.2c00799] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene (HTT). The new insights into HD's cellular and molecular pathways have led to the identification of numerous potent small-molecule therapeutics for HD therapy. The field of HD-targeting small-molecule therapeutics is accelerating, and the approval of these therapeutics to combat HD may be expected in the near future. For instance, preclinical candidates such as naphthyridine-azaquinolone, AN1, AN2, CHDI-00484077, PRE084, EVP4593, and LOC14 have shown promise for further optimization to enter into HD clinical trials. This perspective aims to summarize the advent of small-molecule therapeutics at various stages of clinical development for HD therapy, emphasizing their structure and design, therapeutic effects, and specific mechanisms of action. Further, we have highlighted the key drivers involved in HD pathogenesis to provide insights into the basic principle for designing promising anti-HD therapeutic leads.
Collapse
Affiliation(s)
- Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh, Uttar Pradesh202002, India
| | - Shahnawaz A Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh, Uttar Pradesh202002, India
| |
Collapse
|
5
|
Koczwara KE, Lake NJ, DeSimone AM, Lek M. Neuromuscular disorders: finding the missing genetic diagnoses. Trends Genet 2022; 38:956-971. [PMID: 35908999 DOI: 10.1016/j.tig.2022.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/24/2022]
Abstract
Neuromuscular disorders (NMDs) are a wide-ranging group of diseases that seriously affect the quality of life of affected individuals. The development of next-generation sequencing revolutionized the diagnosis of NMD, enabling the discovery of hundreds of NMD genes and many more pathogenic variants. However, the diagnostic yield of genetic testing in NMD cohorts remains incomplete, indicating a large number of genetic diagnoses are not identified through current methods. Fortunately, recent advancements in sequencing technologies, analytical tools, and high-throughput functional screening provide an opportunity to circumvent current challenges. Here, we discuss reasons for missing genetic diagnoses in NMD, how emerging technologies and tools can overcome these hurdles, and examine future approaches to improving diagnostic yields in NMD.
Collapse
Affiliation(s)
- Katherine E Koczwara
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Nicole J Lake
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Alec M DeSimone
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Monkol Lek
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
6
|
Cell models for Alzheimer’s and Parkinson’s disease: At the interface of biology and drug discovery. Biomed Pharmacother 2022; 149:112924. [DOI: 10.1016/j.biopha.2022.112924] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 11/23/2022] Open
|
7
|
Benchoua A, Lasbareilles M, Tournois J. Contribution of Human Pluripotent Stem Cell-Based Models to Drug Discovery for Neurological Disorders. Cells 2021; 10:cells10123290. [PMID: 34943799 PMCID: PMC8699352 DOI: 10.3390/cells10123290] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
One of the major obstacles to the identification of therapeutic interventions for central nervous system disorders has been the difficulty in studying the step-by-step progression of diseases in neuronal networks that are amenable to drug screening. Recent advances in the field of human pluripotent stem cell (PSC) biology offers the capability to create patient-specific human neurons with defined clinical profiles using reprogramming technology, which provides unprecedented opportunities for both the investigation of pathogenic mechanisms of brain disorders and the discovery of novel therapeutic strategies via drug screening. Many examples not only of the creation of human pluripotent stem cells as models of monogenic neurological disorders, but also of more challenging cases of complex multifactorial disorders now exist. Here, we review the state-of-the art brain cell types obtainable from PSCs and amenable to compound-screening formats. We then provide examples illustrating how these models contribute to the definition of new molecular or functional targets for drug discovery and to the design of novel pharmacological approaches for rare genetic disorders, as well as frequent neurodegenerative diseases and psychiatric disorders.
Collapse
Affiliation(s)
- Alexandra Benchoua
- Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France;
- High Throughput Screening Platform, CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France;
- Correspondence:
| | - Marie Lasbareilles
- Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France;
- UEVE UMR 861, I-STEM, AFM, 91100 Corbeil-Essonnes, France
| | - Johana Tournois
- High Throughput Screening Platform, CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France;
| |
Collapse
|
8
|
Molinari C, Ruga S, Farghali M, Galla R, Fernandez-Godino R, Clemente N, Uberti F. Effects of a New Combination of Natural Extracts on Glaucoma-Related Retinal Degeneration. Foods 2021; 10:1885. [PMID: 34441662 PMCID: PMC8391439 DOI: 10.3390/foods10081885] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/09/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Glaucoma is currently the leading cause of irreversible blindness; it is a neuropathy characterized by structural alterations of the optic nerve, leading to visual impairments. The aim of this work is to develop a new oral formulation able to counteract the early changes connected to glaucomatous degeneration. The composition is based on gastrodin and vitamin D3 combined with vitamin C, blackcurrant, and lycopene. METHODS Cells and tissues of the retina were used to study biological mechanisms involved in glaucoma, to slow down the progression of the disease. Experiments mimicking the conditions of glaucoma were carried out to examine the etiology of retinal degeneration. RESULTS Our results show a significant ability to restore glaucoma-induced damage, by counteracting ROS production and promoting cell survival by inhibiting apoptosis. These effects were confirmed by the intracellular mechanism that was activated following administration of the compound, either before or after the glaucoma induction. In particular, the main results were obtained as a preventive action of glaucoma, showing a beneficial action on all selected markers, both on cells and on eyecup preparations. It is therefore possible to hypothesize both the preventive and therapeutic use of this formulation, in the presence of risk factors, and due to its ability to inhibit the apoptotic cycle and to stimulate cell survival mechanisms, respectively. CONCLUSION This formulation has exhibited an active role in the prevention or restoration of glaucoma damage for the first time.
Collapse
Affiliation(s)
- Claudio Molinari
- Laboratory of Physiology, Department of Translational Medicine, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.M.); (S.R.); (M.F.); (R.G.)
| | - Sara Ruga
- Laboratory of Physiology, Department of Translational Medicine, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.M.); (S.R.); (M.F.); (R.G.)
| | - Mahitab Farghali
- Laboratory of Physiology, Department of Translational Medicine, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.M.); (S.R.); (M.F.); (R.G.)
| | - Rebecca Galla
- Laboratory of Physiology, Department of Translational Medicine, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.M.); (S.R.); (M.F.); (R.G.)
| | - Rosario Fernandez-Godino
- Ocular Genomics Institute-Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02115, USA;
| | - Nausicaa Clemente
- Dipartimento di Scienze della Salute, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, 28100 Novara, Italy;
| | - Francesca Uberti
- Laboratory of Physiology, Department of Translational Medicine, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.M.); (S.R.); (M.F.); (R.G.)
| |
Collapse
|
9
|
Aldewachi H, Al-Zidan RN, Conner MT, Salman MM. High-Throughput Screening Platforms in the Discovery of Novel Drugs for Neurodegenerative Diseases. Bioengineering (Basel) 2021; 8:30. [PMID: 33672148 PMCID: PMC7926814 DOI: 10.3390/bioengineering8020030] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/05/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases (NDDs) are incurable and debilitating conditions that result in progressive degeneration and/or death of nerve cells in the central nervous system (CNS). Identification of viable therapeutic targets and new treatments for CNS disorders and in particular, for NDDs is a major challenge in the field of drug discovery. These difficulties can be attributed to the diversity of cells involved, extreme complexity of the neural circuits, the limited capacity for tissue regeneration, and our incomplete understanding of the underlying pathological processes. Drug discovery is a complex and multidisciplinary process. The screening attrition rate in current drug discovery protocols mean that only one viable drug may arise from millions of screened compounds resulting in the need to improve discovery technologies and protocols to address the multiple causes of attrition. This has identified the need to screen larger libraries where the use of efficient high-throughput screening (HTS) becomes key in the discovery process. HTS can investigate hundreds of thousands of compounds per day. However, if fewer compounds could be screened without compromising the probability of success, the cost and time would be largely reduced. To that end, recent advances in computer-aided design, in silico libraries, and molecular docking software combined with the upscaling of cell-based platforms have evolved to improve screening efficiency with higher predictability and clinical applicability. We review, here, the increasing role of HTS in contemporary drug discovery processes, in particular for NDDs, and evaluate the criteria underlying its successful application. We also discuss the requirement of HTS for novel NDD therapies and examine the major current challenges in validating new drug targets and developing new treatments for NDDs.
Collapse
Affiliation(s)
- Hasan Aldewachi
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield S1 1WB, UK;
- College of Pharmacy, Nineveh University, Mosul 41002, Iraq
| | - Radhwan N. Al-Zidan
- College of Pharmacy, University of Mosul, Mosul 41002, Iraq;
- School of Applied Sciences, Edinburgh Napier University, Edinburgh EH11 4BN, UK
| | - Matthew T. Conner
- School of Sciences, Research Institute in Healthcare Science, University of Wolverhampton, Wolverhampton WV1 1LY, UK;
| | - Mootaz M. Salman
- College of Pharmacy, University of Mosul, Mosul 41002, Iraq;
- Oxford Parkinson’s Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| |
Collapse
|
10
|
Pasteuning-Vuhman S, de Jongh R, Timmers A, Pasterkamp RJ. Towards Advanced iPSC-based Drug Development for Neurodegenerative Disease. Trends Mol Med 2020; 27:263-279. [PMID: 33121873 DOI: 10.1016/j.molmed.2020.09.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases (NDDs) are a heterogeneous group of diseases that are characterized by the progressive loss of neurons leading to motor, sensory, and/or cognitive defects. Currently, NDDs are not curable and treatment focuses on alleviating symptoms and halting disease progression. Phenotypic heterogeneity between individual NDD patients, lack of robust biomarkers, the limited translational potential of experimental models, and other factors have hampered drug development for the treatment of NDDs. This review summarizes and discusses the use of induced pluripotent stem cell (iPSC) approaches for improving drug discovery and testing. It highlights challenges associated with iPSC modeling and also discusses innovative approaches such as brain organoids and microfluidic-based technology which will improve drug development for NDDs.
Collapse
Affiliation(s)
- Svetlana Pasteuning-Vuhman
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Rianne de Jongh
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Annabel Timmers
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
11
|
Khalil AS, Jaenisch R, Mooney DJ. Engineered tissues and strategies to overcome challenges in drug development. Adv Drug Deliv Rev 2020; 158:116-139. [PMID: 32987094 PMCID: PMC7518978 DOI: 10.1016/j.addr.2020.09.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/29/2020] [Accepted: 09/23/2020] [Indexed: 12/16/2022]
Abstract
Current preclinical studies in drug development utilize high-throughput in vitro screens to identify drug leads, followed by both in vitro and in vivo models to predict lead candidates' pharmacokinetic and pharmacodynamic properties. The goal of these studies is to reduce the number of lead drug candidates down to the most likely to succeed in later human clinical trials. However, only 1 in 10 drug candidates that emerge from preclinical studies will succeed and become an approved therapeutic. Lack of efficacy or undetected toxicity represents roughly 75% of the causes for these failures, despite these parameters being the primary exclusion criteria in preclinical studies. Recently, advances in both biology and engineering have created new tools for constructing new preclinical models. These models can complement those used in current preclinical studies by helping to create more realistic representations of human tissues in vitro and in vivo. In this review, we describe current preclinical models to identify their value and limitations and then discuss select areas of research where improvements in preclinical models are particularly needed to advance drug development. Following this, we discuss design considerations for constructing preclinical models and then highlight recent advances in these efforts. Taken together, we aim to review the advances as of 2020 surrounding the prospect of biological and engineering tools for adding enhanced biological relevance to preclinical studies to aid in the challenges of failed drug candidates and the burden this poses on the drug development enterprise and thus healthcare.
Collapse
Affiliation(s)
- Andrew S Khalil
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA.
| |
Collapse
|
12
|
Patel-Murray NL, Adam M, Huynh N, Wassie BT, Milani P, Fraenkel E. A Multi-Omics Interpretable Machine Learning Model Reveals Modes of Action of Small Molecules. Sci Rep 2020; 10:954. [PMID: 31969612 PMCID: PMC6976599 DOI: 10.1038/s41598-020-57691-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/06/2020] [Indexed: 12/12/2022] Open
Abstract
High-throughput screening and gene signature analyses frequently identify lead therapeutic compounds with unknown modes of action (MoAs), and the resulting uncertainties can lead to the failure of clinical trials. We developed an approach for uncovering MoAs through an interpretable machine learning model of transcriptomics, epigenomics, metabolomics, and proteomics. Examining compounds with beneficial effects in models of Huntington's Disease, we found common MoAs for compounds with unrelated structures, connectivity scores, and binding targets. The approach also predicted highly divergent MoAs for two FDA-approved antihistamines. We experimentally validated these effects, demonstrating that one antihistamine activates autophagy, while the other targets bioenergetics. The use of multiple omics was essential, as some MoAs were virtually undetectable in specific assays. Our approach does not require reference compounds or large databases of experimental data in related systems and thus can be applied to the study of agents with uncharacterized MoAs and to rare or understudied diseases.
Collapse
Affiliation(s)
- Natasha L Patel-Murray
- Computational and Systems Biology Graduate Program, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Miriam Adam
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Nhan Huynh
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Brook T Wassie
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Pamela Milani
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
- Broad Institute, Cambridge, MA, 02139, USA.
| |
Collapse
|
13
|
Linsley JW, Reisine T, Finkbeiner S. Cell death assays for neurodegenerative disease drug discovery. Expert Opin Drug Discov 2019; 14:901-913. [PMID: 31179783 DOI: 10.1080/17460441.2019.1623784] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: Neurodegenerative diseases affect millions of people worldwide. Neurodegeneration is gradual over time, characterized by neuronal death that causes deterioration of cognitive or motor functions, ultimately leading to the patient's death. Currently, there are no treatments that effectively slow the progression of any neurodegenerative disease, but improved microscopy assays and models for neurodegeneration could lead the way to the discovery of disease-modifying therapeutics. Areas covered: Herein, the authors describe cell-based assays used to discover drugs with the potential to slow neurodegeneration, and their associated disease models. They focus on microscopy technologies that can be adapted to a high-throughput screening format that both detect cell death and monitor early signs of neurodegeneration and functional changes to identify drugs that the block early stages of neurodegeneration. Expert opinion: Many different phenotypes have been used in screens for the development of therapeutics towards neurodegenerative disease. The context of each phenotype in relation to neurodegeneration must be established to identify therapeutics likely to successfully target and treat disease. The use of improved models of neurodegeneration, statistical analyses, computational models, and improved markers of neuronal death will help in this pursuit and lead to better screening methods to identify therapeutic compounds against neurodegenerative disease.
Collapse
Affiliation(s)
- Jeremy W Linsley
- a Gladstone Center for Systems and Therapeutics , San Francisco , CA , USA
| | - Terry Reisine
- b Independent scientific consultant , Santa Cruz , CA , USA
| | - Steven Finkbeiner
- a Gladstone Center for Systems and Therapeutics , San Francisco , CA , USA.,c Neuroscience Graduate Program, University of California , San Francisco , CA , USA.,d Biomedical Sciences and Neuroscience Graduate Program, University of California , San Francisco , CA , USA.,e Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes , San Francisco , CA , USA.,f Department of Neurology, University of California , San Francisco , CA , USA.,g Department of Physiology, University of California , San Francisco , CA , USA
| |
Collapse
|
14
|
Use of human pluripotent stem cell-derived cells for neurodegenerative disease modeling and drug screening platform. Future Med Chem 2019; 11:1305-1322. [DOI: 10.4155/fmc-2018-0520] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Most neurodegenerative diseases are characterized by a complex and mostly still unresolved pathology. This fact, together with the lack of reliable disease models, has precluded the development of effective therapies counteracting the disease progression. The advent of human pluripotent stem cells has revolutionized the field allowing the generation of disease-relevant neural cell types that can be used for disease modeling, drug screening and, possibly, cell transplantation purposes. In this Review, we discuss the applications of human pluripotent stem cells, the development of efficient protocols for the derivation of the different neural cells and their applicability for robust in vitro disease modeling and drug screening platforms for most common neurodegenerative conditions.
Collapse
|
15
|
Abdolvahabi A, Rasouli S, Croom CM, Plewman DL. High-Throughput Microplate-Based Fluorescence Assays for Studying Stochastic Aggregation of Superoxide Dismutase-1. Methods Mol Biol 2019; 1873:93-108. [PMID: 30341605 DOI: 10.1007/978-1-4939-8820-4_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Investigating in vitro kinetics of superoxide dismutase-1 (SOD1) aggregation with high-throughput microplate-based assays provides valuable information regarding SOD1 pathogenesis in amyotrophic lateral sclerosis (ALS) and opens venues for the development of effective therapies. In this chapter, we first explain the step-by-step purification and demetallation of wild-type (WT) and ALS-variant SOD1 proteins from Saccharomyces cerevisiae (baker's yeast). We then describe the methodology for a microplate-based fluorescence assay that is used to study real-time kinetics of metal-free (apo)-SOD1 aggregation. This technique is highly sensitive, semiautomated, requires minimum modifications to protein, and produces a plethora of data in a short period of time. We also describe a new approach for extracting clinically relevant information from SOD1 aggregation data using Kaplan-Meier estimators.
Collapse
Affiliation(s)
- Alireza Abdolvahabi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Sanaz Rasouli
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
- Institute of Biomedical Studies, Baylor University, Waco, TX, USA
| | - Corbin M Croom
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Devon L Plewman
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| |
Collapse
|
16
|
Esteves S, Duarte-Silva S, Maciel P. Discovery of Therapeutic Approaches for Polyglutamine Diseases: A Summary of Recent Efforts. Med Res Rev 2016; 37:860-906. [PMID: 27870126 DOI: 10.1002/med.21425] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/01/2016] [Accepted: 10/05/2016] [Indexed: 12/19/2022]
Abstract
Polyglutamine (PolyQ) diseases are a group of neurodegenerative disorders caused by the expansion of cytosine-adenine-guanine (CAG) trinucleotide repeats in the coding region of specific genes. This leads to the production of pathogenic proteins containing critically expanded tracts of glutamines. Although polyQ diseases are individually rare, the fact that these nine diseases are irreversibly progressive over 10 to 30 years, severely impairing and ultimately fatal, usually implicating the full-time patient support by a caregiver for long time periods, makes their economic and social impact quite significant. This has led several researchers worldwide to investigate the pathogenic mechanism(s) and therapeutic strategies for polyQ diseases. Although research in the field has grown notably in the last decades, we are still far from having an effective treatment to offer patients, and the decision of which compounds should be translated to the clinics may be very challenging. In this review, we provide a comprehensive and critical overview of the most recent drug discovery efforts in the field of polyQ diseases, including the most relevant findings emerging from two different types of approaches-hypothesis-based candidate molecule testing and hypothesis-free unbiased drug screenings. We hereby summarize and reflect on the preclinical studies as well as all the clinical trials performed to date, aiming to provide a useful framework for increasingly successful future drug discovery and development efforts.
Collapse
Affiliation(s)
- Sofia Esteves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, University of Minho, Guimarães, Braga, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, University of Minho, Guimarães, Braga, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, University of Minho, Guimarães, Braga, Portugal
| |
Collapse
|
17
|
Jahnke HG, Krinke D, Seidel D, Lilienthal K, Schmidt S, Azendorf R, Fischer M, Mack T, Striggow F, Althaus H, Schober A, Robitzki AA. A novel 384-multiwell microelectrode array for the impedimetric monitoring of Tau protein induced neurodegenerative processes. Biosens Bioelectron 2016; 88:78-84. [PMID: 27506337 DOI: 10.1016/j.bios.2016.07.074] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/20/2016] [Accepted: 07/22/2016] [Indexed: 01/13/2023]
Abstract
Over the last decades, countless bioelectronic monitoring systems were developed for the analysis of cells as well as complex tissues. Most studies addressed the sensitivity and specificity of the bioelectronic detection method in comparison to classical molecular biological assays. In contrast, the up scaling as a prerequisite for the practical application of these novel bioelectronic monitoring systems is mostly only discussed theoretically. In this context, we developed a novel 384-multiwell microelectrode array (MMEA) based measurement system for the sensitive label-free real-time monitoring of neurodegenerative processes by impedance spectroscopy. With respect to the needs of productive screening systems for robust and reproducible measurements on high numbers of plates, we focused on reducing the critical contacting of more than 400 electrodes for a 384-MMEA. Therefore, we introduced an on top array of immersive counter electrodes that are individually addressed by a multiplexer and connected all measurement electrodes on the 384-MMEA to a single contact point. More strikingly, our novel approach provided a comparable signal stability and sensitivity similar to an array with integrated counter electrodes. Next, we optimized a SH-SY5Y cell based tauopathy model by introducing a novel 5-fold Tau mutation eliminating the need of artificial tauopathy induction. In combination with our novel 384-MMEA based measurement system, the concentration and time dependent neuroregenerative effect of the kinase inhibitor SRN-003-556 could be quantitatively monitored. Thus, our novel screening system could be a useful tool to identify and develop potential novel therapeutics in the field of Tau-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Heinz-Georg Jahnke
- Centre for Biotechnology and Biomedicine, Molecular biological-biochemical Processing Technology, Leipzig University, Deutscher Platz 5, Leipzig, D-04103 Germany
| | - Dana Krinke
- Centre for Biotechnology and Biomedicine, Molecular biological-biochemical Processing Technology, Leipzig University, Deutscher Platz 5, Leipzig, D-04103 Germany
| | - Diana Seidel
- Centre for Biotechnology and Biomedicine, Molecular biological-biochemical Processing Technology, Leipzig University, Deutscher Platz 5, Leipzig, D-04103 Germany
| | - Katharina Lilienthal
- Institute of Micro, and Nanotechnologies MacroNano®, Nano-biosystem Technology (Microfluidics and Biosensors group), Technische Universität Ilmenau, Gustav-Kirchhoff-Straße 7, Ilmenau, 98693 Germany
| | - Sabine Schmidt
- Centre for Biotechnology and Biomedicine, Molecular biological-biochemical Processing Technology, Leipzig University, Deutscher Platz 5, Leipzig, D-04103 Germany
| | - Ronny Azendorf
- Centre for Biotechnology and Biomedicine, Molecular biological-biochemical Processing Technology, Leipzig University, Deutscher Platz 5, Leipzig, D-04103 Germany
| | - Michael Fischer
- Institute of Micro, and Nanotechnologies MacroNano®, Nano-biosystem Technology (Microfluidics and Biosensors group), Technische Universität Ilmenau, Gustav-Kirchhoff-Straße 7, Ilmenau, 98693 Germany
| | - Till Mack
- KeyNeurotek Pharmaceuticals AG, Zenit Technologiepark, Leipziger Str. 44, Magdeburg, 39120 Germany; Department of Neurodegeneration and Intervention Strategies, German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, D-39120 Germany
| | - Frank Striggow
- KeyNeurotek Pharmaceuticals AG, Zenit Technologiepark, Leipziger Str. 44, Magdeburg, 39120 Germany; Department of Neurodegeneration and Intervention Strategies, German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, Magdeburg, D-39120 Germany
| | - Holger Althaus
- Fraunhofer Institute for Factory Operation and Automation IFF, Magdeburg, Germany
| | - Andreas Schober
- Institute of Micro, and Nanotechnologies MacroNano®, Nano-biosystem Technology (Microfluidics and Biosensors group), Technische Universität Ilmenau, Gustav-Kirchhoff-Straße 7, Ilmenau, 98693 Germany
| | - Andrea A Robitzki
- Centre for Biotechnology and Biomedicine, Molecular biological-biochemical Processing Technology, Leipzig University, Deutscher Platz 5, Leipzig, D-04103 Germany.
| |
Collapse
|
18
|
Macchi F, Deleersnijder A, Van den Haute C, Munck S, Pottel H, Michiels A, Debyser Z, Gerard M, Baekelandt V. High-content analysis of α-synuclein aggregation and cell death in a cellular model of Parkinson's disease. J Neurosci Methods 2015; 261:117-27. [PMID: 26620202 DOI: 10.1016/j.jneumeth.2015.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 11/16/2015] [Accepted: 11/16/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alpha-synuclein (α-SYN) aggregates represent a key feature of Parkinson's disease, but the exact relationship between α-SYN aggregation and neurodegeneration remains incompletely understood. Therefore, the availability of a cellular assay that allows medium-throughput analysis of α-SYN-linked pathology will be of great value for studying the aggregation process and for advancing α-SYN-based therapies. NEW METHOD Here we describe a high-content neuronal cell assay that simultaneously measures oxidative stress-induced α-SYN aggregation and apoptosis. RESULTS We optimized an automated and reproducible assay to quantify both α-SYN aggregation and cell death in human SH-SY5Y neuroblastoma cells. COMPARISON WITH EXISTING METHODS Quantification of α-SYN aggregates in cells has typically relied on manual imaging and counting or cell-free assays, which are time consuming and do not allow a concurrent analysis of cell viability. Our high-content analysis method for quantification of α-SYN aggregation allows simultaneous measurements of multiple cell parameters at a single-cell level in a fast, objective and automated manner. CONCLUSIONS The presented analysis approach offers a rapid, objective and multiparametric approach for the screening of compounds and genes that might alter α-SYN aggregation and/or toxicity.
Collapse
Affiliation(s)
- Francesca Macchi
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Kapucijnenvoer 33, Leuven B-3000, Flanders, Belgium
| | - Angélique Deleersnijder
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Kapucijnenvoer 33, Leuven B-3000, Flanders, Belgium
| | - Chris Van den Haute
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Kapucijnenvoer 33, Leuven B-3000, Flanders, Belgium; Leuven Viral Vector Core, KU Leuven, Leuven B-3000, Flanders, Belgium
| | - Sebastian Munck
- KU Leuven, Department of Human Genetics, Flanders Interuniversity Institute of Biotechnology, Kapucijnenvoer 33, Leuven B-3000, Flanders, Belgium
| | - Hans Pottel
- KU Leuven Campus Kulak Kortrijk, Public Health and Primary Care, Interdisciplinary Research Facility Life Sciences, Etienne Sabbelaan 53, Kortrijk B-8500, Flanders, Belgium
| | - Annelies Michiels
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Kapucijnenvoer 33, Leuven B-3000, Flanders, Belgium; Leuven Viral Vector Core, KU Leuven, Leuven B-3000, Flanders, Belgium
| | - Zeger Debyser
- Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven B-3000, Flanders, Belgium
| | - Melanie Gerard
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Kapucijnenvoer 33, Leuven B-3000, Flanders, Belgium; KU Leuven campus Kulak Kortrijk, Laboratory of Biochemistry, Interdisciplinary Research Facility Life Sciences, Etienne Sabbelaan 53, Kortrijk B-8500, Flanders, Belgium
| | - Veerle Baekelandt
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Kapucijnenvoer 33, Leuven B-3000, Flanders, Belgium.
| |
Collapse
|
19
|
Abstract
High-throughput screens (HTS ) are powerful tools that permit the rapid evaluation of thousands of samples in a cost-effective manner and minimize sample and reagent consumption. Such assays have recently begun to be utilized to evaluate cell death modalities and also the cytoprotective efficacy of compounds against a wide variety of stresses. Here we describe the design, preparation, and undertaking of HTS-appropriate assays that utilize simple and cost-effective fluorophore - and luminescence -based functional readouts of cell viability. These assays permit the examination of 96–384 compounds in a single multiwell plate with highly robust statistical significance at a fraction of the financial and work cost of traditional approaches.
Collapse
|
20
|
Xu J, Lacoske MH, Theodorakis EA. Neurotrophic natural products: chemistry and biology. Angew Chem Int Ed Engl 2014; 53:956-87. [PMID: 24353244 PMCID: PMC3945720 DOI: 10.1002/anie.201302268] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases and spinal cord injury affect approximately 50 million people worldwide, bringing the total healthcare cost to over 600 billion dollars per year. Nervous system growth factors, that is, neurotrophins, are a potential solution to these disorders, since they could promote nerve regeneration. An average of 500 publications per year attests to the significance of neurotrophins in biomedical sciences and underlines their potential for therapeutic applications. Nonetheless, the poor pharmacokinetic profile of neurotrophins severely restricts their clinical use. On the other hand, small molecules that modulate neurotrophic activity offer a promising therapeutic approach against neurological disorders. Nature has provided an impressive array of natural products that have potent neurotrophic activities. This Review highlights the current synthetic strategies toward these compounds and summarizes their ability to induce neuronal growth and rehabilitation. It is anticipated that neurotrophic natural products could be used not only as starting points in drug design but also as tools to study the next frontier in biomedical sciences: the brain activity map project.
Collapse
Affiliation(s)
- Jing Xu
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0358 (USA), Homepage: http://theodorakisgroup.ucsd.edu
| | - Michelle H. Lacoske
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0358 (USA), Homepage: http://theodorakisgroup.ucsd.edu
| | - Emmanuel A. Theodorakis
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0358 (USA), Homepage: http://theodorakisgroup.ucsd.edu
| |
Collapse
|
21
|
Xu J, Lacoske MH, Theodorakis EA. Neurotrophe Naturstoffe - ihre Chemie und Biologie. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201302268] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
22
|
La Rosa S, Benicchi T, Bettinetti L, Ceccarelli I, Diodato E, Federico C, Fiengo P, Franceschini D, Gokce O, Heitz F, Lazzeroni G, Luthi-Carter R, Magnoni L, Miragliotta V, Scali C, Valacchi M. Fused 3-Hydroxy-3-trifluoromethylpyrazoles Inhibit Mutant Huntingtin Toxicity. ACS Med Chem Lett 2013; 4:979-84. [PMID: 24900595 PMCID: PMC4027250 DOI: 10.1021/ml400251g] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 08/08/2013] [Indexed: 11/30/2022] Open
Abstract
Here, we describe the selection and optimization of a chemical series active in both a full-length and a fragment-based Huntington's disease (HD) assay. Twenty-four thousand small molecules were screened in a phenotypic HD assay, identifying a series of compounds bearing a 3-hydroxy-3-trifluoromethylpyrazole moiety as able to revert the toxicity induced by full-length mutant Htt by up to 50%. A chemical exploration around the series led to the identification of compound 4f, which demonstrated to be active in a Htt171-82Q rat primary striatal neuron assay and a PC12-Exon-1 based assay. This compound was selected for testing in R6/2 mice, in which it was well-tolerated and showed a positive effect on body weight and a positive trend in preventing ventricular volume enlargment. These studies provide strong rationale for further testing the potential benefits of 3-hydroxy-3-trifluoromethylpyrazoles in treating HD.
Collapse
Affiliation(s)
- Salvatore La Rosa
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Tiziana Benicchi
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Laura Bettinetti
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Ilaria Ceccarelli
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Enrica Diodato
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Cesare Federico
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Pasquale Fiengo
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Davide Franceschini
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Ozgun Gokce
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL),
Lausanne, Switzerland
| | - Freddy Heitz
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Giulia Lazzeroni
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Ruth Luthi-Carter
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL),
Lausanne, Switzerland
| | - Letizia Magnoni
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | | | - Carla Scali
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| | - Michela Valacchi
- Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy
| |
Collapse
|
23
|
Titus SA, Southall N, Marugan J, Austin CP, Zheng W. High-Throughput Multiplexed Quantitation of Protein Aggregation and Cytotoxicity in a Huntington's Disease Model. CURRENT CHEMICAL GENOMICS 2012; 6:79-86. [PMID: 23346268 PMCID: PMC3551243 DOI: 10.2174/1875397301206010079] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/05/2012] [Accepted: 11/05/2012] [Indexed: 11/22/2022]
Abstract
A hallmark of Huntington’s disease is the presence of a large polyglutamine expansion in the first exon of the Huntingtin protein and the propensity of protein aggregation by the mutant proteins. Aberrant protein aggregation also occurs in other polyglutamine expansion disorders, as well as in other neurodegenerative diseases including Parkinson’s, Alzheimer’s, and prion diseases. However, the pathophysiological role of these aggregates in the cell death that characterizes the diseases remains unclear. Identification of small molecule probes that modulate protein aggregation and cytotoxicity caused by aggregated proteins may greatly facilitate the studies on pathogenesis of these diseases and potentially lead to development of new therapies. Based on a detergent insoluble property of the Huntingtin protein aggregates, we have developed a homogenous assay to rapidly quantitate the levels of protein aggregates in a cellular model of Huntington’s disease. The protein aggregation assay has also been multiplexed with a protease release assay for the measurement of cytotoxicity resulting from aggregated proteins in the same cells. Through a testing screen of a compound library, we have demonstrated that this multiplexed cytotoxicity and protein aggregation assay has ability to identify active compounds that prevent cell death and/or modulate protein aggregation in cells of the Huntington’s disease model. Therefore, this multiplexed screening approach is also useful for development of high-throughput screening assays for other neurodegenerative diseases involving protein aggregation.
Collapse
Affiliation(s)
- Steven A Titus
- National Center for Advancing Translational Sciences, National Institutes of Health. 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
24
|
Abstract
The functional annotation of genomes, construction of molecular networks and novel drug target identification, are important challenges that need to be addressed as a matter of great urgency1-4. Multiple complementary 'omics' approaches have provided clues as to the genetic risk factors and pathogenic mechanisms underlying numerous neurodegenerative diseases, but most findings still require functional validation5. For example, a recent genome wide association study for Parkinson's Disease (PD), identified many new loci as risk factors for the disease, but the underlying causative variant(s) or pathogenic mechanism is not known6, 7. As each associated region can contain several genes, the functional evaluation of each of the genes on phenotypes associated with the disease, using traditional cell biology techniques would take too long. There is also a need to understand the molecular networks that link genetic mutations to the phenotypes they cause. It is expected that disease phenotypes are the result of multiple interactions that have been disrupted. Reconstruction of these networks using traditional molecular methods would be time consuming. Moreover, network predictions from independent studies of individual components, the reductionism approach, will probably underestimate the network complexity8. This underestimation could, in part, explain the low success rate of drug approval due to undesirable or toxic side effects. Gaining a network perspective of disease related pathways using HT/HC cellular screening approaches, and identifying key nodes within these pathways, could lead to the identification of targets that are more suited for therapeutic intervention. High-throughput screening (HTS) is an ideal methodology to address these issues9-12. but traditional methods were one dimensional whole-well cell assays, that used simplistic readouts for complex biological processes. They were unable to simultaneously quantify the many phenotypes observed in neurodegenerative diseases such as axonal transport deficits or alterations in morphology properties13, 14. This approach could not be used to investigate the dynamic nature of cellular processes or pathogenic events that occur in a subset of cells. To quantify such features one has to move to multi-dimensional phenotypes termed high-content screening (HCS)4, 15-17. HCS is the cell-based quantification of several processes simultaneously, which provides a more detailed representation of the cellular response to various perturbations compared to HTS. HCS has many advantages over HTS18, 19, but conducting a high-throughput (HT)-high-content (HC) screen in neuronal models is problematic due to high cost, environmental variation and human error. In order to detect cellular responses on a 'phenomics' scale using HC imaging one has to reduce variation and error, while increasing sensitivity and reproducibility. Herein we describe a method to accurately and reliably conduct shRNA screens using automated cell culturing20 and HC imaging in neuronal cellular models. We describe how we have used this methodology to identify modulators for one particular protein, DJ1, which when mutated causes autosomal recessive parkinsonism21. Combining the versatility of HC imaging with HT methods, it is possible to accurately quantify a plethora of phenotypes. This could subsequently be utilized to advance our understanding of the genome, the pathways involved in disease pathogenesis as well as identify potential therapeutic targets.
Collapse
Affiliation(s)
- Shushant Jain
- Department of Clinical Genetics, VU University Medical Center.
| | | | | |
Collapse
|
25
|
Towards a more robust approach to selecting and prosecuting promising targets and compounds. Future Med Chem 2011; 2:25-34. [PMID: 21426044 DOI: 10.4155/fmc.09.135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
There are many factors that influence predictivity in drug discovery and impact on productivity within the pharmaceutical industry. This article will concentrate on just two aspects; first, the role of investigating target modulation within a (human) disease setting, from target selection through screening to animal models, and second, potential developments in the analysis and probing of the chemical space appropriate for drug discovery and, in particular, steps to improve predictivity thus moving to a more forward-looking process. The activities associated with target selection should develop significantly over the next 5-10 years leading to a more robust association of target modulation with disease modification. In addition, better understanding of the opportunities for target modulators should drive and improve the selection of ligands suitable for therapeutic applications. Within these areas it will be important to move away from a retrospective consideration of druggable targets towards a forward-looking approach based on holistic (disease context) profiling of both (progressable) targets and subsequently their ligands. Improvements in the predictive analysis and probing of the chemical space will be needed to confront both safety and efficacy end points that currently remain major reasons for failure in the clinic. It is hoped that improvements in data visualization together with chemocentric mining of the literature will facilitate better interrogation of development and clinical data, potentially modifying research project plans to better address these key issues.
Collapse
|
26
|
Reinhart PH, Kaltenbach LS, Essrich C, Dunn DE, Eudailey JA, DeMarco CT, Turmel GJ, Whaley JC, Wood A, Cho S, Lo DC. Identification of anti-inflammatory targets for Huntington's disease using a brain slice-based screening assay. Neurobiol Dis 2011; 43:248-56. [PMID: 21458569 DOI: 10.1016/j.nbd.2011.03.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 02/02/2011] [Accepted: 03/23/2011] [Indexed: 10/18/2022] Open
Abstract
Huntington's disease (HD) is a late-onset, neurodegenerative disease for which there are currently no cures nor disease-modifying treatments. Here we report the identification of several potential anti-inflammatory targets for HD using an ex vivo model of HD that involves the acute transfection of human mutant huntingtin-based constructs into rat brain slices. This model recapitulates key components of the human disease, including the formation of intracellular huntingtin protein (HTT)-containing inclusions and the progressive neurodegeneration of striatal neurons-both occurring within the native tissue context of these neurons. Using this "high-throughput biology" screening platform, we conducted a hypothesis-neutral screen of a collection of drug-like compounds which identified several anti-inflammatory targets that provided neuroprotection against HTT fragment-induced neurodegeneration. The nature of these targets provide further support for non-cell autonomous mechanisms mediating significant aspects of neuropathogenesis induced by mutant HTT fragment proteins.
Collapse
Affiliation(s)
- Peter H Reinhart
- Discovery Neuroscience, Wyeth Research, Princeton, NJ 08543, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kaltenbach LS, Bolton MM, Shah B, Kanju PM, Lewis GM, Turmel GJ, Whaley JC, Trask OJ, Lo DC. Composite primary neuronal high-content screening assay for Huntington's disease incorporating non-cell-autonomous interactions. ACTA ACUST UNITED AC 2010; 15:806-19. [PMID: 20581077 DOI: 10.1177/1087057110373392] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disease characterized by progressive cognitive, behavioral, and motor deficits and caused by expansion of a polyglutamine repeat in the Huntingtin protein (Htt). Despite its monogenic nature, HD pathogenesis includes obligatory non-cell-autonomous pathways involving both the cortex and the striatum, and therefore effective recapitulation of relevant HD disease pathways in cell lines and primary neuronal monocultures is intrinsically limited. To address this, the authors developed an automated high-content imaging screen in high-density primary cultures of cortical and striatal neurons together with supporting glial cells. Cortical and striatal neurons are transfected separately with different fluorescent protein markers such that image-based high-content analysis can be used to assay these neuronal populations separately but still supporting their intercellular interactions, including abundant synaptic interconnectivity. This assay was reduced to practice using transfection of a mutant N-terminal Htt domain and validated via a screen of ~400 selected small molecules. Both expected as well as novel candidate targets for HD emerged from this screen; of particular interest were target classes with close relative proximity to clinical testing. These findings suggest that composite primary cultures incorporating increased levels of biological complexity can be used for high-content imaging and "high-context" screening to represent molecular targets that otherwise may be operant only in the complex tissue environment found in vivo during disease pathogenesis.
Collapse
Affiliation(s)
- Linda S Kaltenbach
- Center for Drug Discovery and Department of Neurobiology, Duke University Medical Center, Durham, NC 27704, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Adding more content to screening: reactivation of FOXO as a therapeutic strategy. Clin Transl Oncol 2009; 11:651-8. [DOI: 10.1007/s12094-009-0420-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
29
|
Fecke W, Gianfriddo M, Gaviraghi G, Terstappen GC, Heitz F. Small molecule drug discovery for Huntington's Disease. Drug Discov Today 2009; 14:453-64. [DOI: 10.1016/j.drudis.2009.02.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Revised: 02/06/2009] [Accepted: 02/13/2009] [Indexed: 10/21/2022]
|