1
|
Rodriguez P, Kalia V, Fenollar-Ferrer C, Gibson CL, Gichi Z, Rajoo A, Matier CD, Pezacki AT, Xiao T, Carvelli L, Chang CJ, Miller GW, Khamoui AV, Boerner J, Blakely RD. Glial swip-10 controls systemic mitochondrial function, oxidative stress, and neuronal viability via copper ion homeostasis. Proc Natl Acad Sci U S A 2024; 121:e2320611121. [PMID: 39288174 PMCID: PMC11441482 DOI: 10.1073/pnas.2320611121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 08/01/2024] [Indexed: 09/19/2024] Open
Abstract
Cuprous copper [Cu(I)] is an essential cofactor for enzymes that support many fundamental cellular functions including mitochondrial respiration and suppression of oxidative stress. Neurons are particularly reliant on mitochondrial production of ATP, with many neurodegenerative diseases, including Parkinson's disease, associated with diminished mitochondrial function. The gene MBLAC1 encodes a ribonuclease that targets pre-mRNA of replication-dependent histones, proteins recently found in yeast to reduce Cu(II) to Cu(I), and when mutated disrupt ATP production, elevates oxidative stress, and severely impacts cell growth. Whether this process supports neuronal and/or systemic physiology in higher eukaryotes is unknown. Previously, we identified swip-10, the putative Caenorhabditis elegans ortholog of MBLAC1, establishing a role for glial swip-10 in limiting dopamine (DA) neuron excitability and sustaining DA neuron viability. Here, we provide evidence from computational modeling that SWIP-10 protein structure mirrors that of MBLAC1 and locates a loss of function coding mutation at a site expected to disrupt histone RNA hydrolysis. Moreover, we find through genetic, biochemical, and pharmacological studies that deletion of swip-10 in worms negatively impacts systemic Cu(I) levels, leading to deficits in mitochondrial respiration and ATP production, increased oxidative stress, and neurodegeneration. These phenotypes can be offset in swip-10 mutants by the Cu(I) enhancing molecule elesclomol and through glial expression of wildtype swip-10. Together, these studies reveal a glial-expressed pathway that supports systemic mitochondrial function and neuronal health via regulation of Cu(I) homeostasis, a mechanism that may lend itself to therapeutic strategies to treat devastating neurodegenerative diseases.
Collapse
Affiliation(s)
- Peter Rodriguez
- Department of Biological Sciences, Charles E. Schmidt College of Science, Boca Raton, FL33412
| | - Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY10032
| | - Cristina Fenollar-Ferrer
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, Bethesda, MD20892
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
| | - Chelsea L. Gibson
- Department of Biological Sciences, Charles E. Schmidt College of Science, Boca Raton, FL33412
- Oak Ridge Institute for Science and Education, Oak Ridge, TN37830
| | - Zayna Gichi
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter, FL33458
| | - Andre Rajoo
- Department of Biology, Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL33458
| | - Carson D. Matier
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
| | - Aidan T. Pezacki
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Princeton University, Princeton, NJ08544
| | - Tong Xiao
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Princeton University, Princeton, NJ08544
| | - Lucia Carvelli
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter, FL33458
- Department of Biology, Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL33458
| | - Christopher J. Chang
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Princeton University, Princeton, NJ08544
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY10032
| | - Andy V. Khamoui
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
- Department of Exercise Science and Health Promotion, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL33431
| | - Jana Boerner
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
| | - Randy D. Blakely
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL33458
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter, FL33458
| |
Collapse
|
2
|
Rodriguez P, Blakely RD. Sink or swim: Does a worm paralysis phenotype hold clues to neurodegenerative disease? J Cell Physiol 2024; 239:e31125. [PMID: 37795580 DOI: 10.1002/jcp.31125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023]
Abstract
Receiving a neurodegenerative disease (NDD) diagnosis, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, or amyotrophic lateral sclerosis, is devastating, particularly given the limited options for treatment. Advances in genetic technologies have allowed for efficient modeling of NDDs in animals and brought hope for new disease-modifying medications. The complexity of the mammalian brain and the costs and time needed to identify and develop therapeutic leads limits progress. Modeling NDDs in invertebrates, such as the fruit fly Drosophila melanogaster and the nematode Caenorhabditis elegans, offers orders of magnitude increases in speed of genetic analysis and manipulation, and can be pursued at substantially reduced cost, providing an important, platform complement and inform research with mammalian NDD models. In this review, we describe how our efforts to exploit C. elegans for the study of neural signaling and health led to the discovery of a paralytic phenotype (swimming-induced paralysis) associated with altered dopamine signaling and, surprisingly, to the discovery of a novel gene and pathway whose dysfunction in glial cells triggers neurodegeneration. Research to date on swip-10 and its putative mammalian ortholog MBLAC1, suggests that a tandem analysis will offer insights into NDD mechanisms and insights into novel, disease-modifying therapeutics.
Collapse
Affiliation(s)
- Peter Rodriguez
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Boca Raton, Florida, USA
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Boca Raton, Florida, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| |
Collapse
|
3
|
Singhvi A, Shaham S, Rapti G. Glia Development and Function in the Nematode Caenorhabditis elegans. Cold Spring Harb Perspect Biol 2024:a041346. [PMID: 38565269 PMCID: PMC11445397 DOI: 10.1101/cshperspect.a041346] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The nematode Caenorhabditis elegans is a powerful experimental setting for uncovering fundamental tenets of nervous system organization and function. Its nearly invariant and simple anatomy, coupled with a plethora of methodologies for interrogating single-gene functions at single-cell resolution in vivo, have led to exciting discoveries in glial cell biology and mechanisms of glia-neuron interactions. Findings over the last two decades reinforce the idea that insights from C. elegans can inform our understanding of glial operating principles in other species. Here, we summarize the current state-of-the-art, and describe mechanistic insights that have emerged from a concerted effort to understand C. elegans glia. The remarkable acceleration in the pace of discovery in recent years paints a portrait of striking molecular complexity, exquisite specificity, and functional heterogeneity among glia. Glial cells affect nearly every aspect of nervous system development and function, from generating neurons, to promoting neurite formation, to animal behavior, and to whole-animal traits, including longevity. We discuss emerging questions where C. elegans is poised to fill critical knowledge gaps in our understanding of glia biology.
Collapse
Affiliation(s)
- Aakanksha Singhvi
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, Washington 98109, USA
- Department of Biological Structure, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, New York, New York 10065, USA
| | - Georgia Rapti
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory, Monterotondo, Rome 00015, Italy
- Interdisciplinary Center of Neurosciences, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
4
|
Ceyhan B, Nategh P, Neghabi M, LaMar JA, Konjalwar S, Rodriguez P, Hahn MK, Gross M, Grumbar G, Salleng KJ, Blakely RD, Ranji M. Optical Imaging Demonstrates Tissue-Specific Metabolic Perturbations in Mblac1 Knockout Mice. IEEE JOURNAL OF TRANSLATIONAL ENGINEERING IN HEALTH AND MEDICINE 2024; 12:298-305. [PMID: 38410184 PMCID: PMC10896421 DOI: 10.1109/jtehm.2024.3355962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/10/2023] [Accepted: 01/15/2024] [Indexed: 02/28/2024]
Abstract
OBJECTIVE Metabolic changes have been extensively documented in neurodegenerative brain disorders, including Parkinson's disease and Alzheimer's disease (AD). Mutations in the C. elegans swip-10 gene result in dopamine (DA) dependent motor dysfunction accompanied by DA neuron degeneration. Recently, the putative human ortholog of swip-10 (MBLAC1) was implicated as a risk factor in AD, a disorder that, like PD, has been associated with mitochondrial dysfunction. Interestingly, the AD risk associated with MBLAC1 arises in subjects with cardiovascular morbidity, suggesting a broader functional insult arising from reduced MBLAC1 protein expression and one possibly linked to metabolic alterations. METHODS Our current studies, utilizing Mblac1 knockout (KO) mice, seek to determine whether mitochondrial respiration is affected in the peripheral tissues of these mice. We quantified the levels of mitochondrial coenzymes, NADH, FAD, and their redox ratio (NADH/FAD, RR) in livers and kidneys of wild-type (WT) mice and their homozygous KO littermates of males and females, using 3D optical cryo-imaging. RESULTS Compared to WT, the RR of livers from KO mice was significantly reduced, without an apparent sex effect, driven predominantly by significantly lower NADH levels. In contrast, no genotype and sex differences were observed in kidney samples. Serum analyses of WT and KO mice revealed significantly elevated glucose levels in young and aged KO adults and diminished cholesterol levels in the aged KOs, consistent with liver dysfunction. DISCUSSION/CONCLUSION As seen with C. elegans swip-10 mutants, loss of MBLAC1 protein results in metabolic changes that are not restricted to neural cells and are consistent with the presence of peripheral comorbidities accompanying neurodegenerative disease in cases where MBLAC1 expression changes impact risk.
Collapse
Affiliation(s)
- Busenur Ceyhan
- Biophotonics LaboratoryDepartment of Electrical Engineering and Computer Science, College of Engineering and Computer ScienceFlorida Atlantic UniversityBoca RatonFL33431USA
| | - Parisa Nategh
- Biophotonics LaboratoryDepartment of Electrical Engineering and Computer Science, College of Engineering and Computer ScienceFlorida Atlantic UniversityBoca RatonFL33431USA
| | - Mehrnoosh Neghabi
- Biophotonics LaboratoryDepartment of Electrical Engineering and Computer Science, College of Engineering and Computer ScienceFlorida Atlantic UniversityBoca RatonFL33431USA
| | - Jacob A. LaMar
- Department of Biomedical ScienceCharles E. Schmidt College of MedicineFlorida Atlantic UniversityBoca RatonFL33431USA
| | - Shalaka Konjalwar
- Biophotonics LaboratoryDepartment of Electrical Engineering and Computer Science, College of Engineering and Computer ScienceFlorida Atlantic UniversityBoca RatonFL33431USA
| | - Peter Rodriguez
- Department of Biomedical ScienceCharles E. Schmidt College of MedicineFlorida Atlantic UniversityBoca RatonFL33431USA
| | - Maureen K. Hahn
- Department of Biomedical ScienceCharles E. Schmidt College of MedicineFlorida Atlantic UniversityBoca RatonFL33431USA
- Stiles-Nicholson Brain Institute, Florida Atlantic UniversityJupiterFL33458USA
| | - Matthew Gross
- Department of Biomedical ScienceCharles E. Schmidt College of MedicineFlorida Atlantic UniversityBoca RatonFL33431USA
| | - Gregory Grumbar
- Department of Biomedical ScienceCharles E. Schmidt College of MedicineFlorida Atlantic UniversityBoca RatonFL33431USA
| | - Kenneth J. Salleng
- Division of Research, Comparative MedicineFlorida Atlantic UniversityBoca RatonFL33431USA
| | - Randy D. Blakely
- Department of Biomedical ScienceCharles E. Schmidt College of MedicineFlorida Atlantic UniversityBoca RatonFL33431USA
- Stiles-Nicholson Brain Institute, Florida Atlantic UniversityJupiterFL33458USA
| | - Mahsa Ranji
- Biophotonics LaboratoryDepartment of Electrical Engineering and Computer Science, College of Engineering and Computer ScienceFlorida Atlantic UniversityBoca RatonFL33431USA
- Stiles-Nicholson Brain Institute, Florida Atlantic UniversityJupiterFL33458USA
| |
Collapse
|
5
|
Clark AS, Huayta J, Morton KS, Meyer JN, San-Miguel A. Morphological hallmarks of dopaminergic neurodegeneration are associated with altered neuron function in Caenorhabditis elegans. Neurotoxicology 2024; 100:100-106. [PMID: 38070655 PMCID: PMC10872346 DOI: 10.1016/j.neuro.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/26/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023]
Abstract
Caenorhabditis elegans (C. elegans) is an excellent model system to study neurodegenerative diseases, such as Parkinson's disease, as it enables analysis of both neuron morphology and function in live animals. Multiple structural changes in neurons, such as cephalic dendrite morphological abnormalities, have been considered hallmarks of neurodegeneration in this model, but their relevance to changes in neuron function are not entirely clear. We sought to test whether hallmark morphological changes associated with chemically induced dopaminergic neuron degeneration, such as dendrite blebbing, breakage, and loss, are indicative of neuronal malfunction and result in changes in behavior. We adapted an established dopaminergic neuronal function assay by measuring paralysis in the presence of exogenous dopamine, which revealed clear differences between cat-2 dopamine deficient mutants, wildtype worms, and dat-1 dopamine abundant mutants. Next, we integrated an automated image processing algorithm and a microfluidic device to segregate worm populations by their cephalic dendrite morphologies. We show that nematodes with dopaminergic dendrite degeneration markers, such as blebbing or breakage, paralyze at higher rates in a dopamine solution, providing evidence that dopaminergic neurodegeneration morphologies are correlated with functional neuronal outputs.
Collapse
Affiliation(s)
- Andrew S Clark
- Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC, USA
| | - Javier Huayta
- Nicholas School of the Environment, Duke University, Durham, NC, USA
| | | | - Joel N Meyer
- Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Adriana San-Miguel
- Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
6
|
Sheloukhova L, Watanabe H. Analysis of cnidarian Gcm suggests a neuronal origin of glial EAAT1 function. Sci Rep 2023; 13:14790. [PMID: 37684386 PMCID: PMC10491807 DOI: 10.1038/s41598-023-42046-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023] Open
Abstract
In bilaterian central nervous systems, coordination of neurotransmission by glial cells enables highly sophisticated neural functions. The diversity of transcription factors (TFs) involved in gliogenesis suggests multiple evolutionary origins of various glial cell types of bilaterians. Many of these TFs including the glial cells missing (Gcm) are also present in genomes of Cnidaria, the closest outgroup to Bilateria, but their function remains to be elucidated. In this study, we analyzed the function of Gcm, a multifunctional TF involved in development of glial and non-glial cell types, in the sea anemone, Nematostella vectensis. siRNA-mediated knockdown of Nematostella Gcm altered expression of cell adhesion proteins, glutamate and GABA transporters, ion channels, metabolic enzymes, and zinc finger and Ets-related TFs. NvGcm and mRNAs of downstream genes are expressed in broad neural cell clusters. However, immunostaining of a NvGcm target protein, the glutamate transporter, NvEAAT1, visualized a novel class of cells with flat cell bodies and no clear processes. Together with the finding of unique morphological features of NvEAAT1-functioning cells, these data suggest that extracellular glutamate metabolism, one of major glial functions, is deployed downstream of Gcm in specific neural cell types in Cnidaria.
Collapse
Affiliation(s)
- Larisa Sheloukhova
- Evolutionary Neurobiology Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0412, Japan
| | - Hiroshi Watanabe
- Evolutionary Neurobiology Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0412, Japan.
| |
Collapse
|
7
|
Clark AS, Huayta J, Morton KS, Meyer JN, San-Miguel A. Morphological hallmarks of dopaminergic neurodegeneration are associated with altered neuron function in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.22.554364. [PMID: 37662210 PMCID: PMC10473754 DOI: 10.1101/2023.08.22.554364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Caenorhabditis elegans (C. elegans) is an excellent model system to study neurodegenerative diseases, such as Parkinson's disease, as it enables analysis of both neuron morphology and function in live animals. Multiple structural changes in neurons, such as cephalic dendrite morphological abnormalities, have been considered hallmarks of neurodegeneration in this model, but their relevance to changes in neuron function are not entirely clear. We sought to test whether hallmark morphological changes associated with chemically induced dopaminergic neuron degeneration, such as dendrite blebbing, breakage, and loss, are indicative of neuronal malfunction and result in changes in behavior. We adapted an established dopaminergic neuronal function assay by measuring paralysis in the presence of exogenous dopamine, which revealed clear differences between cat-2 dopamine deficient mutants, wildtype worms, and dat-1 dopamine abundant mutants. Next, we integrated an automated image processing algorithm and a microfluidic device to segregate worm populations by their cephalic dendrite morphologies. We show that nematodes with dopaminergic dendrite degeneration markers, such as blebbing or breakage, paralyze at higher rates in a dopamine solution, providing evidence that dopaminergic neurodegeneration morphologies are correlated with functional neuronal outputs.
Collapse
Affiliation(s)
- Andrew S Clark
- Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Javier Huayta
- Nicholas School of the Environment, Duke University, Durham, North Carolina, USA
| | - Katherine S Morton
- Nicholas School of the Environment, Duke University, Durham, North Carolina, USA
| | - Joel N Meyer
- Nicholas School of the Environment, Duke University, Durham, North Carolina, USA
| | - Adriana San-Miguel
- Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
8
|
Ceyhan B, LaMar J, Nategh P, Neghabi M, Konjalwar S, Rodriguez P, Hahn MK, Blakely RD, Ranji M. Optical Imaging Reveals Liver Metabolic Perturbations in Mblac1 Knockout Mice. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38083729 DOI: 10.1109/embc40787.2023.10341032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Metabolic changes have been extensively documented in brain tissue undergoing neurodegeneration, including Parkinson's disease and Alzheimer's disease (AD). Mutations in the C. elegans swip-10 gene result in dopamine (DA) dependent motor dysfunction accompanied by DA neuron degeneration. Recently, the putative human ortholog of swip-10 (MBLAC1) was implicated as a risk factor in AD, that like PD, has been associated with mitochondrial dysfunction. Interestingly, the AD risk associated with MBLAC1 arises in subjects with cardiovascular morbidity, suggesting the possibility of a broader functional insult arising from reduced MBLAC1 protein expression, and one possibly linked to metabolic alterations. Our current studies, utilizing Mblac1 knockout (KO) mice, seeks to determine whether mitochondrial respiration is affected in peripheral tissues of these animals in this model. To initiate these studies, we quantified the levels of mitochondrial coenzymes, NADH, FAD, and their redox ratio (NADH/FAD, RR) in the livers of wild type (WT) mice and their homozygous KO littermates, using 3D optical cryo-imaging. We found that Mblac1 KO mice exhibited a greater oxidized redox state compared to WT mice. When compared to the WT group, the redox ratio of KO mice was decreased by 46.32%, driven predominantly by significantly lower NADH levels (more oxidized state). We speculate that, as seen with C. elegans swip-10 mutants, that loss of MBLAC1 protein results in deficits in tricarboxylic acid cycle (TCA) production of NADH and FAD TCA that leads to diminished cellular ATP production and oxidative stress. Such observations are consistent with changes that in the central nervous system (CNS) could support neurodegeneration and in the periphery account for comorbidities.
Collapse
|
9
|
Lakosa A, Rahimian A, Tomasi F, Marti F, Reynolds LM, Tochon L, David V, Danckaert A, Canonne C, Tahraoui S, de Chaumont F, Forget B, Maskos U, Besson M. Impact of the gut microbiome on nicotine's motivational effects and glial cells in the ventral tegmental area in male mice. Neuropsychopharmacology 2023; 48:963-974. [PMID: 36932179 PMCID: PMC10156728 DOI: 10.1038/s41386-023-01563-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 02/22/2023] [Accepted: 02/25/2023] [Indexed: 03/19/2023]
Abstract
A link between gut dysbiosis and the pathogenesis of brain disorders has been identified. A role for gut bacteria in drug reward and addiction has been suggested but very few studies have investigated their impact on brain and behavioral responses to addictive drugs so far. In particular, their influence on nicotine's addiction-like processes remains unknown. In addition, evidence shows that glial cells shape the neuronal activity of the mesolimbic system but their regulation, within this system, by the gut microbiome is not established. We demonstrate that a lack of gut microbiota in male mice potentiates the nicotine-induced activation of sub-regions of the mesolimbic system. We further show that gut microbiota depletion enhances the response to nicotine of dopaminergic neurons of the posterior ventral tegmental area (pVTA), and alters nicotine's rewarding and aversive effects in an intra-VTA self-administration procedure. These effects were not associated with gross behavioral alterations and the nicotine withdrawal syndrome was not impacted. We further show that depletion of the gut microbiome modulates the glial cells of the mesolimbic system. Notably, it increases the number of astrocytes selectively in the pVTA, and the expression of postsynaptic density protein 95 in both VTA sub-regions, without altering the density of the astrocytic glutamatergic transporter GLT1. Finally, we identify several sub-populations of microglia in the VTA that differ between its anterior and posterior sub-parts, and show that they are re-organized in conditions of gut microbiota depletion. The present study paves the way for refining our understanding of the pathophysiology of nicotine addiction.
Collapse
Affiliation(s)
- Alina Lakosa
- Institut Pasteur, Université Paris Cité, Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR 3571, Paris, France
| | - Anaïs Rahimian
- Institut Pasteur, Université Paris Cité, Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR 3571, Paris, France
| | - Flavio Tomasi
- Institut Pasteur, Université Paris Cité, Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR 3571, Paris, France
- Neuroscience Paris Seine, Sorbonne Université, INSERM, CNRS, 75005 Paris, France
| | - Fabio Marti
- Plasticité du Cerveau, CNRS UMR 8249, ESPCI Paris, Université PSL, Paris, France
- Neuroscience Paris Seine, Sorbonne Université, INSERM, CNRS, 75005, Paris, France
| | - Lauren M Reynolds
- Plasticité du Cerveau, CNRS UMR 8249, ESPCI Paris, Université PSL, Paris, France
| | - Léa Tochon
- Université de Bordeaux, Bordeaux, France
- CNRS UMR 5287, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Bordeaux, France
| | - Vincent David
- Université de Bordeaux, Bordeaux, France
- CNRS UMR 5287, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Bordeaux, France
| | - Anne Danckaert
- UTechS Photonics Bioimaging/C2RT, Institut Pasteur, Université Paris Cité, 25 rue du Dr Roux, 75724, Paris Cedex 15, France
| | - Candice Canonne
- Institut Pasteur, Université Paris Cité, Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR 3571, Paris, France
| | - Sylvana Tahraoui
- Institut Pasteur, Université Paris Cité, Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR 3571, Paris, France
| | - Fabrice de Chaumont
- Génétique humaine et fonctions cognitives, CNRS UMR 3571, Institut Pasteur, Université Paris Cité, 25 rue du Dr Roux, 75724, Paris Cedex 15, France
| | - Benoît Forget
- Institut Pasteur, Université Paris Cité, Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR 3571, Paris, France
- Génétique humaine et fonctions cognitives, CNRS UMR 3571, Institut Pasteur, Université Paris Cité, 25 rue du Dr Roux, 75724, Paris Cedex 15, France
| | - Uwe Maskos
- Institut Pasteur, Université Paris Cité, Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR 3571, Paris, France
| | - Morgane Besson
- Institut Pasteur, Université Paris Cité, Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR 3571, Paris, France.
| |
Collapse
|
10
|
Olsen L, Levy M, Medley JK, Hassan H, Miller B, Alexander R, Wilcock E, Yi K, Florens L, Weaver K, McKinney SA, Peuß R, Persons J, Kenzior A, Maldonado E, Delventhal K, Gluesenkamp A, Mager E, Coughlin D, Rohner N. Metabolic reprogramming underlies cavefish muscular endurance despite loss of muscle mass and contractility. Proc Natl Acad Sci U S A 2023; 120:e2204427120. [PMID: 36693105 PMCID: PMC9945943 DOI: 10.1073/pnas.2204427120] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 11/15/2022] [Indexed: 01/25/2023] Open
Abstract
Physical inactivity is a scourge to human health, promoting metabolic disease and muscle wasting. Interestingly, multiple ecological niches have relaxed investment into physical activity, providing an evolutionary perspective into the effect of adaptive physical inactivity on tissue homeostasis. One such example, the Mexican cavefish Astyanax mexicanus, has lost moderate-to-vigorous activity following cave colonization, reaching basal swim speeds ~3.7-fold slower than their river-dwelling counterpart. This change in behavior is accompanied by a marked shift in body composition, decreasing total muscle mass and increasing fat mass. This shift persisted at the single muscle fiber level via increased lipid and sugar accumulation at the expense of myofibrillar volume. Transcriptomic analysis of laboratory-reared and wild-caught cavefish indicated that this shift is driven by increased expression of pparγ-the master regulator of adipogenesis-with a simultaneous decrease in fast myosin heavy chain expression. Ex vivo and in vivo analysis confirmed that these investment strategies come with a functional trade-off, decreasing cavefish muscle fiber shortening velocity, time to maximal force, and ultimately maximal swimming speed. Despite this, cavefish displayed a striking degree of muscular endurance, reaching maximal swim speeds ~3.5-fold faster than their basal swim speeds. Multi-omic analysis suggested metabolic reprogramming, specifically phosphorylation of Pgm1-Threonine 19, as a key component enhancing cavefish glycogen metabolism and sustained muscle contraction. Collectively, we reveal broad skeletal muscle changes following cave colonization, displaying an adaptive skeletal muscle phenotype reminiscent to mammalian disuse and high-fat models while simultaneously maintaining a unique capacity for sustained muscle contraction via enhanced glycogen metabolism.
Collapse
Affiliation(s)
- Luke Olsen
- Stowers Institute for Medical Research, Kansas City, MO64110
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS66160
| | - Michaella Levy
- Stowers Institute for Medical Research, Kansas City, MO64110
| | - J. Kyle Medley
- Stowers Institute for Medical Research, Kansas City, MO64110
| | - Huzaifa Hassan
- Stowers Institute for Medical Research, Kansas City, MO64110
| | - Brandon Miller
- Stowers Institute for Medical Research, Kansas City, MO64110
| | | | - Emma Wilcock
- Department of Biology, Widener University, Chester, PA19013
| | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO64110
| | | | - Kyle Weaver
- Stowers Institute for Medical Research, Kansas City, MO64110
| | | | - Robert Peuß
- Institute for Evolution and Biodiversity, University of Münster, Münster48149, Germany
| | - Jenna Persons
- Stowers Institute for Medical Research, Kansas City, MO64110
| | | | - Ernesto Maldonado
- EvoDevo Research Group, Unidad Académica de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México, Puerto Morelos, Quintana Roo77580, Mexico
| | - Kym Delventhal
- Stowers Institute for Medical Research, Kansas City, MO64110
| | - Andrew Gluesenkamp
- Center for Conservation and Research, San Antonio Zoo, San Antonio, TX78212
| | - Edward Mager
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, Denton, TX76203
| | - David Coughlin
- Department of Biology, Widener University, Chester, PA19013
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO64110
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS66160
| |
Collapse
|
11
|
Formisano R, Rosikon KD, Singh A, Dhillon HS. The dopamine membrane transporter plays an active modulatory role in synaptic dopamine homeostasis. J Neurosci Res 2022; 100:1551-1559. [PMID: 34747520 PMCID: PMC9079189 DOI: 10.1002/jnr.24965] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/17/2021] [Accepted: 08/29/2021] [Indexed: 11/11/2022]
Abstract
Modulatory mechanisms of neurotransmitter release and clearance are highly controlled processes whose finely tuned regulation is critical for functioning of the nervous system. Dysregulation of the monoamine neurotransmitter dopamine can lead to several neuropathies. Synaptic modulation of dopamine is known to involve pre-synaptic D2 auto-receptors and acid sensing ion channels. In addition, the dopamine membrane transporter (DAT), which is responsible for clearance of dopamine from the synaptic cleft, is suspected to play an active role in modulating release of dopamine. Using functional imaging on the Caenorhabditis elegans model system, we show that DAT-1 acts as a negative feedback modulator to neurotransmitter vesicle fusion. Results from our fluorescence recovery after photo-bleaching (FRAP) based experiments were followed up with and reaffirmed using swimming-induced paralysis behavioral assays. Utilizing our numerical FRAP data we have developed a mechanistic model to dissect the dynamics of synaptic vesicle fusion, and compare the feedback effects of DAT-1 with the dopamine auto-receptor. Our experimental results and the mechanistic model are of potential broader significance, as similar dynamics are likely to be used by other synaptic modulators including membrane transporters for other neurotransmitters across species.
Collapse
Affiliation(s)
- Rosaria Formisano
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| | - Katarzyna D. Rosikon
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| | - Abhyudai Singh
- Department of Electrical and Computer Engineering, Biomedical Engineering, Data Sciences Institute, University of Delaware, Newark, DE, USA
| | - Harbinder S. Dhillon
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| |
Collapse
|
12
|
Pandey P, Kaur G, Babu K. Crosstalk between neurons and glia through G-protein coupled receptors: Insights from Caenorhabditis elegans. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 193:119-144. [PMID: 36357074 DOI: 10.1016/bs.pmbts.2022.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The past decades have witnessed a dogmatic shift from glia as supporting cells in the nervous system to their active roles in neurocentric functions. Neurons and glia communicate and show bidirectional responses through tripartite synapses. Studies across species indicate that neurotransmitters released by neurons are perceived by glial receptors, which allow for gliotransmitter release. These gliotransmitters can result in activation of neurons via neuronal GPCR receptors. However, studies of these molecular interactions are in their infancy. Caenorhabditis elegans has a conserved neuron-glia architectural repertoire with molecular and functional resemblance to mammals. Further, glia in C. elegans can be manipulated through ablation and mutations allowing for deciphering of glial dependent processes in vivo at single glial resolutions. Here, we will review recent findings from vertebrate and invertebrate organisms with a focus on how C. elegans can be used to advance our understanding of neuron-glia interactions through GPCRs.
Collapse
Affiliation(s)
- Pratima Pandey
- Indian Institute of Science Education and Research, Mohali, Punjab, India.
| | - Gazaldeep Kaur
- National Agri-Food Biotechnology Institute, Mohali, Punjab, India
| | - Kavita Babu
- Indian Institute of Science, Bangalore, Karnataka, India.
| |
Collapse
|
13
|
Serotonin and dopamine modulate aging in response to food odor and availability. Nat Commun 2022; 13:3271. [PMID: 35672307 PMCID: PMC9174215 DOI: 10.1038/s41467-022-30869-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/12/2022] [Indexed: 01/27/2023] Open
Abstract
An organism's ability to perceive and respond to changes in its environment is crucial for its health and survival. Here we reveal how the most well-studied longevity intervention, dietary restriction, acts in-part through a cell non-autonomous signaling pathway that is inhibited by the presence of attractive smells. Using an intestinal reporter for a key gene induced by dietary restriction but suppressed by attractive smells, we identify three compounds that block food odor effects in C. elegans, thereby increasing longevity as dietary restriction mimetics. These compounds clearly implicate serotonin and dopamine in limiting lifespan in response to food odor. We further identify a chemosensory neuron that likely perceives food odor, an enteric neuron that signals through the serotonin receptor 5-HT1A/SER-4, and a dopaminergic neuron that signals through the dopamine receptor DRD2/DOP-3. Aspects of this pathway are conserved in D. melanogaster. Thus, blocking food odor signaling through antagonism of serotonin or dopamine receptors is a plausible approach to mimic the benefits of dietary restriction.
Collapse
|
14
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
15
|
Bowles SN, Johnson CM. Inferences of glia-mediated control in Caenorhabditis elegans. J Neurosci Res 2021; 99:1191-1206. [PMID: 33559247 PMCID: PMC8005477 DOI: 10.1002/jnr.24803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 12/22/2022]
Abstract
Astrocytes modulate synaptic transmission; yet, it remains unclear how glia influence complex behaviors. Here, we explore the effects of Caenorhabditis elegans astrocyte-like cephalic glia (CEPglia ) and the glia-specific bHLH transcription factor HLH-17 on mating behavior and the defecation motor program (DMP). In C. elegans, male mating has been explicitly described through the male tail circuit and is characterized by coordination of multiple independent behaviors to ensure that copulation is achieved. Furthermore, the sex-specific male mating circuitry shares similar components with the DMP, which is complex and rhythmic, and requires a fixed sequence of behaviors to be activated periodically. We found that loss of CEPglia reduced persistence in executing mating behaviors and hindered copulation, while males that lacked HLH-17 demonstrated repetitive prodding behavior that increased the time spent in mating but did not hinder copulation. During the DMP, we found that posterior body wall contractions (pBocs) and enteric muscle contractions (EMCs) were differentially affected by loss of HLH-17 or CEPglia in males and hermaphrodites. pBocs and EMCs required HLH-17 activity in both sexes, whereas loss of CEPglia alone did not affect DMP in males. Our data suggest that CEPglia mediate complex behaviors by signaling to the GABAergic DVB neuron, and that HLH-17 activity influences those discrete steps within those behaviors. Collectively, these data provide evidence of glia as a link in cooperative regulation of complex and rhythmic behavior that, in C. elegans links circuitry in the head and the tail.
Collapse
Affiliation(s)
- Stephanie N. Bowles
- Department of Biology, Georgia State University, Atlanta, GA, 30303, United States
| | - Casonya M. Johnson
- Department of Biology, Georgia State University, Atlanta, GA, 30303, United States
- Department of Biology, James Madison University, Harrisonburg, VA, 22807
| |
Collapse
|
16
|
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: 4.3] [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
|
17
|
Wang M, Witvliet D, Wu M, Kang L, Shao Z. Temperature regulates synaptic subcellular specificity mediated by inhibitory glutamate signaling. PLoS Genet 2021; 17:e1009295. [PMID: 33428618 PMCID: PMC7822552 DOI: 10.1371/journal.pgen.1009295] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 01/22/2021] [Accepted: 12/05/2020] [Indexed: 01/13/2023] Open
Abstract
Environmental factors such as temperature affect neuronal activity and development. However, it remains unknown whether and how they affect synaptic subcellular specificity. Here, using the nematode Caenorhabditis elegans AIY interneurons as a model, we found that high cultivation temperature robustly induces defects in synaptic subcellular specificity through glutamatergic neurotransmission. Furthermore, we determined that the functional glutamate is mainly released by the ASH sensory neurons and sensed by two conserved inhibitory glutamate-gated chloride channels GLC-3 and GLC-4 in AIY. Our work not only presents a novel neurotransmission-dependent mechanism underlying the synaptic subcellular specificity, but also provides a potential mechanistic insight into high-temperature-induced neurological defects.
Collapse
Affiliation(s)
- Mengqing Wang
- Department of Neurosurgery, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Daniel Witvliet
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Mengting Wu
- Department of Neurosurgery, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lijun Kang
- Department of Neurobiology and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhiyong Shao
- Department of Neurosurgery, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Fung W, Wexler L, Heiman MG. Cell-type-specific promoters for C. elegans glia. J Neurogenet 2020; 34:335-346. [PMID: 32696701 PMCID: PMC7855602 DOI: 10.1080/01677063.2020.1781851] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/08/2020] [Indexed: 12/26/2022]
Abstract
Glia shape the development and function of the C. elegans nervous system, especially its sense organs and central neuropil (nerve ring). Cell-type-specific promoters allow investigators to label or manipulate individual glial cell types, and therefore provide a key tool for deciphering glial function. In this technical resource, we compare the specificity, brightness, and consistency of cell-type-specific promoters for C. elegans glia. We identify a set of promoters for the study of seven glial cell types (F16F9.3, amphid and phasmid sheath glia; F11C7.2, amphid sheath glia only; grl-2, amphid and phasmid socket glia; hlh-17, cephalic (CEP) sheath glia; and grl-18, inner labial (IL) socket glia) as well as a pan-glial promoter (mir-228). We compare these promoters to promoters that are expressed more variably in combinations of glial cell types (delm-1 and itx-1). We note that the expression of some promoters depends on external conditions or the internal state of the organism, such as developmental stage, suggesting glial plasticity. Finally, we demonstrate an approach for prospectively identifying cell-type-specific glial promoters using existing single-cell sequencing data, and we use this approach to identify two novel promoters specific to IL socket glia (col-53 and col-177).
Collapse
Affiliation(s)
- Wendy Fung
- These authors contributed equally to this work
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children’s Hospital, Boston MA 02115
| | - Leigh Wexler
- These authors contributed equally to this work
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children’s Hospital, Boston MA 02115
| | - Maxwell G. Heiman
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children’s Hospital, Boston MA 02115
| |
Collapse
|
19
|
Alvarez J, Alvarez-Illera P, García-Casas P, Fonteriz RI, Montero M. The Role of Ca 2+ Signaling in Aging and Neurodegeneration: Insights from Caenorhabditis elegans Models. Cells 2020; 9:cells9010204. [PMID: 31947609 PMCID: PMC7016793 DOI: 10.3390/cells9010204] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 02/06/2023] Open
Abstract
Ca2+ is a ubiquitous second messenger that plays an essential role in physiological processes such as muscle contraction, neuronal secretion, and cell proliferation or differentiation. There is ample evidence that the dysregulation of Ca2+ signaling is one of the key events in the development of neurodegenerative processes, an idea called the "calcium hypothesis" of neurodegeneration. Caenorhabditis elegans (C. elegans) is a very good model for the study of aging and neurodegeneration. In fact, many of the signaling pathways involved in longevity were first discovered in this nematode, and many models of neurodegenerative diseases have also been developed therein, either through mutations in the worm genome or by expressing human proteins involved in neurodegeneration (β-amyloid, α-synuclein, polyglutamine, or others) in defined worm tissues. The worm is completely transparent throughout its whole life, which makes it possible to carry out Ca2+ dynamics studies in vivo at any time, by expressing Ca2+ fluorescent probes in defined worm tissues, and even in specific organelles such as mitochondria. This review will summarize the evidence obtained using this model organism to understand the role of Ca2+ signaling in aging and neurodegeneration.
Collapse
|
20
|
Kudumala S, Sossi S, Carvelli L. Swimming Induced Paralysis to Assess Dopamine Signaling in Caenorhabditis elegans. J Vis Exp 2019. [PMID: 31009010 DOI: 10.3791/59243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The swimming assay described in this protocol is a valid tool to identify proteins regulating the dopaminergic synapses. Similar to mammals, dopamine (DA) controls several functions in C. elegans including learning and motor activity. Conditions that stimulate DA release (e.g., amphetamine (AMPH) treatments) or that prevent DA clearance (e.g., animals lacking the DA transporter (dat-1) which are incapable of reaccumulating DA into the neurons) generate an excess of extracellular DA ultimately resulting in inhibited locomotion. This behavior is particularly evident when animals swim in water. In fact, while wild-type animals continue to swim for an extended period, dat-1 null mutants and wild-type treated with AMPH or inhibitors of the DA transporter sink to the bottom of the well and do not move. This behavior is termed "Swimming Induced Paralysis" (SWIP). Although the SWIP assay is well established, a detailed description of the method is lacking. Here, we describe a step-by-step guide to perform SWIP. To perform the assay, late larval stage-4 animals are placed in a glass spot plate containing control sucrose solution with or without AMPH. Animals are scored for their swimming behavior either manually by visualization under a stereoscope or automatically by recording with a camera mounted on the stereoscope. Videos are then analyzed using a tracking software, which yields a visual representation of thrashing frequency and paralysis in the form of heat maps. Both the manual and automated systems guarantee an easily quantifiable readout of the animals' swimming ability and thus facilitate screening for animals bearing mutations within the dopaminergic system or for auxiliary genes. In addition, SWIP can be used to elucidate the mechanism of action of drugs of abuse such as AMPH.
Collapse
Affiliation(s)
- Sirisha Kudumala
- Harriet Wilkes Honors College, Florida Atlantic University, John D MacArthur Campus
| | - Serena Sossi
- Integrative Biology and Neuroscience program, College of Science, Florida Atlantic University
| | - Lucia Carvelli
- Harriet Wilkes Honors College, Florida Atlantic University, John D MacArthur Campus; Brain Institute, Florida Atlantic University; Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University;
| |
Collapse
|
21
|
Abstract
Glia are abundant components of animal nervous systems. Recognized 170 years ago, concerted attempts to understand these cells began only recently. From these investigations glia, once considered passive filler material in the brain, have emerged as active players in neuron development and activity. Glia are essential for nervous system function, and their disruption leads to disease. The nematode Caenorhabditis elegans possesses glial types similar to vertebrate glia, based on molecular, morphological, and functional criteria, and has become a powerful model in which to study glia and their neuronal interactions. Facile genetic and transgenic methods in this animal allow the discovery of genes required for glial functions, and effects of glia at single synapses can be monitored by tracking neuron shape, physiology, or animal behavior. Here, we review recent progress in understanding glia-neuron interactions in C. elegans. We highlight similarities with glia in other animals, and suggest conserved emerging principles of glial function.
Collapse
Affiliation(s)
- Aakanksha Singhvi
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA;
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, New York, NY 10065, USA;
| |
Collapse
|
22
|
Maeder CI, Kim JI, Liang X, Kaganovsky K, Shen A, Li Q, Li Z, Wang S, Xu XZS, Li JB, Xiang YK, Ding JB, Shen K. The THO Complex Coordinates Transcripts for Synapse Development and Dopamine Neuron Survival. Cell 2018; 174:1436-1449.e20. [PMID: 30146163 DOI: 10.1016/j.cell.2018.07.046] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 05/11/2018] [Accepted: 07/26/2018] [Indexed: 01/07/2023]
Abstract
Synaptic vesicle and active zone proteins are required for synaptogenesis. The molecular mechanisms for coordinated synthesis of these proteins are not understood. Using forward genetic screens, we identified the conserved THO nuclear export complex (THOC) as an important regulator of presynapse development in C. elegans dopaminergic neurons. In THOC mutants, synaptic messenger RNAs are retained in the nucleus, resulting in dramatic decrease of synaptic protein expression, near complete loss of synapses, and compromised dopamine function. CRE binding protein (CREB) interacts with THOC to mark synaptic transcripts for efficient nuclear export. Deletion of Thoc5, a THOC subunit, in mouse dopaminergic neurons causes severe defects in synapse maintenance and subsequent neuronal death in the substantia nigra compacta. These cellular defects lead to abrogated dopamine release, ataxia, and animal death. Together, our results argue that nuclear export mechanisms can select specific mRNAs and be a rate-limiting step for neuronal differentiation and survival.
Collapse
Affiliation(s)
- Celine I Maeder
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Jae-Ick Kim
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xing Liang
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Konstantin Kaganovsky
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ao Shen
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Qin Li
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Zhaoyu Li
- Life Sciences Institute and Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sui Wang
- Department of Opthalmology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - X Z Shawn Xu
- Life Sciences Institute and Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jin Billy Li
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Yang Kevin Xiang
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Jun B Ding
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Kang Shen
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
23
|
Gibson CL, Codreanu SG, Schrimpe-Rutledge AC, Retzlaff CL, Wright J, Mortlock DP, Sherrod SD, McLean JA, Blakely RD. Global untargeted serum metabolomic analyses nominate metabolic pathways responsive to loss of expression of the orphan metallo β-lactamase, MBLAC1. Mol Omics 2018; 14:142-155. [PMID: 29868674 PMCID: PMC6015503 DOI: 10.1039/c7mo00022g] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The C. elegans gene swip-10 encodes an orphan metallo β-lactamase that genetic studies indicate is vital for limiting neuronal excitability and viability. Sequence analysis indicates that the mammalian gene Mblac1 is the likely ortholog of swip-10, with greatest sequence identity localized to the encoded protein's single metallo β-lactamase domain. The substrate for the SWIP-10 protein remains unknown and to date no functional roles have been ascribed to MBLAC1, though we have shown that the protein binds the neuroprotective β-lactam antibiotic, ceftriaxone. To gain insight into the functional role of MBLAC1 in vivo, we used CRISPR/Cas9 methods to disrupt N-terminal coding sequences of the mouse Mblac1 gene, resulting in a complete loss of protein expression in viable, homozygous knockout (KO) animals. Using serum from both WT and KO mice, we performed global, untargeted metabolomic analyses, resolving small molecules via hydrophilic interaction chromatography (HILIC) based ultra-performance liquid chromatography, coupled to mass spectrometry (UPLC-MS/MS). Unsupervised principal component analysis reliably segregated the metabolomes of MBLAC1 KO and WT mice, with 92 features subsequently nominated as significantly different by ANOVA, and for which we made tentative and putative metabolite assignments. Bioinformatic analyses of these molecules nominate validated pathways subserving bile acid biosynthesis and linoleate metabolism, networks known to be responsive to metabolic and oxidative stress. Our findings lead to hypotheses that can guide future targeted studies seeking to identify the substrate for MBLAC1 and how substrate hydrolysis supports the neuroprotective actions of ceftriaxone.
Collapse
Affiliation(s)
- Chelsea L. Gibson
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter FL, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN USA
| | - Simona G. Codreanu
- Department of Chemistry, Vanderbilt University, Nashville, TN USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN USA
| | - Alexandra C. Schrimpe-Rutledge
- Department of Chemistry, Vanderbilt University, Nashville, TN USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN USA
| | - Cassandra L. Retzlaff
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter FL, USA
| | - Jane Wright
- Department of Pharmacology, Vanderbilt University, Nashville, TN USA
| | - Doug P. Mortlock
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN USA
| | - Stacy D. Sherrod
- Department of Chemistry, Vanderbilt University, Nashville, TN USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN USA
| | - John A. McLean
- Department of Chemistry, Vanderbilt University, Nashville, TN USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN USA
| | - Randy D. Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter FL, USA
- Brain Institute, Florida Atlantic University, Jupiter FL, USA
| |
Collapse
|
24
|
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: 2.5] [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
|
25
|
Gibson CL, Balbona JT, Niedzwiecki A, Rodriguez P, Nguyen KCQ, Hall DH, Blakely RD. Glial loss of the metallo β-lactamase domain containing protein, SWIP-10, induces age- and glutamate-signaling dependent, dopamine neuron degeneration. PLoS Genet 2018; 14:e1007269. [PMID: 29590100 PMCID: PMC5891035 DOI: 10.1371/journal.pgen.1007269] [Citation(s) in RCA: 20] [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: 11/14/2017] [Revised: 04/09/2018] [Accepted: 02/22/2018] [Indexed: 12/24/2022] Open
Abstract
Across phylogeny, glutamate (Glu) signaling plays a critical role in regulating neural excitability, thus supporting many complex behaviors. Perturbed synaptic and extrasynaptic Glu homeostasis in the human brain has been implicated in multiple neuropsychiatric and neurodegenerative disorders including Parkinson's disease, where theories suggest that excitotoxic insults may accelerate a naturally occurring process of dopamine (DA) neuron degeneration. In C. elegans, mutation of the glial expressed gene, swip-10, results in Glu-dependent DA neuron hyperexcitation that leads to elevated DA release, triggering DA signaling-dependent motor paralysis. Here, we demonstrate that swip-10 mutations induce premature and progressive DA neuron degeneration, with light and electron microscopy studies demonstrating the presence of dystrophic dendritic processes, as well as shrunken and/or missing cell soma. As with paralysis, DA neuron degeneration in swip-10 mutants is rescued by glial-specific, but not DA neuron-specific expression of wildtype swip-10, consistent with a cell non-autonomous mechanism. Genetic studies implicate the vesicular Glu transporter VGLU-3 and the cystine/Glu exchanger homolog AAT-1 as potential sources of Glu signaling supporting DA neuron degeneration. Degeneration can be significantly suppressed by mutations in the Ca2+ permeable Glu receptors, nmr-2 and glr-1, in genes that support intracellular Ca2+ signaling and Ca2+-dependent proteolysis, as well as genes involved in apoptotic cell death. Our studies suggest that Glu stimulation of nematode DA neurons in early larval stages, without the protective actions of SWIP-10, contributes to insults that ultimately drive DA neuron degeneration. The swip-10 model may provide an efficient platform for the identification of molecular mechanisms that enhance risk for Parkinson's disease and/or the identification of agents that can limit neurodegenerative disease progression.
Collapse
Affiliation(s)
- Chelsea L. Gibson
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States of America
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States of America
| | - Joseph T. Balbona
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States of America
| | - Ashlin Niedzwiecki
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States of America
| | - Peter Rodriguez
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States of America
| | - Ken C. Q. Nguyen
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - David H. Hall
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Randy D. Blakely
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States of America
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States of America
- Department of Psychiatry, Vanderbilt University, Nashville, TN, United States of America
- The Brain Institute, Florida Atlantic University, Jupiter, FL, United States of America
- * E-mail:
| |
Collapse
|
26
|
Retzlaff CL, Kussrow A, Schorkopf T, Saetear P, Bornhop DJ, Hardaway JA, Sturgeon SM, Wright J, Blakely RD. Metallo-β-lactamase Domain-Containing Protein 1 (MBLAC1) Is a Specific, High-Affinity Target for the Glutamate Transporter Inducer Ceftriaxone. ACS Chem Neurosci 2017; 8:2132-2138. [PMID: 28783953 DOI: 10.1021/acschemneuro.7b00232] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Ceftriaxone, a β-lactam antibiotic, has been reported to act independently of its antimicrobial actions to normalize perturbed central nervous system glutamate levels, principally by elevating expression of glial glutamate transporters. Identification of a specific, high-affinity target for ceftriaxone could significantly impact therapeutic development for multiple brain disorders, ranging from neurodegenerative disorders to addiction. Recently, we identified a glial-expressed Caenorhabditis elegans gene, swip-10, that encodes a metallo-β-lactamase domain-containing protein, and limits glutamate-dependent changes in dopamine neuron excitability. Bioinformatic analyses identified MBLAC1 as the likely mammalian orthologue of swip-10. Using cyanogen bromide immobilized ceftriaxone for affinity capture experiments and backscattering interferometry to monitor MBLAC1 binding of unmodified ceftriaxone, we obtained evidence for specific, high affinity (KD = 2.2 μM) binding of ceftriaxone to MBLAC1. We discuss our findings with respect to MBLAC1 as a potentially exclusive, high-affinity binding partner of ceftriaxone in the CNS, and the path forward in the development of novel, MBLAC1-based therapeutics.
Collapse
Affiliation(s)
- Cassandra L. Retzlaff
- Department
of Biomedical Science and Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida 33458, United States
| | | | | | | | | | | | | | | | - Randy D. Blakely
- Department
of Biomedical Science and Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, Florida 33458, United States
| |
Collapse
|
27
|
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: 2.3] [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
|
28
|
Xin W, Edwards N, Bonci A. VTA dopamine neuron plasticity - the unusual suspects. Eur J Neurosci 2016; 44:2975-2983. [PMID: 27711998 PMCID: PMC11466316 DOI: 10.1111/ejn.13425] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/26/2016] [Accepted: 10/03/2016] [Indexed: 12/25/2022]
Abstract
Dopamine neurons in the ventral tegmental area (VTA) are involved in a variety of physiological and pathological conditions, ranging from motivated behaviours to substance use disorders. While many studies have shown that these neurons can express plasticity at excitatory and inhibitory synapses, little is known about how inhibitory inputs and glial activity shape the output of DA neurons and therefore, merit greater discussion. In this review, we will attempt to fill in a bit more of the puzzle, with a focus on inhibitory transmission and astrocyte function. We summarize the findings within the VTA as well as observations made in other brain regions that have important implications for plasticity in general and should be considered in the context of DA neuron plasticity.
Collapse
Affiliation(s)
- Wendy Xin
- Synaptic Plasticity Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas Edwards
- Synaptic Plasticity Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Antonello Bonci
- Synaptic Plasticity Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
29
|
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: 1.5] [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
|
30
|
Kage-Nakadai E, Ohta A, Ujisawa T, Sun S, Nishikawa Y, Kuhara A, Mitani S. Caenorhabditis elegans homologue of Prox1/Prospero is expressed in the glia and is required for sensory behavior and cold tolerance. Genes Cells 2016; 21:936-48. [PMID: 27402188 DOI: 10.1111/gtc.12394] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/11/2016] [Indexed: 02/01/2023]
Abstract
The Caenorhabditis elegans (C. elegans) amphid sensory organ contains only 4 glia-like cells and 24 sensory neurons, providing a simple model for analyzing glia or neuron-glia interactions. To better characterize glial development and function, we carried out RNA interference screening for transcription factors that regulate the expression of an amphid sheath glial cell marker and identified pros-1, which encodes a homeodomain transcription factor homologous to Drosophila prospero/mammalian Prox1, as a positive regulator. The functional PROS-1::EGFP fusion protein was localized in the nuclei of the glia and the excretory cell but not in the amphid sensory neurons. pros-1 deletion mutants exhibited larval lethality, and rescue experiments showed that pros-1 and human Prox1 transgenes were able to rescue the larval lethal phenotype, suggesting that pros-1 is a functional homologue of mammalian Prox1, at least partially. We further found that the structure and functions of sensory neurons, such as the morphology of sensory endings, sensory behavior and sensory-mediated cold tolerance, appeared to be affected by the pros-1 RNAi. Together, our results show that the C. elegans PROS-1 is a transcriptional regulator in the glia but is involved not only in sensory behavior but also in sensory-mediated physiological tolerance.
Collapse
Affiliation(s)
- Eriko Kage-Nakadai
- Department of Physiology, Tokyo Women's Medical University School of Medicine, Tokyo, 162-8666, Japan.,The OCU Advanced Research Institute for Natural Science and Technology, Osaka City University, Osaka, 558-8585, Japan.,Graduate School of Human Life Science, Osaka City University, Osaka, 558-8585, Japan
| | - Akane Ohta
- Laboratory of Molecular and Cellular Regulation, Faculty of Science and Engineering, and Institute for Integrative Neurobiology, Konan University, Kobe, 658-8501, Japan
| | - Tomoyo Ujisawa
- Laboratory of Molecular and Cellular Regulation, Faculty of Science and Engineering, and Institute for Integrative Neurobiology, Konan University, Kobe, 658-8501, Japan
| | - Simo Sun
- Graduate School of Human Life Science, Osaka City University, Osaka, 558-8585, Japan
| | - Yoshikazu Nishikawa
- Graduate School of Human Life Science, Osaka City University, Osaka, 558-8585, Japan
| | - Atsushi Kuhara
- Laboratory of Molecular and Cellular Regulation, Faculty of Science and Engineering, and Institute for Integrative Neurobiology, Konan University, Kobe, 658-8501, Japan
| | - Shohei Mitani
- Department of Physiology, Tokyo Women's Medical University School of Medicine, Tokyo, 162-8666, Japan.
| |
Collapse
|
31
|
Yulan XU, Yadan X, Lijun K. [The effect of glial cells in the function and development of the nervous system in Caenorhabditis elegans]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2016; 45:315-22. [PMID: 27651199 PMCID: PMC10396986 DOI: 10.3785/j.issn.1008-9292.2016.05.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 12/23/2015] [Indexed: 06/06/2023]
Abstract
There are three types of glial cells in Caenorhabditis elegans (C. elegans for short): sheath glia, socket glia and glutamate receptor glia. They are mainly located in four sensory organs including the amphid, the cephalic organ, the outer labial sensilla and the inner labial sensilla. C. elegans glial cells play key roles in dendrite extension, neurite guidance and extension, and are essential for synaptogenesis and maintain the normal morphology and the function of sensory nerve endings as well. A recent study shown that some nematode neurons are derived from the glial cells. Moreover, nematodes glial cells can directly modulate the function of sensory neurons. Some glial cells can also respond to certain external stimuli, such as mechanical stimulation, and adjust the accompanying neuronal activities.The article summarizes the progress on effects of nematodes glial cells on the nervous system development and function.
Collapse
Affiliation(s)
- X U Yulan
- Institute of Neuroscience, Zhengjiang University School of Medicine, Hangzhou 310058, China
| | - Xue Yadan
- Institute of Neuroscience, Zhengjiang University School of Medicine, Hangzhou 310058, China
| | - Kang Lijun
- Institute of Neuroscience, Zhengjiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
32
|
In actio optophysiological analyses reveal functional diversification of dopaminergic neurons in the nematode C. elegans. Sci Rep 2016; 6:26297. [PMID: 27193056 PMCID: PMC4872038 DOI: 10.1038/srep26297] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/29/2016] [Indexed: 11/08/2022] Open
Abstract
Many neuronal groups such as dopamine-releasing (dopaminergic) neurons are functionally divergent, although the details of such divergence are not well understood. Dopamine in the nematode Caenorhabditis elegans modulates various neural functions and is released from four left-right pairs of neurons. The terminal identities of these dopaminergic neurons are regulated by the same genetic program, and previous studies have suggested that they are functionally redundant. In this study, however, we show functional divergence within the dopaminergic neurons of C. elegans. Because dopaminergic neurons of the animals were supposedly activated by mechanical stimulus upon entry into a lawn of their food bacteria, we developed a novel integrated microscope system that can auto-track a freely-moving (in actio) C. elegans to individually monitor and stimulate the neuronal activities of multiple neurons. We found that only head-dorsal pair of dopaminergic neurons (CEPD), but not head-ventral or posterior pairs, were preferentially activated upon food entry. In addition, the optogenetic activation of CEPD neurons alone exhibited effects similar to those observed upon food entry. Thus, our results demonstrated functional divergence in the genetically similar dopaminergic neurons, which may provide a new entry point toward understanding functional diversity of neurons beyond genetic terminal identification.
Collapse
|
33
|
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: 1.1] [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
|
34
|
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: 0.9] [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
|