1
|
Parrales V, Arcile G, Laserre L, Normant S, Le Goff G, Da Costa Noble C, Ouazzani J, Callizot N, Haïk S, Rabhi C, Bizat N. Neuroprotective Effect of Withaferin Derivatives toward MPP + and 6-OHDA Toxicity to Dopaminergic Neurons. ACS Chem Neurosci 2025; 16:802-817. [PMID: 39946298 DOI: 10.1021/acschemneuro.4c00655] [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] [Indexed: 02/21/2025] Open
Abstract
Parkinson's disease is a neurodegenerative proteinopathy that primarily affects mesencephalic dopaminergic neurons. This dopaminergic depletion can be phenotypically reproduced in various experimental models through the administration of two neurotoxins: N-methyl-4-phenylpyridinium (MPP+) and 6-hydroxydopamine (6-OHDA). The mechanisms underlying the cell death processes induced by these toxins remain a subject of debate. In this context, studies suggest that oxidative-stress-related processes may contribute to the dysfunction and death of dopaminergic neurons. Therefore, investigating pharmacological compounds that can counteract these processes remains crucial for developing therapeutic strategies targeting these neuropathological mechanisms. Withania somnifera (L.) Dunal, commonly known as ashwagandha, is a plant whose roots are used in Ayurvedic medicine to treat various ailments, including those affecting the central nervous system. The active compound Withaferin-A (WFA), a steroid lactone from the withanolide group, is reported to possess antioxidant properties. In this study, we explored the potential neuroprotective effects of WFA and two of its molecular derivatives, cr-591 and cr-777, which contain, respectively, an additional cysteine or glutathione chemical group, known for their antiradical properties. We demonstrated that WFA and its two derivatives, cr-591 and cr-777, protect the integrity and function of dopaminergic neurons exposed to the neurotoxins MPP+ and 6-OHDA both in vitro, using primary mesencephalic neuron cultures from rodents, and in vivo, using the nematode Caenorhabditis elegans.
Collapse
Affiliation(s)
- Valeria Parrales
- Paris Brain Institute, Inserm U1127, CNRS Sorbonne University, Hospital Pitié-Salpêtrière, UMR7225, Paris 75013, France
- Laboratoire Ethnodyne, 151 Boulevard Haussmann, Paris 75008, France
| | - Guillaume Arcile
- Institut de Chimie des Substances Naturelles (ICSN, UPR2301), University Paris-Saclay, 1 Avenue de la Terrasse, Gif-sur-Yvette 91198, France
| | - Louise Laserre
- Paris Brain Institute, Inserm U1127, CNRS Sorbonne University, Hospital Pitié-Salpêtrière, UMR7225, Paris 75013, France
| | - Sébastien Normant
- Paris Brain Institute, Inserm U1127, CNRS Sorbonne University, Hospital Pitié-Salpêtrière, UMR7225, Paris 75013, France
- Laboratoire Ethnodyne, 151 Boulevard Haussmann, Paris 75008, France
| | - Géraldine Le Goff
- Institut de Chimie des Substances Naturelles (ICSN, UPR2301), University Paris-Saclay, 1 Avenue de la Terrasse, Gif-sur-Yvette 91198, France
| | | | - Jamal Ouazzani
- Institut de Chimie des Substances Naturelles (ICSN, UPR2301), University Paris-Saclay, 1 Avenue de la Terrasse, Gif-sur-Yvette 91198, France
| | - Noelle Callizot
- Neuro-Sys, 410 Chemin Départemental 60, Gardanne 13120, France
| | - Stéphane Haïk
- Paris Brain Institute, Inserm U1127, CNRS Sorbonne University, Hospital Pitié-Salpêtrière, UMR7225, Paris 75013, France
- AP-HP, Cellule Nationale de Référence des Maladies de Creutzfeldt-Jakob, University Hospital Pitié-Salpêtrière, Paris 75013, France
| | - Chérif Rabhi
- Institut de Chimie des Substances Naturelles (ICSN, UPR2301), University Paris-Saclay, 1 Avenue de la Terrasse, Gif-sur-Yvette 91198, France
- Laboratoire Ethnodyne, 151 Boulevard Haussmann, Paris 75008, France
| | - Nicolas Bizat
- Paris Brain Institute, Inserm U1127, CNRS Sorbonne University, Hospital Pitié-Salpêtrière, UMR7225, Paris 75013, France
- Faculté de Pharmacie de Paris, Paris University, 4 Avenue de l'Observatoire, Paris 75006, France
| |
Collapse
|
2
|
Ide S, Ikeda K. Caenorhabditis elegans for opioid addiction research. Curr Opin Neurobiol 2024; 88:102914. [PMID: 39236640 DOI: 10.1016/j.conb.2024.102914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/16/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
The problem of drug addiction has become a profound societal problem worldwide. A better understanding of the neurobiological basis of addiction and the discovery of more effective treatments are needed. Recent studies have shown that many mechanisms that underlie addiction exist in more primitive organisms, including the nematode Caenorhabditis elegans (C. elegans). C. elegans is also hypothesized to possess a functional opioid-like system, including the endogenous opioid-like peptide NLP-24 and opioid-like receptor NPR-17. Opioids, such as morphine, are thought to cause addiction-like behavior by activating dopamine nerves in C. elegans via the opioid-like system. Accumulating evidence suggests that C. elegans is an excellent animal model for identifying molecular mechanisms of addiction.
Collapse
Affiliation(s)
- Soichiro Ide
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-Ku, Tokyo, Japan.
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-Ku, Tokyo, Japan; Department of Neuropsychopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi-cho, Kodaira, Tokyo, Japan
| |
Collapse
|
3
|
McMillen A, Chew Y. Neural mechanisms of dopamine function in learning and memory in Caenorhabditis elegans. Neuronal Signal 2024; 8:NS20230057. [PMID: 38572143 PMCID: PMC10987485 DOI: 10.1042/ns20230057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/03/2023] [Accepted: 12/11/2023] [Indexed: 04/05/2024] Open
Abstract
Research into learning and memory over the past decades has revealed key neurotransmitters that regulate these processes, many of which are evolutionarily conserved across diverse species. The monoamine neurotransmitter dopamine is one example of this, with countless studies demonstrating its importance in regulating behavioural plasticity. However, dopaminergic neural networks in the mammalian brain consist of hundreds or thousands of neurons, and thus cannot be studied at the level of single neurons acting within defined neural circuits. The nematode Caenorhabditis elegans (C. elegans) has an experimentally tractable nervous system with a completely characterized synaptic connectome. This makes it an advantageous system to undertake mechanistic studies into how dopamine encodes lasting yet flexible behavioural plasticity in the nervous system. In this review, we synthesize the research to date exploring the importance of dopaminergic signalling in learning, memory formation, and forgetting, focusing on research in C. elegans. We also explore the potential for dopamine-specific fluorescent biosensors in C. elegans to visualize dopaminergic neural circuits during learning and memory formation in real-time. We propose that the use of these sensors in C. elegans, in combination with optogenetic and other light-based approaches, will further illuminate the detailed spatiotemporal requirements for encoding behavioural plasticity in an accessible experimental system. Understanding the key molecules and circuit mechanisms that regulate learning and forgetting in more compact invertebrate nervous systems may reveal new druggable targets for enhancing memory storage and delaying memory loss in bigger brains.
Collapse
Affiliation(s)
- Anna McMillen
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, 5042, South Australia, Australia
| | - Yee Lian Chew
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, 5042, South Australia, Australia
| |
Collapse
|
4
|
Hua X, Wang D. Disruption of dopamine metabolism by exposure to 6-PPD quinone in Caenorhabditis elegans. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 337:122649. [PMID: 37777057 DOI: 10.1016/j.envpol.2023.122649] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Caenorhabditis elegans is a useful model for examining metabolic processes and related mechanisms. We here examined the effect of exposure to N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine quinone (6-PPDQ) on dopamine metabolism and underling molecular basis in nematodes. The dopamine content was reduced by 6-PPDQ (1 and 10 μg/L). Meanwhile, dopamine related behaviors (basal slowing response and area restricted searching) were changed by 6-PPDQ (1 and 10 μg/L). Exposure to 6-PPDQ (1 and 10 μg/L) decreased expressions of genes (cat-2 and bas-1) encoding enzymes governing dopamine synthesis and cat-1 encoding dopamine transporter. Development of dopaminergic neurons was also affected by 10 μg/L 6-PPDQ as reflected by decrease in fluorescence intensity, neuronal loss, and defect in dendrite development. Exposure to 6-PPDQ (1 and 10 μg/L) altered expressions of ast-1 and rcat-1 encoding upregulators of cat-2 and bas-1. The dopamine content and expressions of cat-2 and bas-1 were inhibited by RNAi of ast-1 and increased by RNAi of rcat-1 in 6-PPDQ exposed nematodes. Using endpoints of locomotion behavior and brood size, in 6-PPDQ exposed nematodes, the susceptibility to toxicity was caused by RNAi of ast-1, cat-2, bas-1, and cat-1, and the resistance to toxicity was induced by RNAi of rcat-1. Therefore, 6-PPDQ exposure disrupted dopamine metabolism and the altered molecular basis for dopamine metabolism was associated with 6-PPDQ toxicity induction. Moreover, the defects in dopamine related behaviors and toxicity on locomotion and reproduction could be rescued by treatment with 0.1 mM dopamine.
Collapse
Affiliation(s)
- Xin Hua
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Medical School, Southeast University, Nanjing, China
| | - Dayong Wang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Medical School, Southeast University, Nanjing, China.
| |
Collapse
|
5
|
Estrada-Valencia R, de Lima ME, Colonnello A, Rangel-López E, Saraiva NR, de Ávila DS, Aschner M, Santamaría A. The Endocannabinoid System in Caenorhabditis elegans. Rev Physiol Biochem Pharmacol 2023; 184:1-31. [PMID: 34401955 PMCID: PMC8850531 DOI: 10.1007/112_2021_64] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The existence of a formal Endocannabinoid System in C. elegans has been questioned due to data showing the absence of typical cannabinoid receptors in the worm; however, the presence of a full metabolism for endocannabinoids, alternative ligands, and receptors for these agents and a considerable number of orthologous and homologous genes regulating physiological cannabinoid-like signals and responses - several of which are similar to those of mammals - demonstrates a well-structured and functional complex system in nematodes. In this review, we describe and compare similarities and differences between the Endocannabinoid System in mammals and nematodes, highlighting the basis for the integral study of this novel system in the worm.
Collapse
Affiliation(s)
| | - María Eduarda de Lima
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Aline Colonnello
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Edgar Rangel-López
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Nariani Rocha Saraiva
- Laboratório de Bioquímica e Toxicologia em Caenorhabditis elegans, UNIPAMPA, Uruguaiana, Brazil
| | - Daiana Silva de Ávila
- Laboratório de Bioquímica e Toxicologia em Caenorhabditis elegans, UNIPAMPA, Uruguaiana, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico.
| |
Collapse
|
6
|
Parrales-Macias V, Harfouche A, Ferrié L, Haïk S, Michel PP, Raisman-Vozari R, Figadère B, Bizat N, Maciuk A. Effects of a New Natural Catechol- O-methyl Transferase Inhibitor on Two In Vivo Models of Parkinson's Disease. ACS Chem Neurosci 2022; 13:3303-3313. [PMID: 36347018 DOI: 10.1021/acschemneuro.2c00356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
A tetrahydroisoquinoline identified in Mucuna pruriens ((1R,3S)-6,7-dihydroxy-1-methyl-1,2,3,4-tetrahydroisoquinoline-1,3-dicarboxylic acid, compound 4) was synthesized and assessed for its in vitro pharmacological profile and in vivo effects in two animal models of Parkinson's disease. Compound 4 inhibits catechol-O-methyltransferase (COMT) with no affinity for the dopaminergic receptors or the dopamine transporter. It restores dopamine-mediated motor behavior when it is co-administered with L-DOPA to C. elegans worms with 1-methyl-4-phenylpyridinium-damaged dopaminergic neurons. In a 6-hydroxydopamine rat model of Parkinson's disease, its co-administration at 30 mg/kg with L-DOPA enhances the effect of L-DOPA with an intensity similar to that of tolcapone 1 at 30 mg/kg but for a shorter duration. The effect is not dose-dependent. Compound 4 seems not to cross the blood-brain barrier and thus acts as a peripheral COMT inhibitor. COMT inhibition by compound 4 further validates the traditional use of M. pruriens for the treatment of Parkinson's disease, and compound 4 can thus be considered as a promising drug candidate for the development of safe, peripheral COMT inhibitors.
Collapse
Affiliation(s)
- Valeria Parrales-Macias
- Paris Brain Institute - ICM, Inserm, CNRS, Hôpital Pitié Salpêtrière, Sorbonne Université, Paris 75013, France
| | - Abha Harfouche
- CNRS, BioCIS, Université Paris-Saclay, Orsay 91400, France
| | - Laurent Ferrié
- CNRS, BioCIS, Université Paris-Saclay, Orsay 91400, France
| | - Stéphane Haïk
- Paris Brain Institute - ICM, Inserm, CNRS, Hôpital Pitié Salpêtrière, Sorbonne Université, Paris 75013, France
| | - Patrick P Michel
- Paris Brain Institute - ICM, Inserm, CNRS, Hôpital Pitié Salpêtrière, Sorbonne Université, Paris 75013, France
| | - Rita Raisman-Vozari
- Paris Brain Institute - ICM, Inserm, CNRS, Hôpital Pitié Salpêtrière, Sorbonne Université, Paris 75013, France
| | - Bruno Figadère
- CNRS, BioCIS, Université Paris-Saclay, Orsay 91400, France
| | - Nicolas Bizat
- Paris Brain Institute - ICM, Inserm, CNRS, Hôpital Pitié Salpêtrière, Sorbonne Université, Paris 75013, France
| | | |
Collapse
|
7
|
Albrecht PA, Fernandez-Hubeid LE, Deza-Ponzio R, Martins AC, Aschner M, Virgolini MB. Developmental lead exposure affects dopaminergic neuron morphology and modifies basal slowing response in Caenorhabditis elegans: effects of ethanol. Neurotoxicology 2022; 91:349-359. [PMID: 35724878 DOI: 10.1016/j.neuro.2022.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/04/2022] [Accepted: 06/13/2022] [Indexed: 11/28/2022]
Abstract
Lead (Pb) and ethanol (EtOH) are neurotoxicants that affect the dopaminergic (DAergic) system. We first sought to assess the morphology of the DAergic neurons in the Caenorhabditis elegans BY200 strain. The results demonstrated dose-dependent damage in these neurons induced by developmental Pb exposure. Secondly, transgenic worms exposed to 24μM Pb and administered with 200mM EtOH were evaluated in the basal slowing response (BSR). Pb induced impairment in the BSR in the wild-type strain that did not improve in response to EtOH, an effect also observed in strains that lack the DOP-1, DOP-2, and DOP-3 receptors. The animals that overexpress tyrosine hydroxylase (TH), or lack the vesicular transport (VMAT) showed a Pb-induced impairment in the BSR that seemed to improve after EtOH. Interestingly, a dramatic impairment in the BSR was observed in the Pb group in strains lacking the DOP-4 receptor, resembling the response of the TH-deficient strain, an effect that in both cases showed a non-significant reversal by EtOH. These results suggest that the facilitatory effect of EtOH on the impaired BSR observed in Pb-exposed null mutant strains may be the result of a compensatory effect in the altered DAergic synapse present in these animals.
Collapse
Affiliation(s)
- Paula A Albrecht
- Instituto de Farmacología Experimental de Córdoba-Consejo Nacional de Investigaciones Técnicas (IFEC-CONICET) and Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Lucia E Fernandez-Hubeid
- Instituto de Farmacología Experimental de Córdoba-Consejo Nacional de Investigaciones Técnicas (IFEC-CONICET) and Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Romina Deza-Ponzio
- Instituto de Farmacología Experimental de Córdoba-Consejo Nacional de Investigaciones Técnicas (IFEC-CONICET) and Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Airton C Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Miriam B Virgolini
- Instituto de Farmacología Experimental de Córdoba-Consejo Nacional de Investigaciones Técnicas (IFEC-CONICET) and Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina.
| |
Collapse
|
8
|
Paredes GF, Viehboeck T, Markert S, Mausz MA, Sato Y, Liebeke M, König L, Bulgheresi S. Differential regulation of degradation and immune pathways underlies adaptation of the ectosymbiotic nematode Laxus oneistus to oxic-anoxic interfaces. Sci Rep 2022; 12:9725. [PMID: 35697683 PMCID: PMC9192688 DOI: 10.1038/s41598-022-13235-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 05/13/2022] [Indexed: 11/09/2022] Open
Abstract
Eukaryotes may experience oxygen deprivation under both physiological and pathological conditions. Because oxygen shortage leads to a reduction in cellular energy production, all eukaryotes studied so far conserve energy by suppressing their metabolism. However, the molecular physiology of animals that naturally and repeatedly experience anoxia is underexplored. One such animal is the marine nematode Laxus oneistus. It thrives, invariably coated by its sulfur-oxidizing symbiont Candidatus Thiosymbion oneisti, in anoxic sulfidic or hypoxic sand. Here, transcriptomics and proteomics showed that, whether in anoxia or not, L. oneistus mostly expressed genes involved in ubiquitination, energy generation, oxidative stress response, immune response, development, and translation. Importantly, ubiquitination genes were also highly expressed when the nematode was subjected to anoxic sulfidic conditions, together with genes involved in autophagy, detoxification and ribosome biogenesis. We hypothesize that these degradation pathways were induced to recycle damaged cellular components (mitochondria) and misfolded proteins into nutrients. Remarkably, when L. oneistus was subjected to anoxic sulfidic conditions, lectin and mucin genes were also upregulated, potentially to promote the attachment of its thiotrophic symbiont. Furthermore, the nematode appeared to survive oxygen deprivation by using an alternative electron carrier (rhodoquinone) and acceptor (fumarate), to rewire the electron transfer chain. On the other hand, under hypoxia, genes involved in costly processes (e.g., amino acid biosynthesis, development, feeding, mating) were upregulated, together with the worm's Toll-like innate immunity pathway and several immune effectors (e.g., bactericidal/permeability-increasing proteins, fungicides). In conclusion, we hypothesize that, in anoxic sulfidic sand, L. oneistus upregulates degradation processes, rewires the oxidative phosphorylation and reinforces its coat of bacterial sulfur-oxidizers. In upper sand layers, instead, it appears to produce broad-range antimicrobials and to exploit oxygen for biosynthesis and development.
Collapse
Affiliation(s)
- Gabriela F Paredes
- Department of Functional and Evolutionary Ecology, Environmental Cell Biology Group, University of Vienna, Vienna, Austria
| | - Tobias Viehboeck
- Department of Functional and Evolutionary Ecology, Environmental Cell Biology Group, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Ecology and Evolution, Vienna, Austria
- Division of Microbial Ecology, Center for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Stephanie Markert
- Department of Pharmaceutical Biotechnology, Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| | | | - Yui Sato
- Max Planck Institute for Marine Microbiology, Bremen, Germany
| | - Manuel Liebeke
- Max Planck Institute for Marine Microbiology, Bremen, Germany
| | - Lena König
- Department of Functional and Evolutionary Ecology, Environmental Cell Biology Group, University of Vienna, Vienna, Austria
| | - Silvia Bulgheresi
- Department of Functional and Evolutionary Ecology, Environmental Cell Biology Group, University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Refai O, Aggarwal S, Cheng MH, Gichi Z, Salvino JM, Bahar I, Blakely RD, Mortensen OV. Allosteric Modulator KM822 Attenuates Behavioral Actions of Amphetamine in Caenorhabditis elegans through Interactions with the Dopamine Transporter DAT-1. Mol Pharmacol 2022; 101:123-131. [PMID: 34906999 PMCID: PMC8969146 DOI: 10.1124/molpharm.121.000400] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/03/2021] [Indexed: 11/22/2022] Open
Abstract
Aberrant dopamine (DA) signaling is associated with several psychiatric disorders, such as autism, bipolar disorder, addiction, and Parkinson's disease, and several medications that target the DA transporter (DAT) can induce or treat these disorders. In addition, psychostimulants, such as cocaine and D-amphetamine (AMPH), rely on the competitive interactions with the transporter's substrate binding site to produce their rewarding effects. Agents that exhibit noncompetitive, allosteric modulation of DAT remain an important topic of investigation due to their potential therapeutic applications. We previously identified a novel allosteric modulator of human DAT, KM822, that can decrease the affinity of cocaine for DAT and attenuate cocaine-elicited behaviors; however, whether DAT is the sole mediator of KM822 actions in vivo is unproven given the large number of potential off-target sites. Here, we provide in silico and in vitro evidence that the allosteric site engaged by KM822 is conserved between human DAT and Caenorhabditis elegans DAT-1. KM822 binds to a similar pocket in DAT-1 as previously identified in human DAT. In functional dopamine uptake assays, KM822 affects the interaction between AMPH and DAT-1 by reducing the affinity of AMPH for DAT-1. Finally, through a combination of genetic and pharmacological in vivo approaches we provide evidence that KM822 diminishes the behavioral actions of AMPH on swimming-induced paralysis through a direct allosteric modulation of DAT-1. More broadly, our findings demonstrate allosteric modulation of DAT as a behavior modifying strategy and suggests that Caenorhabditis elegans can be operationalized to identify and investigate the interactions of DAT allosteric modulators. SIGNIFICANCE STATEMENT: We previously demonstrated that the dopamine transporter (DAT) allosteric modulator KM822 decreases cocaine affinity for human DAT. Here, using in silico and in vivo genetic approaches, we extend this finding to interactions with amphetamine, demonstrating evolutionary conservation of the DAT allosteric site. In Caenorhabditis elegans, we report that KM822 suppresses amphetamine behavioral effects via specific interactions with DAT-1. Our findings reveal Caenorhabditis elegans as a new tool to study allosteric modulation of DAT and its behavioral consequences.
Collapse
Affiliation(s)
- Osama Refai
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida (O.R., Z.G., R.D.B.); Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida (O.R., R.D.B.); Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.A., O.V.M.); Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (M.H.C., I.B.); and The Wistar Institute, Philadelphia, Pennsylvania (J.M.S.)
| | - Shaili Aggarwal
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida (O.R., Z.G., R.D.B.); Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida (O.R., R.D.B.); Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.A., O.V.M.); Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (M.H.C., I.B.); and The Wistar Institute, Philadelphia, Pennsylvania (J.M.S.)
| | - Mary Hongying Cheng
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida (O.R., Z.G., R.D.B.); Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida (O.R., R.D.B.); Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.A., O.V.M.); Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (M.H.C., I.B.); and The Wistar Institute, Philadelphia, Pennsylvania (J.M.S.)
| | - Zayna Gichi
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida (O.R., Z.G., R.D.B.); Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida (O.R., R.D.B.); Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.A., O.V.M.); Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (M.H.C., I.B.); and The Wistar Institute, Philadelphia, Pennsylvania (J.M.S.)
| | - Joseph M Salvino
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida (O.R., Z.G., R.D.B.); Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida (O.R., R.D.B.); Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.A., O.V.M.); Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (M.H.C., I.B.); and The Wistar Institute, Philadelphia, Pennsylvania (J.M.S.)
| | - Ivet Bahar
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida (O.R., Z.G., R.D.B.); Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida (O.R., R.D.B.); Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.A., O.V.M.); Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (M.H.C., I.B.); and The Wistar Institute, Philadelphia, Pennsylvania (J.M.S.)
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida (O.R., Z.G., R.D.B.); Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida (O.R., R.D.B.); Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.A., O.V.M.); Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (M.H.C., I.B.); and The Wistar Institute, Philadelphia, Pennsylvania (J.M.S.)
| | - Ole V Mortensen
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida (O.R., Z.G., R.D.B.); Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida (O.R., R.D.B.); Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (S.A., O.V.M.); Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (M.H.C., I.B.); and The Wistar Institute, Philadelphia, Pennsylvania (J.M.S.)
| |
Collapse
|
10
|
Kirshenboim I, Aviner B, Itskovits E, Zaslaver A, Broday L. Dopamine-dependent biphasic behaviour under 'deep diving' conditions in Caenorhabditis elegans. Proc Biol Sci 2021; 288:20210128. [PMID: 33715430 PMCID: PMC7944115 DOI: 10.1098/rspb.2021.0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Underwater divers are susceptible to neurological risks due to their exposure to increased pressure. Absorption of elevated partial pressure of inert gases such as helium and nitrogen may lead to nitrogen narcosis. Although the symptoms of nitrogen narcosis are known, the molecular mechanisms underlying these symptoms have not been elucidated. Here, we examined the behaviour of the soil nematode Caenorhabditis elegans under scuba diving conditions. We analysed wild-type animals and mutants in the dopamine pathway under hyperbaric conditions, using several gas compositions and under varying pressure levels. We found that the animals changed their speed on a flat bacterial surface in response to pressure in a biphasic mode that depended on dopamine. Dopamine-deficient cat-2 mutant animals did not exhibit a biphasic response in high pressure, while the extracellular accumulation of dopamine in dat-1 mutant animals mildly influenced this response. Our data demonstrate that in C. elegans, similarly to mammalian systems, dopamine signalling is involved in the response to high pressure. This study establishes C. elegans as a powerful system to elucidate the molecular mechanisms that underly nitrogen toxicity in response to high pressure.
Collapse
Affiliation(s)
- Inbar Kirshenboim
- Department of Cell and Developmental Biology, School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Israel Naval Medical Institute, Israel Defense Forces Medical Corps, Haifa, Israel
| | - Ben Aviner
- Israel Naval Medical Institute, Israel Defense Forces Medical Corps, Haifa, Israel
| | - Eyal Itskovits
- Department of Genetics, Silberman Institute of Life Science, Edmond J. Safra Campus, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Alon Zaslaver
- Department of Genetics, Silberman Institute of Life Science, Edmond J. Safra Campus, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Limor Broday
- Department of Cell and Developmental Biology, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
11
|
Ke T, Prince LM, Bowman AB, Aschner M. Latent alterations in swimming behavior by developmental methylmercury exposure are modulated by the homolog of tyrosine hydroxylase in Caenorhabditis elegans. Neurotoxicol Teratol 2021; 85:106963. [PMID: 33626374 DOI: 10.1016/j.ntt.2021.106963] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/18/2022]
Abstract
Methylmercury (MeHg) is a persistent environmental neurotoxicant that may cause adverse neurodevelopmental effects. Previous studies showed that developmental MeHg exposure caused damage to brain functions that were unmasked after a silent period of years or decades. However, the underlying mechanisms of the latent neurotoxicity associated with MeHg exposure from earlier developmental stages have yet to be fully understood. Herein, we established a Caenorhabditis elegans (C. elegans) model of developmental MeHg latent toxicity. Synchronized L1 stage worms were exposed to MeHg (0, 0.05, 0.5 and 5 μM) for 48 h. Swimming moving speeds at adulthood were analyzed in worms exposed to MeHg exposure at early larvae stages. Worms developmentally exposed to MeHg had a significant decline in swimming moving speed on day 10 adult stage, but not on day 1 or 5 adult stage, even though the mercury level in the worms exposed to 0.05 or 0.5 μM MeHg were below the quantification limit on day 10 adult. Day 10 adult worms treated with MeHg showed a significant decrease in bending angle and bending frequency during swimming. Furthermore, their reduced moving speeds tended to increase during the 300-s swimming experiment. Dopamine signaling is known to be involved in the regulation of worms' moving speed. Accordingly, the moving speed of worms with cat-2 (mammalian tyrosine hydroxylase homolog) mutation or dat-1 deletion were assayed on day 10 adult. The cat-2 mutant worms did not show a decline in moving speeds, body bends or bending angles during swimming on day 10 adult stage. Analyses of moving speeds of worms with dat-1 deletion showed that the moving speeds were further reduced after MeHg exposure. However, the effects of MeHg and dat-1 deletion were not synergistic, as the interaction between these parameters did not attain statistical significance. Altogether, our results suggest that developmental MeHg exposure reduced moving speed, and this latent toxicity was less pronounced in the context of deficient production of dopamine synthesis. Tyrosine hydroxylase plays an important role in regulating dopamine-mediated modulation of neurobehavioral functions. These findings uncovered a pivotal role of dopamine and its metabolism in the latent neurotoxic effects of MeHg.
Collapse
Affiliation(s)
- Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Lisa M Prince
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, United States
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States; IM Sechenov First Moscow State Medical University, Sechenov University, Moscow, Russia.
| |
Collapse
|
12
|
Pandey P, Singh A, Kaur H, Ghosh-Roy A, Babu K. Increased dopaminergic neurotransmission results in ethanol dependent sedative behaviors in Caenorhabditis elegans. PLoS Genet 2021; 17:e1009346. [PMID: 33524034 PMCID: PMC7877767 DOI: 10.1371/journal.pgen.1009346] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 02/11/2021] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
Ethanol is a widely used drug, excessive consumption of which could lead to medical conditions with diverse symptoms. Ethanol abuse causes dysfunction of memory, attention, speech and locomotion across species. Dopamine signaling plays an essential role in ethanol dependent behaviors in animals ranging from C. elegans to humans. We devised an ethanol dependent assay in which mutants in the dopamine autoreceptor, dop-2, displayed a unique sedative locomotory behavior causing the animals to move in circles while dragging the posterior half of their body. Here, we identify the posterior dopaminergic sensory neuron as being essential to modulate this behavior. We further demonstrate that in dop-2 mutants, ethanol exposure increases dopamine secretion and functions in a DVA interneuron dependent manner. DVA releases the neuropeptide NLP-12 that is known to function through cholinergic motor neurons and affect movement. Thus, DOP-2 modulates dopamine levels at the synapse and regulates alcohol induced movement through NLP-12.
Collapse
Affiliation(s)
- Pratima Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
| | - Anuradha Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
| | - Harjot Kaur
- National Brain Research Centre, Gurgaon, India
| | | | - Kavita Babu
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
- Centre for Neuroscience, Indian Institute of Science (IISc), Bangalore, India
| |
Collapse
|
13
|
Ke T, Tsatsakis A, Santamaría A, Antunes Soare FA, Tinkov AA, Docea AO, Skalny A, Bowman AB, Aschner M. Chronic exposure to methylmercury induces puncta formation in cephalic dopaminergic neurons in Caenorhabditis elegans. Neurotoxicology 2020; 77:105-113. [PMID: 31935438 DOI: 10.1016/j.neuro.2020.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 01/09/2023]
Abstract
The neurotransmitter dopamine is a neuromodulator in the positive and negative regulation of brain circuits. Dopamine insufficiency or overload has been implicated in aberrant activities of neural circuits that play key roles in the pathogenesis of neurological and psychiatric diseases. Dopaminergic neurons are vulnerable to environmental insults. The neurotoxin methylmercury (MeHg) produces dopaminergic neuron damage in rodent as well as in Caenorhabditis elegans (C. elegans) models. Previous studies have demonstrated the utility of C. elegans as an alternative and complementary experimental model in dissecting out mechanism of MeHg-induced dopaminergic neurodegeneration. However, a sensitive pathological change that marks early events in neurodegeneration induced by environmental level of MeHg, is still lacking. By establishing a chronic exposure C. elegans model, for the first time, we have shown the propensity of MeHg (5 μM, 10 days) to induce bright puncta of dat-1::mCherry aggreagtes in the dendrites of cephalic (2 CEPs) dopaminergic neurons in a dose- and time-dependent manner, while these changes were not found in other dopaminergic neurons: anterior deirids (2 ADEs) and posterior deirids (2 PDEs), cholinergic neurons (2 AIYs) or glutamatergic neurons (2 PVDs). The bright puncta appear as an aggregation of mCherry proteins accumulating in dendrites. Further staining shows that the puncta were not inclusions in lysosome, or amyloid protein aggregates. In addition, features of the puncta including enlarged sphere shape (0.5-2 μm diameters), bright and accompanying with the shrinkage of the dendrite suggest that the puncta are likely composed of homologous mCherry molecules packaged at the dendritic site for exportation. Moreover, in the glutathione S-transferase 4 (gst-4) transcriptional reporter strain and RT-PCR assay, the expression levels of gst-4 and tubulins (tba-1 and tba-2) genes were not significantly modified under this chronic exposure paradigm, but gst-4 did show significant changes in an one day exposure paradigm. Collectively, these results suggest that CEP dopaminergic neurons are a sensitive target of MeHg, and the current exposure paradigm could be used as a model to investigate mechanism of dopaminergic neurotoxicity.
Collapse
Affiliation(s)
- Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Aristidis Tsatsakis
- Laboratory of Toxicology and Forensic Sciences, Medical School, University of Crete, Heraklion, Greece
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, 14269, Mexico City, Mexico
| | - Félix Alexandre Antunes Soare
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States; Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Alexey A Tinkov
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl 150000, Russia
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy, Faculty of Pharmacy, Craiova, 200349, Romania
| | - Anatoly Skalny
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl 150000, Russia; Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, Moscow 105064, Russia; Orenburg State University, Pobedy Ave., 13, Orenburg 460352, Russia
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States; Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, Moscow 105064, Russia.
| |
Collapse
|
14
|
Robinson SB, Refai O, Hardaway JA, Sturgeon S, Popay T, Bermingham DP, Freeman P, Wright J, Blakely RD. Dopamine-dependent, swimming-induced paralysis arises as a consequence of loss of function mutations in the RUNX transcription factor RNT-1. PLoS One 2019; 14:e0216417. [PMID: 31083672 PMCID: PMC6513266 DOI: 10.1371/journal.pone.0216417] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/21/2019] [Indexed: 11/18/2022] Open
Abstract
Dopamine (DA) is a neurotransmitter with actions across phylogeny that modulate core behaviors such as motor activity, reward, attention, and cognition. Perturbed DA signaling in humans is associated with multiple disorders, including addiction, ADHD, schizophrenia, and Parkinson's disease. The presynaptic DA transporter exerts powerful control on DA signaling by efficient clearance of the neurotransmitter following release. As in vertebrates, Caenorhabditis elegans DAT (DAT-1) constrains DA signaling and loss of function mutations in the dat-1 gene result in slowed crawling on solid media and swimming-induced paralysis (Swip) in water. Previously, we identified a mutant line, vt34, that exhibits robust DA-dependent Swip. vt34 exhibits biochemical and behavioral phenotypes consistent with reduced DAT-1 function though vt34; dat-1 double mutants exhibit an enhanced Swip phenotype, suggesting contributions of the vt34-associated mutation to additional mechanisms that lead to excess DA signaling. SNP mapping and whole genome sequencing of vt34 identified the site of the molecular lesion in the gene B0412.2 that encodes the Runx transcription factor ortholog RNT-1. Unlike dat-1 animals, but similar to other loss of function rnt-1 mutants, vt34 exhibits altered male tail morphology and reduced body size. Deletion mutations in both rnt-1 and the bro-1 gene, which encodes a RNT-1 binding partner also exhibit Swip. Both vt34 and rnt-1 mutations exhibit reduced levels of dat-1 mRNA as well as the tyrosine hydroxylase ortholog cat-2. Although reporter studies indicate that rnt-1 is expressed in DA neurons, its re-expression in DA neurons of vt34 animals fails to fully rescue Swip. Moreover, as shown for vt34, rnt-1 mutation exhibits additivity with dat-1 in generating Swip, as do rnt-1 and bro-1 mutations, and vt34 exhibits altered capacity for acetylcholine signaling at the neuromuscular junction. Together, these findings identify a novel role for rnt-1 in limiting DA neurotransmission and suggest that loss of RNT-1 may disrupt function of both DA neurons and body wall muscle to drive Swip.
Collapse
Affiliation(s)
- Sarah B Robinson
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Osama Refai
- Department of Biomedical Science, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL United States of America
| | - J Andrew Hardaway
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Sarah Sturgeon
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Tessa Popay
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Daniel P Bermingham
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Phyllis Freeman
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, United States of America
| | - Jane Wright
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL United States of America
- Brain Institute, Florida Atlantic University, Jupiter, FL, United States of America
| |
Collapse
|
15
|
Essmann CL, Ryan KR, Elmi M, Bryon-Dodd K, Porter A, Vaughan A, McMullan R, Nurrish S. Activation of RHO-1 in cholinergic motor neurons competes with dopamine signalling to control locomotion. PLoS One 2018; 13:e0204057. [PMID: 30240421 PMCID: PMC6150489 DOI: 10.1371/journal.pone.0204057] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/31/2018] [Indexed: 12/14/2022] Open
Abstract
The small GTPase RhoA plays a crucial role in the regulation of neuronal signalling to generate behaviour. In the developing nervous system RhoA is known to regulate the actin cytoskeleton, however the effectors of RhoA-signalling in adult neurons remain largely unidentified. We have previously shown that activation of the RhoA ortholog (RHO-1) in C. elegans cholinergic motor neurons triggers hyperactivity of these neurons and loopy locomotion with exaggerated body bends. This is achieved in part through increased diacylglycerol (DAG) levels and the recruitment of the synaptic vesicle protein UNC-13 to synaptic release sites, however other pathways remain to be identified. Dopamine, which is negatively regulated by the dopamine re-uptake transporter (DAT), has a central role in modulating locomotion in both humans and C. elegans. In this study we identify a new pathway in which RHO-1 regulates locomotory behaviour by repressing dopamine signalling, via DAT-1, linking these two pathways together. We observed an upregulation of dat-1 expression when RHO-1 is activated and show that loss of DAT-1 inhibits the loopy locomotion phenotype caused by RHO-1 activation. Reducing dopamine signalling in dat-1 mutants through mutations in genes involved in dopamine synthesis or in the dopamine receptor DOP-1 restores the ability of RHO-1 to trigger loopy locomotion in dat-1 mutants. Taken together, we show that negative regulation of dopamine signalling via DAT-1 is necessary for the neuronal RHO-1 pathway to regulate locomotion.
Collapse
Affiliation(s)
- Clara L. Essmann
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Katie R. Ryan
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Muna Elmi
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Kimberley Bryon-Dodd
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Andrew Porter
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Andrew Vaughan
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Rachel McMullan
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Stephen Nurrish
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| |
Collapse
|
16
|
Refai O, Blakely RD. Blockade and reversal of swimming-induced paralysis in C. elegans by the antipsychotic and D2-type dopamine receptor antagonist azaperone. Neurochem Int 2018; 123:59-68. [PMID: 29800604 DOI: 10.1016/j.neuint.2018.05.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 10/16/2022]
Abstract
The catecholamine neurotransmitter dopamine (DA) exerts powerful modulatory control of physiology and behavior across phylogeny. Perturbations of DA signaling in humans are associated with multiple neurodegenerative and behavioral disorders, including Parkinson's disease, attention-deficit/hyperactivity disorder, addiction and schizophrenia. In the nematode C. elegans, DA signaling regulates mating behavior, learning, food seeking and locomotion. Previously, we demonstrated that loss of function mutations in the dat-1 gene that encodes the presynaptic DA transporter (DAT-1) results in a rapid cessation of movement when animals are placed in water, termed Swimming Induced Paralysis (Swip). Loss of function mutations in genes that support DA biosynthesis, DA vesicular packaging and DA action at the extrasynaptic D2-type DA receptor DOP-3 suppress Swip in dat-1 animals, consistent with paralysis as arising from excessive DA signaling. Although animals grown on the vesicular monoamine transporter antagonist reserpine diminish Swip, the drug must be applied chronically, can impact the signaling of multiple biogenic amines, and has been reported to have penetrant, off-target actions. Here, we demonstrate that the antipsychotic drug azaperone potently and rapidly suppresses Swip behavior in either dat-1 mutants, as well as in wildtype animals treated with the DAT-1 antagonist nisoxetine, with genetic experiments consistent with DOP-3 antagonism as the mechanism of Swip suppression. Reversal of Swip in previously paralyzed dat-1 animals by azaperone application demonstrates an otherwise functionally-intact swimming circuit in these mutants. Finally, whereas azaperone suppresses DA-dependent Swip, the drug fails to attenuate the DA-independent paralysis induced by βPEA, aldicarb or genetic disruption of γ-aminobutyric acid (GABA) signaling. We discuss our findings with respect to the use of azaperone as a potent and selective tool in the identification and analysis of presynaptic mechanisms that regulate DA signaling.
Collapse
Affiliation(s)
- Osama Refai
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, FL, USA
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, FL, USA; Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA.
| |
Collapse
|
17
|
The Atypical MAP Kinase SWIP-13/ERK8 Regulates Dopamine Transporters through a Rho-Dependent Mechanism. J Neurosci 2017; 37:9288-9304. [PMID: 28842414 DOI: 10.1523/jneurosci.1582-17.2017] [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: 06/05/2017] [Revised: 07/31/2017] [Accepted: 08/12/2017] [Indexed: 12/26/2022] Open
Abstract
The neurotransmitter dopamine (DA) regulates multiple behaviors across phylogeny, with disrupted DA signaling in humans associated with addiction, attention-deficit/ hyperactivity disorder, schizophrenia, and Parkinson's disease. The DA transporter (DAT) imposes spatial and temporal limits on DA action, and provides for presynaptic DA recycling to replenish neurotransmitter pools. Molecular mechanisms that regulate DAT expression, trafficking, and function, particularly in vivo, remain poorly understood, though recent studies have implicated rho-linked pathways in psychostimulant action. To identify genes that dictate the ability of DAT to sustain normal levels of DA clearance, we pursued a forward genetic screen in Caenorhabditis elegans based on the phenotype swimming-induced paralysis (Swip), a paralytic behavior observed in hermaphrodite worms with loss-of-function dat-1 mutations. Here, we report the identity of swip-13, which encodes a highly conserved ortholog of the human atypical MAP kinase ERK8. We present evidence that SWIP-13 acts presynaptically to insure adequate levels of surface DAT expression and DA clearance. Moreover, we provide in vitro and in vivo evidence supporting a conserved pathway involving SWIP-13/ERK8 activation of Rho GTPases that dictates DAT surface expression and function.SIGNIFICANCE STATEMENT Signaling by the neurotransmitter dopamine (DA) is tightly regulated by the DA transporter (DAT), insuring efficient DA clearance after release. Molecular networks that regulate DAT are poorly understood, particularly in vivo Using a forward genetic screen in the nematode Caenorhabditis elegans, we implicate the atypical mitogen activated protein kinase, SWIP-13, in DAT regulation. Moreover, we provide in vitro and in vivo evidence that SWIP-13, as well as its human counterpart ERK8, regulate DAT surface availability via the activation of Rho proteins. Our findings implicate a novel pathway that regulates DA synaptic availability and that may contribute to risk for disorders linked to perturbed DA signaling. Targeting this pathway may be of value in the development of therapeutics in such disorders.
Collapse
|
18
|
Maulik M, Mitra S, Bult-Ito A, Taylor BE, Vayndorf EM. Behavioral Phenotyping and Pathological Indicators of Parkinson's Disease in C. elegans Models. Front Genet 2017; 8:77. [PMID: 28659967 PMCID: PMC5468440 DOI: 10.3389/fgene.2017.00077] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/22/2017] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder with symptoms that progressively worsen with age. Pathologically, PD is characterized by the aggregation of α-synuclein in cells of the substantia nigra in the brain and loss of dopaminergic neurons. This pathology is associated with impaired movement and reduced cognitive function. The etiology of PD can be attributed to a combination of environmental and genetic factors. A popular animal model, the nematode roundworm Caenorhabditis elegans, has been frequently used to study the role of genetic and environmental factors in the molecular pathology and behavioral phenotypes associated with PD. The current review summarizes cellular markers and behavioral phenotypes in transgenic and toxin-induced PD models of C. elegans.
Collapse
Affiliation(s)
- Malabika Maulik
- Department of Chemistry and Biochemistry, University of Alaska FairbanksFairbanks, AK, United States
| | - Swarup Mitra
- Department of Chemistry and Biochemistry, University of Alaska FairbanksFairbanks, AK, United States
| | - Abel Bult-Ito
- Department of Biology and Wildlife, University of Alaska FairbanksFairbanks, AK, United States
| | - Barbara E Taylor
- Department of Biological Sciences, California State University, Long BeachLong Beach, CA, United States
| | - Elena M Vayndorf
- Institute of Arctic Biology, University of Alaska FairbanksFairbanks, AK, United States
| |
Collapse
|
19
|
Sequence determinants of the Caenhorhabditis elegans dopamine transporter dictating in vivo axonal export and synaptic localization. Mol Cell Neurosci 2016; 78:41-51. [PMID: 27913309 DOI: 10.1016/j.mcn.2016.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 02/06/2023] Open
Abstract
The monoamine neurotransmitter dopamine (DA) acts across phylogeny to modulate both simple and complex behaviors. The presynaptic DA transporter (DAT) is a major determinant of DA signaling capacity in ensuring efficient extracellular DA clearance. In humans, DAT is also a major target for prescribed and abused psychostimulants. Multiple structural determinants of DAT function and regulation have been defined, though largely these findings have arisen from heterologous expression or ex vivo cell culture studies. Loss of function mutations in the gene encoding the Caenhorhabditis elegans DAT (dat-1) produces rapid immobility when animals are placed in water, a phenotype termed swimming-induced paralysis (Swip). The ability of a DA neuron-expressed, GFP-tagged DAT-1 fusion protein (GFP::DAT-1) to localize to synapses and rescue Swip in these animals provides a facile approach to define sequences supporting DAT somatic export and function in vivo. In prior studies, we found that truncation of the last 25 amino acids of the DAT-1 C-terminus (Δ25) precludes Swip rescue, supported by a deficit in GFP::DAT-1 synaptic localization. Here, we further defined the elements within Δ25 required for DAT-1 export and function in vivo. We identified two conserved motifs (584KW585 and 591PYRKR595) where mutation results in a failure of GFP::DAT-1 to be efficiently exported to synapses and restore DAT-1 function. The 584KW585 motif conforms to a sequence proposed to support SEC24 binding, ER export from the endoplasmic reticulum (ER), and surface expression of mammalian DAT proteins, whereas the 591PYRKR595 sequence conforms to a 3R motif identified as a SEC24 binding site in vertebrate G-protein coupled receptors. Consistent with a potential role of SEC24 orthologs in DAT-1 export, we demonstrated DA neuron-specific expression of a sec-24.2 transcriptional reporter. Mutations of the orthologous C-terminal sequences in human DAT (hDAT) significantly reduced transporter surface expression and DA uptake, despite normal hDAT protein expression. Although, hDAT mutants retained SEC24 interactions, as defined in co-immunoprecipitation studies. However, these mutations disrupted the ability of SEC24D to enhance hDAT surface expression. Our studies document an essential role of conserved DAT C-terminal sequences in transporter somatic export and synaptic localization in vivo, that add further support for important roles for SEC24 family members in efficient transporter trafficking.
Collapse
|
20
|
Caito SW, Aschner M. Quantification of Glutathione in Caenorhabditis elegans. ACTA ACUST UNITED AC 2016; 64:6.18.1-6.18.6. [PMID: 26309452 DOI: 10.1002/0471140856.tx0618s64] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Glutathione (GSH) is the most abundant intracellular thiol with diverse functions from redox signaling, xenobiotic detoxification, and apoptosis. The quantification of GSH is an important measure for redox capacity and oxidative stress. This protocol quantifies total GSH from Caenorhabditis elegans, an emerging model organism for toxicology studies. GSH is measured using the 5,5'-dithiobis-(2-nitrobenzoic acid) (DTNB) cycling method originally created for cell and tissue samples but optimized for whole worm extracts. DTNB reacts with GSH to from a 5'-thio-2-nitrobenzoic acid (TNB) chromophore with maximum absorbance of 412 nm. This method is both rapid and sensitive, making it ideal for studies involving a large number of transgenic nematode strains.
Collapse
Affiliation(s)
- Samuel W Caito
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
21
|
Wang T, Du L, Shan L, Dong H, Feng J, Kiessling MC, Angstman NB, Schmitz C, Jia F. A Prospective Case-Control Study of Radial Extracorporeal Shock Wave Therapy for Spastic Plantar Flexor Muscles in Very Young Children With Cerebral Palsy. Medicine (Baltimore) 2016; 95:e3649. [PMID: 27175689 PMCID: PMC4902531 DOI: 10.1097/md.0000000000003649] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
To assess the effects of radial extracorporeal shock wave therapy (rESWT) on plantar flexor muscle spasticity and gross motor function in very young patients with cerebral palsy (CP).The design was case-control study (level of evidence 3).The setting was the Department of Pediatric Neurology and Neurorehabilitation, First Hospital of Jilin University, Changchun, China.Those with a diagnosis of CP and spastic plantar flexor muscles were recruited between April 2014 and April 2015.According to the parents' decision, patients received 1 ESWT session per week for 3 months, with 1500 radial shock waves per ESWT session and leg with positive energy flux density of 0.03 mJ/mm, combined with traditional conservative therapy (rESWT group) or traditional conservative therapy alone (control group).The Modified Ashworth Scale (MAS) (primary outcome measure) and passive range of motion (pROM) measurements were collected at baseline (BL), 1 month (M1), and 3 months (M3) after BL. The Gross Motor Function Measure (GMFM)-88 was collected at BL and M3.Sixty-six patients completed the final review at 3 months and were included in the study. Subjects ranged in age from 12 to 60 months (mean age 27.0 ± 13.6 months; median age 22.0 months; 33.3% female). For the rESWT group (n = 34), mean MAS grades at BL, M1, and M3 were 2.6, 1.9, and 1.5 on the left side and 1.9, 1.7, and 1.2 on the right side. For the control group (n = 32), mean MAS grades at BL, M1, and M3 were 2.5, 2.4, and 2.1 on the left side and 1.8, 1.8, and 1.5 on the right side. The within-subject effects time × side and time × treatment were statistically significant (P < 0.01). Similar results were found for the improvement of mean pROM. GMFM-88 improved from BL to M3, but showed no statistically significant difference between the groups. There were no significant complications.This study demonstrates that the combination of rESWT and traditional conservative therapy is more effective than traditional conservative therapy alone in the treatment of spasticity in very young patients with CP.
Collapse
Affiliation(s)
- Tiantian Wang
- From the Department of Pediatric Neurology and Neurorehabilitation (TW, LD, LS, HD, JF, FJ) ; the Institute of Pediatrics (FJ), First Hospital of Jilin University, Changchun, China; the Neurological Research Center (FJ), First Hospital of Jilin University, Changchun, China; and the Extracorporeal Shock Wave Research Unit (MCK, NBA, CS), Department of Neuroanatomy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Zheng J, Wang M, Wei W, Keller JN, Adhikari B, King JF, King ML, Peng N, Laine RA. Dietary Plant Lectins Appear to Be Transported from the Gut to Gain Access to and Alter Dopaminergic Neurons of Caenorhabditis elegans, a Potential Etiology of Parkinson's Disease. Front Nutr 2016; 3:7. [PMID: 27014695 PMCID: PMC4780318 DOI: 10.3389/fnut.2016.00007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 02/15/2016] [Indexed: 11/13/2022] Open
Abstract
Lectins from dietary plants have been shown to enhance drug absorption in the gastrointestinal tract of rats, be transported trans-synaptically as shown by tracing of axonal and dendritic paths, and enhance gene delivery. Other carbohydrate-binding protein toxins are known to traverse the gut intact in dogs. Post-feeding rhodamine- or TRITC-tagged dietary lectins, the lectins were tracked from gut to dopaminergic neurons (DAergic-N) in transgenic Caenorhabditis elegans (C. elegans) [egIs1(Pdat-1:GFP)] where the mutant has the green fluorescent protein (GFP) gene fused to a dopamine transport protein gene labeling DAergic-N. The lectins were supplemented along with the food organism Escherichia coli (OP50). Among nine tested rhodamine/TRITC-tagged lectins, four, including Phaseolus vulgaris erythroagglutinin (PHA-E), Bandeiraea simplicifolia (BS-I), Dolichos biflorus agglutinin (DBA), and Arachis hypogaea agglutinin (PNA), appeared to be transported from gut to the GFP-DAergic-N. Griffonia Simplicifolia and PHA-E, reduced the number of GFP-DAergic-N, suggesting a toxic activity. PHA-E, BS-I, Pisum sativum (PSA), and Triticum vulgaris agglutinin (Succinylated) reduced fluorescent intensity of GFP-DAergic-N. PHA-E, PSA, Concanavalin A, and Triticum vulgaris agglutinin decreased the size of GFP-DAergic-N, while BS-I increased neuron size. These observations suggest that dietary plant lectins are transported to and affect DAergic-N in C. elegans, which support Braak and Hawkes' hypothesis, suggesting one alternate potential dietary etiology of Parkinson's disease (PD). A recent Danish study showed that vagotomy resulted in 40% lower incidence of PD over 20 years. Differences in inherited sugar structures of gut and neuronal cell surfaces may make some individuals more susceptible in this conceptual disease etiology model.
Collapse
Affiliation(s)
- Jolene Zheng
- School of Nutrition and Food Sciences, Louisiana State University, Baton Rouge, LA, USA
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Mingming Wang
- School of Nutrition and Food Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Wenqian Wei
- Department of Veterinary Science, College of Agriculture, Louisiana State University, Baton Rouge, LA, USA
- School of Life Sciences, Fudan University, Shanghai, China
| | - Jeffrey N. Keller
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Binita Adhikari
- Nicholls State University, Thibodaux, LA, USA
- Louisiana Biomedical Research Network (LBRN) Summer Research Program (2010), Baton Rouge, LA, USA
| | - Jason F. King
- Department of Biological Sciences, Louisiana State University and A&M College, Baton Rouge, LA, USA
- Department of Chemistry, Louisiana State University and A&M College, Baton Rouge, LA, USA
| | - Michael L. King
- Department of Biological Sciences, Louisiana State University and A&M College, Baton Rouge, LA, USA
- Department of Chemistry, Louisiana State University and A&M College, Baton Rouge, LA, USA
| | - Nan Peng
- School of Life Sciences, Fudan University, Shanghai, China
| | - Roger A. Laine
- Department of Biological Sciences, Louisiana State University and A&M College, Baton Rouge, LA, USA
- Department of Chemistry, Louisiana State University and A&M College, Baton Rouge, LA, USA
| |
Collapse
|
23
|
Bermingham DP, Hardaway JA, Snarrenberg CL, Robinson SB, Folkes OM, Salimando GJ, Jinnah H, Blakely RD. Acute blockade of the Caenorhabditis elegans dopamine transporter DAT-1 by the mammalian norepinephrine transporter inhibitor nisoxetine reveals the influence of genetic modifications of dopamine signaling in vivo. Neurochem Int 2016; 98:122-8. [PMID: 26850478 DOI: 10.1016/j.neuint.2016.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 11/16/2022]
Abstract
Modulation of neurotransmission by the catecholamine dopamine (DA) is conserved across phylogeny. In the nematode Caenorhabditis elegans, excess DA signaling triggers Swimming-Induced Paralysis (Swip), a phenotype first described in animals with loss of function mutations in the presynaptic DA transporter (dat-1). Swip has proven to be a phenotype suitable for the identification of novel dat-1 mutations as well as the identification of novel genes that impact DA signaling. Pharmacological manipulations can also induce Swip, though the reagents employed to date lack specificity and potency, limiting their use in evaluation of dat-1 expression and function. Our lab previously established the mammalian norepinephrine transporter (NET) inhibitor nisoxetine to be a potent antagonist of DA uptake conferred by DAT-1 following heterologous expression. Here we demonstrate the ability of low (μM) concentrations of nisoxetine to trigger Swip within minutes of incubation, with paralysis dependent on DA release and signaling, and non-additive with Swip triggered by dat-1 deletion. Using nisoxetine in combination with genetic mutations that impact DA release, we further demonstrate the utility of the drug for demonstrating contributions of presynaptic DA receptors and ion channels to Swip. Together, these findings reveal nisoxetine as a powerful reagent for monitoring multiple dimensions of DA signaling in vivo, thus providing a new resource that can be used to evaluate contributions of dat-1 and other genes linked to DA signaling without the potential for compensations that attend constitutive genetic mutations.
Collapse
Affiliation(s)
- Daniel P Bermingham
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240-7933, USA
| | - J Andrew Hardaway
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240-7933, USA
| | - Chelsea L Snarrenberg
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240-7933, USA
| | - Sarah B Robinson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240-7933, USA
| | - Oakleigh M Folkes
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240-7933, USA
| | - Greg J Salimando
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240-7933, USA
| | - Hussain Jinnah
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240-7933, USA
| | - Randy D Blakely
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240-7933, USA; Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37240-7933, USA.
| |
Collapse
|
24
|
Caenorhabditis elegans as a Model to Study the Molecular and Genetic Mechanisms of Drug Addiction. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 137:229-52. [PMID: 26810004 DOI: 10.1016/bs.pmbts.2015.10.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Drug addiction takes a massive toll on society. Novel animal models are needed to test new treatments and understand the basic mechanisms underlying addiction. Rodent models have identified the neurocircuitry involved in addictive behavior and indicate that rodents possess some of the same neurobiologic mechanisms that mediate addiction in humans. Recent studies indicate that addiction is mechanistically and phylogenetically ancient and many mechanisms that underlie human addiction are also present in invertebrates. The nematode Caenorhabditis elegans has conserved neurobiologic systems with powerful molecular and genetic tools and a rapid rate of development that enables cost-effective translational discovery. Emerging evidence suggests that C. elegans is an excellent model to identify molecular mechanisms that mediate drug-induced behavior and potential targets for medications development for various addictive compounds. C. elegans emit many behaviors that can be easily quantitated including some that involve interactions with the environment. Ethanol (EtOH) is the best-studied drug-of-abuse in C. elegans and at least 50 different genes/targets have been identified as mediating EtOH's effects and polymorphisms in some orthologs in humans are associated with alcohol use disorders. C. elegans has also been shown to display dopamine and cholinergic system-dependent attraction to nicotine and demonstrate preference for cues previously associated with nicotine. Cocaine and methamphetamine have been found to produce dopamine-dependent reward-like behaviors in C. elegans. These behavioral tests in combination with genetic/molecular manipulations have led to the identification of dozens of target genes/systems in C. elegans that mediate drug effects. The one target/gene identified as essential for drug-induced behavioral responses across all drugs of abuse was the cat-2 gene coding for tyrosine hydroxylase, which is consistent with the role of dopamine neurotransmission in human addiction. Overall, C. elegans can be used to model aspects of drug addiction and identify systems and molecular mechanisms that mediate drug effects. The findings are surprisingly consistent with analogous findings in higher-level organisms. Further, model refinement is warranted to improve model validity and increase utility for medications development.
Collapse
|
25
|
Likhite N, Jackson CA, Liang MS, Krzyzanowski MC, Lei P, Wood JF, Birkaya B, Michaels KL, Andreadis ST, Clark SD, Yu MC, Ferkey DM. The protein arginine methyltransferase PRMT5 promotes D2-like dopamine receptor signaling. Sci Signal 2015; 8:ra115. [PMID: 26554819 PMCID: PMC5473623 DOI: 10.1126/scisignal.aad0872] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Protein arginine methylation regulates diverse functions of eukaryotic cells, including gene expression, the DNA damage response, and circadian rhythms. We showed that arginine residues within the third intracellular loop of the human D2 dopamine receptor, which are conserved in the DOP-3 receptor in the nematode Caenorhabditis elegans, were methylated by protein arginine methyltransferase 5 (PRMT5). By mutating these arginine residues, we further showed that their methylation enhanced the D2 receptor-mediated inhibition of cyclic adenosine monophosphate (cAMP) signaling in cultured human embryonic kidney (HEK) 293T cells. Analysis of prmt-5-deficient worms indicated that methylation promoted the dopamine-mediated modulation of chemosensory and locomotory behaviors in C. elegans through the DOP-3 receptor. In addition to delineating a previously uncharacterized means of regulating GPCR (heterotrimeric guanine nucleotide-binding protein-coupled receptor) signaling, these findings may lead to the development of a new class of pharmacological therapies that modulate GPCR signaling by changing the methylation status of these key proteins.
Collapse
Affiliation(s)
- Neah Likhite
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Christopher A Jackson
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Mao-Shih Liang
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Michelle C Krzyzanowski
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Pedro Lei
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Jordan F Wood
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Barbara Birkaya
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Kerry L Michaels
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Stelios T Andreadis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Stewart D Clark
- Department of Pharmacology and Toxicology, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA. Department of Psychology, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA. Research Institute on Addictions, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | - Michael C Yu
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA.
| | - Denise M Ferkey
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA.
| |
Collapse
|
26
|
Abstract
Caenorhabditis elegans has been shown to measure variability in environmental food density, using the information to fine-tune foraging strategies; a compact neural circuit has been identified that responds to large fluctuations in food-related cues and uses dopamine to encode the amount of recently encountered variability.
Collapse
Affiliation(s)
- Michael Hendricks
- Department of Biology, McGill University, Stewart Biology Building W5/11, 1205 av Dr Penfield, Montreal, Quebec H3A 1B1, Canada.
| |
Collapse
|
27
|
Verlinden H, Vleugels R, Verdonck R, Urlacher E, Vanden Broeck J, Mercer A. Pharmacological and signalling properties of a D2-like dopamine receptor (Dop3) in Tribolium castaneum. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2015; 56:9-20. [PMID: 25449128 DOI: 10.1016/j.ibmb.2014.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 11/05/2014] [Accepted: 11/06/2014] [Indexed: 06/04/2023]
Abstract
Dopamine is an important neurotransmitter in the central nervous system of vertebrates and invertebrates. Despite their evolutionary distance, striking parallels exist between deuterostomian and protostomian dopaminergic systems. In both, signalling is achieved via a complement of functionally distinct dopamine receptors. In this study, we investigated the sequence, pharmacology and tissue distribution of a D2-like dopamine receptor from the red flour beetle Tribolium castaneum (TricaDop3) and compared it with related G protein-coupled receptors in other invertebrate species. The TricaDop3 receptor-encoding cDNA shows considerable sequence similarity with members of the Dop3 receptor class. Real time qRT-PCR showed high expression in both the central brain and the optic lobes, consistent with the role of dopamine as neurotransmitter. Activation of TricaDop3 expressed in mammalian cells increased intracellular Ca(2+) signalling and decreased NKH-477 (a forskolin analogue)-stimulated cyclic AMP levels in a dose-dependent manner. We studied the pharmacological profile of the TricaDop3 receptor and demonstrated that the synthetic vertebrate dopamine receptor agonists, 2 - amino- 6,7 - dihydroxy - 1,2,3,4 - tetrahydronaphthalene hydrobromide (6,7-ADTN) and bromocriptine acted as agonists. Methysergide was the most potent of the antagonists tested and showed competitive inhibition in the presence of dopamine. This study offers important information on the Dop3 receptor from Tribolium castaneum that will facilitate functional analyses of dopamine receptors in insects and other invertebrates.
Collapse
Affiliation(s)
- Heleen Verlinden
- Department of Animal Physiology and Neurobiology, Zoological Institute, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium; Department of Zoology, University of Otago, 340 Great King Street, Dunedin, New Zealand.
| | - Rut Vleugels
- Department of Animal Physiology and Neurobiology, Zoological Institute, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium
| | - Rik Verdonck
- Department of Animal Physiology and Neurobiology, Zoological Institute, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium
| | - Elodie Urlacher
- Department of Zoology, University of Otago, 340 Great King Street, Dunedin, New Zealand
| | - Jozef Vanden Broeck
- Department of Animal Physiology and Neurobiology, Zoological Institute, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium
| | - Alison Mercer
- Department of Zoology, University of Otago, 340 Great King Street, Dunedin, New Zealand
| |
Collapse
|
28
|
Chege PM, McColl G. Caenorhabditis elegans: a model to investigate oxidative stress and metal dyshomeostasis in Parkinson's disease. Front Aging Neurosci 2014; 6:89. [PMID: 24904406 PMCID: PMC4032941 DOI: 10.3389/fnagi.2014.00089] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 04/27/2014] [Indexed: 12/04/2022] Open
Abstract
Parkinson's disease (PD) is characterized by progressive motor impairment attributed to progressive loss of dopaminergic (DAergic) neurons in the substantia nigra pars compacta. Additional clinical manifestations include non-motor symptoms such as insomnia, depression, psychosis, and cognitive impairment. PD patients with mild cognitive impairment have an increased risk of developing dementia. The affected brain regions also show perturbed metal ion levels, primarily iron. These observations have led to speculation that metal ion dyshomeostasis plays a key role in the neuronal death of this disease. However, the mechanisms underlying this metal-associated neurodegeneration have yet to be completely elucidated. Mammalian models have traditionally been used to investigate PD pathogenesis. However, alternate animal models are also being adopted, bringing to bear their respective experimental advantage. The nematode, Caenorhabditis elegans, is one such system that has well-developed genetics, is amenable to transgenesis and has relatively low associated experimental costs. C. elegans has a well characterized neuronal network that includes a simple DAergic system. In this review we will discuss mechanisms thought to underlie PD and the use of C. elegans to investigate these processes.
Collapse
Affiliation(s)
| | - Gawain McColl
- The Florey Institute of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| |
Collapse
|
29
|
Hardaway JA, Wang J, Fleming PA, Fleming KA, Whitaker SM, Nackenoff A, Snarrenberg CL, Hardie SL, Zhang B, Blakely RD. An open-source analytical platform for analysis of C. elegans swimming-induced paralysis. J Neurosci Methods 2014; 232:58-62. [PMID: 24792527 DOI: 10.1016/j.jneumeth.2014.04.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/22/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND The nematode Caenhorhabditis elegans offers great power for the identification and characterization of genes that regulate behavior. In support of this effort, analytical methods are required that provide dimensional analyses of subcomponents of behavior. Previously, we demonstrated that loss of the presynaptic dopamine (DA) transporter, dat-1, evokes DA-dependent Swimming-Induced Paralysis (Swip) (Mcdonald et al., 2007), a behavior compatible with forward genetic screens (Hardaway et al., 2012). NEW METHOD Here, we detail the development and implementation of SwimR, a set of tools that provide for an automated, kinetic analysis of C. elegans Swip. SwimR relies on open source programs that can be freely implemented and modified. RESULTS We show that SwimR can display time-dependent alterations of swimming behavior induced by drug-treatment, illustrating this capacity with the dat-1 blocker and tricyclic antidepressant imipramine (IMI). We demonstrate the capacity of SwimR to extract multiple kinetic parameters that are impractical to obtain in manual assays. COMPARISON WITH EXISTING METHODS Standard measurements of C. elegans swimming utilizes manual assessments of the number of animals exhibiting swimming versus paralysis. Our approach deconstructs the time course and rates of movement in an automated fashion, offering a significant increase in the information that can be obtained from swimming behavior. CONCLUSIONS The SwimR platform is a powerful tool for the deconstruction of worm thrashing behavior in the context of both genetic and pharmacological manipulations that can be used to segregate pathways that underlie nematode swimming mechanics.
Collapse
Affiliation(s)
| | - Jing Wang
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA
| | - Paul A Fleming
- Departments of Electrical Engineering and Computer Science, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA
| | - Katherine A Fleming
- Departments of Electrical Engineering and Computer Science, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA
| | | | - Alex Nackenoff
- Department of Pharmacology, Nashville, TN 37232-8548, USA
| | | | | | - Bing Zhang
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA
| | - Randy D Blakely
- Department of Pharmacology, Nashville, TN 37232-8548, USA; Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA.
| |
Collapse
|
30
|
Safratowich BD, Lor C, Bianchi L, Carvelli L. Amphetamine activates an amine-gated chloride channel to generate behavioral effects in Caenorhabditis elegans. J Biol Chem 2013; 288:21630-7. [PMID: 23775081 DOI: 10.1074/jbc.m113.484139] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Amphetamine is a highly addictive psychostimulant, which is thought to generate its effects by promoting release of dopamine through reverse activation of dopamine transporters. However, some amphetamine-mediated behaviors persist in dopamine transporter knock-out animals, suggesting the existence of alternative amphetamine targets. Here we demonstrate the identification of a novel amphetamine target by showing that in Caenorhabditis elegans, a large fraction of the behavioral effects of amphetamine is mediated through activation of the amine-gated chloride channel, LGC-55. These findings bring to light alternative pathways engaged by amphetamine, and urge rethinking of the molecular mechanisms underlying the effects of this highly-addictive psychostimulant.
Collapse
Affiliation(s)
- Bryan D Safratowich
- Department of Pharmacology, Physiology, and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9037, USA
| | | | | | | |
Collapse
|
31
|
Pfister G, Rieb J, Avramov M, Rock T, Griebler C, Schramm KW. Detection of catecholamines in single specimens of groundwater amphipods. Anal Bioanal Chem 2013; 405:5571-82. [DOI: 10.1007/s00216-013-6952-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 03/26/2013] [Accepted: 03/26/2013] [Indexed: 11/25/2022]
|
32
|
Caito SW, Zhang Y, Aschner M. Involvement of AAT transporters in methylmercury toxicity in Caenorhabditis elegans. Biochem Biophys Res Commun 2013; 435:546-50. [PMID: 23669041 DOI: 10.1016/j.bbrc.2013.04.090] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 04/17/2013] [Indexed: 10/26/2022]
Abstract
Methylmercury (MeHg) is a potent neurotoxin that enters mammalian cells as a conjugate with L-cysteine through L-type large neutral amino acid transporter, LAT1, by a molecular mimicry mechanism by structurally resembling L-methionine. Caenorhabditis elegans (C. elegans) has been increasingly used to study the neurotoxic effects of MeHg, but little is known about uptake and transport of MeHg in the worm. This study examined whether MeHg uptake through LAT1 is evolutionarily conserved in nematodes. MeHg toxicity in C. elegans was blocked by pre-treatment of worms with l-methionine, suggesting a role for amino acid transporters in MeHg transport. Knockdown of aat-1, aat-2, and aat-3, worm homologues to LAT1, increased the survival of C. elegans following MeHg treatment and significantly attenuated MeHg content following exposure. These results indicate that MeHg is transported in the worm by a conserved mechanism dependent on functioning amino acid transporters.
Collapse
Affiliation(s)
- Samuel W Caito
- Division of Clinical Pharmacology and Pediatric Toxicology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232-0414, USA
| | | | | |
Collapse
|
33
|
Salam S, Ansari A, Amon S, Rezai P, Selvaganapathy PR, Mishra RK, Gupta BP. A microfluidic phenotype analysis system reveals function of sensory and dopaminergic neuron signaling in C. elegans electrotactic swimming behavior. WORM 2013; 2:e24558. [PMID: 24058875 PMCID: PMC3704449 DOI: 10.4161/worm.24558] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/02/2013] [Accepted: 04/04/2013] [Indexed: 12/21/2022]
Abstract
The nematode (worm) C. elegans is a leading multicellular animal model to study neuronal-basis of behavior. Worms respond to a wide range of stimuli and exhibit characteristic movement patterns. Here we describe the use of a microfluidics setup to probe neuronal activity that relies on the innate response of C. elegans to swim toward the cathode in the presence of a DC electric field (termed "electrotaxis"). Using this setup, we examined mutants affecting sensory and dopaminergic neurons and found that their electrotactic responses were defective. Such animals moved with reduced speed (35-80% slower than controls) with intermittent pauses, abnormal turning and slower body bends. A similar phenotype was observed in worms treated with neurotoxins 6-OHDA (6- hydroxy dopamine), MPTP (1-methyl 4-phenyl 1,2,3,6-tetrahydropyridine) and rotenone (20-60% slower). We also found that neurotoxin effects could be suppressed by pre-exposing worms to a known neuroprotective compound acetaminophen. Collectively, these results show that microfluidic electrotaxis can identify alterations in dopamine and amphid neuronal signaling based on swimming responses of C. elegans. Further characterization has revealed that the electrotactic swimming response is highly sensitive and reliable in detecting neuronal abnormalities. Thus, our microfluidics setup could be used to dissect neuronal function and toxin-induced neurodegeneration. Among other applications, the setup promises to facilitate genetic and chemical screenings to identify factors that mediate neuronal signaling and neuroprotection.
Collapse
Affiliation(s)
- Sangeena Salam
- Department of Biology; McMaster University; Hamilton, ON Canada
| | - Ata Ansari
- Department of Biology; McMaster University; Hamilton, ON Canada
- Department of Psychiatry and Behavioral Neuroscience; McMaster University; Hamilton, ON Canada
| | - Siavash Amon
- Department of Biology; McMaster University; Hamilton, ON Canada
| | - Pouya Rezai
- Department of Mechanical Engineering; McMaster University; Hamilton, ON Canada
| | | | - Ram K. Mishra
- Department of Psychiatry and Behavioral Neuroscience; McMaster University; Hamilton, ON Canada
| | | |
Collapse
|
34
|
Caito S, Fretham S, Martinez-Finley E, Chakraborty S, Avila D, Chen P, Aschner M. Genome-Wide Analyses of Metal Responsive Genes in Caenorhabditis elegans. Front Genet 2012; 3:52. [PMID: 22514555 PMCID: PMC3322339 DOI: 10.3389/fgene.2012.00052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 03/20/2012] [Indexed: 11/22/2022] Open
Abstract
Metals are major contaminants that influence human health. Many metals have physiologic roles, but excessive levels can be harmful. Advances in technology have made toxicogenomic analyses possible to characterize the effects of metal exposure on the entire genome. Much of what is known about cellular responses to metals has come from mammalian systems; however the use of non-mammalian species is gaining wider attention. Caenorhabditis elegans is a small round worm whose genome has been fully sequenced and its development from egg to adult is well characterized. It is an attractive model for high throughput screens due to its short lifespan, ease of genetic mutability, low cost, and high homology with humans. Research performed in C. elegans has led to insights in apoptosis, gene expression, and neurodegeneration, all of which can be altered by metal exposure. Additionally, by using worms one can potentially study mechanisms that underline differential responses to metals in nematodes and humans, allowing for identification of novel pathways and therapeutic targets. In this review, toxicogenomic studies performed in C. elegans exposed to various metals will be discussed, highlighting how this non-mammalian system can be utilized to study cellular processes and pathways induced by metals. Recent work focusing on neurodegeneration in Parkinson’s disease will be discussed as an example of the usefulness of genetic screens in C. elegans and the novel findings that can be produced.
Collapse
Affiliation(s)
- Samuel Caito
- Division of Clinical Pharmacology and Pediatric Toxicology, Department of Pediatrics, Vanderbilt University Medical Center Nashville, TN, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Beggs KT, Tyndall JDA, Mercer AR. Honey bee dopamine and octopamine receptors linked to intracellular calcium signaling have a close phylogenetic and pharmacological relationship. PLoS One 2011; 6:e26809. [PMID: 22096499 PMCID: PMC3214027 DOI: 10.1371/journal.pone.0026809] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 10/04/2011] [Indexed: 12/04/2022] Open
Abstract
Background Three dopamine receptor genes have been identified that are highly conserved among arthropod species. One of these genes, referred to in honey bees as Amdop2, shows a close phylogenetic relationship to the a-adrenergic-like octopamine receptor family. In this study we examined in parallel the functional and pharmacological properties of AmDOP2 and the honey bee octopamine receptor, AmOA1. For comparison, pharmacological properties of the honey bee dopamine receptors AmDOP1 and AmDOP3, and the tyramine receptor AmTYR1, were also examined. Methodology/Principal Findings Using HEK293 cells heterologously expressing honey bee biogenic amine receptors, we found that activation of AmDOP2 receptors, like AmOA1 receptors, initiates a rapid increase in intracellular calcium levels. We found no evidence of calcium signaling via AmDOP1, AmDOP3 or AmTYR1 receptors. AmDOP2- and AmOA1-mediated increases in intracellular calcium were inhibited by 10 µM edelfosine indicating a requirement for phospholipase C-β activity in this signaling pathway. Edelfosine treatment had no effect on AmDOP2- or AmOA1-mediated increases in intracellular cAMP. The synthetic compounds mianserin and epinastine, like cis-(Z)-flupentixol and spiperone, were found to have significant antagonist activity on AmDOP2 receptors. All 4 compounds were effective antagonists also on AmOA1 receptors. Analysis of putative ligand binding sites offers a possible explanation for why epinastine acts as an antagonist at AmDOP2 receptors, but fails to block responses mediated via AmDOP1. Conclusions/Significance Our results indicate that AmDOP2, like AmOA1, is coupled not only to cAMP, but also to calcium-signalling and moreover, that the two signalling pathways are independent upstream of phospholipase C-β activity. The striking similarity between the pharmacological properties of these 2 receptors suggests an underlying conservation of structural properties related to receptor function. Taken together, these results strongly support phylogenetic analyses indicating that the AmDOP2 and AmOA1 receptor genes are immediate paralogs.
Collapse
Affiliation(s)
- Kyle T. Beggs
- Department of Zoology, University of Otago, Dunedin, New Zealand
| | | | - Alison R. Mercer
- Department of Zoology, University of Otago, Dunedin, New Zealand
- * E-mail:
| |
Collapse
|
36
|
Martinez-Finley EJ, Avila DS, Chakraborty S, Aschner M. Insights from Caenorhabditis elegans on the role of metals in neurodegenerative diseases. Metallomics 2011; 3:271-9. [PMID: 21210060 PMCID: PMC3172965 DOI: 10.1039/c0mt00064g] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neurodegeneration is characterized by the cell death or loss of structure and/or function of neurons. Many neurodegenerative diseases including Parkinson's disease (PD) and Alzheimer's disease (AD) are the result of neurodegenerative processes. Metals are essential for many life processes, but they are also culpable for several neurodegenerative mechanisms. In this review, we discuss the role of metals in neurodegenerative diseases with emphasis on the utility of Caenorhabditis elegans (C. elegans) genetic models in deciphering mechanisms associated with the etiology of PD and AD.
Collapse
Affiliation(s)
- Ebany J. Martinez-Finley
- Division of Clinical Pharmacology and Pediatric Toxicology, Vanderbilt University Medical Center, 11425 MRB IV, 2215-B Garland Ave., Nashville, TN 37232-0414, USA; Tel: 615-322-8024
- Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daiana Silva Avila
- Division of Clinical Pharmacology and Pediatric Toxicology, Vanderbilt University Medical Center, 11425 MRB IV, 2215-B Garland Ave., Nashville, TN 37232-0414, USA; Tel: 615-322-8024
| | - Sudipta Chakraborty
- Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Molecular Neuroscience, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael Aschner
- Division of Clinical Pharmacology and Pediatric Toxicology, Vanderbilt University Medical Center, 11425 MRB IV, 2215-B Garland Ave., Nashville, TN 37232-0414, USA; Tel: 615-322-8024
- Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Molecular Neuroscience, Vanderbilt University Medical Center, Nashville, TN, USA
- The Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Division of Pediatric Toxicology, Nashville, TN, USA
| |
Collapse
|
37
|
Aitlhadj L, Ávila DS, Benedetto A, Aschner M, Stürzenbaum SR. Environmental exposure, obesity, and Parkinson's disease: lessons from fat and old worms. ENVIRONMENTAL HEALTH PERSPECTIVES 2011; 119:20-8. [PMID: 20797931 PMCID: PMC3018495 DOI: 10.1289/ehp.1002522] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 08/25/2010] [Indexed: 05/02/2023]
Abstract
BACKGROUND A common link has been exposed, namely, that metal exposure plays a role in obesity and in Parkinson's disease (PD). This link may help to elucidate mechanisms of neurotoxicity. OBJECTIVE We reviewed the utility of the nematode, Caenorhabditis elegans, as a model organism to study neurodegeneration in obesity and Parkinson's disease (PD), with an emphasis on the neurotransmitter, dopamine (DA). DATA SOURCES A PubMed literature search was performed using the terms "obesity" and any of the following: "C. elegans," "central nervous system," "neurodegeneration," "heavy metals," "dopamine" or "Parkinson's disease." We reviewed the identified studies, including others cited therein, to summarize the current evidence of neurodegeneration in obesity and PD, with an emphasis on studies carried out in C. elegans and environmental toxins in the etiology of both diseases. DATA EXTRACTION AND DATA SYNTHESIS Heavy metals and DA have both been linked to diet-induced obesity, which has led to the notion that the mechanism of environmentally induced neurodegeneration in PD may also apply to obesity. C. elegans has been instrumental in expanding our mechanism-based knowledge of PD, and this species is emerging as a good model of obesity. With well-established toxicity and neurogenetic assays, it is now feasible to explore the putative link between metal- and chemical-induced neurodegeneration. CONCLUSIONS One side effect of an aging population is an increase in the prevalence of obesity, metabolic disorders, and neurodegenerative orders, diseases that are likely to co-occur. Environmental toxins, especially heavy metals, may prove to be a previously neglected part of the puzzle.
Collapse
Affiliation(s)
- Layla Aitlhadj
- King’s College London, Pharmaceutical Science Division, London, United Kingdom
| | - Daiana Silva Ávila
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alexandre Benedetto
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael Aschner
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | |
Collapse
|
38
|
Harrington AJ, Hamamichi S, Caldwell GA, Caldwell KA. C. elegans as a model organism to investigate molecular pathways involved with Parkinson's disease. Dev Dyn 2010; 239:1282-95. [PMID: 20108318 DOI: 10.1002/dvdy.22231] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Parkinson's disease (PD) is an age-related movement disorder resulting, in part, from selective loss of dopaminergic neurons. Both invertebrate and mammalian models have been developed to study the cellular mechanisms altered during disease progression; nevertheless there are limitations within each model. Mammalian models remain invaluable in studying PD, but are expensive and time consuming. Here, we review genetic and environmental factors associated with PD, and describe how the nematode roundworm, Caenorhabditis elegans, has been used as a model organism for studying various aspects of this neurodegenerative disease. Both genetic and chemical screens have been conducted in C. elegans to identify molecular pathways, proteins, and small molecules that can impact PD pathology. Lastly, we highlight future areas of investigation, in the context of emerging fields in biology, where the nematode can be exploited to provide mechanistic insights and potential strategies to accelerate the path toward possible therapeutic intervention for PD.
Collapse
Affiliation(s)
- Adam J Harrington
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama 35487-0344, USA
| | | | | | | |
Collapse
|
39
|
Ezak MJ, Ferkey DM. The C. elegans D2-like dopamine receptor DOP-3 decreases behavioral sensitivity to the olfactory stimulus 1-octanol. PLoS One 2010; 5:e9487. [PMID: 20209143 PMCID: PMC2830454 DOI: 10.1371/journal.pone.0009487] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Accepted: 02/10/2010] [Indexed: 11/19/2022] Open
Abstract
We previously found that dopamine signaling modulates the sensitivity of wild-type C. elegans to the aversive odorant 1-octanol. C. elegans lacking the CAT-2 tyrosine hydroxylase enzyme, which is required for dopamine biosynthesis, are hypersensitive in their behavioral avoidance of dilute concentrations of octanol. Dopamine can also modulate the context-dependent response of C. elegans lacking RGS-3 function, a negative regulator of G alpha signaling. rgs-3 mutant animals are defective in their avoidance of 100% octanol when they are assayed in the absence of food (E. coli bacterial lawn), but their response is restored when they are assayed in the presence of food or exogenous dopamine. However, it is not known which receptor might be mediating dopamine's effects on octanol avoidance. Herein we describe a role for the C. elegans D2-like receptor DOP-3 in the regulation of olfactory sensitivity. We show that DOP-3 is required for the ability of food and exogenous dopamine to rescue the octanol avoidance defect of rgs-3 mutant animals. In addition, otherwise wild-type animals lacking DOP-3 function are hypersensitive to dilute octanol, reminiscent of cat-2 mutants. Furthermore, we demonstrate that DOP-3 function in the ASH sensory neurons is sufficient to rescue the hypersensitivity of dop-3 mutant animals, while dop-3 RNAi knockdown in ASH results in octanol hypersensitivity. Taken together, our data suggest that dopaminergic signaling through DOP-3 normally acts to dampen ASH signaling and behavioral sensitivity to octanol.
Collapse
Affiliation(s)
- Meredith J. Ezak
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Denise M. Ferkey
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
| |
Collapse
|
40
|
Voglis G, Tavernarakis N. A synaptic DEG/ENaC ion channel mediates learning in C. elegans by facilitating dopamine signalling. EMBO J 2008; 27:3288-99. [PMID: 19037257 DOI: 10.1038/emboj.2008.252] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Accepted: 11/07/2008] [Indexed: 01/26/2023] Open
Abstract
An important component of learned behaviour is the ability to forecast positive or negative outcomes based on specific sensory cues. Predictive capacity is typically manifested by appropriate behavioural patterning. However, the molecular mechanisms underlying behavioural plasticity are poorly understood. Caenorhabditis elegans displays experience-dependent behavioural responses by associating distinct environmental signals. We find that ASIC-1, a member of the degenerin/epithelial sodium channel family, which localizes at presynaptic terminals of dopaminergic neurons, is required for associative learning in C. elegans. ASIC-1 functions in these neurons to amplify normal dopaminergic signalling, necessary for associative learning. Our results reveal a novel role of DEG/ENaC ion channels in neuronal communication by enhancing the activity of dopaminergic synapses. Similar mechanisms may facilitate synaptic plasticity in vertebrates.
Collapse
Affiliation(s)
- Giannis Voglis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | | |
Collapse
|
41
|
McDonald PW, Hardie SL, Jessen TN, Carvelli L, Matthies DS, Blakely RD. Vigorous motor activity in Caenorhabditis elegans requires efficient clearance of dopamine mediated by synaptic localization of the dopamine transporter DAT-1. J Neurosci 2007; 27:14216-27. [PMID: 18094261 PMCID: PMC6673513 DOI: 10.1523/jneurosci.2992-07.2007] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2007] [Revised: 10/16/2007] [Accepted: 11/13/2007] [Indexed: 11/21/2022] Open
Abstract
The catecholamine dopamine (DA) functions as a powerful modulatory neurotransmitter in both invertebrates and vertebrates. As in man, DA neurons in the nematode Caenorhabditis elegans express a cocaine-sensitive transporter (DAT-1), presumably to regulate synaptic DA signaling and limit DA spillover to extrasynaptic sites, although evidence supporting this is currently lacking. In this report, we describe and validate a novel and readily quantifiable phenotype, swimming-induced paralysis (SWIP) that emerges in DAT-1-deficient nematodes when animals exert maximal physical activity in water. We verify the dependence of SWIP on DA biosynthesis, vesicular packaging, synaptic release, and on the DA receptor DOP-3. Using DAT-1 specific antibodies and GFP::DAT-1 fusions, we demonstrate a synaptic enrichment of DAT-1 that is achieved independently of synaptic targeting of the vesicular monoamine transporter (VMAT). Importantly, dat-1 deletions and point mutations that disrupt DA uptake in cultured C. elegans neurons and/or impact DAT-1 synaptic localization in vivo generate SWIP. SWIP assays, along with in vivo imaging of wild-type and mutant GFP::DAT-1 fusions identify a distal COOH terminal segment of the transporter as essential for efficient somatic export, synaptic localization and in vivo DA clearance. Our studies provide the first description of behavioral perturbations arising from altered trafficking of DATs in vivo in any organism and support a model whereby endogenous DA actions in C. elegans are tightly regulated by synaptic DAT-1.
Collapse
Affiliation(s)
- Paul W McDonald
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548, USA
| | | | | | | | | | | |
Collapse
|
42
|
Schmidt E, Seifert M, Baumeister R. Caenorhabditis elegans as a model system for Parkinson's disease. NEURODEGENER DIS 2007; 4:199-217. [PMID: 17596715 DOI: 10.1159/000101845] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is one of the most common age-related neurodegenerative diseases that is characterized by selective loss of dopaminergic neurons. Despite recent findings from mammalian model systems, molecular mechanisms of the pathophysiology are poorly understood. Given the high conservation of molecular pathways from invertebrates to mammalians, combined with technical advantages, such as high-throughput approaches, Caenorhabditis elegans represents a powerful system for the identification of factors involved in neurodegeneration. In this review we describe that C. elegans can be used to advance our understanding of the genetic mechanisms implicated in these disorders.
Collapse
Affiliation(s)
- Enrico Schmidt
- Bioinformatics and Molecular Genetics (Faculty of Biology), Center for Biochemistry, University of Freiburg, Freiburg, Germany
| | | | | |
Collapse
|