1
|
Saint-Jour E, Allichon MC, Andrianarivelo A, Montalban E, Martin C, Huet L, Heck N, Hagenston AM, Ravenhorst A, Marias M, Gervasi N, Arrivet F, Vilette A, Pinchaud K, Betuing S, Lissek T, Caboche J, Bading H, Vanhoutte P. Nuclear Calcium Signaling in D 1 Receptor-Expressing Neurons of the Nucleus Accumbens Regulates Molecular, Cellular, and Behavioral Adaptations to Cocaine. Biol Psychiatry 2025; 98:34-45. [PMID: 39864789 DOI: 10.1016/j.biopsych.2025.01.013] [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: 06/28/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND The persistence of cocaine-evoked adaptations relies on gene regulations within the reward circuit, especially in the ventral striatum (i.e., nucleus accumbens [NAc]). Notably, activation of the ERK (extracellular signal-regulated kinase) pathway in the striatum is known to trigger a transcriptional program shaping long-term responses to cocaine. Nuclear calcium signaling has also been shown to control multiple forms of transcription-dependent neuroadaptations, but the dynamics and roles of striatal nuclear calcium signaling in preclinical models of addiction remain unknown. METHODS A genetically encoded cell type-specific nuclear calcium probe has been developed to monitor calcium dynamics in the nuclei of striatal neurons, including in freely moving mice. A cell type-specific inhibitor of nuclear calcium signaling combined with 3-dimensional imaging of neuronal morphology, immunostaining, and behavior was used to disentangle the roles of nuclear calcium in NAc medium spiny neurons (MSNs) expressing the dopamine D1 receptor (D1R) or D2 receptor (D2R) on cocaine-evoked responses. RESULTS The D1R-mediated potentiation of calcium influx through glutamate NMDA receptors, which shapes cocaine effects, also drives nuclear calcium transients. Fiber photometry revealed that cocaine-treated mice showed a sustained nuclear calcium increase in NAc D1R-MSNs. Disrupting nuclear calcium in D1R-MSNs, but not D2R-MSNs, blocked cocaine-evoked morphological changes of MSNs and gene expression and blunted cocaine's rewarding effects. CONCLUSIONS Our study unravels the dynamics and roles of cocaine-induced nuclear calcium signaling increases in D1R-MSNs on molecular, cellular, and behavioral adaptations to cocaine and represents a significant breakthrough because it could contribute to the development of innovative strategies with therapeutic potential to alleviate addiction symptoms.
Collapse
Affiliation(s)
- Estefani Saint-Jour
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Marie-Charlotte Allichon
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Andry Andrianarivelo
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Enrica Montalban
- Université Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, Unité Mixte de Recherche 8251, Centre National de la Recherche Scientifique, Paris, France
| | - Claire Martin
- Université Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, Unité Mixte de Recherche 8251, Centre National de la Recherche Scientifique, Paris, France
| | - Lisa Huet
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Nicolas Heck
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Anna M Hagenston
- Heidelberg University, Interdisciplinary Center for Neurosciences, Institute of Neurobiology, Heidelberg, Germany
| | - Aisha Ravenhorst
- Heidelberg University, Interdisciplinary Center for Neurosciences, Institute of Neurobiology, Heidelberg, Germany
| | - Mélanie Marias
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Nicolas Gervasi
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique UMR 7241, Institut National de la Santé et de la Recherche Médicale U1050, Paris Science et Lettre Research University, Paris, France
| | - Faustine Arrivet
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Adèle Vilette
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Katleen Pinchaud
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Sandrine Betuing
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Thomas Lissek
- Heidelberg University, Interdisciplinary Center for Neurosciences, Institute of Neurobiology, Heidelberg, Germany
| | - Jocelyne Caboche
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Hilmar Bading
- Heidelberg University, Interdisciplinary Center for Neurosciences, Institute of Neurobiology, Heidelberg, Germany
| | - Peter Vanhoutte
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France.
| |
Collapse
|
2
|
Montemarano A, Fox LD, Alkhaleel FA, Ostman AE, Sohail H, Pandey S, Murdaugh LB, Fox ME. A Drd1-cre mouse line with nucleus accumbens gene dysregulation exhibits blunted fentanyl seeking. Neuropsychopharmacology 2025:10.1038/s41386-025-02116-0. [PMID: 40316698 DOI: 10.1038/s41386-025-02116-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/04/2025] [Accepted: 04/22/2025] [Indexed: 05/04/2025]
Abstract
The synthetic opioid fentanyl remains abundant in the illicit drug supply, contributing to tens of thousands of overdose deaths every year. Despite this, the neurobiological effects of fentanyl use remain largely understudied. The nucleus accumbens (NAc) is a central locus promoting persistent drug use and relapse, largely dependent on activity of dopamine D1 receptors. NAc D1 receptor-expressing medium spiny neurons (D1-MSNs) undergo molecular and physiological neuroadaptations in response to chronic fentanyl that may promote relapse. Here, we obtained Drd1-cre120Mxu mice to investigate D1-dependent mechanisms of fentanyl relapse. We serendipitously discovered this mouse line has reduced fentanyl seeking, despite similar intravenous fentanyl self-administration, similar sucrose self-administration and seeking, and greater fentanyl-induced locomotion compared to wildtype counterparts. We found drug-naïve Drd1-cre120Mxu mice have elevated D1 receptor expression in NAc and increased sensitivity to the D1 receptor agonist SKF-38393. After fentanyl self-administration, Drd1-cre120Mxu mice exhibit divergent expression of MSN markers, opioid receptors, glutamate receptor subunits, and TrkB which may underly their blunted fentanyl seeking. Finally, we show fentanyl-related behavior is unaltered by chemogenetic manipulation of NAc core D1-MSNs in Drd1-cre120Mxu mice. Conversely, chemogenetic stimulation of ventral mesencephalon-projecting NAc core MSNs (putative D1-MSNs) in wildtype mice recapitulated the blunted fentanyl seeking of Drd1-cre120Mxu mice, supporting a role for aberrant D1-MSN signaling in this behavior. Together, our data uncover alterations in NAc gene expression and function with implications for susceptibility and resistance to developing fentanyl use disorder.
Collapse
Affiliation(s)
- Annalisa Montemarano
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Logan D Fox
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Farrah A Alkhaleel
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Alexandria E Ostman
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Hajra Sohail
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Samiksha Pandey
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Laura B Murdaugh
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Megan E Fox
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA.
- Department of Neuroscience and Experimental Therapeutics, Pennsylvania State University College of Medicine, Hershey, PA, USA.
| |
Collapse
|
3
|
Lissek T. The universal role of adaptive transcription in health and disease. FEBS J 2025; 292:2479-2505. [PMID: 39609264 PMCID: PMC12103072 DOI: 10.1111/febs.17324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 07/25/2024] [Accepted: 11/07/2024] [Indexed: 11/30/2024]
Abstract
In animals, adaptive transcription is a crucial mechanism to connect environmental stimulation to changes in gene expression and subsequent organism remodeling. Adaptive transcriptional programs involving molecules such as CREB, SRF, MEF2, FOS, and EGR1 are central to a wide variety of organism functions, including learning and memory, immune system plasticity, and muscle hypertrophy, and their activation increases cellular resilience and prevents various diseases. Yet, they also form the basis for many maladaptive processes and are involved in the progression of addiction, depression, cancer, cardiovascular disorders, autoimmune conditions, and metabolic dysfunction among others and are thus prime examples for mediating the adaptation-maladaptation dilemma. They are implicated in the therapeutic effects of major treatment modalities such as antidepressants and can have negative effects on treatment, for example, contributing to therapy resistance in cancer. This review examines the universal role of adaptive transcription as a mechanism for the induction of adaptive cell state transitions in health and disease and explores how many medical disorders can be conceptualized as caused by errors in cellular adaptation goals. It also considers the underlying principles in the basic structure of adaptive gene programs such as their division into a core and a directional program. Finally, it analyses how one might best reprogram cells via targeting of adaptive transcription in combination with complex stimulation patterns to leverage endogenous cellular reprogramming dynamics and achieve optimal health of the whole organism.
Collapse
Affiliation(s)
- Thomas Lissek
- Interdisciplinary Center for NeurosciencesHeidelberg UniversityGermany
| |
Collapse
|
4
|
Brida KL, Jorgensen ET, Phillips RA, Newman CE, Tuscher JJ, Morring EK, Zipperly ME, Ianov L, Montgomery KD, Tippani M, Hyde TM, Maynard KR, Martinowich K, Day JJ. Reelin marks cocaine-activated striatal neurons, promotes neuronal excitability, and regulates cocaine reward. SCIENCE ADVANCES 2025; 11:eads4441. [PMID: 40138397 PMCID: PMC12076537 DOI: 10.1126/sciadv.ads4441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/20/2025] [Indexed: 03/29/2025]
Abstract
Drugs of abuse activate defined neuronal populations in reward structures such as the nucleus accumbens (NAc), which promote the enduring synaptic, circuit, and behavioral consequences of drug exposure. While the molecular and cellular effects arising from experience with drugs like cocaine are increasingly well understood, mechanisms that dictate NAc neuronal recruitment remain unknown. Here, we leveraged unbiased single-nucleus transcriptional profiling and targeted in situ detection to identify Reln (encoding the secreted glycoprotein, Reelin) as a marker of cocaine-activated neuronal populations within the rat NAc. A CRISPR interference approach enabling selective Reln knockdown in the adult NAc altered expression of calcium signaling genes, promoted a transcriptional trajectory consistent with loss of cocaine sensitivity, and decreased MSN excitability. Behaviorally, Reln knockdown prevented cocaine locomotor sensitization, abolished cocaine place preference memory, and decreased cocaine self-administration behavior. These results identify Reelin as a critical mechanistic link between neuronal activation and cocaine-induced behavioral adaptations.
Collapse
Affiliation(s)
- Kasey L. Brida
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emily T. Jorgensen
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Robert A. Phillips
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Catherine E. Newman
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J. Tuscher
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emily K. Morring
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan E. Zipperly
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center,
University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kelsey D. Montgomery
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral
Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205,
USA
- Department of Neurology, Johns Hopkins
University School of Medicine, Baltimore, MD 21205, USA
| | - Kristen R. Maynard
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral
Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205,
USA
- Department of Neuroscience, Johns Hopkins
University School of Medicine, Baltimore, MD 21205, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral
Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205,
USA
- Department of Neuroscience, Johns Hopkins
University School of Medicine, Baltimore, MD 21205, USA
- The Kavli Neuroscience Discovery
Institute, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jeremy J. Day
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
5
|
Harahap-Carrillo IS, Fok D, Wong F, Malik G, Maung R, Qiu X, Ojeda-Juárez D, Thaney VE, Sanchez AB, Godzik A, Roberts AJ, Kaul M. Chronic, Low-Dose Methamphetamine Reveals Sexual Dimorphism of Memory Performance, Histopathology, and Gene Expression Affected by HIV-1 Tat Protein in a Transgenic Model of NeuroHIV. Viruses 2025; 17:361. [PMID: 40143289 PMCID: PMC11946854 DOI: 10.3390/v17030361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Methamphetamine (METH) use is frequent among people with HIV (PWH) and appears to increase the risk of neuronal injury and neurocognitive impairment (NCI). This study explored in vivo the effects of a 12 week (long-term), low-dose METH regimen in a transgenic animal model of neuroHIV with inducible expression of HIV-1 transactivator of transcription (Tat). Seven months after transient Tat induction and five months after METH exposure ended, we detected behavioral changes in the Barnes maze (BM) spatial memory task in the Tat and METH groups but not the combined Tat + METH group. The novel object recognition (NOR) task revealed that Tat extinguished discrimination in female animals with and without METH, although METH alone slightly improved NOR. In contrast, in males, Tat, METH, and Tat + METH all compromised NOR. Neuropathological examination detected sex-dependent and brain region-specific changes of pre-synaptic terminals, neurites, and activation of astrocytes and microglia. RNA-sequencing and quantitative reverse transcription polymerase chain reaction indicated that METH and Tat significantly altered gene expression, including factors linked to Alzheimer's disease-like NCI. In summary, chronic low-dose METH exerts long-term effects on behavioral function, neuropathology, and mRNA expression, and modulates the effects of Tat, suggesting sex-dependent and -independent mechanisms may converge in HIV brain injury and NCI.
Collapse
Affiliation(s)
- Indira S. Harahap-Carrillo
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside (UCR), Riverside, CA 92521, USA; (I.S.H.-C.); (D.F.); (F.W.); (G.M.); (R.M.); (X.Q.); (D.O.-J.); (A.G.)
- Neuroscience Graduate Program, College of Natural & Agricultural Sciences, University of California, Riverside (UCR), Riverside, CA 92521, USA
| | - Dominic Fok
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside (UCR), Riverside, CA 92521, USA; (I.S.H.-C.); (D.F.); (F.W.); (G.M.); (R.M.); (X.Q.); (D.O.-J.); (A.G.)
| | - Frances Wong
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside (UCR), Riverside, CA 92521, USA; (I.S.H.-C.); (D.F.); (F.W.); (G.M.); (R.M.); (X.Q.); (D.O.-J.); (A.G.)
| | - Gabriel Malik
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside (UCR), Riverside, CA 92521, USA; (I.S.H.-C.); (D.F.); (F.W.); (G.M.); (R.M.); (X.Q.); (D.O.-J.); (A.G.)
| | - Ricky Maung
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside (UCR), Riverside, CA 92521, USA; (I.S.H.-C.); (D.F.); (F.W.); (G.M.); (R.M.); (X.Q.); (D.O.-J.); (A.G.)
- Translational Methamphetamine AIDS Research Center (TMARC), Department of Psychiatry, University of California, San Diego (UCSD), San Diego, CA 92093, USA;
- Center for Infectious and Inflammatory Disease, Sanford Burnham Prebys Medical Discovery Institute (SBP), La Jolla, CA 92037, USA;
| | - Xinru Qiu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside (UCR), Riverside, CA 92521, USA; (I.S.H.-C.); (D.F.); (F.W.); (G.M.); (R.M.); (X.Q.); (D.O.-J.); (A.G.)
| | - Daniel Ojeda-Juárez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside (UCR), Riverside, CA 92521, USA; (I.S.H.-C.); (D.F.); (F.W.); (G.M.); (R.M.); (X.Q.); (D.O.-J.); (A.G.)
- Center for Infectious and Inflammatory Disease, Sanford Burnham Prebys Medical Discovery Institute (SBP), La Jolla, CA 92037, USA;
| | - Victoria E. Thaney
- Center for Infectious and Inflammatory Disease, Sanford Burnham Prebys Medical Discovery Institute (SBP), La Jolla, CA 92037, USA;
| | - Ana B. Sanchez
- Translational Methamphetamine AIDS Research Center (TMARC), Department of Psychiatry, University of California, San Diego (UCSD), San Diego, CA 92093, USA;
- Center for Infectious and Inflammatory Disease, Sanford Burnham Prebys Medical Discovery Institute (SBP), La Jolla, CA 92037, USA;
| | - Adam Godzik
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside (UCR), Riverside, CA 92521, USA; (I.S.H.-C.); (D.F.); (F.W.); (G.M.); (R.M.); (X.Q.); (D.O.-J.); (A.G.)
| | - Amanda J. Roberts
- The Scripps Research Institute (TSRI), Animal Models Core, La Jolla, CA 92037, USA;
| | - Marcus Kaul
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside (UCR), Riverside, CA 92521, USA; (I.S.H.-C.); (D.F.); (F.W.); (G.M.); (R.M.); (X.Q.); (D.O.-J.); (A.G.)
- Neuroscience Graduate Program, College of Natural & Agricultural Sciences, University of California, Riverside (UCR), Riverside, CA 92521, USA
- Translational Methamphetamine AIDS Research Center (TMARC), Department of Psychiatry, University of California, San Diego (UCSD), San Diego, CA 92093, USA;
- Center for Infectious and Inflammatory Disease, Sanford Burnham Prebys Medical Discovery Institute (SBP), La Jolla, CA 92037, USA;
| |
Collapse
|
6
|
Lakunina A, Socha KZ, Ladd A, Bowen AJ, Chen S, Colonell J, Doshi A, Karsh B, Krumin M, Kulik P, Li A, Neutens P, O'Callaghan J, Olsen M, Putzeys J, Tilmans HA, Ye Z, Welkenhuysen M, Häusser M, Koch C, Ting JT, Neuropixels Opto Consortium, Dutta B, Harris TD, Steinmetz NA, Svoboda K, Siegle JH, Carandini M. Neuropixels Opto: Combining high-resolution electrophysiology and optogenetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.04.636286. [PMID: 39975326 PMCID: PMC11838571 DOI: 10.1101/2025.02.04.636286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
High-resolution extracellular electrophysiology is the gold standard for recording spikes from distributed neural populations, and is especially powerful when combined with optogenetics for manipulation of specific cell types with high temporal resolution. We integrated these approaches into prototype Neuropixels Opto probes, which combine electronic and photonic circuits. These devices pack 960 electrical recording sites and two sets of 14 light emitters onto a 1 cm shank, allowing spatially addressable optogenetic stimulation with blue and red light. In mouse cortex, Neuropixels Opto probes delivered high-quality recordings together with spatially addressable optogenetics, differentially activating or silencing neurons at distinct cortical depths. In mouse striatum and other deep structures, Neuropixels Opto probes delivered efficient optotagging, facilitating the identification of two cell types in parallel. Neuropixels Opto probes represent an unprecedented tool for recording, identifying, and manipulating neuronal populations.
Collapse
Affiliation(s)
- Anna Lakunina
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Karolina Z Socha
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Alexander Ladd
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Anna J Bowen
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Susu Chen
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Jennifer Colonell
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Anjal Doshi
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Bill Karsh
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Michael Krumin
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Pavel Kulik
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Anna Li
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
| | | | | | - Meghan Olsen
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | | | | | - Zhiwen Ye
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
| | | | - Michael Häusser
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | | | - Jonathan T Ting
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Timothy D Harris
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas A Steinmetz
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Karel Svoboda
- Allen Institute for Neural Dynamics, Seattle, WA, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | | | - Matteo Carandini
- UCL Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
7
|
Montemarano A, Fox LD, Alkhaleel FA, Ostman AE, Sohail H, Pandey S, Fox ME. A Drd1-cre mouse line with nucleus accumbens gene dysregulation exhibits blunted fentanyl seeking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638324. [PMID: 40027693 PMCID: PMC11870424 DOI: 10.1101/2025.02.14.638324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The synthetic opioid fentanyl remains abundant in the illicit drug supply, contributing to tens of thousands of overdose deaths every year. Despite this, the neurobiological effects of fentanyl use remain largely understudied. The nucleus accumbens (NAc) is a central locus promoting persistent drug use and relapse, largely dependent on activity of dopamine D1 receptors. NAc D1 receptor-expressing medium spiny neurons (D1-MSNs) undergo molecular and physiological adaptations that contribute to negative affect during fentanyl abstinence, but whether these neuroadaptations also promote fentanyl relapse is unclear. Here, we obtained Drd1-cre 120Mxu mice to investigate D1-dependent mechanisms of fentanyl relapse. We serendipitously discovered this mouse line is resistant to fentanyl seeking, despite similar intravenous fentanyl self-administration, and greater fentanyl-induced locomotion, compared to wildtype counterparts. In drug naïve mice, we found Drd1-cre 120Mxu mice have elevated D1 receptor expression in NAc, alongside increased expression of MSN marker genes Chrm4 and Penk . We show Drd1-cre 120Mxu mice have increased sensitivity to the D1 receptor agonist SKF-38393, and exhibit divergent expression of MSN markers, opioid receptors, glutamate receptor subunits, and TrkB after fentanyl self-administration that may underly blunted fentanyl seeking. Finally, we show fentanyl-related behavior is unaltered by chemogenetic manipulation of D1-MSNs in Drd1-cre 120Mxu mice. Conversely, chemogenetic stimulation of putative D1-MSNs in wildtype mice recapitulated the blunted fentanyl seeking of Drd1-cre 120Mxu mice, supporting a role for aberrant D1-MSN signaling in this behavior. Together, our data uncover alterations in NAc gene expression and function with implications for susceptibility and resistance to developing fentanyl use disorder.
Collapse
|
8
|
Barbosa-Méndez S, Matus-Ortega M, Hernandez-Miramontes R, Salazar-Juarez A. COT-TT vaccine attenuates cocaine-seeking and cocaine-conditioned place preference in rats. Hum Vaccin Immunother 2024; 20:2299068. [PMID: 38228468 PMCID: PMC10793666 DOI: 10.1080/21645515.2023.2299068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024] Open
Abstract
Vaccination active, promising alternative immunological strategy to treat of CUD. Various models of cocaine vaccines have been evaluated in animals and humans with relative success. In this sense, it is necessary to improve or optimize the cocaine vaccines already evaluated. Our laboratory previously reported the efficacy of the tetanus toxoid-conjugated morphine vaccine (M6-TT). The M6-TT vaccine can generate high titers of antibodies and reduce heroin-induced behavioral effects in rodents. So, it would be plausible to assume that if we modify the M6-TT vaccine by changing the hapten and maintaining the rest of the structural elements of the vaccine, we will maintain the properties of the M6-TT vaccine (high antibody titers). The objective of this study was to determine whether the antibodies generated by a tetanus toxoid-conjugated cocaine vaccine (COC-TT) can recognize and capture cocaine and decrease the cocaine-induced reinforcing effects. Male Wistar rats were immunized with the COC-TT. A solid-phase antibody-capture ELISA was used to monitor antibody titer responses after each booster dose in vaccinated animals. The study used cocaine self-administration and place-preference testing to evaluate the cocaine-reinforcing effects. The COC-TT vaccine could generate high levels of anti-cocaine antibodies. The antibodies reduced the cocaine self-administration and cocaine place preference. In addition, they decreased the cocaine-induced Fos protein expression. These findings suggest that the COC-TT vaccine generates a robust immunogenic response capable of reducing the reinforcing effects of cocaine, which supports its possible future use in clinical trials in patients with CUD.
Collapse
Affiliation(s)
- Susana Barbosa-Méndez
- Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría, Ciudad de México, México
| | - Maura Matus-Ortega
- Laboratorio de Neurobiología Molecular y Neuroquímica de las Adicciones, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría, Ciudad de México, México
| | - Ricardo Hernandez-Miramontes
- Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría, Ciudad de México, México
| | - Alberto Salazar-Juarez
- Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría, Ciudad de México, México
| |
Collapse
|
9
|
Ku MJ, Kim CY, Park JW, Lee S, Jeong EY, Jeong JW, Kim WY, Kim JH. Wireless optogenetic stimulation on the prelimbic to the nucleus accumbens core circuit attenuates cocaine-induced behavioral sensitization. Neurobiol Dis 2024; 203:106733. [PMID: 39536953 DOI: 10.1016/j.nbd.2024.106733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/03/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Behavioral sensitization is defined as the heightened and persistent behavioral response to repeated drug exposure as a manifestation of drug craving. Psychomotor stimulants such as cocaine can induce strong behavioral sensitization. In this study, we explored the effects of optogenetic stimulation of the prelimbic (PL) to the nucleus accumbnes (NAc) core on the expression of cocaine-induced behavioral sensitization. Using wireless optogenetics, we selectively stimulated the PL-NAc core circuit, and assessed the effects of this treatment on cocaine-induced locomotor activity and accompanying changes in neuronal activation and dendritic spine density. Our findings revealed that optogenetic stimulation of the PL-NAc core circuit effectively suppressed the cocaine-induced locomotor sensitization, accompanied by a reduction in c-Fos expression within the NAc core. Moreover, optogenetic stimulation led to reduction in dendritic spine density, particularly thin and mushroom spine densities, in the NAc core. This study demonstrates that cocaine-induced locomotor sensitization can be regulated by optogenetic stimulation of the PL-NAc core circuit, providing insights into the crucial role of this circuit in psychomotor stimulant addiction.
Collapse
Affiliation(s)
- Min Jeong Ku
- Department of Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Choong Yeon Kim
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Information & Electronics Research Institute, Daejeon 34141, Republic of Korea
| | - Jong Woo Park
- Department of Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seohyeon Lee
- Department of Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Eun Young Jeong
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jae-Woong Jeong
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; KAIST Institute for NanoCentury, Daejeon 34141, Republic of Korea; KAIST Institute for Health Science and Technology, Daejeon 34141, Republic of Korea.
| | - Wha Young Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Jeong-Hoon Kim
- Department of Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
10
|
Mews P, Van der Zee Y, Gurung A, Estill M, Futamura R, Kronman H, Ramakrishnan A, Ryan M, Reyes AA, Garcia BA, Browne CJ, Sidoli S, Shen L, Nestler EJ. Cell type-specific epigenetic priming of gene expression in nucleus accumbens by cocaine. SCIENCE ADVANCES 2024; 10:eado3514. [PMID: 39365860 PMCID: PMC11451531 DOI: 10.1126/sciadv.ado3514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 09/03/2024] [Indexed: 10/06/2024]
Abstract
A hallmark of addiction is the ability of drugs of abuse to trigger relapse after periods of prolonged abstinence. Here, we describe an epigenetic mechanism whereby chronic cocaine exposure causes lasting chromatin and downstream transcriptional modifications in the nucleus accumbens (NAc), a critical brain region controlling motivation. We link prolonged withdrawal from cocaine to the depletion of the histone variant H2A.Z, coupled with increased genome accessibility and latent priming of gene transcription, in D1 dopamine receptor-expressing medium spiny neurons (D1 MSNs) that relate to aberrant gene expression upon drug relapse. The histone chaperone ANP32E removes H2A.Z from chromatin, and we demonstrate that D1 MSN-selective Anp32e knockdown prevents cocaine-induced H2A.Z depletion and blocks cocaine's rewarding actions. By contrast, very different effects of cocaine exposure, withdrawal, and relapse were found for D2 MSNs. These findings establish histone variant exchange as an important mechanism and clinical target engaged by drugs of abuse to corrupt brain function and behavior.
Collapse
Affiliation(s)
- Philipp Mews
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yentl Van der Zee
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashik Gurung
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Molly Estill
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rita Futamura
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hope Kronman
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meagan Ryan
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Abner A. Reyes
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin A. Garcia
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Caleb J. Browne
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medticine, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
11
|
Barbosa-Méndez S, Salazar-Juárez A. Mirtazapine decreased cocaine-induced c-fos expression and dopamine release in rats. Front Psychiatry 2024; 15:1428730. [PMID: 39188520 PMCID: PMC11346032 DOI: 10.3389/fpsyt.2024.1428730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Introduction Chronic cocaine exposure induces an increase in dopamine release and an increase in the expression of the Fos protein in the rat striatum. It has been suggested that both are necessary for the expression of cocaine-induced alterations in behavior and neural circuitry. Mirtazapine dosing attenuated the cocaine-induced psychomotor and reinforcer effects. Methods The study evaluates the effect of chronic dosing of mirtazapine on cocaine-induced extracellular dopamine levels and Fos protein expression in rats. Male Wistar rats received cocaine (10 mg/Kg; i.p.) during the induction and expression of locomotor sensitization. The mirtazapine (30 mg/Kg; MIR), was administered 30 minutes before cocaine during the cocaine withdrawal. After each treatment, the locomotor activity was recorded for 30 minutes. Animals were sacrificed after treatment administration. Dopamine levels were determined by high-performance liquid chromatographic (HPLC) in the ventral striatum, the prefrontal cortex (PFC), and the ventral tegmental area (VTA) in animals treated with mirtazapine and cocaine. The quantification of c-fos immunoreactive cells was carried out by stereology analysis. Results Mirtazapine generated a decrease in cocaine-induced locomotor activity. In addition, mirtazapine decreased the amount of cocaine-induced dopamine and the number of cells immunoreactive to the Fos protein in the striatum, PFC, and VTA. Discussion These data suggest that mirtazapine could prevent the consolidation of changes in behavior and the cocaine-induced reorganization of neuronal circuits. It would explain the mirtazapine-induced effects on cocaine behavioral sensitization. Thus, these data together could support its possible use for the treatment of patients with cocaine use disorder.
Collapse
|
12
|
Hughes BW, Huebschman JL, Tsvetkov E, Siemsen BM, Snyder KK, Akiki RM, Wood DJ, Penrod RD, Scofield MD, Berto S, Taniguchi M, Cowan CW. NPAS4 supports cocaine-conditioned cues in rodents by controlling the cell type-specific activation balance in the nucleus accumbens. Nat Commun 2024; 15:5971. [PMID: 39117647 PMCID: PMC11310321 DOI: 10.1038/s41467-024-50099-1] [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: 09/26/2022] [Accepted: 06/28/2024] [Indexed: 08/10/2024] Open
Abstract
Powerful associations that link drugs of abuse with cues in the drug-paired environment often serve as prepotent relapse triggers. Drug-associated contexts and cues activate ensembles of nucleus accumbens (NAc) neurons, including D1-class medium spiny neurons (MSNs) that typically promote, and D2-class MSNs that typically oppose, drug seeking. We found that in mice, cocaine conditioning upregulated transiently the activity-regulated transcription factor, Neuronal PAS Domain Protein 4 (NPAS4), in a small subset of NAc neurons. The NPAS4+ NAc ensemble was required for cocaine conditioned place preference. We also observed that NPAS4 functions within NAc D2-, but not D1-, MSNs to support cocaine-context associations and cue-induced cocaine, but not sucrose, seeking. Together, our data show that the NPAS4+ ensemble of NAc neurons is essential for cocaine-context associations in mice, and that NPAS4 itself functions in NAc D2-MSNs to support cocaine-context associations by suppressing drug-induced counteradaptations that oppose relapse-related behaviour.
Collapse
Affiliation(s)
- Brandon W Hughes
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Jessica L Huebschman
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Evgeny Tsvetkov
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Benjamin M Siemsen
- Department of Anesthesiology, Medical University of South Carolina, Charleston, SC, USA
| | - Kirsten K Snyder
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Rose Marie Akiki
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, USA
| | - Daniel J Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, USA
| | - Rachel D Penrod
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Michael D Scofield
- Department of Anesthesiology, Medical University of South Carolina, Charleston, SC, USA
| | - Stefano Berto
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Makoto Taniguchi
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| | - Christopher W Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
13
|
Wang X, Wang T, Kaneko S, Kriukov E, Lam E, Szczepan M, Chen J, Gregg A, Wang X, Fernandez-Gonzalez A, Mitsialis SA, Kourembanas S, Baranov P, Sun Y. Photoreceptors inhibit pathological retinal angiogenesis through transcriptional regulation of Adam17 via c-Fos. Angiogenesis 2024; 27:379-395. [PMID: 38483712 PMCID: PMC11303108 DOI: 10.1007/s10456-024-09912-0] [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: 11/14/2023] [Accepted: 03/03/2024] [Indexed: 04/11/2024]
Abstract
Pathological retinal angiogenesis profoundly impacts visual function in vascular eye diseases, such as retinopathy of prematurity (ROP) in preterm infants and age-related macular degeneration in the elderly. While the involvement of photoreceptors in these diseases is recognized, the underlying mechanisms remain unclear. This study delved into the pivotal role of photoreceptors in regulating abnormal retinal blood vessel growth using an oxygen-induced retinopathy (OIR) mouse model through the c-Fos/A disintegrin and metalloprotease 17 (Adam17) axis. Our findings revealed a significant induction of c-Fos expression in rod photoreceptors, and c-Fos depletion in these cells inhibited pathological neovascularization and reduced blood vessel leakage in the OIR mouse model. Mechanistically, c-Fos directly regulated the transcription of Adam17 a shedding protease responsible for the production of bioactive molecules involved in inflammation, angiogenesis, and cell adhesion and migration. Furthermore, we demonstrated the therapeutic potential by using an adeno-associated virus carrying a rod photoreceptor-specific short hairpin RNA against c-fos which effectively mitigated abnormal retinal blood vessel overgrowth, restored retinal thickness, and improved electroretinographic (ERG) responses. In conclusion, this study highlights the significance of photoreceptor c-Fos in ROP pathology, offering a novel perspective for the treatment of this disease.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tianxi Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Satoshi Kaneko
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emil Kriukov
- Department of Ophthalmology, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Enton Lam
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Manon Szczepan
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jasmine Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Austin Gregg
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xingyan Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - S Alex Mitsialis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Petr Baranov
- Department of Ophthalmology, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Brida KL, Jorgensen ET, Phillips RA, Newman CE, Tuscher JJ, Morring EK, Zipperly ME, Ianov L, Montgomery KD, Tippani M, Hyde TM, Maynard KR, Martinowich K, Day JJ. Reelin marks cocaine-activated striatal ensembles, promotes neuronal excitability, and regulates cocaine reward. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599348. [PMID: 38948801 PMCID: PMC11212904 DOI: 10.1101/2024.06.17.599348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Drugs of abuse activate defined neuronal ensembles in brain reward structures such as the nucleus accumbens (NAc), which are thought to promote the enduring synaptic, circuit, and behavioral consequences of drug exposure. While the molecular and cellular effects arising from experience with drugs like cocaine are increasingly well understood, the mechanisms that sculpt NAc ensemble participation are largely unknown. Here, we leveraged unbiased single-nucleus transcriptional profiling to identify expression of the secreted glycoprotein Reelin (encoded by the Reln gene) as a marker of cocaine-activated neuronal ensembles within the rat NAc. Multiplexed in situ detection confirmed selective expression of the immediate early gene Fos in Reln+ neurons after cocaine experience, and also revealed enrichment of Reln mRNA in Drd1 + medium spiny neurons (MSNs) in both the rat and human brain. Using a novel CRISPR interference strategy enabling selective Reln knockdown in the adult NAc, we observed altered expression of genes linked to calcium signaling, emergence of a transcriptional trajectory consistent with loss of cocaine sensitivity, and a striking decrease in MSN intrinsic excitability. At the behavioral level, loss of Reln prevented cocaine locomotor sensitization, abolished cocaine place preference memory, and decreased cocaine self-administration behavior. Together, these results identify Reelin as a critical mechanistic link between ensemble participation and cocaine-induced behavioral adaptations.
Collapse
|
15
|
Liu X, Wang F, Le Q, Ma L. Cellular and molecular basis of drug addiction: The role of neuronal ensembles in addiction. Curr Opin Neurobiol 2023; 83:102813. [PMID: 37972536 DOI: 10.1016/j.conb.2023.102813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023]
Abstract
Addiction has been conceptualized as a disease of learning and memory. Learned associations between environmental cues and unconditioned rewards induced by drug administration, which play a critical role in addiction, have been shown to be encoded in sparsely distributed populations of neurons called neuronal ensembles. This review aims to highlight how synaptic remodeling and alterations in signaling pathways that occur specifically in neuronal ensembles contribute to the pathogenesis of addiction. Furthermore, a causal link between transcriptional and epigenetic modifications in neuronal ensembles and the development of the addictive state is proposed. Translational studies of molecular and cellular changes in neuronal ensembles that contribute to drug-seeking behavior, will allow the identification of molecular and circuit targets and interventions for substance use disorders.
Collapse
Affiliation(s)
- Xing Liu
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| | - Feifei Wang
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China.
| | - Qiumin Le
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| | - Lan Ma
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| |
Collapse
|
16
|
Phillips RA, Wan E, Tuscher JJ, Reid D, Drake OR, Ianov L, Day JJ. Temporally specific gene expression and chromatin remodeling programs regulate a conserved Pdyn enhancer. eLife 2023; 12:RP89993. [PMID: 37938195 PMCID: PMC10631760 DOI: 10.7554/elife.89993] [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] [Indexed: 11/09/2023] Open
Abstract
Neuronal and behavioral adaptations to novel stimuli are regulated by temporally dynamic waves of transcriptional activity, which shape neuronal function and guide enduring plasticity. Neuronal activation promotes expression of an immediate early gene (IEG) program comprised primarily of activity-dependent transcription factors, which are thought to regulate a second set of late response genes (LRGs). However, while the mechanisms governing IEG activation have been well studied, the molecular interplay between IEGs and LRGs remain poorly characterized. Here, we used transcriptomic and chromatin accessibility profiling to define activity-driven responses in rat striatal neurons. As expected, neuronal depolarization generated robust changes in gene expression, with early changes (1 hr) enriched for inducible transcription factors and later changes (4 hr) enriched for neuropeptides, synaptic proteins, and ion channels. Remarkably, while depolarization did not induce chromatin remodeling after 1 hr, we found broad increases in chromatin accessibility at thousands of sites in the genome at 4 hr after neuronal stimulation. These putative regulatory elements were found almost exclusively at non-coding regions of the genome, and harbored consensus motifs for numerous activity-dependent transcription factors such as AP-1. Furthermore, blocking protein synthesis prevented activity-dependent chromatin remodeling, suggesting that IEG proteins are required for this process. Targeted analysis of LRG loci identified a putative enhancer upstream of Pdyn (prodynorphin), a gene encoding an opioid neuropeptide implicated in motivated behavior and neuropsychiatric disease states. CRISPR-based functional assays demonstrated that this enhancer is both necessary and sufficient for Pdyn transcription. This regulatory element is also conserved at the human PDYN locus, where its activation is sufficient to drive PDYN transcription in human cells. These results suggest that IEGs participate in chromatin remodeling at enhancers and identify a conserved enhancer that may act as a therapeutic target for brain disorders involving dysregulation of Pdyn.
Collapse
Affiliation(s)
- Robert A Phillips
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
| | - Ethan Wan
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
| | - Jennifer J Tuscher
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
| | - David Reid
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
| | - Olivia R Drake
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
| | - Lara Ianov
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
- Civitan International Research Center, University of Alabama at BirminghamBirminghamUnited States
| | - Jeremy J Day
- Department of Neurobiology, University of Alabama at BirminghamBirminghamUnited States
| |
Collapse
|
17
|
Zhang YF, Wu J, Wang Y, Johnson NL, Bhattarai JP, Li G, Wang W, Guevara C, Shoenhard H, Fuccillo MV, Wesson DW, Ma M. Ventral striatal islands of Calleja neurons bidirectionally mediate depression-like behaviors in mice. Nat Commun 2023; 14:6887. [PMID: 37898623 PMCID: PMC10613228 DOI: 10.1038/s41467-023-42662-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 10/17/2023] [Indexed: 10/30/2023] Open
Abstract
The ventral striatum is a reward center implicated in the pathophysiology of depression. It contains islands of Calleja, clusters of dopamine D3 receptor-expressing granule cells, predominantly in the olfactory tubercle (OT). These OT D3 neurons regulate self-grooming, a repetitive behavior manifested in affective disorders. Here we show that chronic restraint stress (CRS) induces robust depression-like behaviors in mice and decreases excitability of OT D3 neurons. Ablation or inhibition of these neurons leads to depression-like behaviors, whereas their activation ameliorates CRS-induced depression-like behaviors. Moreover, activation of OT D3 neurons has a rewarding effect, which diminishes when grooming is blocked. Finally, we propose a model that explains how OT D3 neurons may influence dopamine release via synaptic connections with OT spiny projection neurons (SPNs) that project to midbrain dopamine neurons. Our study reveals a crucial role of OT D3 neurons in bidirectionally mediating depression-like behaviors, suggesting a potential therapeutic target.
Collapse
Affiliation(s)
- Yun-Feng Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, 100101, Beijing, China.
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| | - Jialiang Wu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Yingqi Wang
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Natalie L Johnson
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, 32610, USA
| | - Janardhan P Bhattarai
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Guanqing Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, 100101, Beijing, China
- College of Life Sciences, Hebei University, Baoding, 071002, Hebei, China
| | - Wenqiang Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, 100101, Beijing, China
- College of Life Sciences, Hebei University, Baoding, 071002, Hebei, China
| | - Camilo Guevara
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Hannah Shoenhard
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Marc V Fuccillo
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Daniel W Wesson
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, 32610, USA
| | - Minghong Ma
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
18
|
Gupta S, Guevara CA, Tielemans A, Huntley GW, Benson DL. Parkinson's-linked LRRK2-G2019S derails AMPAR trafficking, mobility and composition in striatum with cell-type and subunit specificity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562231. [PMID: 37905106 PMCID: PMC10614818 DOI: 10.1101/2023.10.13.562231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Parkinson's (PD) is a multi-factorial disease that affects multiple brain systems and circuits. While defined by motor symptoms caused by degeneration of brainstem dopamine neurons, debilitating non-motor abnormalities in fronto-striatal based cognitive function are common, appear early and are initially independent of dopamine. Young adult mice expressing the PD-associated G2019S missense mutation in Lrrk2 also exhibit deficits in fronto-striatal-based cognitive tasks. In mice and humans, cognitive functions require dynamic adjustments in glutamatergic synapse strength through cell-surface trafficking of AMPA-type glutamate receptors (AMPARs), but it is unknown how LRRK2 mutation impacts dynamic features of AMPAR trafficking in striatal projection neurons (SPNs). Here, we used Lrrk2 G2019S knockin mice to show that surface AMPAR subunit stoichiometry is altered biochemically and functionally in mutant SPNs to favor incorporation of GluA1 over GluA2. GluA1-containing AMPARs were resistant to internalization from the cell surface, leaving an excessive accumulation of GluA1 on the surface within and outside synapses. This negatively impacted trafficking dynamics that normally support synapse strengthening, as GluA1-containing AMPARs failed to increase at synapses in response to a potentiating stimulus and showed significantly reduced surface mobility. Surface GluA2-containing AMPARs were expressed at normal levels in synapses, indicating subunit-selective impairment. Abnormal surface accumulation of GluA1 was independent of PKA activity and was limited to D 1 R SPNs. Since LRRK2 mutation is thought to be part of a common PD pathogenic pathway, our data suggest that sustained, striatal cell-type specific changes in AMPAR composition and trafficking contribute to cognitive or other impairments associated with PD. SIGNIFICANCE STATEMENT Mutations in LRRK2 are common genetic risks for PD. Lrrk2 G2019S mice fail to exhibit long-term potentiation at corticostriatal synapses and show significant deficits in frontal-striatal based cognitive tasks. While LRRK2 has been implicated generally in protein trafficking, whether G2019S derails AMPAR trafficking at synapses on striatal neurons (SPNs) is unknown. We show that surface GluA1-AMPARs fail to internalize and instead accumulate excessively within and outside synapses. This effect is selective to D 1 R SPNs and negatively impacts synapse strengthening as GluA1-AMPARs fail to increase at the surface in response to potentiation and show limited surface mobility. Thus, LRRK2-G2019S narrows the effective range of plasticity mechanisms, supporting the idea that cognitive symptoms reflect an imbalance in AMPAR trafficking mechanisms within cell-type specific projections.
Collapse
|
19
|
Wang X, Sun S, Chen H, Yun B, Zhang Z, Wang X, Wu Y, Lv J, He Y, Li W, Chen L. Identification of key genes and therapeutic drugs for cocaine addiction using integrated bioinformatics analysis. Front Neurosci 2023; 17:1201897. [PMID: 37469839 PMCID: PMC10352680 DOI: 10.3389/fnins.2023.1201897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/05/2023] [Indexed: 07/21/2023] Open
Abstract
Introduction Cocaine is a highly addictive drug that is abused due to its excitatory effect on the central nervous system. It is critical to reveal the mechanisms of cocaine addiction and identify key genes that play an important role in addiction. Methods In this study, we proposed a centrality algorithm integration strategy to identify key genes in a protein-protein interaction (PPI) network constructed by deferential genes from cocaine addiction-related datasets. In order to investigate potential therapeutic drugs for cocaine addiction, a network of targeted relationships between nervous system drugs and key genes was established. Results Four key genes (JUN, FOS, EGR1, and IL6) were identified and well validated using CTD database correlation analysis, text mining, independent dataset analysis, and enrichment analysis methods, and they might serve as biomarkers of cocaine addiction. A total of seventeen drugs have been identified from the network of targeted relationships between nervous system drugs and key genes, of which five (disulfiram, cannabidiol, dextroamphetamine, diazepam, and melatonin) have been shown in the literature to play a role in the treatment of cocaine addiction. Discussion This study identified key genes and potential therapeutic drugs for cocaine addiction, which provided new ideas for the research of the mechanism of cocaine addiction.
Collapse
|
20
|
Phillips RA, Tuscher JJ, Fitzgerald ND, Wan E, Zipperly ME, Duke CG, Ianov L, Day JJ. Distinct subpopulations of D1 medium spiny neurons exhibit unique transcriptional responsiveness to cocaine. Mol Cell Neurosci 2023; 125:103849. [PMID: 36965548 PMCID: PMC10898607 DOI: 10.1016/j.mcn.2023.103849] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/13/2023] [Accepted: 03/18/2023] [Indexed: 03/27/2023] Open
Abstract
Drugs of abuse increase extracellular concentrations of dopamine in the nucleus accumbens (NAc), resulting in transcriptional alterations that drive long-lasting cellular and behavioral adaptations. While decades of research have focused on the transcriptional mechanisms by which drugs of abuse influence neuronal physiology and function, few studies have comprehensively defined NAc cell type heterogeneity in transcriptional responses to drugs of abuse. Here, we used single nucleus RNA-seq (snRNA-seq) to characterize the transcriptome of over 39,000 NAc cells from male and female adult Sprague-Dawley rats following acute or repeated cocaine experience. This dataset identified 16 transcriptionally distinct cell populations, including two populations of medium spiny neurons (MSNs) that express the Drd1 dopamine receptor (D1-MSNs). Critically, while both populations expressed classic marker genes of D1-MSNs, only one population exhibited a robust transcriptional response to cocaine. Validation of population-selective transcripts using RNA in situ hybridization revealed distinct spatial compartmentalization of these D1-MSN populations within the NAc. Finally, analysis of published NAc snRNA-seq datasets from non-human primates and humans demonstrated conservation of MSN subtypes across rat and higher order mammals, and further highlighted cell type-specific transcriptional differences across the NAc and broader striatum. These results highlight the utility in using snRNA-seq to characterize both cell type heterogeneity and cell type-specific responses to cocaine and provides a useful resource for cross-species comparisons of NAc cell composition.
Collapse
Affiliation(s)
- Robert A Phillips
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J Tuscher
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - N Dalton Fitzgerald
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ethan Wan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan E Zipperly
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Corey G Duke
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremy J Day
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
21
|
Wang Z, Gu C, Qiu P. Editorial: Neural pathophysiological mechanisms of neuropsychiatric disorders underlying substance abuse. Front Cell Neurosci 2023; 17:1194734. [PMID: 37180948 PMCID: PMC10167276 DOI: 10.3389/fncel.2023.1194734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 05/16/2023] Open
Affiliation(s)
- Zhuo Wang
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Cihang Gu
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Pingming Qiu
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Phillips RA, Tuscher JJ, Fitzgerald ND, Wan E, Zipperly ME, Duke CG, Ianov L, Day JJ. Distinct subpopulations of D1 medium spiny neurons exhibit unique transcriptional responsiveness to cocaine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523845. [PMID: 36711527 PMCID: PMC9882178 DOI: 10.1101/2023.01.12.523845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Drugs of abuse increase extracellular concentrations of dopamine in the nucleus accumbens (NAc), resulting in transcriptional alterations that drive long-lasting cellular and behavioral adaptations. While decades of research have focused on the transcriptional mechanisms by which drugs of abuse influence neuronal physiology and function, few studies have comprehensively defined NAc cell type heterogeneity in transcriptional responses to drugs of abuse. Here, we used single nucleus RNA-seq (snRNA-seq) to characterize the transcriptome of over 39,000 NAc cells from male and female adult Sprague-Dawley rats following acute or repeated cocaine experience. This dataset identified 16 transcriptionally distinct cell populations, including two populations of medium spiny neurons (MSNs) that express the Drd1 dopamine receptor (D1-MSNs). Critically, while both populations expressed classic marker genes of D1-MSNs, only one population exhibited a robust transcriptional response to cocaine. Validation of population-selective transcripts using RNA in situ hybridization revealed distinct spatial compartmentalization of these D1-MSN populations within the NAc. Finally, analysis of published NAc snRNA-seq datasets from non-human primates and humans demonstrated conservation of MSN subtypes across rat and higher order mammals, and further highlighted cell type-specific transcriptional differences across the NAc and broader striatum. These results highlight the utility in using snRNA-seq to characterize both cell type heterogeneity and cell type-specific responses to cocaine and provides a useful resource for cross-species comparisons of NAc cell composition.
Collapse
Affiliation(s)
- Robert A. Phillips
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J. Tuscher
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - N. Dalton Fitzgerald
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ethan Wan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan E. Zipperly
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Corey G. Duke
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremy J. Day
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
23
|
Kunder N, de la Peña JB, Lou TF, Chase R, Suresh P, Lawson J, Shukla T, Black B, Campbell ZT. The RNA-Binding Protein HuR Is Integral to the Function of Nociceptors in Mice and Humans. J Neurosci 2022; 42:9129-9141. [PMID: 36270801 PMCID: PMC9761683 DOI: 10.1523/jneurosci.1630-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
HuR is an RNA-binding protein implicated in RNA processing, stability, and translation. Previously, we examined protein synthesis in dorsal root ganglion (DRG) neurons treated with inflammatory mediators using ribosome profiling. We found that the HuR consensus binding element was enriched in transcripts with elevated translation. HuR is expressed in the soma of nociceptors and their axons. Pharmacologic inhibition of HuR with the small molecule CMLD-2 reduced the activity of mouse and human sensory neurons. Peripheral administration of CMLD-2 in the paw or genetic elimination of HuR from sensory neurons diminished behavioral responses associated with NGF- and IL-6-induced allodynia in male and female mice. Genetic disruption of HuR altered the proximity of mRNA decay factors near a key neurotrophic factor (TrkA). Collectively, the data suggest that HuR is required for local control of mRNA stability and reveals a new biological function for a broadly conserved post-transcriptional regulatory factor.SIGNIFICANCE STATEMENT Nociceptors undergo long-lived changes in excitability, which may contribute to chronic pain. Noxious cues that promote pain lead to rapid induction of protein synthesis. The underlying mechanisms that confer specificity to mRNA control in nociceptors are unclear. Here, we identify a conserved RNA-binding protein called HuR as a key regulatory factor in sensory neurons. Using a combination of genetics and pharmacology, we demonstrate that HuR is required for signaling in nociceptors. In doing so, we report an important mechanism of mRNA control in sensory neurons that ensures appropriate nociceptive responses to inflammatory mediators.
Collapse
Affiliation(s)
- Nikesh Kunder
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - June Bryan de la Peña
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53792
| | - Tzu-Fang Lou
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Rebecca Chase
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Prarthana Suresh
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Jennifer Lawson
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts 01854
| | - Tarjani Shukla
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53792
| | - Bryan Black
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts 01854
| | - Zachary T Campbell
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53792
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53792
| |
Collapse
|
24
|
Abstract
Motherhood goes through preparation, onset and maintenance phases until the natural weaning. A variety of changes in hormonal/neurohormonal systems and brain circuits are involved in the maternal behavior. Hormones, neuropeptides, and neurotransmitters involved in maternal behavior act via G-protein-coupled receptors, many of which in turn activate plasma membrane enzymes including phospholipase C (PLC) β isoforms. In this study, we examined the effect of PLCβ1 knockout (KO) on maternal behavior. There was little difference between PLCβ1-KO and wild-type (WT) dams in the relative time spent in maternal behavior during the period between 24 h prepartum and 12 h postpartum (-24 h ∼ PPH 12). After PPH 18, however, PLCβ1-KO dams neglected their pups so that they all died in 2-3 days. In the pup retrieval test, latency was not different during the period within PPH 12, but after PPH 18, PLCβ1-KO dams could not finish pup retrieval in a given time. During both periods, FosB expression in the nucleus accumbens (NAcc) of PLCβ1-KO dams was significantly lower than WT, but not different in the medial preoptic area (mPOA). Given that mPOA activity is required for initiation of maternal behavior, and that NAcc is known to be involved in maternal motivation and maintenance of maternal behavior, our results suggest that PLCβ1 signaling is essential for transition from the onset to maintenance phase of maternal behavior.
Collapse
Affiliation(s)
- Hea-jin Kim
- Brain Science Institute (BSI), Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Jaewon Jang
- Brain Science Institute (BSI), Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea,Department of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Hae-Young Koh
- Brain Science Institute (BSI), Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea, Hae-Young Koh Brain Science Institute (BSI), Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| |
Collapse
|
25
|
Onimus O, Valjent E, Fisone G, Gangarossa G. Haloperidol-Induced Immediate Early Genes in Striatopallidal Neurons Requires the Converging Action of cAMP/PKA/DARPP-32 and mTOR Pathways. Int J Mol Sci 2022; 23:ijms231911637. [PMID: 36232936 PMCID: PMC9569967 DOI: 10.3390/ijms231911637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022] Open
Abstract
Antipsychotics share the common pharmacological feature of antagonizing the dopamine 2 receptor (D2R), which is abundant in the striatum and involved in both the therapeutic and side effects of this drug’s class. The pharmacological blockade of striatal D2R, by disinhibiting the D2R-containing medium-sized spiny neurons (MSNs), leads to a plethora of molecular, cellular and behavioral adaptations, which are central in the action of antipsychotics. Here, we focused on the cell type-specific (D2R-MSNs) regulation of some striatal immediate early genes (IEGs), such as cFos, Arc and Zif268. Taking advantage of transgenic mouse models, pharmacological approaches and immunofluorescence analyses, we found that haloperidol-induced IEGs in the striatum required the synergistic activation of A2a (adenosine) and NMDA (glutamate) receptors. At the intracellular signaling level, we found that the PKA/DARPP-32 and mTOR pathways synergistically cooperate to control the induction of IEGs by haloperidol. By confirming and further expanding previous observations, our results provide novel insights into the regulatory mechanisms underlying the molecular/cellular action of antipsychotics in the striatum.
Collapse
Affiliation(s)
- Oriane Onimus
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Emmanuel Valjent
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, Inserm, 34094 Montpellier, France
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Giuseppe Gangarossa
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
- Correspondence:
| |
Collapse
|
26
|
Atehortua Martinez LA, Curis E, Mekdad N, Larrieu C, Courtin C, Jourdren L, Blugeon C, Laplanche JL, Megarbane B, Marie-Claire C, Benturquia N. Individual differences in cocaine-induced conditioned place preference in male rats: Behavioral and transcriptomic evidence. J Psychopharmacol 2022; 36:1161-1175. [PMID: 36121009 PMCID: PMC9548661 DOI: 10.1177/02698811221123047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Substance use disorder emerges in a small proportion of drug users and has the characteristics of a chronic relapsing pathology. AIMS Our study aimed to demonstrate and characterize the variability in the expression of the rewarding effects of cocaine in the conditioned place preference (CPP) paradigm. METHODS A cocaine-CPP paradigm in male Sprague-Dawley rats with an extinction period of 12 days and reinstatement was conducted. A statistical model was developed to distinguish rats expressing or not a cocaine-induced place preference. RESULTS Two groups of rats were identified: rats that did express rewarding effects (CPP expression (CPPE), score >102 s) and rats that did not (no CPP expression (nCPPE), score between -85 and 59 s). These two groups did not show significant differences in a battery of behavioral tests. To identify differentially expressed genes in the CPPE and nCPPE groups, a whole-transcriptome ribonucleic acid-sequencing analysis was performed in the nucleus accumbens (NAc) 24 h after the CPP test. Four immediate early genes (Fos, Egr2, Nr4a1, and Zbtb37) were differentially expressed in the NAc of CPPE rats after expression of CPP. Variability in cocaine-induced place preference persisted in the CPPE and nCPPE groups after the extinction and reinstatement phases. Transcriptomic differences observed after reinstatement were distinct from those observed immediately after expression of CPP. CONCLUSION These new findings provide insights into the identification of mechanisms underlying interindividual variability in the response to cocaine's rewarding effects.
Collapse
Affiliation(s)
- Luisa Alessandra Atehortua Martinez
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Emmanuel Curis
- UR 7537 BioSTM, Université Paris Cité, Paris, France
- Laboratoire d’Hématologie, Hôpital Lariboisière, APHP, Paris, France
| | - Nawel Mekdad
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Claire Larrieu
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Cindie Courtin
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Laurent Jourdren
- Genomic Facility, Institut de Biologie de l’École Normale Supérieure, École Normale Supérieure, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, PSL Research University, Paris, France
| | - Corinne Blugeon
- Genomic Facility, Institut de Biologie de l’École Normale Supérieure, École Normale Supérieure, Centre National pour la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, PSL Research University, Paris, France
| | - Jean-Louis Laplanche
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Bruno Megarbane
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Cynthia Marie-Claire
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| | - Nadia Benturquia
- Institut National de la Santé et de la Recherche Médicale UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Paris, France
| |
Collapse
|
27
|
Wang S, Li M, Su L, Wang Y, Ma D, Wang H, Zhu J, Chen T. Knockout of Dopamine D3 Receptor Gene Blocked Methamphetamine-Induced Distinct Changes of Dopaminergic and Glutamatergic Synapse in the Nucleus Accumbens Shell of Mice. Front Cell Neurosci 2022; 16:893190. [PMID: 35711471 PMCID: PMC9195588 DOI: 10.3389/fncel.2022.893190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/03/2022] [Indexed: 12/15/2022] Open
Abstract
Structural plasticity changes in the brain are thought to underlie, at least partially, drug-induced persistent changes in behavior. Our previous study reported that increased synaptic density in the nucleus accumbens shell (NAcsh) correlates with and may contribute to behavioral sensitization induced by methamphetamine (METH). However, the distinct changes of dopaminergic and glutamatergic synapses and the modulating effects of dopamine D3 receptor remain unclear. In the current study, we used immunohistochemistry electron-microscopy and immunofluorescence to detect the changes of dopamine D1, D2, and glutamate NR2B-positive synapses and cells in the NAcsh of METH-sensitized wild type (WT) and knockout of dopamine D3 receptor gene (D3–/–) mice. We found that METH induced long-term behavioral sensitization in WT mice, which was accompanied by an increased number and rate of dopamine D1 receptor-positive synapses and cells, as well as glutamate NR2B-positive synapses and cells. In contrast, the number and rate of dopamine D2 receptor-positive synapses and cells were significantly decreased in the NAcsh of METH-sensitized WT mice. D3–/– mice exhibited attenuated acute locomotor responses and behavioral sensitization to METH compared with WT mice. Moreover, the knockout of dopamine D3 receptor gene inhibited METH-induced changes of dopaminergic and glutamatergic synapses in the NAcsh of METH-sensitized mice. Taken together, our results suggest that METH induced distinct changes of dopaminergic and glutamatergic synapses and cells in the NAcsh of mice, which was blocked by the knockout of dopamine D3 receptor gene, and may contribute to, at least partially, METH-induced behavior sensitization as well as the modulating effect of the dopamine D3 receptor.
Collapse
Affiliation(s)
- Shuai Wang
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an, China
| | - Ming Li
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an, China
| | - Linlan Su
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an, China
| | - Yu Wang
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an, China
| | - Dongliang Ma
- Programme in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Hongyan Wang
- Programme in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jie Zhu
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Jie Zhu,
| | - Teng Chen
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an, China
- Teng Chen,
| |
Collapse
|
28
|
Puri NM, Romano GR, Lin TY, Mai QN, Irannejad R. The organic cation Transporter 2 regulates dopamine D1 receptor signaling at the Golgi apparatus. eLife 2022; 11:75468. [PMID: 35467530 PMCID: PMC9098220 DOI: 10.7554/elife.75468] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Dopamine is a key catecholamine in the brain and kidney, where it is involved in a number of physiological functions such as locomotion, cognition, emotion, endocrine regulation, and renal function. As a membrane-impermeant hormone and neurotransmitter, dopamine is thought to signal by binding and activating dopamine receptors, members of the G protein coupled receptor (GPCR) family, only on the plasma membrane. Here, using novel nanobody-based biosensors, we demonstrate for the first time that the dopamine D1 receptor (D1DR), the primary mediator of dopaminergic signaling in the brain and kidney, not only functions on the plasma membrane but becomes activated at the Golgi apparatus in the presence of its ligand. We present evidence that activation of the Golgi pool of D1DR is dependent on organic cation transporter 2 (OCT2), a dopamine transporter, providing an explanation for how the membrane-impermeant dopamine accesses subcellular pools of D1DR. We further demonstrate that dopamine activates Golgi-D1DR in murine striatal medium spiny neurons, and this activity depends on OCT2 function. We also introduce a new approach to selectively interrogate compartmentalized D1DR signaling by inhibiting Gαs coupling using a nanobody-based chemical recruitment system. Using this strategy, we show that Golgi-localized D1DRs regulate cAMP production and mediate local protein kinase A activation. Together, our data suggest that spatially compartmentalized signaling hubs are previously unappreciated regulatory aspects of D1DR signaling. Our data provide further evidence for the role of transporters in regulating subcellular GPCR activity.
Collapse
Affiliation(s)
- Natasha M Puri
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Giovanna R Romano
- Biochemistry Department, Weill Cornell Medicine, New York, United States
| | - Ting-Yu Lin
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Quynh N Mai
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Roshanak Irannejad
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
29
|
SWI/SNF chromatin remodeler complex within the reward pathway is required for behavioral adaptations to stress. Nat Commun 2022; 13:1807. [PMID: 35379786 PMCID: PMC8980038 DOI: 10.1038/s41467-022-29380-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 02/22/2022] [Indexed: 01/01/2023] Open
Abstract
Enduring behavioral changes upon stress exposure involve changes in gene expression sustained by epigenetic modifications in brain circuits, including the mesocorticolimbic pathway. Brahma (BRM) and Brahma Related Gene 1 (BRG1) are ATPase subunits of the SWI/SNF complexes involved in chromatin remodeling, a process essential to enduring plastic changes in gene expression. Here, we show that in mice, social defeat induces changes in BRG1 nuclear distribution. The inactivation of the Brg1/Smarca4 gene within dopamine-innervated regions or the constitutive inactivation of the Brm/Smarca2 gene leads to resilience to repeated social defeat and decreases the behavioral responses to cocaine without impacting midbrain dopamine neurons activity. Within striatal medium spiny neurons, Brg1 gene inactivation reduces the expression of stress- and cocaine-induced immediate early genes, increases levels of heterochromatin and at a global scale decreases chromatin accessibility. Altogether these data demonstrate the pivotal function of SWI/SNF complexes in behavioral and transcriptional adaptations to salient environmental challenges. Repeated exposure to social stressors in rodents results in behavioural changes. Here the authors show that behavioural adaptations to stress are associated with nuclear organization changes through SWI/SNF chromatin remodeler in specific neuronal populations of the mesolimbic system.
Collapse
|
30
|
Grimm C, Frässle S, Steger C, von Ziegler L, Sturman O, Shemesh N, Peleg-Raibstein D, Burdakov D, Bohacek J, Stephan KE, Razansky D, Wenderoth N, Zerbi V. Optogenetic activation of striatal D1R and D2R cells differentially engages downstream connected areas beyond the basal ganglia. Cell Rep 2021; 37:110161. [PMID: 34965430 DOI: 10.1016/j.celrep.2021.110161] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 10/20/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
The basal ganglia (BG) are a group of subcortical nuclei responsible for motor and executive function. Central to BG function are striatal cells expressing D1 (D1R) and D2 (D2R) dopamine receptors. D1R and D2R cells are considered functional antagonists that facilitate voluntary movements and inhibit competing motor patterns, respectively. However, whether they maintain a uniform function across the striatum and what influence they exert outside the BG is unclear. Here, we address these questions by combining optogenetic activation of D1R and D2R cells in the mouse ventrolateral caudoputamen with fMRI. Striatal D1R/D2R stimulation evokes distinct activity within the BG-thalamocortical network and differentially engages cerebellar and prefrontal regions. Computational modeling of effective connectivity confirms that changes in D1R/D2R output drive functional relationships between these regions. Our results suggest a complex functional organization of striatal D1R/D2R cells and hint toward an interconnected fronto-BG-cerebellar network modulated by striatal D1R and D2R cells.
Collapse
Affiliation(s)
- Christina Grimm
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland; Neuroscience Center Zurich, ETH Zürich and University of Zurich, Zürich, Switzerland
| | - Stefan Frässle
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zürich and ETH Zürich, Zürich, Switzerland
| | - Céline Steger
- Neuroscience Center Zurich, ETH Zürich and University of Zurich, Zürich, Switzerland; Center for MR Research, University Children's Hospital Zurich, Zürich, Switzerland
| | - Lukas von Ziegler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland; Neuroscience Center Zurich, ETH Zürich and University of Zurich, Zürich, Switzerland
| | - Oliver Sturman
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland; Neuroscience Center Zurich, ETH Zürich and University of Zurich, Zürich, Switzerland
| | - Noam Shemesh
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Daria Peleg-Raibstein
- Laboratory of Neurobehavioral Dynamics, Department of Health Sciences and Technology, Institute for Neuroscience, ETH Zürich, Zürich, Switzerland; Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zürich, Switzerland
| | - Denis Burdakov
- Laboratory of Neurobehavioral Dynamics, Department of Health Sciences and Technology, Institute for Neuroscience, ETH Zürich, Zürich, Switzerland; Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zürich, Switzerland; Neuroscience Center Zurich, ETH Zürich and University of Zurich, Zürich, Switzerland
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland; Neuroscience Center Zurich, ETH Zürich and University of Zurich, Zürich, Switzerland
| | - Klaas Enno Stephan
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zürich and ETH Zürich, Zürich, Switzerland
| | - Daniel Razansky
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zürich, Zürich, Switzerland; Institute of Biological and Medical Imaging (IBMI), Technical University of Munich and Helmholtz Center Munich, Munich, Germany; Neuroscience Center Zurich, ETH Zürich and University of Zurich, Zürich, Switzerland
| | - Nicole Wenderoth
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland; Neuroscience Center Zurich, ETH Zürich and University of Zurich, Zürich, Switzerland
| | - Valerio Zerbi
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland; Neuroscience Center Zurich, ETH Zürich and University of Zurich, Zürich, Switzerland.
| |
Collapse
|
31
|
Lissek T, Andrianarivelo A, Saint‐Jour E, Allichon M, Bauersachs HG, Nassar M, Piette C, Pruunsild P, Tan Y, Forget B, Heck N, Caboche J, Venance L, Vanhoutte P, Bading H. Npas4 regulates medium spiny neuron physiology and gates cocaine-induced hyperlocomotion. EMBO Rep 2021; 22:e51882. [PMID: 34661342 PMCID: PMC8647009 DOI: 10.15252/embr.202051882] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 09/11/2021] [Accepted: 09/22/2021] [Indexed: 12/01/2022] Open
Abstract
We show here that the transcription factor Npas4 is an important regulator of medium spiny neuron spine density and electrophysiological parameters and that it determines the magnitude of cocaine-induced hyperlocomotion in mice. Npas4 is induced by synaptic stimuli that cause calcium influx, but not dopaminergic or PKA-stimulating input, in mouse medium spiny neurons and human iPSC-derived forebrain organoids. This induction is independent of ubiquitous kinase pathways such as PKA and MAPK cascades, and instead depends on calcineurin and nuclear calcium signalling. Npas4 controls a large regulon containing transcripts for synaptic molecules, such as NMDA receptors and VDCC subunits, and determines in vivo MSN spine density, firing rate, I/O gain function and paired-pulse facilitation. These functions at the molecular and cellular levels control the locomotor response to drugs of abuse, as Npas4 knockdown in the nucleus accumbens decreases hyperlocomotion in response to cocaine in male mice while leaving basal locomotor behaviour unchanged.
Collapse
Affiliation(s)
- Thomas Lissek
- Interdisciplinary Center for NeurosciencesDepartment of NeurobiologyHeidelberg UniversityHeidelbergGermany
| | - Andry Andrianarivelo
- INSERM, UMR‐S 1130Neuroscience Paris SeineInstitute of Biology Paris SeineParisFrance
- CNRSUMR 8246Neuroscience Paris SeineParisFrance
- Sorbonne UniversitéUPMC Université Paris 06UM CR18Neuroscience Paris SeineParisFrance
| | - Estefani Saint‐Jour
- INSERM, UMR‐S 1130Neuroscience Paris SeineInstitute of Biology Paris SeineParisFrance
- CNRSUMR 8246Neuroscience Paris SeineParisFrance
- Sorbonne UniversitéUPMC Université Paris 06UM CR18Neuroscience Paris SeineParisFrance
| | - Marie‐Charlotte Allichon
- INSERM, UMR‐S 1130Neuroscience Paris SeineInstitute of Biology Paris SeineParisFrance
- CNRSUMR 8246Neuroscience Paris SeineParisFrance
- Sorbonne UniversitéUPMC Université Paris 06UM CR18Neuroscience Paris SeineParisFrance
| | - Hanke Gwendolyn Bauersachs
- Interdisciplinary Center for NeurosciencesDepartment of NeurobiologyHeidelberg UniversityHeidelbergGermany
| | - Merie Nassar
- Center for Interdisciplinary Research in Biology (CIRB)College de FranceCNRS UMR7241INSERM U1050Université PSLParisFrance
| | - Charlotte Piette
- Center for Interdisciplinary Research in Biology (CIRB)College de FranceCNRS UMR7241INSERM U1050Université PSLParisFrance
| | - Priit Pruunsild
- Interdisciplinary Center for NeurosciencesDepartment of NeurobiologyHeidelberg UniversityHeidelbergGermany
| | - Yan‐Wei Tan
- Interdisciplinary Center for NeurosciencesDepartment of NeurobiologyHeidelberg UniversityHeidelbergGermany
| | - Benoit Forget
- INSERM, UMR‐S 1130Neuroscience Paris SeineInstitute of Biology Paris SeineParisFrance
- CNRSUMR 8246Neuroscience Paris SeineParisFrance
- Sorbonne UniversitéUPMC Université Paris 06UM CR18Neuroscience Paris SeineParisFrance
| | - Nicolas Heck
- INSERM, UMR‐S 1130Neuroscience Paris SeineInstitute of Biology Paris SeineParisFrance
- CNRSUMR 8246Neuroscience Paris SeineParisFrance
- Sorbonne UniversitéUPMC Université Paris 06UM CR18Neuroscience Paris SeineParisFrance
| | - Jocelyne Caboche
- INSERM, UMR‐S 1130Neuroscience Paris SeineInstitute of Biology Paris SeineParisFrance
- CNRSUMR 8246Neuroscience Paris SeineParisFrance
- Sorbonne UniversitéUPMC Université Paris 06UM CR18Neuroscience Paris SeineParisFrance
| | - Laurent Venance
- Center for Interdisciplinary Research in Biology (CIRB)College de FranceCNRS UMR7241INSERM U1050Université PSLParisFrance
| | - Peter Vanhoutte
- INSERM, UMR‐S 1130Neuroscience Paris SeineInstitute of Biology Paris SeineParisFrance
- CNRSUMR 8246Neuroscience Paris SeineParisFrance
- Sorbonne UniversitéUPMC Université Paris 06UM CR18Neuroscience Paris SeineParisFrance
| | - Hilmar Bading
- Interdisciplinary Center for NeurosciencesDepartment of NeurobiologyHeidelberg UniversityHeidelbergGermany
| |
Collapse
|
32
|
Kim HJ, Koh HY. Training-Dependent Change in Content of Association in Appetitive Pavlovian Conditioning. Front Behav Neurosci 2021; 15:750131. [PMID: 34899203 PMCID: PMC8656303 DOI: 10.3389/fnbeh.2021.750131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/11/2021] [Indexed: 12/01/2022] Open
Abstract
In appetitive Pavlovian conditioning, experience with a conditional relationship between a cue [conditioned stimulus (CS)] and a reward [unconditioned stimulus (US)] bestows CS with the ability to promote adaptive behavior patterns. Different features of US (e.g., identity-specific sensory, general motivational) can be encoded by CS based on the nature of the CS-US relationship experienced (e.g., temporal factors such as training amount) and the content of association may determine the influence of CS over behavior (e.g., mediated learning, conditioned reinforcement). The content of association changed with varying conditioning factors, thereby altering behavioral consequences, however, has never been addressed in relevant brain signals evoked by CS. Our previous study found that phospholipase C β1-knockout (PLCβ1-KO) mice display persistent mediated learning over the extended course of odor-sugar conditioning, and that wild-type (WT) mice lose mediated learning sensitivity after extended training. In this study, in order to see whether this behavioral difference between these two genotypes comes from a difference in the course of association content, we examined whether odor CS can evoke the taste sensory representation of an absent sugar US after minimal- and extended training in these mice. In contrast to WT, which lost CS-evoked neural activation (c-Fos expression) in the gustatory cortex after extended training, KO mice displayed persistent association with the sensory feature of sugar, suggesting that sensory encoding is reliably linked to mediated learning sensitivity and there is a training-dependent change in the content of association in WT. PLCβ1 knockdown in the left medial prefrontal cortex (mPFC) resulted in mediated learning sensitivity and CS-evoked gustatory cortical activation after extended training, proposing a molecular component of the neural system underlying this Pavlovian conditioning process. We also discuss how disruption of this process is implicated for hallucination-like behaviors (impaired reality testing).
Collapse
Affiliation(s)
- Hea-jin Kim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, South Korea
| | - Hae-Young Koh
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, South Korea
| |
Collapse
|
33
|
Campos-Ordoñez T, Alcalá E, Ibarra-Castañeda N, Buriticá J, González-Pérez Ó. Chronic exposure to cyclohexane induces stereotypic circling, hyperlocomotion, and anxiety-like behavior associated with atypical c-Fos expression in motor- and anxiety-related brain regions. Behav Brain Res 2021; 418:113664. [PMID: 34780858 DOI: 10.1016/j.bbr.2021.113664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/29/2021] [Accepted: 11/07/2021] [Indexed: 12/27/2022]
Abstract
Recreational abuse of solvents continues, despite cyclohexane (CHX) is used as a safe replacement in gasoline or adhesive formulations. Increasing evidence indicates that CHX inhalation affects brain functioning; however, scanty information is available about its effects on behavior and brain activity upon drug removal. In this study, we used CD1 adult mice to mimic an intoxication period of recreational drugs for 30 days. During the CHX exposure (~30,000 ppm), we analyzed exploratory and biphasic behaviors, stereotypic circling, and locomotion. After CHX removal (24 h or a month later), we assessed anxiety-like behaviors and quantified c-Fos cells in motor- and anxiety-related brain regions. Our findings indicate that the repeated inhalation of CHX produced steady hyperactivity and reduced ataxia, sedation, and seizures as the exposure to CHX progressed. Also, CHX decreased grooming and rearing behaviors. In the first week of CHX inhalation, a stereotypic circling behavior emerged, and locomotion increased gradually. One month after CHX withdrawal, mice showed low activity in the center zone of the open field and more buried marbles. Twenty-four hours after CHX removal, c-Fos expression was low in the dorsal striatum, ventral striatum, motor cortex, dorsomedial prefrontal cortex, basolateral amygdala, lateral hypothalamus, and ventral hippocampus. One month later, c-Fos expression remained low in the ventral striatum and lateral hypothalamus but increased in the dorsomedial prefrontal cortex and primary motor cortex. This study provides a comprehensive behavioral characterization and novel histological evidence of the CHX effects on the brain when is administered in a recreational-like mode.
Collapse
Affiliation(s)
- Tania Campos-Ordoñez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima, Mexico; Centro de Estudios e Investigaciones en Comportamiento, University of Guadalajara, Jalisco, Mexico.
| | - Emmanuel Alcalá
- Centro de Estudios e Investigaciones en Comportamiento, University of Guadalajara, Jalisco, Mexico; Research Laboratory on Optimal Design, Devices and Advanced Materials, Department of Mathematics and Physics, ITESO, Jalisco, Mexico
| | - Nereida Ibarra-Castañeda
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima, Mexico; Medical Science PhD Program, School of Medicine, University of Colima, Colima, Mexico
| | - Jonathan Buriticá
- Centro de Estudios e Investigaciones en Comportamiento, University of Guadalajara, Jalisco, Mexico
| | - Óscar González-Pérez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima, Mexico.
| |
Collapse
|
34
|
Nakamura Y, Longueville S, Nishi A, Hervé D, Girault JA, Nakamura Y. Dopamine D1 receptor-expressing neurons activity is essential for locomotor and sensitizing effects of a single injection of cocaine. Eur J Neurosci 2021; 54:5327-5340. [PMID: 34273137 DOI: 10.1111/ejn.15394] [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: 12/20/2020] [Revised: 07/05/2021] [Accepted: 07/13/2021] [Indexed: 11/29/2022]
Abstract
Dopamine D1 receptors play an important role in the effects of cocaine. Here, we investigated the role of neurons which express these receptors (D1-neurons) in the acute locomotor effects of cocaine and the locomotor sensitization observed after a second injection of this drug, using the previously established two-injection protocol of sensitization. We inhibited D1-neurons using double transgenic mice conditionally expressing the inhibitory Gi-coupled designer receptor exclusively activated by designer drugs (Gi-DREADD) in D1-neurons. Chemogenetic inhibition of D1-neurons by a low dose of clozapine (0.1 mg/kg) decreased the cocaine-induced expression of Fos in striatal neurons. It diminished the basal locomotor activity and acute hyper-locomotion induced by cocaine (20 mg/kg). Clozapine 0.1 mg/kg had no effect by itself and did not alter cocaine effects in wild-type mice. Inhibition of D1-neurons during the first cocaine administration prevented the sensitization of the locomotor response in response to a second cocaine administration 10 days later. On Day 11, inhibition of D1-neurons by clozapine stimulation of Gi-DREADD blocked cocaine-induced locomotion including in sensitized mice, whereas on Day 12, in the absence of clozapine and D1-neurons inhibition, all mice displayed a sensitized response to cocaine. These results show that chemogenetic inhibition of D1-neurons decreases spontaneous and cocaine-induced locomotor activity. It prevents sensitization induction and blocks sensitized locomotion in a two-injection protocol of sensitization but does not reverse established sensitization. Our study further supports the central role of D1-neurons in mediating the acute locomotor effects of cocaine and its sensitization.
Collapse
Affiliation(s)
- Yukari Nakamura
- INSERM UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne University, Paris, France.,Institut du Fer à Moulin, Paris, France.,Department of Pharmacology, Kurume University School of Medicine, Kurume, Japan
| | - Sophie Longueville
- INSERM UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne University, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Akinori Nishi
- Department of Pharmacology, Kurume University School of Medicine, Kurume, Japan
| | - Denis Hervé
- INSERM UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne University, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Jean-Antoine Girault
- INSERM UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne University, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Yuki Nakamura
- INSERM UMR-S 1270, Paris, France.,Faculty of Sciences and Engineering, Sorbonne University, Paris, France.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
35
|
Carullo NVN, Hinds JE, Revanna JS, Tuscher JJ, Bauman AJ, Day JJ. A Cre-Dependent CRISPR/dCas9 System for Gene Expression Regulation in Neurons. eNeuro 2021; 8:ENEURO.0188-21.2021. [PMID: 34321217 PMCID: PMC8376295 DOI: 10.1523/eneuro.0188-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/02/2021] [Accepted: 07/09/2021] [Indexed: 01/02/2023] Open
Abstract
Site-specific genetic and epigenetic targeting of distinct cell populations is a central goal in molecular neuroscience and is crucial to understand the gene regulatory mechanisms that underlie complex phenotypes and behaviors. While recent technological advances have enabled unprecedented control over gene expression, many of these approaches are focused on selected model organisms and/or require labor-intensive customization for different applications. The simplicity and modularity of clustered regularly interspaced short palindromic repeats (CRISPR)-based systems have transformed genome editing and expanded the gene regulatory toolbox. However, there are few available tools for cell-selective CRISPR regulation in neurons. We designed, validated, and optimized CRISPR activation (CRISPRa) and CRISPR interference (CRISPRi) systems for Cre recombinase-dependent gene regulation. Unexpectedly, CRISPRa systems based on a traditional double-floxed inverted open reading frame (DIO) strategy exhibited leaky target gene induction even without Cre. Therefore, we developed an intron-containing Cre-dependent CRISPRa system (SVI-DIO-dCas9-VPR) that alleviated leaky gene induction and outperformed the traditional DIO system at endogenous genes in HEK293T cells and rat primary neuron cultures. Using gene-specific CRISPR sgRNAs, we demonstrate that SVI-DIO-dCas9-VPR can activate numerous rat or human genes (GRM2, Tent5b, Fos, Sstr2, and Gadd45b) in a Cre-specific manner. To illustrate the versatility of this tool, we created a parallel CRISPRi construct that successfully inhibited expression from a luciferase reporter in HEK293T cells only in the presence of Cre. These results provide a robust framework for Cre-dependent CRISPR-dCas9 approaches across different model systems, and enable cell-specific targeting when combined with common Cre driver lines or Cre delivery via viral vectors.
Collapse
Affiliation(s)
- Nancy V N Carullo
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jenna E Hinds
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jasmin S Revanna
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jennifer J Tuscher
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Allison J Bauman
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jeremy J Day
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
36
|
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
|
37
|
Han Y, Xia G, He Y, He Y, Farias M, Xu Y, Wu Q. A hindbrain dopaminergic neural circuit prevents weight gain by reinforcing food satiation. SCIENCE ADVANCES 2021; 7:eabf8719. [PMID: 34039606 DOI: 10.1126/sciadv.abf8719] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/06/2021] [Indexed: 06/12/2023]
Abstract
The neural circuitry mechanism that underlies dopaminergic (DA) control of innate feeding behavior is largely uncharacterized. Here, we identified a subpopulation of DA neurons situated in the caudal ventral tegmental area (cVTA) directly innervating DRD1-expressing neurons within the lateral parabrachial nucleus (LPBN). This neural circuit potently suppresses food intake via enhanced satiation response. Notably, this cohort of DAcVTA neurons is activated immediately before the cessation of each feeding bout. Acute inhibition of these DA neurons before bout termination substantially suppresses satiety and prolongs the consummatory feeding. Activation of postsynaptic DRD1LPBN neurons inhibits feeding, whereas genetic deletion of Drd1 within the LPBN causes robust increase in food intake and subsequent weight gain. Furthermore, the DRD1LPBN signaling manifests the central mechanism in methylphenidate-induced hypophagia. In conclusion, our study illuminates a hindbrain DAergic circuit that controls feeding through dynamic regulation in satiety response and meal structure.
Collapse
Affiliation(s)
- Yong Han
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Guobin Xia
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yanlin He
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yang He
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Monica Farias
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Qi Wu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
38
|
Burrus CJ, McKinstry SU, Kim N, Ozlu MI, Santoki AV, Fang FY, Ma A, Karadeniz YB, Worthington AK, Dragatsis I, Zeitlin S, Yin HH, Eroglu C. Striatal Projection Neurons Require Huntingtin for Synaptic Connectivity and Survival. Cell Rep 2021; 30:642-657.e6. [PMID: 31968243 DOI: 10.1016/j.celrep.2019.12.069] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 11/07/2019] [Accepted: 12/17/2019] [Indexed: 12/19/2022] Open
Abstract
Huntington's disease (HD) is caused by an autosomal dominant polyglutamine expansion mutation of Huntingtin (HTT). HD patients suffer from progressive motor, cognitive, and psychiatric impairments, along with significant degeneration of the striatal projection neurons (SPNs) of the striatum. HD is widely accepted to be caused by a toxic gain-of-function of mutant HTT. However, whether loss of HTT function, because of dominant-negative effects of the mutant protein, plays a role in HD and whether HTT is required for SPN health and function are not known. Here, we delete Htt from specific subpopulations of SPNs using the Cre-Lox system and find that SPNs require HTT for motor regulation, synaptic development, cell health, and survival during aging. Our results suggest that loss of HTT function in SPNs could play a critical role in HD pathogenesis.
Collapse
Affiliation(s)
- Caley J Burrus
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Spencer U McKinstry
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Namsoo Kim
- Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA
| | - M Ilcim Ozlu
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Aditya V Santoki
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Francia Y Fang
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Annie Ma
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yonca B Karadeniz
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Atesh K Worthington
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ioannis Dragatsis
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, TN 38163, USA
| | - Scott Zeitlin
- Department of Neuroscience, University of Virginia, School of Medicine, Charlottesville, VA 22908, USA
| | - Henry H Yin
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA; Duke Institute for Brain Sciences, Durham, NC 27710, USA
| | - Cagla Eroglu
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Duke Institute for Brain Sciences, Durham, NC 27710, USA; Regeneration Next Initiative, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
39
|
Assali DR, Sidikpramana M, Villa AP, Falkenstein J, Steele AD. Type 1 dopamine receptor (D1R)-independent circadian food anticipatory activity in mice. PLoS One 2021; 16:e0242897. [PMID: 33556069 PMCID: PMC7869994 DOI: 10.1371/journal.pone.0242897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/27/2021] [Indexed: 01/11/2023] Open
Abstract
Circadian rhythms are entrained by light and influenced by non-photic stimuli, such as feeding. The activity preceding scheduled mealtimes, food anticipatory activity (FAA), is elicited in rodents fed a limited amount at scheduled times. FAA is thought to be the output of an unidentified food entrained oscillator. Previous studies, using gene deletion and receptor pharmacology, implicated dopamine type receptor 1 (D1R) signaling in the dorsal striatum as necessary for FAA in mice. To further understand the role of D1R in promoting FAA, we utilized the Cre-lox system to create cell type-specific deletions of D1R, conditionally deleting D1R in GABA neurons using Vgat-ires-Cre line. This conditional deletion mutant had attenuated FAA, but the amount was higher than expected based on prior results using a constitutive knockout of D1R, D1R KODrago. This result prompted us to re-test the original D1R KODrago line, which expressed less FAA than controls, but only moderately so. To determine if genetic drift had diminished the effect of D1R deletion on FAA, we re-established the D1R KODrago knockout line from cryopreserved samples. The reestablished D1R KODrago-cryo had a clear impairment of FAA compared to controls, but still developed increased activity preceding mealtime across the 4 weeks of timed feeding. Finally, we tested a different deletion allele of D1R created by the Knockout Mouse Project. This line of D1R KOKOMP mice had a significant impairment in the acquisition of FAA, but eventually reached similar levels of premeal activity compared to controls after 4 weeks of timed feeding. Taken together, our results suggest that D1R signaling promotes FAA, but other dopamine receptors likely contribute to FAA given that mice lacking the D1 receptor still retain some FAA.
Collapse
Affiliation(s)
- Dina R. Assali
- Department of Biological Sciences, California State Polytechnic University Pomona, Pomona, CA, United States of America
| | - Michael Sidikpramana
- Department of Biological Sciences, California State Polytechnic University Pomona, Pomona, CA, United States of America
| | - Andrew P. Villa
- Department of Biological Sciences, California State Polytechnic University Pomona, Pomona, CA, United States of America
| | - Jeffrey Falkenstein
- Department of Biological Sciences, California State Polytechnic University Pomona, Pomona, CA, United States of America
| | - Andrew D. Steele
- Department of Biological Sciences, California State Polytechnic University Pomona, Pomona, CA, United States of America
- * E-mail:
| |
Collapse
|
40
|
Mohammed RA, Mansour SM. Sodium hydrogen sulfide upregulates cystathionine β-synthase and protects striatum against 3-nitropropionic acid-induced neurotoxicity in rats. J Pharm Pharmacol 2021; 73:310-321. [PMID: 33793881 DOI: 10.1093/jpp/rgaa072] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/29/2020] [Indexed: 01/12/2023]
Abstract
OBJECTIVES Hydrogen sulfide (H2S) is a neuromodulator that plays a protective role in multiple neurodegenerative diseases including Alzheimer's (AD) and Parkinson's (PD). However, the precise mechanisms underlying its effects against Huntington's disease (HD) are still questioned.This study aimed to examine the neuroprotective effects of sodium hydrogen sulfide (NaHS; H2S donor) against 3-nitropropionic acid (3NP)-induced HD like pathology in rats. Methods: Male Wistar rats were randomly allocated into four groups; (1) normal control receiving saline; (2) NaHS control receiving (0.5 mg/kg/day, i.p.) for 14 days; (3,4) receiving 3NP (10 mg/kg/day, i.p.) for 14 days, with NaHS 30 min later in group 4. KEY FINDINGS NaHS improved cognitive and locomotor deficits induced by 3NP as confirmed by the striatal histopathological findings. These former events were biochemically supported by the increment in cystathionine β-synthase (CBS) gene expression, reduction of glutamate (Glu), dopamine (DA), malondialdehyde (MDA), tumour necrosis factor-alpha (TNF-α), cytochrome-c, cleaved caspase-3 and pc-FOS indicating antioxidant, anti-inflammatory as well as anti-apoptotic effects. Furthermore, NaHS pretreatment improved cholinergic dysfunction and increased brain-derived neurotropic factor (BDNF) and nuclear factor erythroid-2-related factor 2 (Nrf2). CONCLUSIONS These findings suggest that appropriate protection with H2S donors might represent a novel approach to slow down HD-like symptoms.
Collapse
Affiliation(s)
- Reham A Mohammed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Suzan M Mansour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.,Department of Pharmacology, Toxicology and Biochemistry, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt, Cairo, Egypt
| |
Collapse
|
41
|
Establishment of a Cre-rat resource for creating conditional and physiological relevant models of human diseases. Transgenic Res 2021; 30:91-104. [PMID: 33481207 PMCID: PMC7854434 DOI: 10.1007/s11248-020-00226-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/28/2020] [Indexed: 10/25/2022]
Abstract
The goal of this study is to establish a Cre/loxP rat resource for conditional and physiologically predictive rat models of human diseases. The laboratory rat (R. norvegicus) is a central experimental animal in several fields of biomedical research, such as cardiovascular diseases, aging, infectious diseases, autoimmunity, cancer models, transplantation biology, inflammation, cancer risk assessment, industrial toxicology, pharmacology, behavioral and addiction studies, and neurobiology. Up till recently, the ability of creating genetically modified rats has been limited compared to that in the mouse mainly due to lack of genetic manipulation tools and technologies in the rat. Recent advances in nucleases, such as CRISPR/Cas9 (clustered regularly-interspaced short palindromic repeats/CRISPR associated protein 9), as well as TARGATT™ integrase system enables fast, efficient and site-specific introduction of exogenous genetic elements into the rat genome. Here, we report the generation of a collection of tissue-specific, inducible transgenic Cre rats as tool models using TARGATT™, CRISPR/Cas9 and random transgenic approach. More specifically, we generated Cre driver rat models that allow controlled gene expression or knockout (conditional models) both temporally and spatially through the Cre-ERT2/loxP system. A total of 10 Cre rat lines and one Cre reporter/test line were generated, including eight (8) Cre lines for neural specific and two (2) lines for cardiovascular specific Cre expression. All of these lines have been deposited with the Rat Resource and Research Center and provide a much-needed resource for the bio-medical community who employ rat models for their studies of human diseases.
Collapse
|
42
|
Matsuno H, Tsuchimine S, Fukuzato N, O'Hashi K, Kunugi H, Sohya K. Sirtuin 6 is a regulator of dendrite morphogenesis in rat hippocampal neurons. Neurochem Int 2021; 145:104959. [PMID: 33444676 DOI: 10.1016/j.neuint.2021.104959] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/15/2020] [Accepted: 01/05/2021] [Indexed: 01/14/2023]
Abstract
Sirtuin 6 (SIRT6), a member of the Sirtuin family, acts as nicotinamide adenine dinucleotide (NAD)-dependent protein deacetylase, mono-adenosine diphosphate (ADP)-ribosyltransferase, and fatty acid deacylase, and plays critical roles in inflammation, aging, glycolysis, and DNA repair. Accumulating evidence has suggested that SIRT6 is involved in brain functions such as neuronal differentiation, neurogenesis, and learning and memory. However, the precise molecular roles of SIRT6 during neuronal circuit formation are not yet well understood. In this study, we tried to elucidate molecular roles of SIRT6 on neurite development by using primary-cultured hippocampal neurons. We observed that SIRT6 was abundantly localized in the nucleus, and its expression was markedly increased during neurite outgrowth and synaptogenesis. By using shRNA-mediated SIRT6-knockdown, we show that both dendritic length and the number of dendrite branches were significantly reduced in the SIRT6-knockdown neurons. Microarray and subsequent gene ontology analysis revealed that reducing SIRT6 caused the downregulation of immediate early genes (IEGs) and alteration of several biological processes including MAPK (ERK1/2) signaling. We found that nuclear accumulation of phosphorylated ERK1/2 was significantly reduced in SIRT6-knockdown neurons. Overexpression of SIRT6 promoted dendritic length and branching, but the mutants lacking deacetylase activity had no significant effect on the dendritic morphology. Collectively, the presented findings reveal a role of SIRT6 in dendrite morphogenesis, and suggest that SIRT6 may act as an important regulator of ERK1/2 signaling pathway that mediates IEG expression, which leads to dendritic development.
Collapse
Affiliation(s)
- Hitomi Matsuno
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan.
| | - Shoko Tsuchimine
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Noriko Fukuzato
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Kazunori O'Hashi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan; Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan; Department of Psychiatry, Teikyo University School of Medicine, 2-11-1, Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Kazuhiro Sohya
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan.
| |
Collapse
|
43
|
Wang F, Xie N, Zhou J, Dai M, Zhang Q, Hardiman PJ, Qu F. Molecular mechanisms underlying altered neurobehavioural development of female offspring of mothers with polycystic ovary syndrome: FOS-mediated regulation of neurotrophins in placenta. EBioMedicine 2020; 60:102993. [PMID: 32949999 PMCID: PMC7501055 DOI: 10.1016/j.ebiom.2020.102993] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/31/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This study explored the mechanisms underlying altered neurobehavioural development of female offspring born to mothers with polycystic ovary syndrome (PCOS). METHODS In total, 20 women with PCOS and 32 healthy women who underwent caesarean deliveries with a single female foetus were recruited. Infants were assessed with Dubowitz scoring. Swan71 cell line with stable FOS overexpression was used to verify the regulatory effects of FOS on brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) expression. Learning and memory in female first-generation (F1) and second-generation (F2) offspring in a rat model of PCOS was tested using the Morris water maze at puberty and adulthood. Transcriptome analysis of pubertal hippocampi and hypothalami of female F1 offspring was conducted. FINDINGS Total score and behaviour subscales of Dubowitz scoring were significantly lower in female infants of women with PCOS. FOS and NGF protein levels were downregulated in placental villi of the PCOS group. FOS played a key role in BDNF inhibition and enhancing NGF in Swan71 cells. PCOS female F1 rats exhibited lower target crossing times during puberty when compared to controls. Transcriptome analysis revealed significant changes in hippocampal and hypothalamic neuronal pathways in female F1 rats at puberty. INTERPRETATION FOS regulation of neurotrophins in the placenta negatively affects neurobehavioural development of female offspring of PCOS mothers. FUNDING This study was funded by the National Key R&D Program of China (2018YFC1004900 to F.Q. and F.W.) and the National Natural Science Foundation of China (81874480 to F.Q.; 81873837 to F.W.).
Collapse
Affiliation(s)
- Fangfang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Ningning Xie
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Jue Zhou
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Minchen Dai
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Paul J Hardiman
- Institute for Women's Health, University College London, London NW3 2PF, United Kingdom
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China; Institute for Women's Health, University College London, London NW3 2PF, United Kingdom.
| |
Collapse
|
44
|
Brami-Cherrier K, Lewis RG, Cervantes M, Liu Y, Tognini P, Baldi P, Sassone-Corsi P, Borrelli E. Cocaine-mediated circadian reprogramming in the striatum through dopamine D2R and PPARγ activation. Nat Commun 2020; 11:4448. [PMID: 32895370 PMCID: PMC7477550 DOI: 10.1038/s41467-020-18200-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Substance abuse disorders are linked to alteration of circadian rhythms, although the molecular and neuronal pathways implicated have not been fully elucidated. Addictive drugs, such as cocaine, induce a rapid increase of dopamine levels in the brain. Here, we show that acute administration of cocaine triggers reprogramming in circadian gene expression in the striatum, an area involved in psychomotor and rewarding effects of drugs. This process involves the activation of peroxisome protein activator receptor gamma (PPARγ), a nuclear receptor involved in inflammatory responses. PPARγ reprogramming is altered in mice with cell-specific ablation of the dopamine D2 receptor (D2R) in the striatal medium spiny neurons (MSNs) (iMSN-D2RKO). Administration of a specific PPARγ agonist in iMSN-D2RKO mice elicits substantial rescue of cocaine-dependent control of circadian genes. These findings have potential implications for development of strategies to treat substance abuse disorders. Drugs of abuse have been shown to perturb circadian rhythms. Here, the authors show in mice that cocaine exposure modulates circadian gene expression in the striatum through a previously unappreciated pathway that involves dopamine D2 receptors and the nuclear receptor PPARγ.
Collapse
Affiliation(s)
- Karen Brami-Cherrier
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA, 92697, USA
| | - Robert G Lewis
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA, 92697, USA
| | - Marlene Cervantes
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, University of California Irvine, Irvine, CA, 92697, USA
| | - Yu Liu
- Institute for Genomics and Bioinformatics, Department of Computer Science, University of California Irvine, Irvine, CA, 92697, USA
| | - Paola Tognini
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, University of California Irvine, Irvine, CA, 92697, USA
| | - Pierre Baldi
- Institute for Genomics and Bioinformatics, Department of Computer Science, University of California Irvine, Irvine, CA, 92697, USA
| | - Paolo Sassone-Corsi
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, University of California Irvine, Irvine, CA, 92697, USA.
| | - Emiliana Borrelli
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
45
|
Belkacemi L, Darmani NA. Dopamine receptors in emesis: Molecular mechanisms and potential therapeutic function. Pharmacol Res 2020; 161:105124. [PMID: 32814171 DOI: 10.1016/j.phrs.2020.105124] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/20/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022]
Abstract
Dopamine is a member of the catecholamine family and is associated with multiple physiological functions. Together with its five receptor subtypes, dopamine is closely linked to neurological disorders such as schizophrenia, Parkinson's disease, depression, attention deficit-hyperactivity, and restless leg syndrome. Unfortunately, several dopamine receptor-based agonists used to treat some of these diseases cause nausea and vomiting as impending side-effects. The high degree of cross interactions of dopamine receptor ligands with many other targets including G-protein coupled receptors, transporters, enzymes, and ion-channels, add to the complexity of discovering new targets for the treatment of nausea and vomiting. Using activation status of signaling cascades as mechanism-based biomarkers to foresee drug sensitivity combined with the development of dopamine receptor-based biased agonists may hold great promise and seems as the next step in drug development for the treatment of such multifactorial diseases. In this review, we update the present knowledge on dopamine and dopamine receptors and their potential roles in nausea and vomiting. The pre- and clinical evidence provided in this review supports the implication of both dopamine and dopamine receptor agonists in the incidence of emesis. Besides the conventional dopaminergic antiemetic drugs, potential novel antiemetic targeting emetic protein signaling cascades may offer superior selectivity profile and potency.
Collapse
Affiliation(s)
- Louiza Belkacemi
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Nissar A Darmani
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, 91766, USA.
| |
Collapse
|
46
|
Savell KE, Tuscher JJ, Zipperly ME, Duke CG, Phillips RA, Bauman AJ, Thukral S, Sultan FA, Goska NA, Ianov L, Day JJ. A dopamine-induced gene expression signature regulates neuronal function and cocaine response. SCIENCE ADVANCES 2020; 6:eaba4221. [PMID: 32637607 PMCID: PMC7314536 DOI: 10.1126/sciadv.aba4221] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 05/08/2020] [Indexed: 05/21/2023]
Abstract
Drugs of abuse elevate dopamine levels in the nucleus accumbens (NAc) and alter transcriptional programs believed to promote long-lasting synaptic and behavioral adaptations. Here, we leveraged single-nucleus RNA-sequencing to generate a comprehensive molecular atlas of cell subtypes in the NAc, defining both sex-specific and cell type-specific responses to acute cocaine experience in a rat model system. Using this transcriptional map, we identified an immediate early gene expression program that is up-regulated following cocaine experience in vivo and dopamine receptor activation in vitro. Multiplexed induction of this gene program with a large-scale CRISPR-dCas9 activation strategy initiated a secondary synapse-centric transcriptional profile, altered striatal physiology in vitro, and enhanced cocaine sensitization in vivo. Together, these results define the transcriptional response to cocaine with cellular precision and demonstrate that drug-responsive gene programs can potentiate both physiological and behavioral adaptations to drugs of abuse.
Collapse
Affiliation(s)
- Katherine E. Savell
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J. Tuscher
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan E. Zipperly
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Corey G. Duke
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Robert A. Phillips
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Allison J. Bauman
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Saakshi Thukral
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Faraz A. Sultan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nicholas A. Goska
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremy J. Day
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
47
|
Salery M, Trifilieff P, Caboche J, Vanhoutte P. From Signaling Molecules to Circuits and Behaviors: Cell-Type-Specific Adaptations to Psychostimulant Exposure in the Striatum. Biol Psychiatry 2020; 87:944-953. [PMID: 31928716 DOI: 10.1016/j.biopsych.2019.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022]
Abstract
Addiction is characterized by a compulsive pattern of drug seeking and consumption and a high risk of relapse after withdrawal that are thought to result from persistent adaptations within brain reward circuits. Drugs of abuse increase dopamine (DA) concentration in these brain areas, including the striatum, which shapes an abnormal memory trace of drug consumption that virtually highjacks reward processing. Long-term neuronal adaptations of gamma-aminobutyric acidergic striatal projection neurons (SPNs) evoked by drugs of abuse are critical for the development of addiction. These neurons form two mostly segregated populations, depending on the DA receptor they express and their output projections, constituting the so-called direct (D1 receptor) and indirect (D2 receptor) SPN pathways. Both SPN subtypes receive converging glutamate inputs from limbic and cortical regions, encoding contextual and emotional information, together with DA, which mediates reward prediction and incentive values. DA differentially modulates the efficacy of glutamate synapses onto direct and indirect SPN pathways by recruiting distinct striatal signaling pathways, epigenetic and genetic responses likely involved in the transition from casual drug use to addiction. Herein we focus on recent studies that have assessed psychostimulant-induced alterations in a cell-type-specific manner, from remodeling of input projections to the characterization of specific molecular events in each SPN subtype and their impact on long-lasting behavioral adaptations. We discuss recent evidence revealing the complex and concerted action of both SPN populations on drug-induced behavioral responses, as these studies can contribute to the design of future strategies to alleviate specific behavioral components of addiction.
Collapse
Affiliation(s)
- Marine Salery
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pierre Trifilieff
- NutriNeuro, Unité Mixte de Recherche (UMR) 1286, Institut National de la Recherche Agronomique, Bordeaux Institut Polytechnique, University of Bordeaux, Bordeaux, France
| | - Jocelyne Caboche
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Sorbonne Université, Faculty of Sciences, Paris, France; Centre National de la Recherche Scientifique, UMR8246, Paris, France; Institut National de la Santé et de la Recherche Médicale, U1130, Paris France.
| | - Peter Vanhoutte
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Sorbonne Université, Faculty of Sciences, Paris, France; Centre National de la Recherche Scientifique, UMR8246, Paris, France; Institut National de la Santé et de la Recherche Médicale, U1130, Paris France
| |
Collapse
|
48
|
Yang AK, Mendoza JA, Lafferty CK, Lacroix F, Britt JP. Hippocampal Input to the Nucleus Accumbens Shell Enhances Food Palatability. Biol Psychiatry 2020; 87:597-608. [PMID: 31699294 DOI: 10.1016/j.biopsych.2019.09.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/28/2019] [Accepted: 09/12/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND Insight into the neural basis of hedonic processing has come from studies of food palatability in rodents. Pharmacological manipulations of the nucleus accumbens shell (NAcSh) have repeatedly been demonstrated to increase hedonic taste reactivity, yet the contribution of specific NAcSh circuit components is unknown. METHODS Bidirectional optogenetic manipulations were targeted to the principal NAcSh projection neurons and afferent pathways in mice during free feeding assays. Number of licks per bout of consumption was used as a measure of food palatability as it was confirmed to track sucrose concentration and subjective flavor preferences. RESULTS Photoinhibition of NAcSh neurons, whether general or cell-type specific, was found to alter consumption without affecting its hedonic impact. Among the principal excitatory afferent pathways, we showed that ventral hippocampal (vHipp) input alone enhances palatability upon low-frequency photostimulation time-locked to consumption. This enhancement in palatability was independent of opioid signaling and not recapitulated by NAcSh or dopamine neuron photostimulation. We further demonstrated that vHipp input photostimulation is sufficient to condition a flavor preference, while its inhibition impedes sucrose-driven flavor preference conditioning. CONCLUSIONS These results demonstrate a novel contribution of vHipp-NAcSh pathway activity to palatability that may relate to its innervation of a particular region or neuronal ensemble in the NAcSh. These findings are consistent with the evidence that vHipp-NAcSh activity is relevant to the pathophysiology of anhedonia and depression as well as the increasing appreciation of hippocampal involvement in people's food pleasantness ratings, hunger, and weight.
Collapse
Affiliation(s)
- Angela K Yang
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Quebec, Canada
| | - Jesse A Mendoza
- Department of Psychology, McGill University, Montreal, Quebec, Canada; Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Quebec, Canada
| | - Christopher K Lafferty
- Department of Psychology, McGill University, Montreal, Quebec, Canada; Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Quebec, Canada
| | - Franca Lacroix
- Department of Psychology, McGill University, Montreal, Quebec, Canada
| | - Jonathan P Britt
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Psychology, McGill University, Montreal, Quebec, Canada; Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, Quebec, Canada.
| |
Collapse
|
49
|
Halawa AA, El-Adl MA, Marghani BH. Postmortem Heat Stress upregulates Thanatotranscriptome of Genes encode Inflammation, Apoptosis and Neuronal Stress in Brain of Rats at Short Postmortem Intervals. AUST J FORENSIC SCI 2019. [DOI: 10.1080/00450618.2019.1682669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Amal A. Halawa
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed A. El-Adl
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Basma H. Marghani
- Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
50
|
Wang Q, Hu Y, Wan J, Dong B, Sun J. Lactate: A Novel Signaling Molecule in Synaptic Plasticity and Drug Addiction. Bioessays 2019; 41:e1900008. [PMID: 31270822 DOI: 10.1002/bies.201900008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/20/2019] [Indexed: 12/21/2022]
Abstract
l-Lactate is emerging as a crucial regulatory nexus for energy metabolism in the brain and signaling transduction in synaptic plasticity, memory processes, and drug addiction instead of being merely a waste by-product of anaerobic glycolysis. In this review, the role of lactate in various memory processes, synapse plasticity and drug addiction on the basis of recent studies is summarized and discussed. To this end, three main parts are presented: first, lactate as an energy substrate in energy metabolism of the brain is described; second, lactate as a novel signaling molecule in synaptic plasticity, neural circuits, memory, and drug addiction is described; and third, in light of the above descriptions, it is plausible to speculate that lactate is predominantly a signaling molecule in specific memory processes and partly acts as an energy substrate. The future perspective in lactate signaling involving microglia and associated precise signaling pathways in the brain is highlighted.
Collapse
Affiliation(s)
- Qiuting Wang
- Key Laboratory for Experimental Teratology of the Ministry of Education, Department of Anatomy, School of Basic Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Ying Hu
- Department of Paediatrics, Jinan Zhangqiu District Hospital of TCM, Jinan, Shandong, 250200, China
| | - Jiale Wan
- Key Laboratory for Experimental Teratology of the Ministry of Education, Department of Anatomy, School of Basic Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China.,Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China
| | - Jinhao Sun
- Key Laboratory for Experimental Teratology of the Ministry of Education, Department of Anatomy, School of Basic Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|