1
|
Maniam S, Maniam S. Screening Techniques for Drug Discovery in Alzheimer's Disease. ACS OMEGA 2024; 9:6059-6073. [PMID: 38371787 PMCID: PMC10870277 DOI: 10.1021/acsomega.3c07046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 02/20/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by progressive and irreversible impairment of memory and other cognitive functions of the aging brain. Pathways such as amyloid beta neurotoxicity, tau pathogenesis and neuroinflammatory have been used to understand AD, despite not knowing the definite molecular mechanism which causes this progressive disease. This review attempts to summarize the small molecules that target these pathways using various techniques involving high-throughput screening, molecular modeling, custom bioassays, and spectroscopic detection tools. Novel and evolving screening methods developed to advance drug discovery initiatives in AD research are also highlighted.
Collapse
Affiliation(s)
- Sandra Maniam
- Department
of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia
| | - Subashani Maniam
- School
of Science, STEM College, RMIT University, Melbourne, Victoria 3001, Australia
| |
Collapse
|
2
|
Stellon D, Talbot J, Hewitt AW, King AE, Cook AL. Seeing Neurodegeneration in a New Light Using Genetically Encoded Fluorescent Biosensors and iPSCs. Int J Mol Sci 2023; 24:1766. [PMID: 36675282 PMCID: PMC9861453 DOI: 10.3390/ijms24021766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Neurodegenerative diseases present a progressive loss of neuronal structure and function, leading to cell death and irrecoverable brain atrophy. Most have disease-modifying therapies, in part because the mechanisms of neurodegeneration are yet to be defined, preventing the development of targeted therapies. To overcome this, there is a need for tools that enable a quantitative assessment of how cellular mechanisms and diverse environmental conditions contribute to disease. One such tool is genetically encodable fluorescent biosensors (GEFBs), engineered constructs encoding proteins with novel functions capable of sensing spatiotemporal changes in specific pathways, enzyme functions, or metabolite levels. GEFB technology therefore presents a plethora of unique sensing capabilities that, when coupled with induced pluripotent stem cells (iPSCs), present a powerful tool for exploring disease mechanisms and identifying novel therapeutics. In this review, we discuss different GEFBs relevant to neurodegenerative disease and how they can be used with iPSCs to illuminate unresolved questions about causes and risks for neurodegenerative disease.
Collapse
Affiliation(s)
- David Stellon
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, TAS 7000, Australia
| | - Jana Talbot
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, TAS 7000, Australia
| | - Alex W. Hewitt
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Anna E. King
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, TAS 7000, Australia
| | - Anthony L. Cook
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, TAS 7000, Australia
| |
Collapse
|
3
|
Potekhina ES, Bass DY, Kelmanson IV, Fetisova ES, Ivanenko AV, Belousov VV, Bilan DS. Drug Screening with Genetically Encoded Fluorescent Sensors: Today and Tomorrow. Int J Mol Sci 2020; 22:E148. [PMID: 33375682 PMCID: PMC7794770 DOI: 10.3390/ijms22010148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Genetically-encoded fluorescent sensors have been actively developed over the last few decades and used in live imaging and drug screening. Real-time monitoring of drug action in a specific cellular compartment, organ, or tissue type; the ability to screen at the single-cell resolution; and the elimination of false-positive results caused by low drug bioavailability that is not detected by in vitro testing methods are a few of the obvious benefits of using genetically-encoded fluorescent sensors in drug screening. In combination with high-throughput screening (HTS), some genetically-encoded fluorescent sensors may provide high reproducibility and robustness to assays. We provide a brief overview of successful, perspective, and hopeful attempts at using genetically encoded fluorescent sensors in HTS of modulators of ion channels, Ca2+ homeostasis, GPCR activity, and for screening cytotoxic, anticancer, and anti-parasitic compounds. We discuss the advantages of sensors in whole organism drug screening models and the perspectives of the combination of human disease modeling by CRISPR techniques with genetically encoded fluorescent sensors for drug screening.
Collapse
Affiliation(s)
- Ekaterina S. Potekhina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Dina Y. Bass
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Ilya V. Kelmanson
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Elena S. Fetisova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
| | - Alexander V. Ivanenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Vsevolod V. Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency, 117997 Moscow, Russia
| | - Dmitry S. Bilan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
4
|
Effect specificity of the early intervention with moxibustion at “BL23” in Alzheimer's disease rats modeled by ovariectomy and D-Galactose injection 艾灸“肾俞”早期干预去卵巢合D-半乳糖注射AD样大鼠效应中的特异性. WORLD JOURNAL OF ACUPUNCTURE-MOXIBUSTION 2020. [DOI: 10.1016/j.wjam.2020.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
5
|
Yu N, Liu Y, Wang S, Tang X, Ge P, Nan J, Zhang J, Yang B. Pressure-controlled microfluidic sub-picoliter ultramicro-volume syringes based on integrated micro-nanostructure arrays. LAB ON A CHIP 2019; 19:3368-3374. [PMID: 31549119 DOI: 10.1039/c9lc00730j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Ultramicro-volume syringes were fabricated by integrating micro-nanostructure arrays in microchannels for quantitatively dispensing sub-picoliter volumes of liquids. Using this system, liquids were dispensed in volume increments as low as 0.5 pL with 96% accuracy. Specifically, the controllable synthesis of nanocrystals was achieved using a lab-on-chip platform that was integrated with the syringes.
Collapse
Affiliation(s)
- Nianzuo Yu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Czeredys M, Vigont VA, Boeva VA, Mikoshiba K, Kaznacheyeva EV, Kuznicki J. Huntingtin-Associated Protein 1A Regulates Store-Operated Calcium Entry in Medium Spiny Neurons From Transgenic YAC128 Mice, a Model of Huntington's Disease. Front Cell Neurosci 2018; 12:381. [PMID: 30455632 PMCID: PMC6231533 DOI: 10.3389/fncel.2018.00381] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/05/2018] [Indexed: 12/31/2022] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disease that is caused by polyglutamine expansion within the huntingtin (HTT) gene. One of the cellular activities that is dysregulated in HD is store-operated calcium entry (SOCE), a process by which Ca2+ release from the endoplasmic reticulum (ER) induces Ca2+ influx from the extracellular space. HTT-associated protein-1 (HAP1) is a binding partner of HTT. The aim of the present study was to examine the role of HAP1A protein in regulating SOCE in YAC128 mice, a transgenic model of HD. After Ca2+ depletion from the ER by the activation of inositol-(1,4,5)triphosphate receptor type 1 (IP3R1), we detected an increase in the activity of SOC channels when HAP1 protein isoform HAP1A was overexpressed in medium spiny neurons (MSNs) from YAC128 mice. A decrease in the activity of SOC channels in YAC128 MSNs was observed when HAP1 protein was silenced. In YAC128 MSNs that overexpressed HAP1A, an increase in activity of IP3R1 was detected while the ionomycin-sensitive ER Ca2+ pool decreased. 6-Bromo-N-(2-phenylethyl)-2,3,4,9-tetrahydro-1H-carbazol-1-amine hydrochloride (C20H22BrClN2), identified in our previous studies as a SOCE inhibitor, restored the elevation of SOCE in YAC128 MSN cultures that overexpressed HAP1A. The IP3 sponge also restored the elevation of SOCE and increased the release of Ca2+ from the ER in YAC128 MSN cultures that overexpressed HAP1A. The overexpression of HAP1A in the human neuroblastoma cell line SK-N-SH (i.e., a cellular model of HD (SK-N-SH HTT138Q)) led to the appearance of a pool of constitutively active SOC channels and an increase in the expression of STIM2 protein. Our results showed that HAP1A causes the activation of SOC channels in HD models by affecting IP3R1 activity.
Collapse
Affiliation(s)
- Magdalena Czeredys
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw (IIMCB), Warsaw, Poland
| | - Vladimir A Vigont
- Institute of Cytology, Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Vasilisa A Boeva
- Institute of Cytology, Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute (BSI), Saitama, Japan
| | - Elena V Kaznacheyeva
- Institute of Cytology, Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Jacek Kuznicki
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw (IIMCB), Warsaw, Poland
| |
Collapse
|
7
|
Cornaglia M, Lehnert T, Gijs MAM. Microfluidic systems for high-throughput and high-content screening using the nematode Caenorhabditis elegans. LAB ON A CHIP 2017; 17:3736-3759. [PMID: 28840220 DOI: 10.1039/c7lc00509a] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In a typical high-throughput drug screening (HTS) process, up to millions of chemical compounds are applied to cells cultured in well plates, aiming to find molecules that exhibit a robust dose-response, as evidenced for example by a fluorescence signal. In high-content screening (HCS), one goes a step further by linking the tested compounds to phenotypic information, obtained, for instance, from microscopic cell images, thereby creating richer data sets that also require more advanced analysis methods. The nematode Caenorhabditis elegans came into the screening picture due to the wide availability of its mutants and human disease models, its relatively easy culture and short life cycle. Being a whole-organism model, it allows drug testing under physiological conditions at multi-tissue levels and provides additional observable phenotypes with respect to cell models, related, for instance, to development, aging, behavior or motility. Worm-based HTS studies in liquid environments on microwell plates have been demonstrated, while microfluidic devices allowed surpassing the performance of plates by enabling more versatile and accurate assays, precise and dynamic dosing of compounds, and readouts down to single-animal resolution. In this review, we discuss microfluidic devices for C. elegans analysis and related studies, published in the period from 2012 to 2017. After an introduction to the different screening approaches, we first focus on microfluidic systems with potential for screening applications. Various enabling technologies, e.g. electrophysiological on-chip recordings or laser axotomy, have been implemented, as well as techniques for reversible worm immobilization and high-resolution imaging, combined with algorithms for automated experimentation and analysis. Several devices for developmental or behavioral assays, and worm sorting based on different phenotypes, have been proposed too. In a subsequent section, we review the application of microfluidic-based systems for medium- and high-throughput screens, including neurobiology and neurodegeneration studies, aging and developmental assays, toxicity and pathogenesis screens, as well as behavioral and motility assays. A thorough analysis of this work reveals a trend towards microfluidic systems more and more capable of offering high-quality analyses of large worm populations, based on multi-phenotypic and/or longitudinal readouts, with clear potential for their application in larger HTS/HCS contexts.
Collapse
Affiliation(s)
- Matteo Cornaglia
- Laboratory of Microsystems, École Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland.
| | | | | |
Collapse
|
8
|
Lee H, Radu C, Han JW, Grailhe R. Assay Development for High Content Quantification of Sod1 Mutant Protein Aggregate Formation in Living Cells. J Vis Exp 2017. [PMID: 29053667 DOI: 10.3791/56425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that can be caused by inherited mutations in the gene encoding copper-zinc superoxide dismutase 1 (SOD1). The structural instability of SOD1 and the detection of SOD1-positive inclusions in familial-ALS patients supports a potential causal role for misfolded and/or aggregated SOD1 in ALS pathology. In this study, we describe the development of a cell-based assay designed to quantify the dynamics of SOD1 aggregation in living cells by high content screening approaches. Using lentiviral vectors, we generated stable cell lines expressing wild-type and mutant A4V SOD1 tagged with yellow fluorescent protein and found that both proteins were expressed in the cytosol without any sign of aggregation. Interestingly, only SOD1 A4V stably expressed in HEK-293, but not in U2OS or SH-SY5Y cell lines, formed aggregates upon proteasome inhibitor treatment. We show that it is possible to quantify aggregation based on dose-response analysis of various proteasome inhibitors, and to track aggregate-formation kinetics by time-lapse microscopy. Our approach introduces the possibility of quantifying the effect of ALS mutations on the role of SOD1 in aggregate formation as well as screening for small molecules that prevent SOD1 A4V aggregation.
Collapse
Affiliation(s)
- Honggun Lee
- Automation & Logistics Management, Screening Sciences & Novel Assay Technologies, Institut Pasteur Korea
| | - Constantin Radu
- Automation & Logistics Management, Screening Sciences & Novel Assay Technologies, Institut Pasteur Korea
| | | | - Regis Grailhe
- Technology Development Platform, Institut Pasteur Korea;
| |
Collapse
|
9
|
Carrico ZM, Le G, Malinow R. A fluorescence assay for detecting amyloid-β using the cytomegalovirus enhancer/promoter. J Biol Methods 2017; 4. [PMID: 29457040 PMCID: PMC5813829 DOI: 10.14440/jbm.2017.200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Robust assays for detecting the effects of elevated concentrations of amyloid-β (Aβ) may facilitate Alzheimer’s disease research. An appropriate assay would be high-throughput and enable identification of drugs and genetic mutations that block the effects of Aβ, potentially leading to treatments for Alzheimer’s disease. We discovered that the commonly used cytomegalovirus (CMV) enhancer/promoter is sensitive to the effects of Aβ. By combining the CMV enhancer/promoter with a fluorescent protein, we created a reporter system that produces changes in intracellular fluorescence in response to Aβ. Using hippocampal neurons, we quantified the ability of a CMV-fluorescent protein recombinant reporter to detect both exogenously applied and overexpressed Aβ. This is the first report of a high-throughput enhancer/promoter-based Aβ detection method. The reporter is able to detect the effects of elevated concentrations of Aβ in a high-throughput fashion, providing a new tool for Alzheimer’s disease research and important knowledge about the commonly used CMV enhancer/promoter.
Collapse
Affiliation(s)
- Zachary M Carrico
- Center for Neural Circuits and Behavior, Departments of Neuroscience and Biology, University of California at San Diego, La Jolla, CA 92093, USA
| | - Geneva Le
- Center for Neural Circuits and Behavior, Departments of Neuroscience and Biology, University of California at San Diego, La Jolla, CA 92093, USA
| | - Roberto Malinow
- Center for Neural Circuits and Behavior, Departments of Neuroscience and Biology, University of California at San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
10
|
Bassett JJ, Monteith GR. Genetically Encoded Calcium Indicators as Probes to Assess the Role of Calcium Channels in Disease and for High-Throughput Drug Discovery. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 79:141-171. [PMID: 28528667 DOI: 10.1016/bs.apha.2017.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The calcium ion (Ca2+) is an important signaling molecule implicated in many cellular processes, and the remodeling of Ca2+ homeostasis is a feature of a variety of pathologies. Typical methods to assess Ca2+ signaling in cells often employ small molecule fluorescent dyes, which are sometimes poorly suited to certain applications such as assessment of cellular processes, which occur over long periods (hours or days) or in vivo experiments. Genetically encoded calcium indicators are a set of tools available for the measurement of Ca2+ changes in the cytosol and subcellular compartments, which circumvent some of the inherent limitations of small molecule Ca2+ probes. Recent advances in genetically encoded calcium sensors have greatly increased their ability to provide reliable monitoring of Ca2+ changes in mammalian cells. New genetically encoded calcium indicators have diverse options in terms of targeting, Ca2+ affinity and fluorescence spectra, and this will further enhance their potential use in high-throughput drug discovery and other assays. This review will outline the methods available for Ca2+ measurement in cells, with a focus on genetically encoded calcium sensors. How these sensors will improve our understanding of the deregulation of Ca2+ handling in disease and their application to high-throughput identification of drug leads will also be discussed.
Collapse
Affiliation(s)
- John J Bassett
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - Gregory R Monteith
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia; Mater Research, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
11
|
T Lymphocytes and Inflammatory Mediators in the Interplay between Brain and Blood in Alzheimer's Disease: Potential Pools of New Biomarkers. J Immunol Res 2017; 2017:4626540. [PMID: 28293644 PMCID: PMC5331319 DOI: 10.1155/2017/4626540] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/22/2016] [Accepted: 01/23/2017] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the main cause of dementia. The disease is among the leading medical concerns of the modern world, because only symptomatic therapies are available, and no reliable, easily accessible biomarkers exist for AD detection and monitoring. Therefore extensive research is conducted to elucidate the mechanisms of AD pathogenesis, which seems to be heterogeneous and multifactorial. Recently much attention has been given to the neuroinflammation and activation of glial cells in the AD brain. Reports also highlighted the proinflammatory role of T lymphocytes infiltrating the AD brain. However, in AD molecular and cellular alterations involving T cells and immune mediators occur not only in the brain, but also in the blood and the cerebrospinal fluid (CSF). Here we review alterations concerning T lymphocytes and related immune mediators in the AD brain, CSF, and blood and the mechanisms by which peripheral T cells cross the blood brain barrier and the blood-CSF barrier. This knowledge is relevant for better AD therapies and for identification of novel biomarkers for improved AD diagnostics in the blood and the CSF. The data will be reviewed with the special emphasis on possibilities for development of AD biomarkers.
Collapse
|
12
|
Czeredys M, Maciag F, Methner A, Kuznicki J. Tetrahydrocarbazoles decrease elevated SOCE in medium spiny neurons from transgenic YAC128 mice, a model of Huntington's disease. Biochem Biophys Res Commun 2016; 483:1194-1205. [PMID: 27553284 DOI: 10.1016/j.bbrc.2016.08.106] [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: 06/15/2016] [Revised: 08/08/2016] [Accepted: 08/18/2016] [Indexed: 10/21/2022]
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disease caused by a polyglutamine expansion within the huntingtin (HTT) gene. One of the cellular functions that is dysregulated in HD is store-operated calcium entry (SOCE), a process in which the depletion of Ca2+ from the endoplasmic reticulum (ER) induces Ca2+ influx from the extracellular space. We detected an enhanced activity of SOC channels in medium spiny neurons (MSNs) from YAC128 mice, a transgenic model of HD, and investigated whether this could be reverted by tetrahydrocarbazoles. The compound 6-bromo-N-(2-phenylethyl)-2,3,4,9-tetrahydro-1H-carbazol-1-amine hydrochloride was indeed able to restore the disturbed Ca2+ homeostasis and stabilize SOCE in YAC128 MSN cultures. We also detected a beneficial effect of this compound on the mitochondrial membrane potential. Since dysregulated Ca2+ homeostasis is believed to be one of the pathological hallmarks of HD, this compound might be a lead structure for HD treatment.
Collapse
Affiliation(s)
- Magdalena Czeredys
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Trojdena 4, 02-109, Warsaw, Poland.
| | - Filip Maciag
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Trojdena 4, 02-109, Warsaw, Poland
| | - Axel Methner
- Department of Neurology, University Medical Center and Focus Program Translational Neuroscience (FTN) of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, D-55131, Mainz, Germany
| | - Jacek Kuznicki
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Trojdena 4, 02-109, Warsaw, Poland
| |
Collapse
|
13
|
Mattiazzi Usaj M, Styles EB, Verster AJ, Friesen H, Boone C, Andrews BJ. High-Content Screening for Quantitative Cell Biology. Trends Cell Biol 2016; 26:598-611. [PMID: 27118708 DOI: 10.1016/j.tcb.2016.03.008] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 12/25/2022]
Abstract
High-content screening (HCS), which combines automated fluorescence microscopy with quantitative image analysis, allows the acquisition of unbiased multiparametric data at the single cell level. This approach has been used to address diverse biological questions and identify a plethora of quantitative phenotypes of varying complexity in numerous different model systems. Here, we describe some recent applications of HCS, ranging from the identification of genes required for specific biological processes to the characterization of genetic interactions. We review the steps involved in the design of useful biological assays and automated image analysis, and describe major challenges associated with each. Additionally, we highlight emerging technologies and future challenges, and discuss how the field of HCS might be enhanced in the future.
Collapse
Affiliation(s)
| | - Erin B Styles
- The Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
| | - Adrian J Verster
- The Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
| | - Helena Friesen
- The Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
| | - Charles Boone
- The Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
| | - Brenda J Andrews
- The Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada.
| |
Collapse
|
14
|
Abstract
Image-based screening is used to measure a variety of phenotypes in cells and whole organisms. Combined with perturbations such as RNA interference, small molecules, and mutations, such screens are a powerful method for gaining systematic insights into biological processes. Screens have been applied to study diverse processes, such as protein-localization changes, cancer cell vulnerabilities, and complex organismal phenotypes. Recently, advances in imaging and image-analysis methodologies have accelerated large-scale perturbation screens. Here, we describe the state of the art for image-based screening experiments and delineate experimental approaches and image-analysis approaches as well as discussing challenges and future directions, including leveraging CRISPR/Cas9-mediated genome engineering.
Collapse
Affiliation(s)
- Michael Boutros
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ) and Department of Cell and Molecular Biology, Heidelberg University, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany.
| | - Florian Heigwer
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ) and Department of Cell and Molecular Biology, Heidelberg University, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Christina Laufer
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ) and Department of Cell and Molecular Biology, Heidelberg University, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| |
Collapse
|
15
|
Majewski L, Kuznicki J. SOCE in neurons: Signaling or just refilling? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1940-52. [DOI: 10.1016/j.bbamcr.2015.01.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/22/2015] [Accepted: 01/26/2015] [Indexed: 01/14/2023]
|