1
|
Wang X, Wang L, Wu M, Zheng Y, Wang R, Shao T, Zeng S, Li A, Li R, Yue Q. Portable and intelligent ratio fluorometry and colorimetry for dual-mode detection of dopamine based on B, N-codoped carbon dots and machine learning. Talanta 2025; 294:128288. [PMID: 40344845 DOI: 10.1016/j.talanta.2025.128288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/01/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
A dual-mode approach was developed for dopamine (DA) assay based on boron (B) and nitrogen (N) co-doped carbon dots (B, N-CDs). This platform enabled highly sensitive and specific detection of DA in biological samples through collaborative ratio fluorometry and colorimetry. B, N-CDs were synthesized via a one-pot hydrothermal method using 3-aminobenzylboric acid (3-APBA) and proline (Pro) as precursors. B, N-CDs exhibited dark blue fluorescence under ultraviolet (UV) light excitation with a quantum yield (QY) of 0.05. In the presence of DA, the fluorescence of B, N-CDs displayed bright blue with the QY at 0.22. The original fluorescence peak intensity at 420 nm of B, N-CDs decreased, while a new peak at 465 nm increased significantly with the increasing concentration of DA. In this case, a ratio fluorometry for DA detection was constructed using B, N-CDs as fluorescence probes. On the other way, the color of B, N-CDs solution changed from colorless to brown with the addition of DA. A colorimetry for DA sensing was established based on the absorbance enhancement of B, N-CDs. There is excellent linear relationship within the concentration range of 2.5-500 μM for DA sensing with limit of detection (LOD) at 0.22 μM (ratio fluorometry) and 1.04 μM (colorimetry) and the relative standard deviations (RSD%) are 0.2170 and 0.02131, respectively. To enable the real-time visual and portable quantification of DA, two intelligent methods were explored by a program named RGB color analysis in a smartphone and machine learning. This dual-mode sensing strategy combined high sensitivity, wide linear range and ease of operation, offering a novel solution for rapid analysis of DA in complex biological substrates.
Collapse
Affiliation(s)
- Xiaoshuang Wang
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Lijun Wang
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Mengnan Wu
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Yihao Zheng
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Ruirui Wang
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Tong Shao
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Suyuan Zeng
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Aifeng Li
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Rui Li
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252059, China.
| | - Qiaoli Yue
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252059, China.
| |
Collapse
|
2
|
Cording KR, Tu EM, Wang H, Agopyan-Miu AHCW, Bateup HS. Cntnap2 loss drives striatal neuron hyperexcitability and behavioral inflexibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.09.593387. [PMID: 38766169 PMCID: PMC11100810 DOI: 10.1101/2024.05.09.593387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by two major diagnostic criteria - persistent deficits in social communication and interaction, and the presence of restricted, repetitive patterns of behavior (RRBs). Evidence from both human and animal model studies of ASD suggest that alteration of striatal circuits, which mediate motor learning, action selection, and habit formation, may contribute to the manifestation of RRBs. CNTNAP2 is a syndromic ASD risk gene, and loss of function of Cntnap2 in mice is associated with RRBs. How loss of Cntnap2 impacts striatal neuron function is largely unknown. In this study, we utilized Cntnap2 -/- mice to test whether altered striatal neuron activity contributes to aberrant motor behaviors relevant to ASD. We find that Cntnap2 -/- mice exhibit increased cortical drive of direct pathway striatal projection neurons (dSPNs). This enhanced drive is likely due to increased intrinsic excitability of dSPNs, which make them more responsive to cortical inputs. We find that Cntnap2 -/- mice exhibit spontaneous repetitive behaviors, increased motor routine learning, perseveration, and cognitive inflexibility. Increased corticostriatal drive of the direct pathway may therefore contribute to the acquisition of repetitive, inflexible behaviors in Cntnap2 mice.
Collapse
Affiliation(s)
- Katherine R. Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA USA
| | - Emilie M. Tu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Hongli Wang
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA USA
| | | | - Helen S. Bateup
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| |
Collapse
|
3
|
Carbonell-Roig J, Aaltonen A, Wilson K, Molinari M, Cartocci V, McGuirt A, Mosharov E, Kehr J, Lieberman OJ, Sulzer D, Borgkvist A, Santini E. Dysregulated acetylcholine-mediated dopamine neurotransmission in the eIF4E Tg mouse model of autism spectrum disorders. Cell Rep 2024; 43:114997. [PMID: 39607825 DOI: 10.1016/j.celrep.2024.114997] [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: 02/05/2024] [Revised: 09/18/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024] Open
Abstract
Autism spectrum disorder (ASD) consists of diverse neurodevelopmental conditions where core behavioral symptoms are critical for diagnosis. Altered dopamine (DA) neurotransmission in the striatum has been suggested to contribute to the behavioral features of ASD. Here, we examine DA neurotransmission in a mouse model of ASD characterized by elevated expression of eukaryotic initiation factor 4E (eIF4E), a key regulator of cap-dependent translation, using a comprehensive approach that encompasses genetics, behavior, synaptic physiology, and imaging. The results indicate that increased eIF4E expression leads to behavioral inflexibility and impaired striatal DA release. The loss of normal DA neurotransmission is due to a defect in nicotinic receptor signaling that regulates calcium dynamics in dopaminergic axons. These findings provide a mechanistic understanding of ASD symptoms and offer a foundation for targeted therapeutic interventions by revealing the intricate interplay between eIF4E, DA neurotransmission, and behavioral flexibility.
Collapse
Affiliation(s)
| | - Alina Aaltonen
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Karin Wilson
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Maya Molinari
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Veronica Cartocci
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Avery McGuirt
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Eugene Mosharov
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Jan Kehr
- Pronexus Analytical AB, 16733 Stockholm-Bromma, Sweden
| | - Ori J Lieberman
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA; Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA 94143, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Anders Borgkvist
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden.
| | - Emanuela Santini
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden.
| |
Collapse
|
4
|
Rajabi P, Noori AS, Sargolzaei J. Autism spectrum disorder and various mechanisms behind it. Pharmacol Biochem Behav 2024; 245:173887. [PMID: 39378931 DOI: 10.1016/j.pbb.2024.173887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024]
Abstract
Autism Spectrum Disorder (ASD) is a complex and heterogeneous neurodevelopmental condition characterized by a range of social, communicative, and behavioral challenges. This comprehensive review delves into key aspects of ASD. Clinical Overview and genetic features provide a foundational understanding of ASD, highlighting the clinical presentation and genetic underpinnings that contribute to its complexity. We explore the intricate neurobiological mechanisms at play in ASD, including structural and functional differences that may underlie the condition's hallmark traits. Emerging research has shed light on the role of the immune system and neuroinflammation in ASD. This section investigates the potential links between immunological factors and ASD, offering insights into the condition's pathophysiology. We examine how atypical functional connectivity and alterations in neurotransmitter systems may contribute to the unique cognitive and behavioral features of ASD. In the pursuit of effective interventions, this section reviews current therapeutic strategies, ranging from behavioral and educational interventions to pharmacological approaches, providing a glimpse into the diverse and evolving landscape of ASD treatment. This holistic exploration of mechanisms in ASD aims to contribute to our evolving understanding of the condition and to guide the development of more targeted and personalized interventions for individuals living with ASD.
Collapse
Affiliation(s)
- Parisa Rajabi
- Department of Psychiatry, Arak University of Medical Sciences, Arak, Iran
| | - Ali Sabbah Noori
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| | - Javad Sargolzaei
- Department of Biology, Faculty of Science, Arak University, Arak, Iran.
| |
Collapse
|
5
|
Rybiczka-Tešulov M, Garritsen O, Venø MT, Wieg L, Dijk RV, Rahimi K, Gomes-Duarte A, Wit MD, van de Haar LL, Michels L, van Kronenburg NCH, van der Meer C, Kjems J, Vangoor VR, Pasterkamp RJ. Circular RNAs regulate neuron size and migration of midbrain dopamine neurons during development. Nat Commun 2024; 15:6773. [PMID: 39117691 PMCID: PMC11310423 DOI: 10.1038/s41467-024-51041-1] [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: 05/04/2023] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Midbrain dopamine (mDA) neurons play an essential role in cognitive and motor behaviours and are linked to different brain disorders. However, the molecular mechanisms underlying their development, and in particular the role of non-coding RNAs (ncRNAs), remain incompletely understood. Here, we establish the transcriptomic landscape and alternative splicing patterns of circular RNAs (circRNAs) at key developmental timepoints in mouse mDA neurons in vivo using fluorescence-activated cell sorting followed by short- and long-read RNA sequencing. In situ hybridisation shows expression of several circRNAs during early mDA neuron development and post-transcriptional silencing unveils roles for different circRNAs in regulating mDA neuron morphology. Finally, in utero electroporation and time-lapse imaging implicate circRmst, a circRNA with widespread morphological effects, in the migration of developing mDA neurons in vivo. Together, these data for the first time suggest a functional role for circRNAs in developing mDA neurons and characterise poorly defined aspects of mDA neuron development.
Collapse
Affiliation(s)
- Mateja Rybiczka-Tešulov
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Oxana Garritsen
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Morten T Venø
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus C, Denmark
- Omiics ApS, Aarhus N, Denmark
| | - Laura Wieg
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Roland van Dijk
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- VectorY Therapeutics, Matrix Innovation Center VI, Amsterdam, The Netherlands
| | - Karim Rahimi
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus C, Denmark
- Department of Genetics, Blavatnik Institute, Harvard Medical School, MA, Boston, USA
| | - Andreia Gomes-Duarte
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- VectorY Therapeutics, Matrix Innovation Center VI, Amsterdam, The Netherlands
| | - Marina de Wit
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Lieke L van de Haar
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Berlin Institute for Medical Systems Biology, Max Delbrück Center, Berlin, Germany
| | - Lars Michels
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- VectorY Therapeutics, Matrix Innovation Center VI, Amsterdam, The Netherlands
| | - Nicky C H van Kronenburg
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Christiaan van der Meer
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus C, Denmark
| | - Vamshidhar R Vangoor
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
6
|
Lottes EN, Ciger F, Bhattacharjee S, Timmins EA, Tete B, Tran T, Matta J, Patel AA, Cox DN. CCT and Cullin1 Regulate the TORC1 Pathway to Promote Dendritic Arborization in Health and Disease. Cells 2024; 13:1029. [PMID: 38920658 PMCID: PMC11201622 DOI: 10.3390/cells13121029] [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: 01/27/2024] [Revised: 06/03/2024] [Accepted: 06/09/2024] [Indexed: 06/27/2024] Open
Abstract
The development of cell-type-specific dendritic arbors is integral to the proper functioning of neurons within their circuit networks. In this study, we examine the regulatory relationship between the cytosolic chaperonin CCT, key insulin pathway genes, and an E3 ubiquitin ligase (Cullin1) in dendritic development. CCT loss of function (LOF) results in dendritic hypotrophy in Drosophila Class IV (CIV) multi-dendritic larval sensory neurons, and CCT has recently been shown to fold components of the TOR (Target of Rapamycin) complex 1 (TORC1) in vitro. Through targeted genetic manipulations, we confirm that an LOF of CCT and the TORC1 pathway reduces dendritic complexity, while overexpression of key TORC1 pathway genes increases the dendritic complexity in CIV neurons. Furthermore, both CCT and TORC1 LOF significantly reduce microtubule (MT) stability. CCT has been previously implicated in regulating proteinopathic aggregation, thus, we examine CIV dendritic development in disease conditions as well. The expression of mutant Huntingtin leads to dendritic hypotrophy in a repeat-length-dependent manner, which can be rescued by Cullin1 LOF. Together, our data suggest that Cullin1 and CCT influence dendritic arborization through the regulation of TORC1 in both health and disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
7
|
Quan C, Zhou S, Zhang Y, Kulyar MFEA, Gong S, Nawaz S, Ahmed AE, Mo Q, Li J. The autophagy-mediated mechanism via TSC1/mTOR signaling pathway in thiram-induced tibial dyschondroplasia of broilers. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 928:172305. [PMID: 38593872 DOI: 10.1016/j.scitotenv.2024.172305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/11/2024]
Abstract
Thiram is a member of the dithiocarbamate family and is widely used in agriculture, especially in low-income countries. Its residues lead to various diseases, among which tibial dyschondroplasia (TD) in broiler chickens is the most common. Recent studies have also demonstrated that thiram residues may harm human health. Our previous study showed that the activity of the mTOR (mammalian target of rapamycin) signaling pathway has changed after thiram exposure. In the current study, we investigated the effect of autophagy via the mTOR signaling pathway after thiram exposure in vitro and in vivo. Our results showed that thiram inhibited the protein expression of mTOR signaling pathway-related genes such as p-4EBP1 and p-S6K1. The analysis showed a significant increase in the expression of key autophagy-related proteins, including LC3, ULK1, ATG5, and Beclin1. Further investigation proved that the effects of thiram were mediated through the downregulation of mTOR. The mTOR agonist MHY-1485 reverse the upregulation of autophagy caused by thiram in vitro. Moreover, our experiment using knockdown of TSC1 resulted in chondrocytes expressing lower levels of autophagy. In conclusion, our results demonstrate that thiram promotes autophagy via the mTOR signaling pathway in chondrogenesis, providing a potential pharmacological target for the prevention of TD.
Collapse
Affiliation(s)
- Chuxian Quan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Shimeng Zhou
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Yan Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | | | - Saisai Gong
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Shah Nawaz
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Ahmed Ezzat Ahmed
- Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Quan Mo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, P.R. China.
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, P.R. China.
| |
Collapse
|
8
|
Belghazi M, Iborra C, Toutendji O, Lasserre M, Debanne D, Goaillard JM, Marquèze-Pouey B. High-Resolution Proteomics Unravel a Native Functional Complex of Cav1.3, SK3, and Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels in Midbrain Dopaminergic Neurons. Cells 2024; 13:944. [PMID: 38891076 PMCID: PMC11172389 DOI: 10.3390/cells13110944] [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: 03/04/2024] [Revised: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Pacemaking activity in substantia nigra dopaminergic neurons is generated by the coordinated activity of a variety of distinct somatodendritic voltage- and calcium-gated ion channels. We investigated whether these functional interactions could arise from a common localization in macromolecular complexes where physical proximity would allow for efficient interaction and co-regulations. For that purpose, we immunopurified six ion channel proteins involved in substantia nigra neuron autonomous firing to identify their molecular interactions. The ion channels chosen as bait were Cav1.2, Cav1.3, HCN2, HCN4, Kv4.3, and SK3 channel proteins, and the methods chosen to determine interactions were co-immunoprecipitation analyzed through immunoblot and mass spectrometry as well as proximity ligation assay. A macromolecular complex composed of Cav1.3, HCN, and SK3 channels was unraveled. In addition, novel potential interactions between SK3 channels and sclerosis tuberous complex (Tsc) proteins, inhibitors of mTOR, and between HCN4 channels and the pro-degenerative protein Sarm1 were uncovered. In order to demonstrate the presence of these molecular interactions in situ, we used proximity ligation assay (PLA) imaging on midbrain slices containing the substantia nigra, and we could ascertain the presence of these protein complexes specifically in substantia nigra dopaminergic neurons. Based on the complementary functional role of the ion channels in the macromolecular complex identified, these results suggest that such tight interactions could partly underly the robustness of pacemaking in dopaminergic neurons.
Collapse
Affiliation(s)
- Maya Belghazi
- CRN2M Centre de Recherche Neurobiologie-Neurophysiologie, CNRS, UMR7286, Aix-Marseille Université, 13015 Marseille, France;
- Institut de Microbiologie de la Méditerranée (IMM), CNRS, Aix-Marseille Université, 13009 Marseille, France
| | - Cécile Iborra
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| | - Ophélie Toutendji
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| | - Manon Lasserre
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| | - Dominique Debanne
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| | - Jean-Marc Goaillard
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
- Institut de Neurosciences de la Timone, CNRS, Aix-Marseille Université, 13005 Marseille, France
| | - Béatrice Marquèze-Pouey
- Ion Channel and Synaptic Neurobiology, INSERM, UMR1072, Aix-Marseille Université, 13015 Marseille, France; (C.I.); (O.T.); (M.L.); (D.D.); (J.-M.G.)
| |
Collapse
|
9
|
Tan B, Browne CJ, Nöbauer T, Vaziri A, Friedman JM, Nestler EJ. Drugs of abuse hijack a mesolimbic pathway that processes homeostatic need. Science 2024; 384:eadk6742. [PMID: 38669575 PMCID: PMC11077477 DOI: 10.1126/science.adk6742] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/26/2024] [Indexed: 04/28/2024]
Abstract
Drugs of abuse are thought to promote addiction in part by "hijacking" brain reward systems, but the underlying mechanisms remain undefined. Using whole-brain FOS mapping and in vivo single-neuron calcium imaging, we found that drugs of abuse augment dopaminoceptive ensemble activity in the nucleus accumbens (NAc) and disorganize overlapping ensemble responses to natural rewards in a cell type-specific manner. Combining FOS-Seq, CRISPR-perturbation, and single-nucleus RNA sequencing, we identified Rheb as a molecular substrate that regulates cell type-specific signal transduction in NAc while enabling drugs to suppress natural reward consumption. Mapping NAc-projecting regions activated by drugs of abuse revealed input-specific effects on natural reward consumption. These findings characterize the dynamic, molecular and circuit basis of a common reward pathway, wherein drugs of abuse interfere with the fulfillment of innate needs.
Collapse
Affiliation(s)
- Bowen Tan
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University; New York, NY 10065, USA
| | - Caleb J. Browne
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health; Toronto, ON, M5T 1R8, Canada
| | - Tobias Nöbauer
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University; New York, NY 10065, USA
| | - Alipasha Vaziri
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University; New York, NY 10065, USA
- The Kavli Neural Systems Institute, The Rockefeller University; New York, NY 10065, USA
| | - Jeffrey M. Friedman
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University; New York, NY 10065, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| |
Collapse
|
10
|
Karalis V, Wood D, Teaney NA, Sahin M. The role of TSC1 and TSC2 proteins in neuronal axons. Mol Psychiatry 2024; 29:1165-1178. [PMID: 38212374 DOI: 10.1038/s41380-023-02402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/13/2024]
Abstract
Tuberous Sclerosis Complex 1 and 2 proteins, TSC1 and TSC2 respectively, participate in a multiprotein complex with a crucial role for the proper development and function of the nervous system. This complex primarily acts as an inhibitor of the mechanistic target of rapamycin (mTOR) kinase, and mutations in either TSC1 or TSC2 cause a neurodevelopmental disorder called Tuberous Sclerosis Complex (TSC). Neurological manifestations of TSC include brain lesions, epilepsy, autism, and intellectual disability. On the cellular level, the TSC/mTOR signaling axis regulates multiple anabolic and catabolic processes, but it is not clear how these processes contribute to specific neurologic phenotypes. Hence, several studies have aimed to elucidate the role of this signaling pathway in neurons. Of particular interest are axons, as axonal defects are associated with severe neurocognitive impairments. Here, we review findings regarding the role of the TSC1/2 protein complex in axons. Specifically, we will discuss how TSC1/2 canonical and non-canonical functions contribute to the formation and integrity of axonal structure and function.
Collapse
Affiliation(s)
- Vasiliki Karalis
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Delaney Wood
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
- Human Neuron Core, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Nicole A Teaney
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Mustafa Sahin
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA.
- Human Neuron Core, Boston Children's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
11
|
Zeylan M, Senyuz S, Picón-Pagès P, García-Elías A, Tajes M, Muñoz FJ, Oliva B, Garcia-Ojalvo J, Barbu E, Vicente R, Nattel S, Ois A, Puig-Pijoan A, Keskin O, Gursoy A. Shared Proteins and Pathways of Cardiovascular and Cognitive Diseases: Relation to Vascular Cognitive Impairment. J Proteome Res 2024; 23:560-573. [PMID: 38252700 PMCID: PMC10846560 DOI: 10.1021/acs.jproteome.3c00289] [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: 05/12/2023] [Revised: 09/29/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024]
Abstract
One of the primary goals of systems medicine is the detection of putative proteins and pathways involved in disease progression and pathological phenotypes. Vascular cognitive impairment (VCI) is a heterogeneous condition manifesting as cognitive impairment resulting from vascular factors. The precise mechanisms underlying this relationship remain unclear, which poses challenges for experimental research. Here, we applied computational approaches like systems biology to unveil and select relevant proteins and pathways related to VCI by studying the crosstalk between cardiovascular and cognitive diseases. In addition, we specifically included signals related to oxidative stress, a common etiologic factor tightly linked to aging, a major determinant of VCI. Our results show that pathways associated with oxidative stress are quite relevant, as most of the prioritized vascular cognitive genes and proteins were enriched in these pathways. Our analysis provided a short list of proteins that could be contributing to VCI: DOLK, TSC1, ATP1A1, MAPK14, YWHAZ, CREB3, HSPB1, PRDX6, and LMNA. Moreover, our experimental results suggest a high implication of glycative stress, generating oxidative processes and post-translational protein modifications through advanced glycation end-products (AGEs). We propose that these products interact with their specific receptors (RAGE) and Notch signaling to contribute to the etiology of VCI.
Collapse
Affiliation(s)
- Melisa
E. Zeylan
- Computational
Sciences and Engineering, Graduate School of Science and Engineering, Koç University, Istanbul 34450, Türkiye
| | - Simge Senyuz
- Computational
Sciences and Engineering, Graduate School of Science and Engineering, Koç University, Istanbul 34450, Türkiye
| | - Pol Picón-Pagès
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Anna García-Elías
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Marta Tajes
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Francisco J. Muñoz
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Baldomero Oliva
- Laboratory
of Structural Bioinformatics (GRIB), Department of Medicine and Life
Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Jordi Garcia-Ojalvo
- Laboratory
of Dynamical Systems Biology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Eduard Barbu
- Institute
of Computer Science, University of Tartu, Tartu, 50090, Estonia
| | - Raul Vicente
- Institute
of Computer Science, University of Tartu, Tartu, 50090, Estonia
| | - Stanley Nattel
- Department
of Medicine and Research Center, Montreal Heart Institute and Université
de Montréal; Institute of Pharmacology, West German Heart and
Vascular Center, University Duisburg-Essen,
Germany; IHU LIRYC and Fondation Bordeaux Université, Bordeaux 33000, France
| | - Angel Ois
- Department
of Neurology, Hospital Del Mar. Hospital
Del Mar - Medical Research Institute and Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Albert Puig-Pijoan
- Department
of Neurology, Hospital Del Mar. Hospital
Del Mar - Medical Research Institute and Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Ozlem Keskin
- Department
of Chemical and Biological Engineering, Koç University, Istanbul 34450, Türkiye
| | - Attila Gursoy
- Department
of Computer Engineering, Koç University, Istanbul 34450, Türkiye
| |
Collapse
|
12
|
Carbonell-Roig J, Aaltonen A, Cartocci V, McGuirt A, Mosharov E, Kehr J, Lieberman OJ, Sulzer D, Borgkvist A, Santini E. Dysregulated acetylcholine-mediated dopamine neurotransmission in the eIF4E Tg mouse model of autism spectrum disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.29.577831. [PMID: 38352367 PMCID: PMC10862723 DOI: 10.1101/2024.01.29.577831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2024]
Abstract
Autism Spectrum Disorders (ASD) consist of diverse neurodevelopmental conditions where core behavioral symptoms are critical for diagnosis. Altered dopamine neurotransmission in the striatum has been suggested to contribute to the behavioral features of ASD. Here, we examine dopamine neurotransmission in a mouse model of ASD characterized by elevated expression of the eukaryotic initiation factor 4E (eIF4E), a key regulator of cap-dependent translation, using a comprehensive approach that encompasses genetics, behavior, synaptic physiology, and imaging. The results indicate that increased eIF4E expression leads to behavioral inflexibility and impaired striatal dopamine release. The loss of normal dopamine neurotransmission is due to a defective nicotinic receptor signaling that regulates calcium dynamics in dopaminergic axons. These findings reveal an intricate interplay between eIF4E, DA neurotransmission, and behavioral flexibility, provide a mechanistic understanding of ASD symptoms and offer a foundation for targeted therapeutic interventions.
Collapse
|
13
|
Kao HY, Yao Y, Yang T, Ziobro J, Zylinski M, Mir MY, Hu S, Cao R, Borna NN, Banerjee R, Parent JM, Wang S, Leventhal DK, Li P, Wang Y. Sudden Unexpected Death in Epilepsy and Respiratory Defects in a Mouse Model of DEPDC5-Related Epilepsy. Ann Neurol 2023; 94:812-824. [PMID: 37606181 PMCID: PMC10592102 DOI: 10.1002/ana.26773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023]
Abstract
OBJECTIVES DEPDC5 is a common causative gene in familial focal epilepsy with or without malformations of cortical development. Its pathogenic variants also confer a significantly higher risk for sudden unexpected death in epilepsy (SUDEP), providing opportunities to investigate the pathophysiology intersecting neurodevelopment, epilepsy, and cardiorespiratory function. There is an urgent need to gain a mechanistic understanding of DEPDC5-related epilepsy and SUDEP, identify biomarkers for patients at high risk, and develop preventive interventions. METHODS Depdc5 was specifically deleted in excitatory or inhibitory neurons in the mouse brain to determine neuronal subtypes that drive epileptogenesis and SUDEP. Electroencephalogram (EEG), cardiac, and respiratory recordings were performed to determine cardiorespiratory phenotypes associated with SUDEP. Baseline respiratory function and the response to hypoxia challenge were also studied in these mice. RESULTS Depdc5 deletion in excitatory neurons in cortical layer 5 and dentate gyrus caused frequent generalized tonic-clonic seizures and SUDEP in young adult mice, but Depdc5 deletion in cortical interneurons did not. EEG suppression immediately following ictal offset was observed in fatal and non-fatal seizures, but low amplitude rhythmic theta frequency activity was lost only in fatal seizures. In addition, these mice developed baseline respiratory dysfunction prior to SUDEP, during which ictal apnea occurred long before terminal cardiac asystole. INTERPRETATION Depdc5 deletion in excitatory neurons is sufficient to cause DEPDC5-related epilepsy and SUDEP. Ictal apnea and respiratory dysregulation play critical roles in SUDEP. Our study also provides a novel mouse model to investigate the underlying mechanisms of DEPDC5-related epilepsy and SUDEP. ANN NEUROL 2023;94:812-824.
Collapse
Affiliation(s)
- Hsin-Yi Kao
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Yilong Yao
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Tao Yang
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Julie Ziobro
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Mary Zylinski
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Mohd Yaqub Mir
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Shuntong Hu
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Runnan Cao
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Rajat Banerjee
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Jack M. Parent
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Michgian Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Shuo Wang
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Daniel K. Leventhal
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Parkinson Disease Foundation Research Center of Excellence, University of Michigan, Ann Arbor, MI, USA
| | - Peng Li
- Michgian Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biologic and Material Sciences, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Yu Wang
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Michgian Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
14
|
Lottes EN, Ciger FH, Bhattacharjee S, Timmins-Wilde EA, Tete B, Tran T, Matta J, Patel AA, Cox DN. CCT and Cullin1 regulate the TORC1 pathway to promote dendritic arborization in health and disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.31.551324. [PMID: 37577581 PMCID: PMC10418059 DOI: 10.1101/2023.07.31.551324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The development of cell-type-specific dendritic arbors is integral to the proper functioning of neurons within their circuit networks. In this study, we examine the regulatory relationship between the cytosolic chaperonin CCT, key insulin pathway genes, and an E3 ubiquitin ligase (Cullin1) in homeostatic dendritic development. CCT loss of function (LOF) results in dendritic hypotrophy in Drosophila Class IV (CIV) multidendritic larval sensory neurons, and CCT has recently been shown to fold components of the TOR (Target of Rapamycin) complex 1 (TORC1), in vitro. Through targeted genetic manipulations, we have confirmed that LOF of CCT and the TORC1 pathway reduces dendritic complexity, while overexpression of key TORC1 pathway genes increases dendritic complexity in CIV neurons. Both CCT and TORC1 LOF significantly reduce microtubule (MT) stability. CCT has been previously implicated in regulating proteinopathic aggregation, thus we examined CIV dendritic development in disease conditions as well. Expression of mutant Huntingtin leads to dendritic hypotrophy in a repeat-length-dependent manner, which can be rescued by TORC1 disinhibition via Cullin1 LOF. Together, our data suggest that Cullin1 and CCT influence dendritic arborization through regulation of TORC1 in both health and disease.
Collapse
Affiliation(s)
- Erin N. Lottes
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | - Feyza H. Ciger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | | | | | - Benoit Tete
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | - Tommy Tran
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | - Jais Matta
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | - Atit A. Patel
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| |
Collapse
|
15
|
Santiago NA, He B, Howard SL, Beaudin S, Strupp BJ, Smith DR. Developmental Manganese Exposure Causes Lasting Attention Deficits Accompanied by Dysregulation of mTOR Signaling and Catecholaminergic Gene Expression in Brain Prefrontal Cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.16.549215. [PMID: 37503220 PMCID: PMC10370122 DOI: 10.1101/2023.07.16.549215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Elevated manganese (Mn) exposure is associated with attentional deficits in children, and is an environmental risk factor for attention deficit hyperactivity disorder (ADHD). We have shown that developmental Mn exposure causes lasting attention and sensorimotor deficits in a rat model of early childhood Mn exposure, and that these deficits are associated with a hypofunctioning catecholaminergic system in the prefrontal cortex (PFC), though the mechanistic basis for these deficits is not well understood. To address this, male Long-Evans rats were exposed orally to Mn (50 mg/kg/d) over PND 1-21 and attentional function was assessed in adulthood using the 5-Choice Serial Reaction Time Task. Targeted catecholaminergic system and epigenetic gene expression, followed by unbiased differential DNA methylation and gene regulation expression transcriptomics in the PFC, were performed in young adult littermates. Results show that developmental Mn exposure causes lasting focused attention deficits that are associated with reduced gene expression of tyrosine hydroxylase, dopamine transporter, and DNA methyltransferase 3a. Further, developmental Mn exposure causes broader lasting methylation and gene expression dysregulation associated with epigenetic regulation, inflammation, cell development, and hypofunctioning catecholaminergic neuronal systems. Pathway enrichment analyses uncovered mTOR and Wnt signaling pathway genes as significant transcriptomic regulators of the Mn altered transcriptome, and Western blot of total, C1 and C2 phospho-mTOR confirmed mTOR pathway dysregulation. Our findings deepen our understanding of the mechanistic basis of how developmental Mn exposure leads to lasting catecholaminergic dysfunction and attention deficits, which may aid future therapeutic interventions of environmental exposure associated disorders. Significance Statement Attention deficit hyperactivity disorder (ADHD) is associated with environmental risk factors, including exposure to neurotoxic agents. Here we used a rodent model of developmental manganese (Mn) exposure producing lasting attention deficits to show broad epigenetic and gene expression changes in the prefrontal cortex, and to identify disrupted mTOR and Wnt signaling pathways as a novel mechanism for how developmental Mn exposure may induce lasting attention and catecholaminergic system impairments. Importantly, our findings establish early development as a critical period of susceptibility to lasting deficits in attentional function caused by elevated environmental toxicant exposure. Given that environmental health threats disproportionately impact communities of color and low socioeconomic status, our findings can aid future studies to assess therapeutic interventions for vulnerable populations.
Collapse
|
16
|
Abu Bakar N, Wan Ibrahim WN, Zulkiflli AR, Saleh Hodin NA, Kim TY, Ling YS, Md Ajat MM, Shaari K, Shohaimi S, Nasruddin NS, Mohd Faudzi SM, Kim CH. Embryonic mercury exposure in zebrafish: Alteration of metabolites and gene expression, related to visual and behavioral impairments. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 256:114862. [PMID: 37004432 DOI: 10.1016/j.ecoenv.2023.114862] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/05/2023] [Accepted: 03/30/2023] [Indexed: 06/19/2023]
Abstract
The widespread presence of mercury, a heavy metal found in the environment and used in numerous industries and domestic, raises concerns about its potential impact on human health. Nevertheless, the adverse effects of this environmental toxicant at low concentrations are often underestimated. There are emerging studies showing that accumulation of mercury in the eye may contribute to visual impairment and a comorbidity between autism spectrum disorders (ASD) trait and visual impairment. However, the underlying mechanism of visual impairment in humans and rodents is challenging. In response to this issue, zebrafish larvae with a cone-dominated retinal visual system were exposed to 100 nM mercury chloride (HgCl2), according to our previous study, followed by light-dark stimulation, a social assay, and color preference to examine the functionality of the visual system in relation to ASD-like behavior. Exposure of embryos to HgCl2 from gastrulation to hatching increased locomotor activity in the dark, reduced shoaling and exploratory behavior, and impaired color preference. Defects in microridges as the first barrier may serve as primary tools for HgCl2 toxicity affecting vision. Depletion of polyunsaturated fatty acids (PUFAs), linoleic acid, arachidonic acid (ARA), alpha-linoleic acid, docosahexaenoic acid (DHA), stearic acid, L-phenylalanine, isoleucine, L-lysine, and N-acetylputrescine, along with the increase of gamma-aminobutyric acid (GABA), sphingosine-1-phosphate, and citrulline assayed by liquid chromatography-mass spectrometry (LC-MS) suggest that these metabolites serve as biomarkers of retinal impairments that affect vision and behavior. Although suppression of adsl, shank3a, tsc1b, and nrxn1a gene expression was observed, among these tsc1b showed more positive correlation with ASD. Collectively, these results contribute new insights into the possible mechanism of mercury toxicity give rise to visual, cognitive, and social deficits in zebrafish.
Collapse
Affiliation(s)
- Noraini Abu Bakar
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Wan Norhamidah Wan Ibrahim
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Natural Medicines and Product Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Abdul Rahman Zulkiflli
- Natural Medicines and Product Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Nur Atikah Saleh Hodin
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Tae-Yoon Kim
- Department of Biology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Yee Soon Ling
- Biotechnology Research Institute, Universiti Malaysia Sabah, Jalan UMS, 88400 Kota Kinabalu, Sabah, Malaysia
| | - Mohd Mokrish Md Ajat
- Natural Medicines and Product Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Malaysia
| | - Khozirah Shaari
- Natural Medicines and Product Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Shamarina Shohaimi
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Nurrul Shaqinah Nasruddin
- Centre for Craniofacial Diagnostics, Faculty of Dentistry, Universiti Kebangsaan Malaysia (UKM), 50300 Kuala Lumpur, Malaysia
| | - Siti Munirah Mohd Faudzi
- Natural Medicines and Product Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
17
|
Vidyadhara DJ, Somayaji M, Wade N, Yücel B, Zhao H, Shashaank N, Ribaudo J, Gupta J, Lam TT, Sames D, Greene LE, Sulzer DL, Chandra SS. Dopamine transporter and synaptic vesicle sorting defects underlie auxilin-associated Parkinson's disease. Cell Rep 2023; 42:112231. [PMID: 36920906 PMCID: PMC10127800 DOI: 10.1016/j.celrep.2023.112231] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 12/22/2022] [Accepted: 02/22/2023] [Indexed: 03/16/2023] Open
Abstract
Auxilin participates in the uncoating of clathrin-coated vesicles (CCVs), thereby facilitating synaptic vesicle (SV) regeneration at presynaptic sites. Auxilin (DNAJC6/PARK19) loss-of-function mutations cause early-onset Parkinson's disease (PD). Here, we utilized auxilin knockout (KO) mice to elucidate the mechanisms through which auxilin deficiency and clathrin-uncoating deficits lead to PD. Auxilin KO mice display cardinal features of PD, including progressive motor deficits, α-synuclein pathology, nigral dopaminergic loss, and neuroinflammation. Significantly, treatment with L-DOPA ameliorated motor deficits. Unbiased proteomic and neurochemical analyses of auxilin KO brains indicated dopamine dyshomeostasis. We validated these findings by demonstrating slower dopamine reuptake kinetics in vivo, an effect associated with dopamine transporter misrouting into axonal membrane deformities in the dorsal striatum. Defective SV protein sorting and elevated synaptic autophagy also contribute to ineffective dopamine sequestration and compartmentalization, ultimately leading to neurodegeneration. This study provides insights into how presynaptic endocytosis deficits lead to dopaminergic vulnerability and pathogenesis of PD.
Collapse
Affiliation(s)
- D J Vidyadhara
- Department of Neurology, Yale University, New Haven, CT, USA; Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Mahalakshmi Somayaji
- Department of Psychiatry, Columbia University, New York, NY, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Nigel Wade
- Department of Neurology, Yale University, New Haven, CT, USA; Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Betül Yücel
- Department of Neurology, Yale University, New Haven, CT, USA; Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Helen Zhao
- Department of Neurology, Yale University, New Haven, CT, USA
| | - N Shashaank
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA; Department of Computer Science, Columbia University, New York, NY, USA; New York Genome Center, New York, NY, USA
| | - Joseph Ribaudo
- Department of Neurology, Yale University, New Haven, CT, USA
| | - Jyoti Gupta
- Department of Neuroscience, Yale University, New Haven, CT, USA
| | - TuKiet T Lam
- Keck MS and Proteomics Resource, Departments of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Dalibor Sames
- Department of Chemistry and NeuroTechnology Center, Columbia University, New York, NY, USA
| | - Lois E Greene
- Laboratory of Cell Biology, NHLBI, National Institutes of Health, Bethesda, MD, USA
| | - David L Sulzer
- Department of Psychiatry, Columbia University, New York, NY, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA; Departments of Neurology and Pharmacology, Columbia University, New York, NY, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Sreeganga S Chandra
- Department of Neurology, Yale University, New Haven, CT, USA; Department of Neuroscience, Yale University, New Haven, CT, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University, New Haven, CT, USA.
| |
Collapse
|
18
|
Jiang G, Long Z, Wang Y, Wang Y, Xue P, Chen M, Yang K, Li W. Inhibition of mammalian target of rapamycin complex 1 in the brain microvascular endothelium ameliorates diabetic Aβ brain deposition and cognitive impairment via the sterol-regulatory element-binding protein 1/lipoprotein receptor-associated protein 1 signaling pathway. CNS Neurosci Ther 2023. [PMID: 36890627 DOI: 10.1111/cns.14133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/02/2022] [Accepted: 02/15/2023] [Indexed: 03/10/2023] Open
Abstract
AIMS Mammalian target of rapamycin complex 1 (mTORC1) is highly activated in diabetes, and the decrease of low-density lipoprotein receptor-associated protein 1 (LRP1) in brain microvascular endothelial cells (BMECs) is a key factor leading to amyloid-β (Aβ) deposition in the brain and diabetic cognitive impairment, but the relationship between them is still unknown. METHODS In vitro, BMECs were cultured with high glucose, and the activation of mTORC1 and sterol-regulatory element-binding protein 1 (SREBP1) was observed. mTORC1 was inhibited by rapamycin and small interfering RNA (siRNA) in BMECs. Betulin and siRNA inhibited SREBP1, observed the mechanism of mTORC1-mediated effects on Aβ efflux in BMECs through LRP1 under high-glucose conditions. Constructed cerebrovascular endothelial cell-specific Raptor-knockout (Raptorfl/+ ) mice to investigate the role of mTORC1 in regulating LRP1-mediated Aβ efflux and diabetic cognitive impairment at the tissue level. RESULTS mTORC1 activation was observed in HBMECs cultured in high glucose, and this change was confirmed in diabetic mice. Inhibiting mTORC1 corrected the reduction in Aβ efflux under high-glucose stimulation. In addition, high glucose activated the expression of SREBP1, and inhibiting of mTORC1 reduced the activation and expression of SREBP1. After inhibiting the activity of SREBP1, the presentation of LRP1 was improved, and the decrease of Aβ efflux mediated by high glucose was corrected. Raptorfl/+ diabetic mice had significantly inhibited activation of mTORC1 and SREBP1, increased LRP1 expression, increased Aβ efflux, and improved cognitive impairment. CONCLUSION Inhibiting mTORC1 in the brain microvascular endothelium ameliorates diabetic Aβ brain deposition and cognitive impairment via the SREBP1/LRP1 signaling pathway, suggesting that mTORC1 may be a potential target for the treatment of diabetic cognitive impairment.
Collapse
Affiliation(s)
- Gege Jiang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenzhen Long
- Xiang Yang No. 1 People Hospital, Affiliated Hospital of Hubei University Medicine, XiangYang, China
| | - Yaoling Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaofeng Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Xue
- Department of Geriatrics, Li-Yuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minfang Chen
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kang Yang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Li
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Anastasaki C, Gao Y, Gutmann DH. Neurons as stromal drivers of nervous system cancer formation and progression. Dev Cell 2023; 58:81-93. [PMID: 36693322 PMCID: PMC9883043 DOI: 10.1016/j.devcel.2022.12.011] [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: 05/16/2022] [Revised: 06/24/2022] [Accepted: 12/27/2022] [Indexed: 01/24/2023]
Abstract
Similar to their pivotal roles in nervous system development, neurons have emerged as critical regulators of cancer initiation, maintenance, and progression. Focusing on nervous system tumors, we describe the normal relationships between neurons and other cell types relevant to normal nerve function, and discuss how disruptions of these interactions promote tumor evolution, focusing on electrical (gap junctions) and chemical (synaptic) coupling, as well as the establishment of new paracrine relationships. We also review how neuron-tumor communication contributes to some of the complications of cancer, including neuropathy, chemobrain, seizures, and pain. Finally, we consider the implications of cancer neuroscience in establishing risk for tumor penetrance and in the design of future anti-tumoral treatments.
Collapse
Affiliation(s)
- Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yunqing Gao
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
20
|
Abstract
Recent advances in genomics have revealed a wide spectrum of genetic variants associated with neurodevelopmental disorders at an unprecedented scale. An increasing number of studies have consistently identified mutations-both inherited and de novo-impacting the function of specific brain circuits. This suggests that, during brain development, alterations in distinct neural circuits, cell types, or broad regulatory pathways ultimately shaping synapses might be a dysfunctional process underlying these disorders. Here, we review findings from human studies and animal model research to provide a comprehensive description of synaptic and circuit mechanisms implicated in neurodevelopmental disorders. We discuss how specific synaptic connections might be commonly disrupted in different disorders and the alterations in cognition and behaviors emerging from imbalances in neuronal circuits. Moreover, we review new approaches that have been shown to restore or mitigate dysfunctional processes during specific critical windows of brain development. Considering the heterogeneity of neurodevelopmental disorders, we also highlight the recent progress in developing improved clinical biomarkers and strategies that will help to identify novel therapeutic compounds and opportunities for early intervention.
Collapse
Affiliation(s)
- David Exposito-Alonso
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom;
- Current affiliation: Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA;
| | - Beatriz Rico
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom;
| |
Collapse
|
21
|
Lin WC, Liu C, Kosillo P, Tai LH, Galarce E, Bateup HS, Lammel S, Wilbrecht L. Transient food insecurity during the juvenile-adolescent period affects adult weight, cognitive flexibility, and dopamine neurobiology. Curr Biol 2022; 32:3690-3703.e5. [PMID: 35863352 PMCID: PMC10519557 DOI: 10.1016/j.cub.2022.06.089] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 04/01/2022] [Accepted: 06/29/2022] [Indexed: 10/17/2022]
Abstract
A major challenge for neuroscience, public health, and evolutionary biology is to understand the effects of scarcity and uncertainty on the developing brain. Currently, a significant fraction of children and adolescents worldwide experience insecure access to food. The goal of our work was to test in mice whether the transient experience of insecure versus secure access to food during the juvenile-adolescent period produced lasting differences in learning, decision-making, and the dopamine system in adulthood. We manipulated feeding schedules in mice from postnatal day (P)21 to P40 as food insecure or ad libitum and found that when tested in adulthood (after P60), males with different developmental feeding history showed significant differences in multiple metrics of cognitive flexibility in learning and decision-making. Adult females with different developmental feeding history showed no differences in cognitive flexibility but did show significant differences in adult weight. We next applied reinforcement learning models to these behavioral data. The best fit models suggested that in males, developmental feeding history altered how mice updated their behavior after negative outcomes. This effect was sensitive to task context and reward contingencies. Consistent with these results, in males, we found that the two feeding history groups showed significant differences in the AMPAR/NMDAR ratio of excitatory synapses on nucleus-accumbens-projecting midbrain dopamine neurons and evoked dopamine release in dorsal striatal targets. Together, these data show in a rodent model that transient differences in feeding history in the juvenile-adolescent period can have significant impacts on adult weight, learning, decision-making, and dopamine neurobiology.
Collapse
Affiliation(s)
- Wan Chen Lin
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Christine Liu
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Polina Kosillo
- Department of Molecular and Cellular Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Lung-Hao Tai
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Ezequiel Galarce
- Robert Wood Johnson Foundation Health and Society Scholar, University of California Berkeley, Berkeley, CA 94720, USA
| | - Helen S Bateup
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cellular Biology, University of California Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Stephan Lammel
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cellular Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Linda Wilbrecht
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; Department of Psychology, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
22
|
Abu Bakar N, Wan Ibrahim WN, Che Abdullah CA, Ramlan NF, Shaari K, Shohaimi S, Mediani A, Nasruddin NS, Kim CH, Mohd Faudzi SM. Embryonic Arsenic Exposure Triggers Long-Term Behavioral Impairment with Metabolite Alterations in Zebrafish. TOXICS 2022; 10:493. [PMID: 36136458 PMCID: PMC9502072 DOI: 10.3390/toxics10090493] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 05/10/2023]
Abstract
Arsenic trioxide (As2O3) is a ubiquitous heavy metal in the environment. Exposure to this toxin at low concentrations is unremarkable in developing organisms. Nevertheless, understanding the underlying mechanism of its long-term adverse effects remains a challenge. In this study, embryos were initially exposed to As2O3 from gastrulation to hatching under semi-static conditions. Results showed dose-dependent increased mortality, with exposure to 30-40 µM As2O3 significantly reducing tail-coiling and heart rate at early larval stages. Surviving larvae after 30 µM As2O3 exposure showed deficits in motor behavior without impairment of anxiety-like responses at 6 dpf and a slight impairment in color preference behavior at 11 dpf, which was later evident in adulthood. As2O3 also altered locomotor function, with a loss of directional and color preference in adult zebrafish, which correlated with changes in transcriptional regulation of adsl, shank3a, and tsc1b genes. During these processes, As2O3 mainly induced metabolic changes in lipids, particularly arachidonic acid, docosahexaenoic acid, prostaglandin, and sphinganine-1-phosphate in the post-hatching period of zebrafish. Overall, this study provides new insight into the potential mechanism of arsenic toxicity leading to long-term learning impairment in zebrafish and may benefit future risk assessments of other environmental toxins of concern.
Collapse
Affiliation(s)
- Noraini Abu Bakar
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Wan Norhamidah Wan Ibrahim
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Che Azurahanim Che Abdullah
- Department of Physics, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
- The Institute of Advanced Technology (ITMA), Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Nurul Farhana Ramlan
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Khozirah Shaari
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Shamarina Shohaimi
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Ahmed Mediani
- Institute of Systems Biology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Malaysia
| | - Nurrul Shaqinah Nasruddin
- Centre for Craniofacial Diagnostics, Faculty of Dentistry, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur 50300, Malaysia
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon 34134, Korea
| | - Siti Munirah Mohd Faudzi
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
| |
Collapse
|
23
|
Raptor downregulation rescues neuronal phenotypes in mouse models of Tuberous Sclerosis Complex. Nat Commun 2022; 13:4665. [PMID: 35945201 PMCID: PMC9363483 DOI: 10.1038/s41467-022-31961-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 07/08/2022] [Indexed: 12/16/2022] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a neurodevelopmental disorder caused by mutations in the TSC1 or TSC2 genes, which encode proteins that negatively regulate mTOR complex 1 (mTORC1) signaling. Current treatment strategies focus on mTOR inhibition with rapamycin and its derivatives. While effective at improving some aspects of TSC, chronic rapamycin inhibits both mTORC1 and mTORC2 and is associated with systemic side-effects. It is currently unknown which mTOR complex is most relevant for TSC-related brain phenotypes. Here we used genetic strategies to selectively reduce neuronal mTORC1 or mTORC2 activity in mouse models of TSC. We find that reduction of the mTORC1 component Raptor, but not the mTORC2 component Rictor, rebalanced mTOR signaling in Tsc1 knock-out neurons. Raptor reduction was sufficient to improve several TSC-related phenotypes including neuronal hypertrophy, macrocephaly, impaired myelination, network hyperactivity, and premature mortality. Raptor downregulation represents a promising potential therapeutic intervention for the neurological manifestations of TSC.
Collapse
|
24
|
Kosillo P, Ahmed KM, Aisenberg EE, Karalis V, Roberts BM, Cragg SJ, Bateup HS. Dopamine neuron morphology and output are differentially controlled by mTORC1 and mTORC2. eLife 2022; 11:e75398. [PMID: 35881440 PMCID: PMC9328766 DOI: 10.7554/elife.75398] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 07/08/2022] [Indexed: 02/07/2023] Open
Abstract
The mTOR pathway is an essential regulator of cell growth and metabolism. Midbrain dopamine neurons are particularly sensitive to mTOR signaling status as activation or inhibition of mTOR alters their morphology and physiology. mTOR exists in two distinct multiprotein complexes termed mTORC1 and mTORC2. How each of these complexes affect dopamine neuron properties, and whether they have similar or distinct functions is unknown. Here, we investigated this in mice with dopamine neuron-specific deletion of Rptor or Rictor, which encode obligatory components of mTORC1 or mTORC2, respectively. We find that inhibition of mTORC1 strongly and broadly impacts dopamine neuron structure and function causing somatodendritic and axonal hypotrophy, increased intrinsic excitability, decreased dopamine production, and impaired dopamine release. In contrast, inhibition of mTORC2 has more subtle effects, with selective alterations to the output of ventral tegmental area dopamine neurons. Disruption of both mTOR complexes leads to pronounced deficits in dopamine release demonstrating the importance of balanced mTORC1 and mTORC2 signaling for dopaminergic function.
Collapse
Affiliation(s)
- Polina Kosillo
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Kamran M Ahmed
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Erin E Aisenberg
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
| | - Vasiliki Karalis
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Bradley M Roberts
- Department of Physiology, Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
| | - Stephanie J Cragg
- Department of Physiology, Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
- Chan Zuckerberg Biohub, San FranciscoSan FranciscoUnited States
| |
Collapse
|
25
|
D’Aiello B, Battisti A, Lazzaro G, Pani P, De Rossi P, Di Vara S, Pretelli I, Costanzo F, Vicari S, Menghini D. Comparing the Effect of Methylphenidate and Anodal tDCS on Inhibitory Control and Working-Memory in Children and Adolescents with Attention Deficit/Hyperactivity Disorder: A Study Protocol for a Randomized, within-Subject Trial. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19084575. [PMID: 35457447 PMCID: PMC9030177 DOI: 10.3390/ijerph19084575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder characterized by inappropriate levels of attention, hyperactivity, and impulsivity that interfere with individual functioning. The international guidelines recommend targeting ADHD-related neurochemical brain abnormalities by intervening via drug treatment, such as methylphenidate (MPH), as first choice. Drug treatments are usually associated with a huge amount of cost for families and the healthcare system, suspension for low compliance, poor long-term efficacy, and side effects. Transcranial direct current stimulation (tDCS) has been suggested as a possible noninvasive means to safely manipulate brain activity and, in turn, improve behavior and cognition in developmental ages. Several studies have shown that tDCS has the potential to improve ADHD-related cognitive deficits, but the effect of tDCS compared with MPH has never been evaluated. The aim of the present within-subject, sham-controlled, randomized proof-of-concept study is to demonstrate the positive effect of one-session anodal tDCS analogous to the MPH drug on inhibitory control and working memory in children and adolescents with ADHD. We strongly believe that this study protocol will serve to accelerate research into low-cost, drug-free, feasible interventions for ADHD.
Collapse
Affiliation(s)
- Barbara D’Aiello
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (B.D.); (A.B.); (G.L.); (P.D.R.); (S.D.V.); (I.P.); (F.C.); (S.V.)
- Department of Human Science, LUMSA University, 00193 Rome, Italy
| | - Andrea Battisti
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (B.D.); (A.B.); (G.L.); (P.D.R.); (S.D.V.); (I.P.); (F.C.); (S.V.)
- Department of Human Science, LUMSA University, 00193 Rome, Italy
| | - Giulia Lazzaro
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (B.D.); (A.B.); (G.L.); (P.D.R.); (S.D.V.); (I.P.); (F.C.); (S.V.)
| | - Pierpaolo Pani
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy;
| | - Pietro De Rossi
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (B.D.); (A.B.); (G.L.); (P.D.R.); (S.D.V.); (I.P.); (F.C.); (S.V.)
| | - Silvia Di Vara
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (B.D.); (A.B.); (G.L.); (P.D.R.); (S.D.V.); (I.P.); (F.C.); (S.V.)
| | - Italo Pretelli
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (B.D.); (A.B.); (G.L.); (P.D.R.); (S.D.V.); (I.P.); (F.C.); (S.V.)
| | - Floriana Costanzo
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (B.D.); (A.B.); (G.L.); (P.D.R.); (S.D.V.); (I.P.); (F.C.); (S.V.)
| | - Stefano Vicari
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (B.D.); (A.B.); (G.L.); (P.D.R.); (S.D.V.); (I.P.); (F.C.); (S.V.)
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Centro di Riabilitazione, Casa San Giuseppe, Opera Don Guanella, 00165 Rome, Italy
| | - Deny Menghini
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (B.D.); (A.B.); (G.L.); (P.D.R.); (S.D.V.); (I.P.); (F.C.); (S.V.)
- Correspondence:
| |
Collapse
|
26
|
Terashima H, Minatohara K, Maruoka H, Okabe S. Imaging neural circuit pathology of autism spectrum disorders: autism-associated genes, animal models and the application of in vivo two-photon imaging. Microscopy (Oxf) 2022; 71:i81-i99. [DOI: 10.1093/jmicro/dfab039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/11/2021] [Accepted: 11/08/2021] [Indexed: 11/12/2022] Open
Abstract
Abstract
Recent advances in human genetics identified genetic variants involved in causing autism spectrum disorders (ASDs). Mouse models that mimic mutations found in patients with ASD exhibit behavioral phenotypes consistent with ASD symptoms. These mouse models suggest critical biological factors of ASD etiology. Another important implication of ASD genetics is the enrichment of ASD risk genes in molecules involved in developing synapses and regulating neural circuit function. Sophisticated in vivo imaging technologies applied to ASD mouse models identify common synaptic impairments in the neocortex, with genetic-mutation-specific defects in local neural circuits. In this article, we review synapse- and circuit-level phenotypes identified by in vivo two-photon imaging in multiple mouse models of ASD and discuss the contributions of altered synapse properties and neural circuit activity to ASD pathogenesis.
Collapse
Affiliation(s)
- Hiroshi Terashima
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Keiichiro Minatohara
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hisato Maruoka
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shigeo Okabe
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
27
|
Vanclooster S, Bissell S, van Eeghen AM, Chambers N, De Waele L, Byars AW, Capal JK, Cukier S, Davis P, Flinn J, Gardner-Lubbe S, Gipson T, Heunis TM, Hook D, Kingswood JC, Krueger DA, Kumm AJ, Sahin M, Schoeters E, Smith C, Srivastava S, Takei M, Waltereit R, Jansen AC, de Vries PJ. The research landscape of tuberous sclerosis complex-associated neuropsychiatric disorders (TAND)-a comprehensive scoping review. J Neurodev Disord 2022; 14:13. [PMID: 35151277 PMCID: PMC8853020 DOI: 10.1186/s11689-022-09423-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Tuberous sclerosis complex (TSC)-associated neuropsychiatric disorders (TAND) is an umbrella term for the behavioural, psychiatric, intellectual, academic, neuropsychological and psychosocial manifestations of TSC. Although TAND affects 90% of individuals with TSC during their lifetime, these manifestations are relatively under-assessed, under-treated and under-researched. We performed a comprehensive scoping review of all TAND research to date (a) to describe the existing TAND research landscape and (b) to identify knowledge gaps to guide future TAND research. METHODS The study was conducted in accordance with stages outlined within the Arksey and O'Malley scoping review framework. Ten research questions relating to study characteristics, research design and research content of TAND levels and clusters were examined. RESULTS Of the 2841 returned searches, 230 articles published between 1987 and 2020 were included (animal studies = 30, case studies = 47, cohort studies = 153), with more than half published since the term TAND was coined in 2012 (118/230; 51%). Cohort studies largely involved children and/or adolescents (63%) as opposed to older adults (16%). Studies were represented across 341 individual research sites from 45 countries, the majority from the USA (89/341; 26%) and the UK (50/341; 15%). Only 48 research sites (14%) were within low-middle income countries (LMICs). Animal studies and case studies were of relatively high/high quality, but cohort studies showed significant variability. Of the 153 cohort studies, only 16 (10%) included interventions. None of these were non-pharmacological, and only 13 employed remote methodologies (e.g. telephone interviews, online surveys). Of all TAND clusters, the autism spectrum disorder-like cluster was the most widely researched (138/230; 60%) and the scholastic cluster the least (53/200; 27%). CONCLUSIONS Despite the recent increase in TAND research, studies that represent participants across the lifespan, LMIC research sites and non-pharmacological interventions were identified as future priorities. The quality of cohort studies requires improvement, to which the use of standardised direct behavioural assessments may contribute. In human studies, the academic level in particular warrants further investigation. Remote technologies could help to address many of the TAND knowledge gaps identified.
Collapse
Affiliation(s)
- Stephanie Vanclooster
- Department of Public Health, Mental Health and Wellbeing Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Stacey Bissell
- Cerebra Network for Neurodevelopmental Disorders, University of Birmingham, Birmingham, UK
| | - Agnies M. van Eeghen
- Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam, The Netherlands
- TAND Expert Centre, ‘s Heeren Loo, Hoofddorp, The Netherlands
| | - Nola Chambers
- Division of Child & Adolescent Psychiatry, Centre for Autism Research in Africa (CARA), University of Cape Town, Cape Town, South Africa
| | - Liesbeth De Waele
- Department of Paediatric Neurology, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Anna W. Byars
- Department of Pediatrics, Division of Neurology, Cincinnati Children’s Hospital Medical Center/University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Jamie K. Capal
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Sebastián Cukier
- Argentine Program for Children, Adolescents and Adults with Autism Spectrum Disorders (PANAACEA), Buenos Aires, Argentina
| | - Peter Davis
- Department of Neurology, Harvard Medical School & Boston Children’s Hospital, Boston, MA USA
| | | | | | - Tanjala Gipson
- Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, TN USA
- Le Bonheur Children’s Hospital and Boling Center for Developmental Disabilities, Memphis, TN USA
| | - Tosca-Marie Heunis
- Department of Public Health, Mental Health and Wellbeing Research Group, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | - Darcy A. Krueger
- TSC Clinic Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, Clinical Pediatrics and Neurology, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Aubrey J. Kumm
- Division of Child & Adolescent Psychiatry, Centre for Autism Research in Africa (CARA), University of Cape Town, Cape Town, South Africa
| | - Mustafa Sahin
- Department of Neurology, Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| | | | | | - Shoba Srivastava
- Division of Child & Adolescent Psychiatry, Centre for Autism Research in Africa (CARA), University of Cape Town, Cape Town, South Africa
- Tuberous Sclerosis Alliance India, Mumbai, India
| | - Megumi Takei
- Japanese Society of Tuberous Sclerosis Complex, Tokyo, Japan
| | - Robert Waltereit
- Child and Adolescent Psychiatry, University Medical Center Göttingen, Georg-August University, Göttingen, Germany
| | - Anna C. Jansen
- Department of Public Health, Mental Health and Wellbeing Research Group, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Pediatrics, Pediatric Neurology Unit, Antwerp University Hospital, Edegem, Belgium
| | - Petrus J. de Vries
- Division of Child & Adolescent Psychiatry, Centre for Autism Research in Africa (CARA), University of Cape Town, Cape Town, South Africa
| |
Collapse
|
28
|
Dichter GS, Rodriguez-Romaguera J. Anhedonia and Hyperhedonia in Autism and Related Neurodevelopmental Disorders. Curr Top Behav Neurosci 2022; 58:237-254. [PMID: 35397066 DOI: 10.1007/7854_2022_312] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although autism spectrum disorder (ASD) is defined by impaired social communication and restricted and repetitive behaviors and interests, ASD is also characterized by impaired motivational processes. The "social motivation theory of autism" describes how social motivation disruptions in ASD in early childhood may impede the drive to engage in reciprocal social behaviors and ultimately interfere with the development of neural networks critical for social communication (Chevallier et al., Trends Cogn Sci 16:231-239, 2012b). Importantly, clinical studies and preclinical research using model organisms for ASD indicate that motivational impairments in ASD are not constrained to social rewards but are evident in response to a range of nonsocial rewards as well. Additionally, translational studies on certain genetically defined neurodevelopmental disorders associated with ASD indicate that these syndromic forms of ASD are also characterized by motivational deficits and mesolimbic dopamine impairments. In this chapter we summarize clinical and preclinical research relevant to reward processing impairments in ASD and related neurodevelopmental disorders. We also propose a nosology to describe reward processing impairments in these disorders that uses a three-axes model. In this triaxial nosology, the first axis defines the direction of the reward response (i.e., anhedonic, hyperhedonic); the second axis defines the construct of the reward process (e.g., reward liking, reward wanting); and the third axis defines the context of the reward response (e.g., social, nonsocial). A more precise nosology for describing reward processing impairments in ASD and related neurodevelopmental disorders will aid in the translation of preclinical research to clinical investigations which will ultimately help to speed up the development of interventions that target motivational systems for ASD and related neurodevelopmental disorders.
Collapse
Affiliation(s)
- Gabriel S Dichter
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Department of Psychiatry, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Jose Rodriguez-Romaguera
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Department of Psychiatry, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
29
|
EWAS of Monozygotic Twins Implicate a Role of mTOR Pathway in Pathogenesis of Tic Spectrum Disorder. Genes (Basel) 2021; 12:genes12101510. [PMID: 34680906 PMCID: PMC8535383 DOI: 10.3390/genes12101510] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 11/17/2022] Open
Abstract
Tic spectrum disorder (TSD) is an umbrella term which includes Gilles de la Tourette syndrome (GTS) and chronic tic disorder (CTD). They are considered highly heritable, yet the genetic components remain largely unknown. In this study we aimed to investigate disease-associated DNA methylation differences to identify genes and pathways which may be implicated in TSD aetiology. For this purpose, we performed an exploratory analysis of the genome-wide DNA methylation patterns in whole blood samples of 16 monozygotic twin pairs, of which eight were discordant and six concordant for TSD, while two pairs were asymptomatic. Although no sites reached genome-wide significance, we identified several sites and regions with a suggestive significance, which were located within or in the vicinity of genes with biological functions associated with neuropsychiatric disorders. The two top genes identified (TSC1 and CRYZ/TYW3) and the enriched pathways and components (phosphoinosides and PTEN pathways, and insulin receptor substrate binding) are related to, or have been associated with, the PI3K/AKT/mTOR pathway. Genes in this pathway have previously been associated with GTS, and mTOR signalling has been implicated in a range of neuropsychiatric disorders. It is thus possible that altered mTOR signalling plays a role in the complex pathogenesis of TSD.
Collapse
|
30
|
Benthall KN, Cording KR, Agopyan-Miu AHCW, Wong CD, Chen EY, Bateup HS. Loss of Tsc1 from striatal direct pathway neurons impairs endocannabinoid-LTD and enhances motor routine learning. Cell Rep 2021; 36:109511. [PMID: 34380034 PMCID: PMC8404511 DOI: 10.1016/j.celrep.2021.109511] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 05/28/2021] [Accepted: 07/21/2021] [Indexed: 02/08/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a neurodevelopmental disorder that often presents with psychiatric conditions, including autism spectrum disorder (ASD). ASD is characterized by restricted, repetitive, and inflexible behaviors, which may result from abnormal activity in striatal circuits that mediate motor learning and action selection. To test whether altered striatal activity contributes to aberrant motor behaviors in the context of TSC, we conditionally deleted Tsc1 from direct or indirect pathway striatal projection neurons (dSPNs or iSPNs, respectively). We find that dSPN-specific loss of Tsc1 impairs endocannabinoid-mediated long-term depression (eCB-LTD) at cortico-dSPN synapses and strongly enhances corticostriatal synaptic drive, which is not observed in iSPNs. dSPN-Tsc1 KO, but not iSPN-Tsc1 KO, mice show enhanced motor learning, a phenotype observed in several mouse models of ASD. These findings demonstrate that dSPNs are particularly sensitive to Tsc1 loss and suggest that enhanced corticostriatal activation may contribute to altered motor behaviors in TSC.
Collapse
Affiliation(s)
- Katelyn N Benthall
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Katherine R Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Corinna D Wong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emily Y Chen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
31
|
Kosillo P, Bateup HS. Dopaminergic Dysregulation in Syndromic Autism Spectrum Disorders: Insights From Genetic Mouse Models. Front Neural Circuits 2021; 15:700968. [PMID: 34366796 PMCID: PMC8343025 DOI: 10.3389/fncir.2021.700968] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder defined by altered social interaction and communication, and repetitive, restricted, inflexible behaviors. Approximately 1.5-2% of the general population meet the diagnostic criteria for ASD and several brain regions including the cortex, amygdala, cerebellum and basal ganglia have been implicated in ASD pathophysiology. The midbrain dopamine system is an important modulator of cellular and synaptic function in multiple ASD-implicated brain regions via anatomically and functionally distinct dopaminergic projections. The dopamine hypothesis of ASD postulates that dysregulation of dopaminergic projection pathways could contribute to the behavioral manifestations of ASD, including altered reward value of social stimuli, changes in sensorimotor processing, and motor stereotypies. In this review, we examine the support for the idea that cell-autonomous changes in dopaminergic function are a core component of ASD pathophysiology. We discuss the human literature supporting the involvement of altered dopamine signaling in ASD including genetic, brain imaging and pharmacologic studies. We then focus on genetic mouse models of syndromic neurodevelopmental disorders in which single gene mutations lead to increased risk for ASD. We highlight studies that have directly examined dopamine neuron number, morphology, physiology, or output in these models. Overall, we find considerable support for the idea that the dopamine system may be dysregulated in syndromic ASDs; however, there does not appear to be a consistent signature and some models show increased dopaminergic function, while others have deficient dopamine signaling. We conclude that dopamine dysregulation is common in syndromic forms of ASD but that the specific changes may be unique to each genetic disorder and may not account for the full spectrum of ASD-related manifestations.
Collapse
Affiliation(s)
- Polina Kosillo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Helen S. Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| |
Collapse
|
32
|
Lin R, Learman LN, Na CH, Renuse S, Chen KT, Chen PY, Lee GH, Xiao B, Resnick SM, Troncoso JC, Szumlinski KK, Linden DJ, Park JM, Savonenko A, Pandey A, Worley PF. Persistently Elevated mTOR Complex 1-S6 Kinase 1 Disrupts DARPP-32-Dependent D 1 Dopamine Receptor Signaling and Behaviors. Biol Psychiatry 2021; 89:1058-1072. [PMID: 33353667 PMCID: PMC8076344 DOI: 10.1016/j.biopsych.2020.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/17/2020] [Accepted: 10/22/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND The serine-threonine kinase mTORC1 (mechanistic target of rapamycin complex 1) is essential for normal cell function but is aberrantly activated in the brain in both genetic-developmental and sporadic diseases and is associated with a spectrum of neuropsychiatric symptoms. The underlying molecular mechanisms of cognitive and neuropsychiatric symptoms remain controversial. METHODS The present study examines behaviors in transgenic models that express Rheb, the most proximal known activator of mTORC1, and profiles striatal phosphoproteomics in a model with persistently elevated mTORC1 signaling. Biochemistry, immunohistochemistry, electrophysiology, and behavior approaches are used to examine the impact of persistently elevated mTORC1 on D1 dopamine receptor (D1R) signaling. The effect of persistently elevated mTORC1 was confirmed using D1-Cre to elevate mTORC1 activity in D1R neurons. RESULTS We report that persistently elevated mTORC1 signaling blocks canonical D1R signaling that is dependent on DARPP-32 (dopamine- and cAMP-regulated neuronal phosphoprotein). The immediate downstream effector of mTORC1, ribosomal S6 kinase 1 (S6K1), phosphorylates and activates DARPP-32. Persistent elevation of mTORC1-S6K1 occludes dynamic D1R signaling downstream of DARPP-32 and blocks multiple D1R responses, including dynamic gene expression, D1R-dependent corticostriatal plasticity, and D1R behavioral responses including sociability. Candidate biomarkers of mTORC1-DARPP-32 occlusion are increased in the brain of human disease subjects in association with elevated mTORC1-S6K1, supporting a role for this mechanism in cognitive disease. CONCLUSIONS The mTORC1-S6K1 intersection with D1R signaling provides a molecular framework to understand the effects of pathological mTORC1 activation on behavioral symptoms in neuropsychiatric disease.
Collapse
Affiliation(s)
- Raozhou Lin
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Lisa N. Learman
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chan-Hyun Na
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Santosh Renuse
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First ST SW, Rochester, MN 55905, USA.,Center for Individualized Medicine, Mayo Clinic, 200 First ST SW, Rochester, MN 55905, USA
| | - Kevin T. Chen
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Po Yu Chen
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Gum-Hwa Lee
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Bo Xiao
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 21224, USA
| | - Juan C. Troncoso
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences and the Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
| | - David J. Linden
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Joo-Min Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Alena Savonenko
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First ST SW, Rochester, MN 55905, USA.,Center for Individualized Medicine, Mayo Clinic, 200 First ST SW, Rochester, MN 55905, USA
| | - Paul F. Worley
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Corresponding author. Phone: 410-502-5489
| |
Collapse
|
33
|
Karalis V, Bateup HS. Current Approaches and Future Directions for the Treatment of mTORopathies. Dev Neurosci 2021; 43:143-158. [PMID: 33910214 PMCID: PMC8440338 DOI: 10.1159/000515672] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/13/2021] [Indexed: 11/19/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) is a kinase at the center of an evolutionarily conserved signaling pathway that orchestrates cell growth and metabolism. mTOR responds to an array of intra- and extracellular stimuli and in turn controls multiple cellular anabolic and catabolic processes. Aberrant mTOR activity is associated with numerous diseases, with particularly profound impact on the nervous system. mTOR is found in two protein complexes, mTOR complex 1 (mTORC1) and 2 (mTORC2), which are governed by different upstream regulators and have distinct cellular actions. Mutations in genes encoding for mTOR regulators result in a collection of neurodevelopmental disorders known as mTORopathies. While these disorders can affect multiple organs, neuropsychiatric conditions such as epilepsy, intellectual disability, and autism spectrum disorder have a major impact on quality of life. The neuropsychiatric aspects of mTORopathies have been particularly challenging to treat in a clinical setting. Current therapeutic approaches center on rapamycin and its analogs, drugs that are administered systemically to inhibit mTOR activity. While these drugs show some clinical efficacy, adverse side effects, incomplete suppression of mTOR targets, and lack of specificity for mTORC1 or mTORC2 may limit their utility. An increased understanding of the neurobiology of mTOR and the underlying molecular, cellular, and circuit mechanisms of mTOR-related disorders will facilitate the development of improved therapeutics. Animal models of mTORopathies have helped unravel the consequences of mTOR pathway mutations in specific brain cell types and developmental stages, revealing an array of disease-related phenotypes. In this review, we discuss current progress and potential future directions for the therapeutic treatment of mTORopathies with a focus on findings from genetic mouse models.
Collapse
Affiliation(s)
- Vasiliki Karalis
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| |
Collapse
|
34
|
Martinez-Pinteño A, Rodriguez N, Blázquez A, Plana MT, Varela E, Gassó P, Lafuente A, Lazaro L, Mas S. DNA Methylation of Fluoxetine Response in Child and Adolescence: Preliminary Results. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2021; 14:459-467. [PMID: 33907441 PMCID: PMC8064712 DOI: 10.2147/pgpm.s289480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/29/2020] [Indexed: 12/28/2022]
Abstract
Purpose The search for predictors of antidepressant response is gaining increasing attention, with epigenetic markers attracting a great deal of interest. We performed a genome-wide study assessing baseline differences in DNA methylation between Responders and Non-Responders. Patients and Methods Twenty-two children and adolescents, receiving fluoxetine treatment for the first time, were classified as Responders or Non-Responders according to CGI-I score after 8 weeks of fluoxetine treatment. Genome-wide DNA methylation was profiled using the Illumina Infinium MethylationEPIC BeadChip Kit and analyzed using the Chip Analysis Methylation Pipeline (ChAMP). Results We identified 21 CpG sites significantly (FDR<0.05) associated with fluoxetine response that showed meaningful differences (Δβ> ±0.2) in methylation level between Responders and Non-Responders. Two genes, RHOJ (Ras Homolog Family Member J) and OR2L13 (Olfactory Receptor family 2 subfamily L member 13), presented more than one significant CpG sites. Conclusion Our findings provide new insights into the molecular mechanisms underlying the complex phenotype of antidepressant response, indicating that methylation at specific genes could be a promising biomarker that needs further replication in large cohorts.
Collapse
Affiliation(s)
- Albert Martinez-Pinteño
- Department of Basic Clinal Practice, Pharmacology Unit, University of Barcelona, Barcelona, Spain
| | - Natalia Rodriguez
- Department of Basic Clinal Practice, Pharmacology Unit, University of Barcelona, Barcelona, Spain
| | - Ana Blázquez
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Maria Teresa Plana
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Eva Varela
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Patricia Gassó
- Department of Basic Clinal Practice, Pharmacology Unit, University of Barcelona, Barcelona, Spain.,Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Amalia Lafuente
- Department of Basic Clinal Practice, Pharmacology Unit, University of Barcelona, Barcelona, Spain.,Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
| | - Luisa Lazaro
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic de Barcelona, Barcelona, Spain.,Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain.,Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Sergi Mas
- Department of Basic Clinal Practice, Pharmacology Unit, University of Barcelona, Barcelona, Spain.,Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
35
|
Wang X, Chang X, Zhan H, Li C, Zhang Q, Li S, Sun Y. Curcumin combined with Baicalin attenuated ethanol-induced hepatitis by suppressing p38 MAPK and TSC1/ eIF-2α/ATF4 pathways in rats. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2020.100851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
López‐Gambero AJ, Rodríguez de Fonseca F, Suárez J. Energy sensors in drug addiction: A potential therapeutic target. Addict Biol 2021; 26:e12936. [PMID: 32638485 DOI: 10.1111/adb.12936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 01/05/2023]
Abstract
Addiction is defined as the repeated exposure and compulsive seek of psychotropic drugs that, despite the harmful effects, generate relapse after the abstinence period. The psychophysiological processes associated with drug addiction (acquisition/expression, withdrawal, and relapse) imply important alterations in neurotransmission and changes in presynaptic and postsynaptic plasticity and cellular structure (neuroadaptations) in neurons of the reward circuits (dopaminergic neuronal activity) and other corticolimbic regions. These neuroadaptation mechanisms imply important changes in neuronal energy balance and protein synthesis machinery. Scientific literature links drug-induced stimulation of dopaminergic and glutamatergic pathways along with presence of neurotrophic factors with alterations in synaptic plasticity and membrane excitability driven by metabolic sensors. Here, we provide current knowledge of the role of molecular targets that constitute true metabolic/energy sensors such as AMPK, mTOR, ERK, or KATP in the development of the different phases of addiction standing out the main brain regions (ventral tegmental area, nucleus accumbens, hippocampus, and amygdala) constituting the hubs in the development of addiction. Because the available treatments show very limited effectiveness, evaluating the drug efficacy of AMPK and mTOR specific modulators opens up the possibility of testing novel pharmacotherapies for an individualized approach in drug abuse.
Collapse
Affiliation(s)
- Antonio Jesús López‐Gambero
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental Hospital Regional Universitario de Málaga Málaga Spain
| | - Juan Suárez
- Instituto de Investigación Biomédica de Málaga (IBIMA), UGC Salud Mental Hospital Regional Universitario de Málaga Málaga Spain
| |
Collapse
|
37
|
Altered baseline and amphetamine-mediated behavioral profiles in dopamine transporter Cre (DAT-Ires-Cre) mice compared to tyrosine hydroxylase Cre (TH-Cre) mice. Psychopharmacology (Berl) 2020; 237:3553-3568. [PMID: 32778904 PMCID: PMC10120402 DOI: 10.1007/s00213-020-05635-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023]
Abstract
RATIONALE Transgenic mouse lines expressing Cre-recombinase under the regulation of either dopamine transporter (DAT) or tyrosine hydroxylase (TH) promoters are commonly used to study the dopamine (DA) system. While use of the TH promoter appears to have less liability to changes in native gene expression, transgene insertion in the DAT locus results in reduced DAT expression and function. This confound is sometimes overlooked in genetically targeted behavioral experiments. OBJECTIVES We sought to evaluate the suitability of DAT-Ires-Cre and TH-Cre transgenic lines for behavioral pharmacology experiments with DA agonists. We hypothesized that DAT-Ires-Cre expression would impact DAT-mediated behaviors, but no impact of TH-Cre expression would be observed. METHODS DAT-Ires-Cre and TH-Cre mice bred on mixed 129S6/C57BL/6 and pure C57BL/6 backgrounds were evaluated for novelty-induced, baseline, and amphetamine (AMPH)-induced locomotion, and for AMPH and D1 agonist (SKF-38393)-induced preservative behaviors. RESULTS DAT-Ires-Cre mice on both mixed 129S6/C57BL/6 and pure C57BL/6 backgrounds displayed increased novelty-induced activity and decreased AMPH-induced locomotion, with mixed results for AMPH-induced stereotypy. TH-Cre mice on both backgrounds showed typical baseline activity and AMPH-induced stereotypy, with a difference in AMPH-induced locomotion observed only on the mixed background. Both transgenic lines displayed unaltered SKF-38393-induced grooming behavior. CONCLUSIONS Our findings indicate that the DAT-Ires-Cre transgenic line may lead to confounds for experiments that are dependent on DAT expression. The TH-Cre transgenic line studied here may be a more useful option, depending on background strain, because of its lack of baseline and drug-induced phenotypes. These data highlight the importance of appropriate controls in studies employing transgenic mice.
Collapse
|
38
|
Scarpin MR, Leiboff S, Brunkard JO. Parallel global profiling of plant TOR dynamics reveals a conserved role for LARP1 in translation. eLife 2020; 9:e58795. [PMID: 33054972 PMCID: PMC7584452 DOI: 10.7554/elife.58795] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
Target of rapamycin (TOR) is a protein kinase that coordinates eukaryotic metabolism. In mammals, TOR specifically promotes translation of ribosomal protein (RP) mRNAs when amino acids are available to support protein synthesis. The mechanisms controlling translation downstream from TOR remain contested, however, and are largely unexplored in plants. To define these mechanisms in plants, we globally profiled the plant TOR-regulated transcriptome, translatome, proteome, and phosphoproteome. We found that TOR regulates ribosome biogenesis in plants at multiple levels, but through mechanisms that do not directly depend on 5' oligopyrimidine tract motifs (5'TOPs) found in mammalian RP mRNAs. We then show that the TOR-LARP1-5'TOP signaling axis is conserved in plants and regulates expression of a core set of eukaryotic 5'TOP mRNAs, as well as new, plant-specific 5'TOP mRNAs. Our study illuminates ancestral roles of the TOR-LARP1-5'TOP metabolic regulatory network and provides evolutionary context for ongoing debates about the molecular function of LARP1.
Collapse
Affiliation(s)
- M Regina Scarpin
- Department of Plant and Microbial Biology, University of California at BerkeleyBerkeleyUnited States
- Plant Gene Expression Center, U.S. Department of Agriculture Agricultural Research ServiceAlbanyUnited States
| | - Samuel Leiboff
- Department of Plant and Microbial Biology, University of California at BerkeleyBerkeleyUnited States
- Plant Gene Expression Center, U.S. Department of Agriculture Agricultural Research ServiceAlbanyUnited States
- Department of Botany and Plant Pathology, Oregon State UniversityCorvallisUnited States
| | - Jacob O Brunkard
- Department of Plant and Microbial Biology, University of California at BerkeleyBerkeleyUnited States
- Plant Gene Expression Center, U.S. Department of Agriculture Agricultural Research ServiceAlbanyUnited States
- Laboratory of Genetics, University of Wisconsin—MadisonMadisonUnited States
| |
Collapse
|
39
|
Wundrach D, Martinetti LE, Stafford AM, Bilinovich SM, Angara K, Prokop JW, Crandall SR, Vogt D. A Human TSC1 Variant Screening Platform in Gabaergic Cortical Interneurons for Genotype to Phenotype Assessments. Front Mol Neurosci 2020; 13:573409. [PMID: 33071758 PMCID: PMC7539171 DOI: 10.3389/fnmol.2020.573409] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/28/2020] [Indexed: 11/23/2022] Open
Abstract
The TSC1 and TSC2 genes are connected to multiple syndromes from Tuberous Sclerosis Complex (TSC) to autism spectrum disorder (ASD), with uncertainty if genetic variants cause all or subsets of phenotypes based on the location and type of change. For TSC1, few have addressed if non-TSC associated genetic variants have direct contributions to changes in neurological genotype-to-phenotype impacts, including elevated rates of ASD and seizures. Dominant variants cause TSC, yet TSC1 has many heritable variants not dominant for TSC that are poorly understood in neurological function, with some associated with ASD. Herein, we examined how missense variants in TSC1, R336W, T360N, T393I, S403L, and H732Y, impacted the development of cortical inhibitory interneurons, cell-types whose molecular, cellular, and physiological properties are altered after the loss of mouse TSC1. We found these variants complemented a known phenotype caused by loss of TSC1, increased cell size. However, distinct variants, particularly S403L showed deficits in complementing an increase in parvalbumin levels and exhibited smaller amplitude after hyperpolarizations. Overall, these data show that subtle phenotypes can be induced by some TSC1 missense variants and provide an in vivo system to assess TSC1 variants’ neurological impact better.
Collapse
Affiliation(s)
- Dean Wundrach
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, United States
| | - Luis E Martinetti
- Department of Physiology, Michigan State University, East Lansing, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States
| | - April M Stafford
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, United States
| | - Stephanie M Bilinovich
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, United States
| | - Kartik Angara
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, United States
| | - Jeremy W Prokop
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, United States.,Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States.,Center for Research in Autism, Intellectual and other Neurodevelopmental Disabilities, Michigan State University, East Lansing, MI, United States
| | - Shane R Crandall
- Department of Physiology, Michigan State University, East Lansing, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States
| | - Daniel Vogt
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Center for Research in Autism, Intellectual and other Neurodevelopmental Disabilities, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
40
|
Kapur M, Ganguly A, Nagy G, Adamson SI, Chuang JH, Frankel WN, Ackerman SL. Expression of the Neuronal tRNA n-Tr20 Regulates Synaptic Transmission and Seizure Susceptibility. Neuron 2020; 108:193-208.e9. [PMID: 32853550 DOI: 10.1016/j.neuron.2020.07.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/07/2020] [Accepted: 07/19/2020] [Indexed: 12/31/2022]
Abstract
The mammalian genome has hundreds of nuclear-encoded tRNAs, but the contribution of individual tRNA genes to cellular and organismal function remains unknown. Here, we demonstrate that mutations in a neuronally enriched arginine tRNA, n-Tr20, increased seizure threshold and altered synaptic transmission. n-Tr20 expression also modulated seizures caused by an epilepsy-linked mutation in Gabrg2, a gene encoding a GABAA receptor subunit. Loss of n-Tr20 altered translation initiation by activating the integrated stress response and suppressing mTOR signaling, the latter of which may contribute to altered neurotransmission in mutant mice. Deletion of a highly expressed isoleucine tRNA similarly altered these signaling pathways in the brain, suggesting that regulation of translation initiation is a conserved response to tRNA loss. Our data indicate that loss of a single member of a tRNA family results in multiple cellular phenotypes, highlighting the disease-causing potential of tRNA mutations.
Collapse
Affiliation(s)
- Mridu Kapur
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, Section of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Archan Ganguly
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, Section of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gabor Nagy
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Scott I Adamson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, Institute for Systems Genomics, UConn Health, Farmington, CT 06030, USA
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Wayne N Frankel
- Institute for Genomic Medicine, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Susan L Ackerman
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, Section of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
41
|
Brunkard JO. Exaptive Evolution of Target of Rapamycin Signaling in Multicellular Eukaryotes. Dev Cell 2020; 54:142-155. [PMID: 32649861 PMCID: PMC7346820 DOI: 10.1016/j.devcel.2020.06.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/12/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022]
Abstract
Target of rapamycin (TOR) is a protein kinase that coordinates metabolism with nutrient and energy availability in eukaryotes. TOR and its primary interactors, RAPTOR and LST8, have been remarkably evolutionarily static since they arose in the unicellular last common ancestor of plants, fungi, and animals, but the upstream regulatory mechanisms and downstream effectors of TOR signaling have evolved considerable diversity in these separate lineages. Here, I focus on the roles of exaptation and adaptation in the evolution of novel signaling axes in the TOR network in multicellular eukaryotes, concentrating especially on amino acid sensing, cell-cell signaling, and cell differentiation.
Collapse
Affiliation(s)
- Jacob O Brunkard
- Department of Plant and Microbial Biology, University of California at Berkeley, Berkeley, CA 94720, USA; Plant Gene Expression Center, U.S. Department of Agriculture Agricultural Research Service, Albany, CA 94710, USA; Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
42
|
von Herrmann KM, Anderson FL, Martinez EM, Young AL, Havrda MC. Slc6a3-dependent expression of a CAPS-associated Nlrp3 allele results in progressive behavioral abnormalities and neuroinflammation in aging mice. J Neuroinflammation 2020; 17:213. [PMID: 32680528 PMCID: PMC7368774 DOI: 10.1186/s12974-020-01866-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/04/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND An association between neuroinflammation and age-related neurologic disorders has been established but the molecular mechanisms and cell types involved have not been thoroughly characterized. Activity of the proinflammatory NLRP3 inflammasome is implicated in Alzheimer's and Parkinson's disease and our recent studies in patients suggest that dopaminergic neurons within the degenerating mesencephalon express NLRP3 throughout the progression of PD. Here, we directly test the impact of enhanced inflammasome activity in mesencephalic neurons by characterizing motor function, tissue integrity, and neuroinflammation in aging mice harboring hyperactivating mutations within the endogenous murine Nlrp3 locus, enabled only in cells expressing the dopaminergic neuron-specific Slc6a3 promoter. METHODS We compared mice harboring inducible alleles encoding the cryopyrin-associated periodic syndrome activating mutations Nlrp3A350V and Nlrp3L351P inserted into the endogenous mouse Nlrp3 locus. Tissue specific expression was driven by breeding these animals with mice expressing Cre recombinase under the control of the dopaminergic neuron-specific Slc6a3 promoter. The experimental mice, designed to express hyperactive NLRP3 only when the endogenous mouse Nlrp3 promotor is active in dopaminergic neurons, were analyzed throughout 18 months of aging using longitudinal motor function assessments. Biochemical and histologic analyses of mesencephalic tissues were conducted in 1- and 18-month-old animals. RESULTS We observed progressive and significant deficits in motor function in animals expressing Nlrp3L351P, compared with animals expressing Nlrp3WT and Nlrp3A350V. Age-dependent neuroinflammatory changes in the mesencephalon were noted in all animals. Analysis of GFAP-immunoreactive astrocytes in the substantia nigra revealed a significant increase in astrocyte number in animals expressing Nlrp3L351P compared with Nlrp3WT and Nlrp3A350V. Further analysis of Nlrp3L351P striatal tissues indicated genotype specific gliosis, elevated Il1b expression, and both morphologic and gene expression indicators of proinflammatory A1 astrocytes. CONCLUSIONS Dopaminergic neurons have the potential to accumulate NLRP3 inflammasome activators with age, including reactive oxygen species, dopamine metabolites, and misfolded proteins. Results indicate the Nlrp3 locus is active in dopaminergic neurons in aging mice, and that the hyperactive Nlrp3L351P allele can drive neuroinflammatory changes in association with progressive behavioral deficits. Findings suggest neuronal NLRP3 inflammasome activity may contribute to neuroinflammation observed during normal aging and the progression of neurologic disorders.
Collapse
Affiliation(s)
- Katharine M von Herrmann
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth College, 1 Rope Ferry Road, Hanover, NH, 03755, USA
| | - Faith L Anderson
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth College, 1 Rope Ferry Road, Hanover, NH, 03755, USA
| | - Eileen M Martinez
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth College, 1 Rope Ferry Road, Hanover, NH, 03755, USA
| | - Alison L Young
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth College, 1 Rope Ferry Road, Hanover, NH, 03755, USA
| | - Matthew C Havrda
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth College, 1 Rope Ferry Road, Hanover, NH, 03755, USA.
| |
Collapse
|
43
|
Feliciano DM. The Neurodevelopmental Pathogenesis of Tuberous Sclerosis Complex (TSC). Front Neuroanat 2020; 14:39. [PMID: 32765227 PMCID: PMC7381175 DOI: 10.3389/fnana.2020.00039] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/10/2020] [Indexed: 12/22/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a model disorder for understanding brain development because the genes that cause TSC are known, many downstream molecular pathways have been identified, and the resulting perturbations of cellular events are established. TSC, therefore, provides an intellectual framework to understand the molecular and biochemical pathways that orchestrate normal brain development. The TSC1 and TSC2 genes encode Hamartin and Tuberin which form a GTPase activating protein (GAP) complex. Inactivating mutations in TSC genes (TSC1/TSC2) cause sustained Ras homologue enriched in brain (RHEB) activation of the mammalian isoform of the target of rapamycin complex 1 (mTORC1). TOR is a protein kinase that regulates cell size in many organisms throughout nature. mTORC1 inhibits catabolic processes including autophagy and activates anabolic processes including mRNA translation. mTORC1 regulation is achieved through two main upstream mechanisms. The first mechanism is regulation by growth factor signaling. The second mechanism is regulation by amino acids. Gene mutations that cause too much or too little mTORC1 activity lead to a spectrum of neuroanatomical changes ranging from altered brain size (micro and macrocephaly) to cortical malformations to Type I neoplasias. Because somatic mutations often underlie these changes, the timing, and location of mutation results in focal brain malformations. These mutations, therefore, provide gain-of-function and loss-of-function changes that are a powerful tool to assess the events that have gone awry during development and to determine their functional physiological consequences. Knowledge about the TSC-mTORC1 pathway has allowed scientists to predict which upstream and downstream mutations should cause commensurate neuroanatomical changes. Indeed, many of these predictions have now been clinically validated. A description of clinical imaging and histochemical findings is provided in relation to laboratory models of TSC that will allow the reader to appreciate how human pathology can provide an understanding of the fundamental mechanisms of development.
Collapse
Affiliation(s)
- David M Feliciano
- Department of Biological Sciences, Clemson University, Clemson, SC, United States
| |
Collapse
|
44
|
Neurodevelopmental Disorders: From Genetics to Functional Pathways. Trends Neurosci 2020; 43:608-621. [PMID: 32507511 DOI: 10.1016/j.tins.2020.05.004] [Citation(s) in RCA: 323] [Impact Index Per Article: 64.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/27/2020] [Accepted: 05/18/2020] [Indexed: 12/21/2022]
Abstract
Neurodevelopmental disorders (NDDs) are a class of disorders affecting brain development and function and are characterized by wide genetic and clinical variability. In this review, we discuss the multiple factors that influence the clinical presentation of NDDs, with particular attention to gene vulnerability, mutational load, and the two-hit model. Despite the complex architecture of mutational events associated with NDDs, the various proteins involved appear to converge on common pathways, such as synaptic plasticity/function, chromatin remodelers and the mammalian target of rapamycin (mTOR) pathway. A thorough understanding of the mechanisms behind these pathways will hopefully lead to the identification of candidates that could be targeted for treatment approaches.
Collapse
|