1
|
Nolan SO, Melugin PR, Erickson KR, Adams WR, Farahbakhsh ZZ, Mcgonigle CE, Kwon MH, Costa VD, Hackett TA, Cuzon Carlson VC, Constantinidis C, Lapish CC, Grant KA, Siciliano CA. Recurrent activity propagates through labile ensembles in macaque dorsolateral prefrontal microcircuits. Curr Biol 2025; 35:431-443.e4. [PMID: 39765226 PMCID: PMC11832050 DOI: 10.1016/j.cub.2024.11.069] [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: 10/20/2023] [Revised: 06/03/2024] [Accepted: 11/27/2024] [Indexed: 01/12/2025]
Abstract
Human and non-human primate studies clearly implicate the dorsolateral prefrontal cortex (dlPFC) as critical for advanced cognitive functions.1,2 It is thought that intracortical synaptic architectures within the dlPFC are the integral neurobiological substrate that gives rise to these processes.3,4,5,6,7 In the prevailing model, each cortical column makes up one fundamental processing unit composed of dense intrinsic connectivity, conceptualized as the "canonical" cortical microcircuit.3,8 Each cortical microcircuit receives sensory and cognitive information from upstream sources, which are represented by sustained activity within the microcircuit, referred to as persistent or recurrent activity.4,9 Via recurrent connections within the microcircuit, activity propagates for a variable length of time, thereby allowing temporary storage and computations to occur locally before ultimately passing a transformed representation to a downstream output.4,5,10 Competing theories regarding how microcircuit activity is coordinated have proven difficult to reconcile in vivo, where intercortical and intracortical computations cannot be fully dissociated.5,9,11,12 Here, using high-density calcium imaging of macaque dlPFC, we isolated intracortical computations by interrogating microcircuit networks ex vivo. Using peri-sulcal stimulation to evoke recurrent activity in deep layers, we found that activity propagates through stochastically assembled intracortical networks wherein orderly, predictable, low-dimensional collective dynamics arise from ensembles with highly labile cellular memberships. Microcircuit excitability covaried with individual cognitive performance, thus anchoring heuristic models of abstract cortical functions within quantifiable constraints imposed by the underlying synaptic architecture. Our findings argue against engram or localist architectures, together demonstrating that generation of high-fidelity population-level signals from distributed, labile networks is an intrinsic feature of dlPFC microcircuitry.
Collapse
Affiliation(s)
- Suzanne O Nolan
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Patrick R Melugin
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Kirsty R Erickson
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Wilson R Adams
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Zahra Z Farahbakhsh
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Colleen E Mcgonigle
- Department of Psychology, Indiana University Indianapolis, Indianapolis, IN 46202, USA
| | - Michelle H Kwon
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Vincent D Costa
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Division of Developmental and Cognitive Neuroscience, Emory National Primate Research Center, Atlanta, GA 30329, USA
| | - Troy A Hackett
- Department of Hearing and Speech Sciences, Department of Psychology, Vanderbilt University School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Verginia C Cuzon Carlson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Christos Constantinidis
- Department of Biomedical Engineering, Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
| | - Christopher C Lapish
- Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA.
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37232, USA; Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
2
|
Barbera G, Thapa R, Adhikari N, Li Y, Lin DT. Imaging distinct neuronal populations with a dual channel miniscope. Front Neurosci 2024; 18:1445020. [PMID: 39717702 PMCID: PMC11663848 DOI: 10.3389/fnins.2024.1445020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/14/2024] [Indexed: 12/25/2024] Open
Abstract
Miniature fluorescence microscopes (miniscopes) are one of the most powerful and versatile tools for recording large scale neural activity in freely moving rodents with single cell resolution. Recent advances in the design of genetically encoded calcium indicators (GECIs) allow to target distinct neuronal populations with non-overlapping emission spectral profiles. However, conventional miniscopes are limited to a single excitation, single focal plane imaging, which does not allow to compensate for chromatic aberration and image from two spectrally distinct calcium indicators. In this paper we present an open-source dual channel miniscope capable of simultaneous imaging of genetically or functionally distinct neuronal populations. Chromatic aberrations are corrected using an electrowetting lens (EWL), which allows fast focal plane change between frames. To demonstrate the capabilities of the dual channel miniscope, we labeled layer specific excitatory neurons or inhibitory interneurons in the medial prefrontal cortex (mPFC) with a red fluorescence protein, and simultaneously imaged neural activity of distinct neuronal populations of freely moving mice via a green GECI.
Collapse
Affiliation(s)
- Giovanni Barbera
- Intramural Research Program, National Institute on Drug Abuse National Institutes of Health, Baltimore, MD, United States
| | - Rashmi Thapa
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| | - Navin Adhikari
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| | - Yun Li
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| | - Da-Ting Lin
- Intramural Research Program, National Institute on Drug Abuse National Institutes of Health, Baltimore, MD, United States
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
3
|
Zhang L, Liu G, Peng Y, Gao J, Tian M. Role of Neural Circuits in Cognitive Impairment. Neurochem Res 2024; 50:49. [PMID: 39644416 DOI: 10.1007/s11064-024-04309-3] [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: 10/06/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Cognitive impairment refers to abnormalities in learning, memory and cognitive judgment, mainly manifested as symptoms such as decreased memory, impaired orientation and reduced computational ability. As the fundamental unit of information processing in the brain, neural circuits have recently attracted great attention due to their functions in regulating pain, emotion and behavior. Furthermore, a growing number of studies have suggested that neural circuits play an important role in cognitive impairment. Neural circuits can affect perception, attention and decision-making, they can also regulate language skill, thinking and memory. Pathological conditions crucially affecting the integrity and preservation of neural circuits and their connectivity will heavily impact cognitive abilities. Nowadays, technological developments have led to many novel methods for studying neural circuits, such as brain imaging, optogenetic techniques, and chemical genetics approaches. Therefore, neural circuits show great promise as a potential target in mitigating cognitive impairment. In this review we discuss the pathogenesis of cognitive impairment and the regulation and detection of neural circuits, thus highlighting the role of neural circuits in cognitive impairment. Hence, therapeutic agents against cognitive impairment may be developed that target neural circuits important in cognition.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Guodong Liu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Yaonan Peng
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Jinqi Gao
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, PR China
| | - Mi Tian
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, PR China.
| |
Collapse
|
4
|
Kielbinski M, Bernacka J. Fiber photometry in neuroscience research: principles, applications, and future directions. Pharmacol Rep 2024; 76:1242-1255. [PMID: 39235662 PMCID: PMC11582208 DOI: 10.1007/s43440-024-00646-w] [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: 07/01/2024] [Revised: 08/16/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
In recent years, fluorescent sensors are enjoying a surge of popularity in the field of neuroscience. Through the development of novel genetically encoded sensors as well as improved methods of detection and analysis, fluorescent sensing has risen as a new major technique in neuroscience alongside molecular, electrophysiological, and imaging methods, opening up new avenues for research. Combined with multiphoton microscopy and fiber photometry, these sensors offer unique advantages in terms of cellular specificity, access to multiple targets - from calcium dynamics to neurotransmitter release to intracellular processes - as well as high capability for in vivo interrogation of neurobiological mechanisms underpinning behavior. Here, we provide a brief overview of the method, present examples of its integration with other tools in recent studies ranging from cellular to systems neuroscience, and discuss some of its principles and limitations, with the aim of introducing new potential users to this rapidly developing and potent technique.
Collapse
Affiliation(s)
- Michal Kielbinski
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | - Joanna Bernacka
- Cancer Neurophysiology Group, Łukasiewicz - PORT, Polish Center for Technology Development, Stabłowicka 147, Wrocław, 54-066, Poland
| |
Collapse
|
5
|
Chen G, Dang D, Zhang C, Qin L, Yan T, Wang W, Liang W. Recent advances in neurotechnology-based biohybrid robots. SOFT MATTER 2024; 20:7993-8011. [PMID: 39328163 DOI: 10.1039/d4sm00768a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Biohybrid robots retain the innate biological characteristics and behavioral traits of animals, making them valuable in applications such as disaster relief, exploration of unknown terrains, and medical care. This review aims to comprehensively discuss the evolution of biohybrid robots, their key technologies and applications, and the challenges they face. By analyzing studies conducted on terrestrial, aquatic, and aerial biohybrid robots, we gain a deeper understanding of how these technologies have made significant progress in simulating natural organisms, improving mechanical performance, and intelligent control. Additionally, we address challenges associated with the application of electrical stimulation technology, the precision of neural signal monitoring, and the ethical considerations for biohybrid robots. We highlight the importance of future research focusing on developing more sophisticated and biocompatible control methods while prioritizing animal welfare. We believe that exploring multimodal monitoring and stimulation technologies holds the potential to enhance the performance of biohybrid robots. These efforts are expected to pave the way for biohybrid robotics technology to introduce greater innovation and well-being to human society in the future.
Collapse
Affiliation(s)
- Guiyong Chen
- School of Mechanical Engineering, Shenyang Jianzhu University, Shenyang 110168, People's Republic of China.
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, People's Republic of China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, People's Republic of China
| | - Dan Dang
- School of Sciences, Shenyang Jianzhu University, Shenyang 110168, People's Republic of China.
| | - Chuang Zhang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, People's Republic of China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, People's Republic of China
| | - Ling Qin
- School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China
| | - Tao Yan
- Department of Anesthesiology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Beijing 100021, People's Republic of China
- Chinese Academy of Medical Sciences, Beijing 100021, People's Republic of China
- Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Wenxue Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, People's Republic of China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, People's Republic of China
| | - Wenfeng Liang
- School of Mechanical Engineering, Shenyang Jianzhu University, Shenyang 110168, People's Republic of China.
| |
Collapse
|
6
|
Engel L, Wolff AR, Blake M, Collins VL, Sinha S, Saunders BT. Dopamine neurons drive spatiotemporally heterogeneous striatal dopamine signals during learning. Curr Biol 2024; 34:3086-3101.e4. [PMID: 38925117 PMCID: PMC11279555 DOI: 10.1016/j.cub.2024.05.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/25/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024]
Abstract
Environmental cues, through Pavlovian learning, become conditioned stimuli that invigorate and guide animals toward rewards. Dopamine (DA) neurons in the ventral tegmental area (VTA) and substantia nigra (SNc) are crucial for this process, via engagement of a reciprocally connected network with their striatal targets. Critically, it remains unknown how dopamine neuron activity itself engages dopamine signals throughout the striatum, across learning. Here, we investigated how optogenetic Pavlovian cue conditioning of VTA or SNc dopamine neurons directs cue-evoked behavior and shapes subregion-specific striatal dopamine dynamics. We used a fluorescent biosensor to monitor dopamine in the nucleus accumbens (NAc) core and shell, dorsomedial striatum (DMS), and dorsolateral striatum (DLS). We demonstrate spatially heterogeneous, learning-dependent dopamine changes across striatal regions. Although VTA stimulation-evoked robust dopamine release in NAc core, shell, and DMS, predictive cues preferentially recruited dopamine release in NAc core, starting early in training, and DMS, late in training. Negative prediction error signals, reflecting a violation in the expectation of dopamine neuron activation, only emerged in the NAc core and DMS. Despite the development of vigorous movement late in training, conditioned dopamine signals did not emerge in the DLS, even during Pavlovian conditioning with SNc dopamine neuron activation, which elicited robust DLS dopamine release. Together, our studies show a broad dissociation in the fundamental prediction and reward-related information generated by VTA and SNc dopamine neuron populations and signaled by dopamine across the striatum. Further, they offer new insight into how larger-scale adaptations across the striatal network emerge during learning to coordinate behavior.
Collapse
Affiliation(s)
- Liv Engel
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Amy R Wolff
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Madelyn Blake
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Val L Collins
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Sonal Sinha
- Krieger School of Arts & Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Benjamin T Saunders
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
7
|
Bender BN, Stringfield SJ, Torregrossa MM. Changes in dorsomedial striatum activity during expression of goal-directed vs. habit-like cue-induced cocaine seeking. ADDICTION NEUROSCIENCE 2024; 11:100149. [PMID: 38957402 PMCID: PMC11218864 DOI: 10.1016/j.addicn.2024.100149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
A preclinical model of cue exposure therapy, cue extinction, reduces cue-induced cocaine seeking that is goal-directed but not habit-like. Goal-directed and habitual behaviors differentially rely on the dorsomedial striatum (DMS) and dorsolateral striatum (DLS), but the effects of cue extinction on dorsal striatal responses to cue-induced drug seeking are unknown. We used fiber photometry in rats trained to self-administer cocaine paired with an audiovisual cue to examine how dorsal striatal intracellular calcium and extracellular dopamine activity differs between goal-directed and habit-like cue-induced cocaine seeking and how it is impacted by cue extinction. After minimal fixed-ratio training, rats showed enhanced DMS and DLS calcium responses to cue-reinforced compared to unreinforced lever presses. After rats were trained on goal-promoting fixed ratio schedules or habit-promoting second-order schedules of reinforcement, different patterns of dorsal striatal calcium and dopamine responses to cue-reinforced lever presses emerged. Rats trained on habit-promoting second-order schedules showed reduced DMS calcium responses and enhanced DLS dopamine responses to cue-reinforced lever presses. Cue extinction reduced calcium responses during subsequent drug seeking in the DMS, but not in the DLS. Therefore, cue extinction may reduce goal-directed behavior through its effects on the DMS, whereas habit-like behavior and the DLS are unaffected.
Collapse
Affiliation(s)
- Brooke N. Bender
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15213, United States
| | - Sierra J. Stringfield
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States
| | - Mary M. Torregrossa
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15213, United States
| |
Collapse
|
8
|
Inami C, Haruta M, Ohta Y, Tanaka M, So M, Sobue K, Akay Y, Kume K, Ohta J, Akay M, Ohsawa M. Real-time monitoring of cortical brain activity in response to acute pain using wide-area Ca 2+ imaging. Biochem Biophys Res Commun 2024; 708:149800. [PMID: 38522402 DOI: 10.1016/j.bbrc.2024.149800] [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: 02/15/2024] [Accepted: 03/16/2024] [Indexed: 03/26/2024]
Abstract
Previous human and rodent studies indicated that nociceptive stimuli activate many brain regions that is involved in the somatosensory and emotional sensation. Although these studies have identified several important brain regions involved in pain perception, it has been a challenge to observe neural activity directly and simultaneously in these multiple brain regions during pain perception. Using a transgenic mouse expressing G-CaMP7 in majority of astrocytes and a subpopulation of excitatory neurons, we recorded the brain activity in the mouse cerebral cortex during acute pain stimulation. Both of hind paw pinch and intraplantar administration of formalin caused strong transient increase of the fluorescence in several cortical regions, including primary somatosensory, motor and retrosplenial cortex. This increase of the fluorescence intensity was attenuated by the pretreatment with morphine. The present study provides important insight into the cortico-cortical network during pain perception.
Collapse
Affiliation(s)
- Chihiro Inami
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Makito Haruta
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama, Ikoma-shi, Nara, Japan
| | - Yasumi Ohta
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama, Ikoma-shi, Nara, Japan
| | - Motoshi Tanaka
- Department of Anesthesiology and Intensive Care Medicine, Graduate School of Medicine. Nagoya City University, 1 Kawasumi, Mizuho-ku, Nagoya, 467-8601, Japan
| | - MinHye So
- Department of Anesthesiology and Intensive Care Medicine, Graduate School of Medicine. Nagoya City University, 1 Kawasumi, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Kazuya Sobue
- Department of Anesthesiology and Intensive Care Medicine, Graduate School of Medicine. Nagoya City University, 1 Kawasumi, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Yasemin Akay
- Biomedical Engineering Department, University of Houston, 3517 Cullen Blvd, Houston, TX, 77204, USA
| | - Kazuhiko Kume
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Jun Ohta
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama, Ikoma-shi, Nara, Japan
| | - Metin Akay
- Biomedical Engineering Department, University of Houston, 3517 Cullen Blvd, Houston, TX, 77204, USA
| | - Masahiro Ohsawa
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan; Department of Integrative Neuroscience, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan; Laboratory of Systems Pharmacology, Faculty of Pharma-Sciences, Teikyo University, Itabashi, Tokyo, 173-8603, Japan.
| |
Collapse
|
9
|
Nghiem TAE, Lee B, Chao THH, Branigan NK, Mistry PK, Shih YYI, Menon V. Space wandering in the rodent default mode network. Proc Natl Acad Sci U S A 2024; 121:e2315167121. [PMID: 38557177 PMCID: PMC11009630 DOI: 10.1073/pnas.2315167121] [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: 09/02/2023] [Accepted: 01/17/2024] [Indexed: 04/04/2024] Open
Abstract
The default mode network (DMN) is a large-scale brain network known to be suppressed during a wide range of cognitive tasks. However, our comprehension of its role in naturalistic and unconstrained behaviors has remained elusive because most research on the DMN has been conducted within the restrictive confines of MRI scanners. Here, we use multisite GCaMP (a genetically encoded calcium indicator) fiber photometry with simultaneous videography to probe DMN function in awake, freely exploring rats. We examined neural dynamics in three core DMN nodes-the retrosplenial cortex, cingulate cortex, and prelimbic cortex-as well as the anterior insula node of the salience network, and their association with the rats' spatial exploration behaviors. We found that DMN nodes displayed a hierarchical functional organization during spatial exploration, characterized by stronger coupling with each other than with the anterior insula. Crucially, these DMN nodes encoded the kinematics of spatial exploration, including linear and angular velocity. Additionally, we identified latent brain states that encoded distinct patterns of time-varying exploration behaviors and found that higher linear velocity was associated with enhanced DMN activity, heightened synchronization among DMN nodes, and increased anticorrelation between the DMN and anterior insula. Our findings highlight the involvement of the DMN in collectively and dynamically encoding spatial exploration in a real-world setting. Our findings challenge the notion that the DMN is primarily a "task-negative" network disengaged from the external world. By illuminating the DMN's role in naturalistic behaviors, our study underscores the importance of investigating brain network function in ecologically valid contexts.
Collapse
Affiliation(s)
| | - Byeongwook Lee
- Department of Psychiatry & Behavioral Sciences, Stanford University, Palo Alto, CA94304
| | - Tzu-Hao Harry Chao
- Center for Animal MRI, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Nicholas K. Branigan
- Department of Psychiatry & Behavioral Sciences, Stanford University, Palo Alto, CA94304
| | - Percy K. Mistry
- Department of Psychiatry & Behavioral Sciences, Stanford University, Palo Alto, CA94304
| | - Yen-Yu Ian Shih
- Center for Animal MRI, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC27514
| | - Vinod Menon
- Department of Psychiatry & Behavioral Sciences, Stanford University, Palo Alto, CA94304
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, CA94304
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA94305
| |
Collapse
|
10
|
Engel L, Wolff AR, Blake M, Collins VL, Sinha S, Saunders BT. Dopamine neurons drive spatiotemporally heterogeneous striatal dopamine signals during learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.01.547331. [PMID: 38585717 PMCID: PMC10996462 DOI: 10.1101/2023.07.01.547331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Environmental cues, through Pavlovian learning, become conditioned stimuli that invigorate and guide animals toward acquisition of rewards. Dopamine neurons in the ventral tegmental area (VTA) and substantia nigra (SNC) are crucial for this process. Dopamine neurons are embedded in a reciprocally connected network with their striatal targets, the functional organization of which remains poorly understood. Here, we investigated how learning during optogenetic Pavlovian cue conditioning of VTA or SNC dopamine neurons directs cue-evoked behavior and shapes subregion-specific striatal dopamine dynamics. We used a fluorescent dopamine biosensor to monitor dopamine in the nucleus accumbens (NAc) core and shell, dorsomedial striatum (DMS), and dorsolateral striatum (DLS). We demonstrate spatially heterogeneous, learning-dependent dopamine changes across striatal regions. While VTA stimulation evoked robust dopamine release in NAc core, shell, and DMS, cues predictive of this activation preferentially recruited dopamine release in NAc core, starting early in training, and DMS, late in training. Corresponding negative prediction error signals, reflecting a violation in the expectation of dopamine neuron activation, only emerged in the NAc core and DMS, and not the shell. Despite development of vigorous movement late in training, conditioned dopamine signals did not similarly emerge in the DLS, even during Pavlovian conditioning with SNC dopamine neuron activation, which elicited robust DLS dopamine release. Together, our studies show broad dissociation in the fundamental prediction and reward-related information generated by different dopamine neuron populations and signaled by dopamine across the striatum. Further, they offer new insight into how larger-scale plasticity across the striatal network emerges during Pavlovian learning to coordinate behavior.
Collapse
Affiliation(s)
- Liv Engel
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
- Current Address: Department of Psychology, University of Toronto
| | - Amy R Wolff
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| | - Madelyn Blake
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| | - Val L Collins
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| | | | - Benjamin T Saunders
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| |
Collapse
|
11
|
Timme NM, Ardinger CE, Weir SDC, Zelaya-Escobar R, Kruger R, Lapish CC. Non-consummatory behavior signals predict aversion-resistant alcohol drinking in head-fixed mice. Neuropharmacology 2024; 242:109762. [PMID: 37871677 PMCID: PMC10872650 DOI: 10.1016/j.neuropharm.2023.109762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/05/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023]
Abstract
A key facet of alcohol use disorder is continuing to drink alcohol despite negative consequences (so called "aversion-resistant drinking"). In this study, we sought to assess the degree to which head-fixed mice exhibit aversion-resistant drinking and to leverage behavioral analysis techniques available in head-fixture to relate non-consummatory behaviors to aversion-resistant drinking. We assessed aversion-resistant drinking in head-fixed female and male C57BL/6 J mice. We adulterated 20% (v/v) alcohol with varying concentrations of the bitter tastant quinine to measure the degree to which mice would continue to drink despite this aversive stimulus. We recorded high-resolution video of the mice during head-fixed drinking, tracked body parts with machine vision tools, and analyzed body movements in relation to consumption. Female and male head-fixed mice exhibited heterogenous levels of aversion-resistant drinking. Additionally, non-consummatory behaviors, such as paw movement and snout movement, were related to the intensity of aversion-resistant drinking. These studies demonstrate that head-fixed mice exhibit aversion-resistant drinking and that non-consummatory behaviors can be used to assess perceived aversiveness in this paradigm. Furthermore, these studies lay the groundwork for future experiments that will utilize advanced electrophysiological techniques to record from large populations of neurons during aversion-resistant drinking to understand the neurocomputational processes that drive this clinically relevant behavior. This article is part of the Special Issue on "PFC circuit function in psychiatric disease and relevant models".
Collapse
Affiliation(s)
- Nicholas M Timme
- Department of Psychology, Indiana University - Purdue University Indianapolis, 402 N. Blackford St, LD 124, Indianapolis, IN, 46202, USA.
| | - Cherish E Ardinger
- Department of Psychology, Indiana University - Purdue University Indianapolis, 402 N. Blackford St, LD 124, Indianapolis, IN, 46202, USA
| | - Seth D C Weir
- Department of Psychology, Indiana University - Purdue University Indianapolis, 402 N. Blackford St, LD 124, Indianapolis, IN, 46202, USA
| | - Rachel Zelaya-Escobar
- Department of Psychology, Indiana University - Purdue University Indianapolis, 402 N. Blackford St, LD 124, Indianapolis, IN, 46202, USA
| | - Rachel Kruger
- Department of Psychology, Indiana University - Purdue University Indianapolis, 402 N. Blackford St, LD 124, Indianapolis, IN, 46202, USA
| | - Christopher C Lapish
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, 635 Barnhill Drive, MSB 5035, Indianapolis, IN, 46202, USA; Stark Neuroscience Institute, Indiana University School of Medicine, 320 W. 15th St, NB 414, Indianapolis, IN, 46202, USA
| |
Collapse
|
12
|
Zhou ZC, Gordon-Fennell A, Piantadosi SC, Ji N, Smith SL, Bruchas MR, Stuber GD. Deep-brain optical recording of neural dynamics during behavior. Neuron 2023; 111:3716-3738. [PMID: 37804833 PMCID: PMC10843303 DOI: 10.1016/j.neuron.2023.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/24/2023] [Accepted: 09/06/2023] [Indexed: 10/09/2023]
Abstract
In vivo fluorescence recording techniques have produced landmark discoveries in neuroscience, providing insight into how single cell and circuit-level computations mediate sensory processing and generate complex behaviors. While much attention has been given to recording from cortical brain regions, deep-brain fluorescence recording is more complex because it requires additional measures to gain optical access to harder to reach brain nuclei. Here we discuss detailed considerations and tradeoffs regarding deep-brain fluorescence recording techniques and provide a comprehensive guide for all major steps involved, from project planning to data analysis. The goal is to impart guidance for new and experienced investigators seeking to use in vivo deep fluorescence optical recordings in awake, behaving rodent models.
Collapse
Affiliation(s)
- Zhe Charles Zhou
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Adam Gordon-Fennell
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Sean C Piantadosi
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Na Ji
- Department of Physics, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Spencer LaVere Smith
- Department of Electrical and Computer Engineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Michael R Bruchas
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.
| | - Garret D Stuber
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
13
|
Ghosh S, Dahiya M, Kumar A, Bheri M, Pandey GK. Calcium imaging: a technique to monitor calcium dynamics in biological systems. PHYSIOLOGY AND MOLECULAR BIOLOGY OF PLANTS : AN INTERNATIONAL JOURNAL OF FUNCTIONAL PLANT BIOLOGY 2023; 29:1777-1811. [PMID: 38222278 PMCID: PMC10784449 DOI: 10.1007/s12298-023-01405-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 01/16/2024]
Abstract
Calcium ion (Ca2+) is a multifaceted signaling molecule that acts as an important second messenger. During the course of evolution, plants and animals have developed Ca2+ signaling in order to respond against diverse stimuli, to regulate a large number of physiological and developmental pathways. Our understanding of Ca2+ signaling and its components in physiological phenomena ranging from lower to higher organisms, and from single cell to multiple tissues has grown exponentially. The generation of Ca2+ transients or signatures for various stress factor is a well-known mechanism adopted in plant and animal systems. However, the decoding of such remarkable signatures is an uphill task and is always an interesting goal for the scientific community. In the past few decades, studies on the concentration and dynamics of intracellular Ca2+ are significantly increasing and have become a trend in modern biology. The advancement in approaches from Ca2+ binding dyes to in vivo Ca2+ imaging through the use of Ca2+ biosensors to achieve spatio-temporal resolution in micro and milliseconds range, provide us phenomenal opportunities to study live cell Ca2+ imaging or dynamics. Here, we describe the usage, improvement and advancement of Ca2+ based dyes, genetically encoded probes and sensors to achieve extraordinary Ca2+ imaging in plants and animals. Graphical abstract
Collapse
Affiliation(s)
- Soma Ghosh
- Department of Plant Molecular Biology, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021 India
| | - Monika Dahiya
- Department of Plant Molecular Biology, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021 India
| | - Amit Kumar
- Department of Plant Molecular Biology, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021 India
| | - Malathi Bheri
- Department of Plant Molecular Biology, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021 India
| | - Girdhar K. Pandey
- Department of Plant Molecular Biology, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021 India
| |
Collapse
|
14
|
Wright EC, Luo PX, Zakharenkov HC, Serna Godoy A, Lake AA, Prince ZD, Sekar S, Culkin HI, Ramirez AV, Dwyer T, Kapoor A, Corbett C, Tian L, Fox AS, Trainor BC. Sexual differentiation of neural mechanisms of stress sensitivity during puberty. Proc Natl Acad Sci U S A 2023; 120:e2306475120. [PMID: 37847733 PMCID: PMC10614610 DOI: 10.1073/pnas.2306475120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/12/2023] [Indexed: 10/19/2023] Open
Abstract
Anxiety disorders are a major public health concern and current treatments are inadequate for many individuals. Anxiety is more common in women than men and this difference arises during puberty. Sex differences in physiological stress responses may contribute to this variability. During puberty, gonadal hormones shape brain structure and function, but the extent to which these changes affect stress sensitivity is unknown. We examined how pubertal androgens shape behavioral and neural responses to social stress in California mice (Peromyscus californicus), a model species for studying sex differences in stress responses. In adults, social defeat reduces social approach and increases social vigilance in females but not males. We show this sex difference is absent in juveniles, and that prepubertal castration sensitizes adult males to social defeat. Adult gonadectomy does not alter behavioral responses to defeat, indicating that gonadal hormones act during puberty to program behavioral responses to stress in adulthood. Calcium imaging in the medioventral bed nucleus of the stria terminalis (BNST) showed that social threats increased neural activity and that prepubertal castration generalized these responses to less threatening social contexts. These results support recent hypotheses that the BNST responds to immediate threats. Prepubertal treatment with the nonaromatizable androgen dihydrotestosterone acts in males and females to reduce the effects of defeat on social approach and vigilance in adults. These data indicate that activation of androgen receptors during puberty is critical for programming behavioral responses to stress in adulthood.
Collapse
Affiliation(s)
- Emily C. Wright
- Department of Psychology, University of California, Davis, CA95616
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA95616
| | - Pei X. Luo
- Department of Psychology, University of California, Davis, CA95616
| | | | | | - Alyssa A. Lake
- Department of Psychology, University of California, Davis, CA95616
| | - Zhana D. Prince
- Department of Psychology, University of California, Davis, CA95616
| | - Shwetha Sekar
- Department of Psychology, University of California, Davis, CA95616
| | - Hannah I. Culkin
- Department of Psychology, University of California, Davis, CA95616
| | | | - Tjien Dwyer
- Department of Psychology, University of California, Davis, CA95616
| | - Amita Kapoor
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI53715
| | - Cody Corbett
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI53715
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA95616
| | - Andrew S. Fox
- Department of Psychology, University of California, Davis, CA95616
- California National Primate Research Center, University of California, Davis, CA95616
| | - Brian C. Trainor
- Department of Psychology, University of California, Davis, CA95616
| |
Collapse
|
15
|
Timme NM, Ardinger CE, Weir SDC, Zelaya-Escobar R, Kruger R, Lapish CC. Non-Consummatory Behavior Signals Predict Aversion-Resistant Alcohol Drinking in Head-Fixed Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.20.545767. [PMID: 37873153 PMCID: PMC10592797 DOI: 10.1101/2023.06.20.545767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
A key facet of alcohol use disorder is continuing to drink alcohol despite negative consequences (so called "aversion-resistant drinking"). In this study, we sought to assess the degree to which head-fixed mice exhibit aversion-resistant drinking and to leverage behavioral analysis techniques available in head-fixture to relate non-consummatory behaviors to aversion-resistant drinking. We assessed aversion-resistant drinking in head-fixed female and male C57BL/6J mice. We adulterated 20% (v/v) alcohol with varying concentrations of the bitter tastant quinine to measure the degree to which mice would continue to drink despite this aversive stimulus. We recorded high-resolution video of the mice during head-fixed drinking, tracked body parts with machine vision tools, and analyzed body movements in relation to consumption. Female and male head-fixed mice exhibited heterogenous levels of aversion-resistant drinking. Additionally, non-consummatory behaviors, such as paw movement and snout movement, were related to the intensity of aversion-resistant drinking. These studies demonstrate that head-fixed mice exhibit aversion-resistant drinking and that non-consummatory behaviors can be used to assess perceived aversiveness in this paradigm. Furthermore, these studies lay the groundwork for future experiments that will utilize advanced electrophysiological techniques to record from large populations of neurons during aversion-resistant drinking to understand the neurocomputational processes that drive this clinically relevant behavior.
Collapse
Affiliation(s)
- Nicholas M. Timme
- Department of Psychology, Indiana University – Purdue University Indianapolis, 402 N. Blackford St, LD 124, Indianapolis, IN, 46202, USA
| | - Cherish E. Ardinger
- Department of Psychology, Indiana University – Purdue University Indianapolis, 402 N. Blackford St, LD 124, Indianapolis, IN, 46202, USA
| | - Seth D. C. Weir
- Department of Psychology, Indiana University – Purdue University Indianapolis, 402 N. Blackford St, LD 124, Indianapolis, IN, 46202, USA
| | - Rachel Zelaya-Escobar
- Department of Psychology, Indiana University – Purdue University Indianapolis, 402 N. Blackford St, LD 124, Indianapolis, IN, 46202, USA
| | - Rachel Kruger
- Department of Psychology, Indiana University – Purdue University Indianapolis, 402 N. Blackford St, LD 124, Indianapolis, IN, 46202, USA
| | - Christopher C. Lapish
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, 635 Barnhill Drive, MSB 5035, Indianapolis, IN, 46202, USA
- Stark Neuroscience Institute, Indiana University School of Medicine, 320 W. 15 St, NB 414, Indianapolis, IN 46202, USA
| |
Collapse
|
16
|
Bender BN, Stringfield SJ, Torregrossa MM. Changes in dorsomedial striatum activity mediate expression of goal-directed vs. habit-like cue-induced cocaine seeking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550364. [PMID: 37546826 PMCID: PMC10402009 DOI: 10.1101/2023.07.24.550364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
A preclinical model of cue exposure therapy, cue extinction, reduces cue-induced cocaine seeking when drug seeking is goal-directed but not habitual. Goal-directed and habitual behaviors differentially rely on the dorsomedial striatum (DMS) and dorsolateral striatum (DLS), but the effects of cue extinction on dorsal striatal responses to cue-induced drug seeking are unknown. We used fiber photometry to examine how dorsal striatal intracellular calcium and extracellular dopamine activity differs between goal-directed and habitual cue-induced cocaine seeking and how it is impacted by cue extinction. Rats trained to self-administer cocaine paired with an audiovisual cue on schedules of reinforcement that promote goal-directed or habitual cocaine seeking had different patterns of dorsal striatal calcium and dopamine responses to cue-reinforced lever presses. Cue extinction reduced calcium and dopamine responses during subsequent drug seeking in the DMS, but not in the DLS. Therefore, cue extinction may reduce goal-directed behavior through its effects on the DMS, whereas habitual behavior and the DLS are unaffected.
Collapse
|
17
|
Kalelkar A, Sipe G, Costa ARCE, Lorenzo IM, Nguyen M, Linares-Garcia I, Vazey E, Huda R. A paradigm for ethanol consumption in head-fixed mice during prefrontal cortical two-photon calcium imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.20.549846. [PMID: 37503061 PMCID: PMC10370124 DOI: 10.1101/2023.07.20.549846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The prefrontal cortex (PFC) is a hub for higher-level cognitive behaviors and is a key target for neuroadaptations in alcohol use disorders. Preclinical models of ethanol consumption are instrumental for understanding how acute and repeated drinking affects PFC structure and function. Recent advances in genetically encoded sensors of neuronal activity and neuromodulator release combined with functional microscopy (multiphoton and one-photon widefield imaging) allow multimodal in-vivo PFC recordings at subcellular and cellular scales. While these methods could enable a deeper understanding of the relationship between alcohol and PFC function/dysfunction, they require animals to be head-fixed. Here, we present a method in mice for binge-like ethanol consumption during head-fixation. Male and female mice were first acclimated to ethanol by providing home cage access to 20% ethanol (v/v) for 4 or 8 days. After home cage drinking, mice consumed ethanol from a lick spout during head-fixation. We used two-photon calcium imaging during the head-fixed drinking paradigm to record from a large population of PFC neurons (>1000) to explore how acute ethanol affects their activity. Drinking modulated activity rates in a subset of neurons on slow (minutes) and fast (seconds) time scales but the majority of neurons were unaffected. Moreover, ethanol intake did not significantly affect network level interactions in the PFC as assessed through inter-neuronal pairwise correlations. By establishing a method for binge-like drinking in head-fixed mice, we lay the groundwork for leveraging advanced microscopy technologies to study alcohol-induced neuroadaptations in PFC and other brain circuits.
Collapse
Affiliation(s)
- Anagha Kalelkar
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University – New Brunswick, 604 Allison Road, Piscataway NJ, 08904, USA
| | - Grayson Sipe
- Department of Brain and Cognitive Science, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, 43 Vassar Street, Cambridge MA, 02139, USA
| | - Ana Raquel Castro E Costa
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University – New Brunswick, 604 Allison Road, Piscataway NJ, 08904, USA
| | - Ilka M. Lorenzo
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University – New Brunswick, 604 Allison Road, Piscataway NJ, 08904, USA
| | - My Nguyen
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University – New Brunswick, 604 Allison Road, Piscataway NJ, 08904, USA
| | - Ivan Linares-Garcia
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University – New Brunswick, 604 Allison Road, Piscataway NJ, 08904, USA
| | - Elena Vazey
- Department of Biology, The University of Massachusetts Amherst, 611 North Pleasant Street, Amherst MA, 01003, USA
| | - Rafiq Huda
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University – New Brunswick, 604 Allison Road, Piscataway NJ, 08904, USA
| |
Collapse
|
18
|
Brown AR, Branthwaite HE, Farahbakhsh ZZ, Mukerjee S, Melugin PR, Song K, Noamany H, Siciliano CA. Structured tracking of alcohol reinforcement (STAR) for basic and translational alcohol research. Mol Psychiatry 2023; 28:1585-1598. [PMID: 36849824 PMCID: PMC10208967 DOI: 10.1038/s41380-023-01994-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/26/2023] [Accepted: 02/07/2023] [Indexed: 03/01/2023]
Abstract
There is inherent tension between methodologies developed to address basic research questions in model species and those intended for preclinical to clinical translation: basic investigations require flexibility of experimental design as hypotheses are rapidly tested and revised, whereas preclinical models emphasize standardized protocols and specific outcome measures. This dichotomy is particularly relevant in alcohol research, which spans a diverse range of basic sciences in addition to intensive efforts towards understanding the pathophysiology of alcohol use disorder (AUD). To advance these goals there is a great need for approaches that facilitate synergy across basic and translational areas of nonhuman alcohol research. In male and female mice, we establish a modular alcohol reinforcement paradigm: Structured Tracking of Alcohol Reinforcement (STAR). STAR provides a robust platform for quantitative assessment of AUD-relevant behavioral domains within a flexible framework that allows direct crosstalk between translational and mechanistically oriented studies. To achieve cross-study integration, despite disparate task parameters, a straightforward multivariate phenotyping analysis is used to classify subjects based on propensity for heightened alcohol consumption and insensitivity to punishment. Combining STAR with extant preclinical alcohol models, we delineate longitudinal phenotype dynamics and reveal putative neuro-biomarkers of heightened alcohol use vulnerability via neurochemical profiling of cortical and brainstem tissues. Together, STAR allows quantification of time-resolved biobehavioral processes essential for basic research questions simultaneous with longitudinal phenotyping of clinically relevant outcomes, thereby providing a framework to facilitate cohesion and translation in alcohol research.
Collapse
Affiliation(s)
- Alex R Brown
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Hannah E Branthwaite
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Zahra Z Farahbakhsh
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Snigdha Mukerjee
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Patrick R Melugin
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Keaton Song
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Habiba Noamany
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
19
|
Bijoch Ł, Włodkowska U, Kasztelanic R, Pawłowska M, Pysz D, Kaczmarek L, Lapkiewicz R, Buczyński R, Czajkowski R. Novel Design and Application of High-NA Fiber Imaging Bundles for In Vivo Brain Imaging with Two-Photon Scanning Fluorescence Microscopy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12831-12841. [PMID: 36880640 PMCID: PMC10020965 DOI: 10.1021/acsami.2c22985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/21/2023] [Indexed: 06/18/2023]
Abstract
Here, we provide experimental verification supporting the use of short-section imaging bundles for two-photon microscopy imaging of the mouse brain. The 8 mm long bundle is made of a pair of heavy-metal oxide glasses with a refractive index contrast of 0.38 to ensure a high numerical aperture NA = 1.15. The bundle is composed of 825 multimode cores, ordered in a hexagonal lattice with a pixel size of 14 μm and a total diameter of 914 μm. We demonstrate successful imaging through custom-made bundles with 14 μm resolution. As the input, we used a 910 nm Ti-sapphire laser with 140 fs pulse and a peak power of 9 × 104 W. The excitation beam and fluorescent image were transferred through the fiber imaging bundle. As test samples, we used 1 μm green fluorescent latex beads, ex vivo hippocampal neurons expressing green fluorescent protein and cortical neurons in vivo expressing the fluorescent reporter GCaMP6s or immediate early gene Fos fluorescent reporter. This system can be used for minimal-invasive in vivo imaging of the cerebral cortex, hippocampus, or deep brain areas as a part of a tabletop system or an implantable setup. It is a low-cost solution, easy to integrate and operate for high-throughput experiments.
Collapse
Affiliation(s)
- Łukasz Bijoch
- BRAINCITY, Nencki Institute of Experimental Biology PAS, Pasteura 3, 02-093 Warszawa, Poland
| | - Urszula Włodkowska
- Nencki
Institute of Experimental Biology PAS, Pasteura 3, 02-093 Warszawa, Poland
| | - Rafał Kasztelanic
- Faculty
of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland
- Institute
of Microelectronics and Photonics, Lukasiewicz
Research Network, Al.
Lotników 32/46, 02-668 Warsaw, Poland
| | - Monika Pawłowska
- BRAINCITY, Nencki Institute of Experimental Biology PAS, Pasteura 3, 02-093 Warszawa, Poland
- Faculty
of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland
| | - Dariusz Pysz
- Institute
of Microelectronics and Photonics, Lukasiewicz
Research Network, Al.
Lotników 32/46, 02-668 Warsaw, Poland
| | - Leszek Kaczmarek
- BRAINCITY, Nencki Institute of Experimental Biology PAS, Pasteura 3, 02-093 Warszawa, Poland
| | - Radek Lapkiewicz
- Faculty
of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland
| | - Ryszard Buczyński
- Faculty
of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland
- Institute
of Microelectronics and Photonics, Lukasiewicz
Research Network, Al.
Lotników 32/46, 02-668 Warsaw, Poland
| | - Rafał Czajkowski
- Nencki
Institute of Experimental Biology PAS, Pasteura 3, 02-093 Warszawa, Poland
| |
Collapse
|
20
|
Abstract
Ethanol (EtOH) has effects on numerous cellular molecular targets, and alterations in synaptic function are prominent among these effects. Acute exposure to EtOH activates or inhibits the function of proteins involved in synaptic transmission, while chronic exposure often produces opposing and/or compensatory/homeostatic effects on the expression, localization, and function of these proteins. Interactions between different neurotransmitters (e.g., neuropeptide effects on release of small molecule transmitters) can also influence both acute and chronic EtOH actions. Studies in intact animals indicate that the proteins affected by EtOH also play roles in the neural actions of the drug, including acute intoxication, tolerance, dependence, and the seeking and drinking of EtOH. The present chapter is an update of our previous Lovinger and Roberto (Curr Top Behav Neurosci 13:31-86, 2013) chapter and reviews the literature describing these acute and chronic synaptic effects of EtOH with a focus on adult animals and their relevance for synaptic transmission, plasticity, and behavior.
Collapse
Affiliation(s)
- David M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism (NIAAA), Rockville, MD, USA
| | - Marisa Roberto
- Molecular Medicine Department, Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
21
|
Marini P, Cowie P, Ayar A, Bewick GS, Barrow J, Pertwee RG, MacKenzie A, Tucci P. M3 Receptor Pathway Stimulates Rapid Transcription of the CB1 Receptor Activation through Calcium Signalling and the CNR1 Gene Promoter. Int J Mol Sci 2023; 24:ijms24021308. [PMID: 36674826 PMCID: PMC9867084 DOI: 10.3390/ijms24021308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/17/2022] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
In this study, we have investigated a possible mechanism that enables CB1/M3 receptor cross-talk, using SH-SY5Y cells as a model system. Our results show that M3 receptor activation initiates signaling that rapidly upregulates the CNR1 gene, resulting in a greatly potentiated CB1 receptor response to agonists. Calcium homeostasis plays an essential intermediary role in this functional CB1/M3 receptor cross-talk. We show that M3 receptor-triggered calcium release greatly increases CB1 receptor expression via both transcriptional and translational activity, by enhancing CNR1 promoter activity. The co-expression of M3 and CB1 receptors in brain areas such as the nucleus accumbens and amygdala support the hypothesis that the altered synaptic plasticity observed after exposure to cannabinoids involves cross-talk with the M3 receptor subtype. In this context, M3 receptors and their interaction with the cannabinoid system at the transcriptional level represent a potential pharmacogenomic target not only for the develop of new drugs for addressing addiction and tolerance. but also to understand the mechanisms underpinning response stratification to cannabinoids.
Collapse
Affiliation(s)
- Pietro Marini
- Institute of Education in Healthcare and Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Philip Cowie
- The Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Ahmet Ayar
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, 61080 Trabzon, Turkey
| | - Guy S. Bewick
- The Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - John Barrow
- Institute of Education in Healthcare and Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Roger G. Pertwee
- The Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Alasdair MacKenzie
- The Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
- Correspondence:
| |
Collapse
|
22
|
Aghi K, Goetz TG, Pfau DR, Sun SED, Roepke TA, Guthman EM. Centering the Needs of Transgender, Nonbinary, and Gender-Diverse Populations in Neuroendocrine Models of Gender-Affirming Hormone Therapy. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:1268-1279. [PMID: 35863692 PMCID: PMC10472479 DOI: 10.1016/j.bpsc.2022.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/20/2022] [Accepted: 07/06/2022] [Indexed: 02/07/2023]
Abstract
Most studies attempting to address the health care needs of the millions of transgender, nonbinary, and/or gender-diverse (TNG) individuals rely on human subjects, overlooking the benefits of translational research in animal models. Researchers have identified many ways in which gonadal steroid hormones regulate neuronal gene expression, connectivity, activity, and function across the brain to control behavior. However, these discoveries primarily benefit cisgender populations. Research into the effects of exogenous hormones such as estradiol, testosterone, and progesterone has a direct translational benefit for TNG individuals on gender-affirming hormone therapies (GAHTs). Despite this potential, endocrinological health care for TNG individuals remains largely unimproved. Here, we outline important areas of translational research that could address the unique health care needs of TNG individuals on GAHT. We highlight key biomedical questions regarding GAHT that can be investigated using animal models. We discuss how contemporary research fails to address the needs of GAHT users and identify equitable practices for cisgender scientists engaging with this work. We conclude that if necessary and important steps are taken to address these issues, translational research on GAHTs will greatly benefit the health care outcomes of TNG people.
Collapse
Affiliation(s)
- Krisha Aghi
- Helen Wills Neuroscience Institute, University of California, Berkeley, California
| | - Teddy G Goetz
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel R Pfau
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan; Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Simón E D Sun
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York; Center for Applied Transgender Studies, Chicago, Illinois
| | - Troy A Roepke
- Department of Animal Sciences, School of Biological and Environmental Sciences, Rutgers University, New Brunswick
| | - Eartha Mae Guthman
- Center for Applied Transgender Studies, Chicago, Illinois; Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey.
| |
Collapse
|
23
|
Nall RW, Chalhoub RM, Kalivas PW. Drug versus non-drug behaviors: A dual-reward model of sex differences and neurobiological mechanisms in rats. J Exp Anal Behav 2022; 117:457-471. [PMID: 35297047 PMCID: PMC10775707 DOI: 10.1002/jeab.752] [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: 10/15/2021] [Revised: 02/02/2022] [Accepted: 02/09/2022] [Indexed: 11/05/2022]
Abstract
Substance Use Disorders (SUDs) are an impactful problem characterized by chronic relapse and engagement in drug-related behaviors at the expense of non-drug behaviors. Brain regions implicated in drug and non-drug-related behaviors often overlap, complicating investigations of neurobiological mechanisms underlying SUDs. Here we presented a within-subject model for studying self-administration, reinforcer competition, extinction, and cued reinstatement of cocaine- and food-seeking in rats. Due to differences in cocaine- and food-reinforced behavior, we transformed data to proportions of baseline, revealing increased resistance to extinction and disproportionately greater cued reinstatement of cocaine seeking relative to food seeking. Consistent with previous reports, females showed greater preference for cocaine reinforcement than males, though these findings failed to reach statistical significance. To demonstrate the model's utility for investigating neurobiological mechanisms, we included proof-of-concept calcium imaging data demonstrating the utility of the behavioral model for detecting cellular activity patterns associated with cocaine- and food-seeking behaviors. Future studies utilizing this model should improve understanding of the development and expression of pathological behaviors characteristic of SUDs in humans, sex differences in these behaviors, and their neurobiological correlates. Thus, the model has utility for improving understanding of SUDs, leading to novel treatments to reduce the pathological behaviors associated with SUDs.
Collapse
Affiliation(s)
- Rusty W. Nall
- Medical University of South Carolina
- Jacksonville State University
| | | | | |
Collapse
|
24
|
Neurogenesis mediated plasticity is associated with reduced neuronal activity in CA1 during context fear memory retrieval. Sci Rep 2022; 12:7016. [PMID: 35488117 PMCID: PMC9054819 DOI: 10.1038/s41598-022-10947-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/14/2022] [Indexed: 12/17/2022] Open
Abstract
Postnatal hippocampal neurogenesis has been demonstrated to affect learning and memory in numerous ways. Several studies have now demonstrated that increased neurogenesis can induce forgetting of memories acquired prior to the manipulation of neurogenesis and, as a result of this forgetting can also facilitate new learning. However, the mechanisms mediating neurogenesis-induced forgetting are not well understood. Here, we used a subregion-based analysis of the immediate early gene c-Fos as well as in vivo fiber photometry to determine changes in activity corresponding with neurogenesis induced forgetting. We found that increasing neurogenesis led to reduced CA1 activity during context memory retrieval. We also demonstrate here that perineuronal net expression in areas CA1 is bidirectionally altered by the levels or activity of postnatally generated neurons in the dentate gyrus. These results suggest that neurogenesis may induce forgetting by disrupting perineuronal nets in CA1 which may otherwise protect memories from degradation.
Collapse
|
25
|
Lovinger DM, Mateo Y, Johnson KA, Engi SA, Antonazzo M, Cheer JF. Local modulation by presynaptic receptors controls neuronal communication and behaviour. Nat Rev Neurosci 2022; 23:191-203. [PMID: 35228740 PMCID: PMC10709822 DOI: 10.1038/s41583-022-00561-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2022] [Indexed: 12/15/2022]
Abstract
Central nervous system neurons communicate via fast synaptic transmission mediated by ligand-gated ion channel (LGIC) receptors and slower neuromodulation mediated by G protein-coupled receptors (GPCRs). These receptors influence many neuronal functions, including presynaptic neurotransmitter release. Presynaptic LGIC and GPCR activation by locally released neurotransmitters influences neuronal communication in ways that modify effects of somatic action potentials. Although much is known about presynaptic receptors and their mechanisms of action, less is known about when and where these receptor actions alter release, especially in vivo. This Review focuses on emerging evidence for important local presynaptic receptor actions and ideas for future studies in this area.
Collapse
Affiliation(s)
- David M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA.
| | - Yolanda Mateo
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Kari A Johnson
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Sheila A Engi
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mario Antonazzo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joseph F Cheer
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
26
|
A Conditioned Place Preference for Heroin Is Signaled by Increased Dopamine and Direct Pathway Activity and Decreased Indirect Pathway Activity in the Nucleus Accumbens. J Neurosci 2022; 42:2011-2024. [PMID: 35031576 PMCID: PMC8916759 DOI: 10.1523/jneurosci.1451-21.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 11/21/2022] Open
Abstract
Repeated pairing of a drug with a neutral stimulus, such as a cue or context, leads to the attribution of the drug's reinforcing properties to that stimulus, and exposure to that stimulus in the absence of the drug can elicit drug-seeking. A principal role for the NAc in the response to drug-associated stimuli has been well documented. Direct and indirect pathway medium spiny neurons (dMSNs and iMSNs) have been shown to bidirectionally regulate cue-induced heroin-seeking in rats expressing addiction-like phenotypes, and a shift in NAc activity toward the direct pathway has been shown in mice following cocaine conditioned place preference (CPP). However, how NAc signaling guides heroin CPP, and whether heroin alters the balance of signaling between dMSNs and iMSNs, remains unknown. Moreover, the role of NAc dopamine signaling in heroin reinforcement is unclear. Here, we integrate fiber photometry for in vivo monitoring of dopamine and dMSN/iMSN calcium activity with a heroin CPP procedure in rats to begin to address these questions. We identify a sensitization-like response to heroin in the NAc, with prominent iMSN activity during initial heroin exposure and prominent dMSN activity following repeated heroin exposure. We demonstrate a ramp in dopamine activity, dMSN activation, and iMSN inactivation preceding entry into a heroin-paired context, and a decrease in dopamine activity, dMSN inactivation, and iMSN activation preceding exit from a heroin-paired context. Finally, we show that buprenorphine is sufficient to prevent the development of heroin CPP and reduce Fos activation in the NAc after conditioning. Together, these data support the hypothesis that an imbalance in NAc activity contributes to the development of drug-cue associations that can drive addiction processes.SIGNIFICANCE STATEMENT The attribution of the reinforcing effects of drugs to neutral stimuli (e.g., cues and contexts) contributes to the long-standing nature of addiction, as re-exposure to drug-associated stimuli can reinstate drug-seeking and -taking even after long periods of abstinence. The NAc has an established role in encoding the value of drug-associated stimuli, and dopamine release into the NAc is known to modulate the reinforcing effects of drugs, including heroin. Using fiber photometry, we show that entering a heroin-paired context is driven by dopamine signaling and NAc direct pathway activation, whereas exiting a heroin-paired context is driven by NAc indirect pathway activation. This study provides further insight into the role of NAc microcircuitry in encoding the reinforcing properties of heroin.
Collapse
|
27
|
Namkung H, Thomas KL, Hall J, Sawa A. Parsing neural circuits of fear learning and extinction across basic and clinical neuroscience: Towards better translation. Neurosci Biobehav Rev 2022; 134:104502. [PMID: 34921863 DOI: 10.1016/j.neubiorev.2021.12.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/22/2022]
Abstract
Over the past decades, studies of fear learning and extinction have advanced our understanding of the neurobiology of threat and safety learning. Animal studies can provide mechanistic/causal insights into human brain regions and their functional connectivity involved in fear learning and extinction. Findings in humans, conversely, may further enrich our understanding of neural circuits in animals by providing macroscopic insights at the level of brain-wide networks. Nevertheless, there is still much room for improvement in translation between basic and clinical research on fear learning and extinction. Through the lens of neural circuits, in this article, we aim to review the current knowledge of fear learning and extinction in both animals and humans, and to propose strategies to fill in the current knowledge gap for the purpose of enhancing clinical benefits.
Collapse
Affiliation(s)
- Ho Namkung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Kerrie L Thomas
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK; School of Biosciences, Cardiff University, Cardiff, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK; School of Medicine, Cardiff University, Cardiff, UK
| | - Akira Sawa
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21287, USA.
| |
Collapse
|
28
|
The effect of self-administered methamphetamine on GABAergic interneuron populations and functional connectivity of the nucleus accumbens and prefrontal cortex. Psychopharmacology (Berl) 2022; 239:2903-2919. [PMID: 35920922 PMCID: PMC9385811 DOI: 10.1007/s00213-022-06175-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 06/08/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Methamphetamine (METH, "ice") is a potent and addictive psychostimulant. Abuse of METH perturbs neurotransmitter systems and induces neurotoxicity; however, the neurobiological mechanisms which underlie addiction to METH are not fully understood, limiting the efficacy of available treatments. Here we investigate METH-induced changes to neuronal nitric oxide synthase (nNOS), parvalbumin and calretinin-expressing GABAergic interneuron populations within the nucleus accumbens (NAc), prefrontal cortex (PFC) and orbitofrontal cortex (OFC). We hypothesise that dysfunction or loss of these GABAergic interneuron populations may disrupt the excitatory/inhibitory balance within the brain. METHODS Male Long Evans rats (N = 32) were trained to lever press for intravenous METH or received yoked saline infusions. Following 14 days of behavioural extinction, animals were given a non-contingent injection of saline or METH (1 mg/kg, IP) to examine drug-primed reinstatement to METH-seeking behaviours. Ninety minutes post-IP injection, animals were culled and brain sections were analysed for Fos, nNOS, parvalbumin and calretinin immunoreactivity in eight distinct subregions of the NAc, PFC and OFC. RESULTS METH exposure differentially affected GABAergic populations, with METH self-administration increasing nNOS immunoreactivity at distinct locations in the prelimbic cortex and decreasing parvalbumin immunoreactivity in the NAc. METH self-administration triggered reduced calretinin immunoreactivity, whilst acute METH administration produced a significant increase in calretinin immunoreactivity. As expected, non-contingent METH-priming treatment increased Fos immunoreactivity in subregions of the NAc and PFC. CONCLUSION Here we report that METH exposure in this model may alter the function of GABAergic interneurons in more subtle ways, such as alterations in neuronal firing or synaptic connectivity.
Collapse
|
29
|
Vickstrom CR, Snarrenberg ST, Friedman V, Liu QS. Application of optogenetics and in vivo imaging approaches for elucidating the neurobiology of addiction. Mol Psychiatry 2022; 27:640-651. [PMID: 34145393 PMCID: PMC9190069 DOI: 10.1038/s41380-021-01181-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/26/2021] [Indexed: 02/05/2023]
Abstract
The neurobiology of addiction has been an intense topic of investigation for more than 50 years. Over this time, technological innovation in methods for studying brain function rapidly progressed, leading to increasingly sophisticated experimental approaches. To understand how specific brain regions, cell types, and circuits are affected by drugs of abuse and drive behaviors characteristic of addiction, it is necessary both to observe and manipulate neural activity in addiction-related behavioral paradigms. In pursuit of this goal, there have been several key technological advancements in in vivo imaging and neural circuit modulation in recent years, which have shed light on the cellular and circuit mechanisms of addiction. Here we discuss some of these key technologies, including circuit modulation with optogenetics, in vivo imaging with miniaturized single-photon microscopy (miniscope) and fiber photometry, and how the application of these technologies has garnered novel insights into the neurobiology of addiction.
Collapse
|
30
|
Vassilev P, Fonseca E, Hernandez G, Pantoja-Urban AH, Giroux M, Nouel D, Van Leer E, Flores C. Custom-Built Operant Conditioning Setup for Calcium Imaging and Cognitive Testing in Freely Moving Mice. eNeuro 2022; 9:ENEURO.0430-21.2022. [PMID: 35105659 PMCID: PMC8856704 DOI: 10.1523/eneuro.0430-21.2022] [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: 10/08/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 11/21/2022] Open
Abstract
Operant chambers are widely used in animal research to study cognition, motivation, and learning processes. Paired with the rapidly developing technologies for brain imaging and manipulations of brain activity, operant conditioning chambers are a powerful tool for neuroscience research. The behavioral testing and imaging setups that are commercially available are often quite costly. Here, we present a custom-built operant chamber that can be constructed in a few days by an unexperienced user with relatively inexpensive, widely available materials. The advantages of our operant setup compared with other open-source and closed-source solutions are its relatively low cost, its support of complex behavioral tasks, its user-friendly setup, and its validated functionality with video imaging of behavior and calcium imaging using the UCLA Miniscope. Using this setup, we replicate our previously published findings showing that mice exposed to social defeat stress in adolescence have inhibitory control impairments in the Go/No-Go task when they reach adulthood. We also present calcium imaging data of medial prefrontal cortex (mPFC) neuronal activity acquired during Go/No-Go testing in freely moving mice and show that neuronal population activity increases from day 1 to day 14 of the task. We propose that our operant chamber is a cheaper alternative to its commercially available counterparts and offers a better balance between versatility and user-friendly setup than other open-source alternatives.
Collapse
Affiliation(s)
- Philip Vassilev
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Montréal, QC, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, QC, H4H 1R3, Canada
| | - Esmeralda Fonseca
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08540
| | - Giovanni Hernandez
- Douglas Mental Health University Institute, Montreal, QC, H4H 1R3, Canada
| | | | - Michel Giroux
- Douglas Mental Health University Institute, Montreal, QC, H4H 1R3, Canada
| | - Dominique Nouel
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08540
| | - Elise Van Leer
- Douglas Mental Health University Institute, Montreal, QC, H4H 1R3, Canada
| | - Cecilia Flores
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Montréal, QC, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, QC, H4H 1R3, Canada
| |
Collapse
|
31
|
Broussard GJ, Petreanu L. Eavesdropping wires: Recording activity in axons using genetically encoded calcium indicators. J Neurosci Methods 2021; 360:109251. [PMID: 34119572 PMCID: PMC8363211 DOI: 10.1016/j.jneumeth.2021.109251] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/31/2021] [Accepted: 06/05/2021] [Indexed: 12/23/2022]
Abstract
Neurons broadcast electrical signals to distal brain regions through extensive axonal arbors. Genetically encoded calcium sensors permit the direct observation of action potential activity at axonal terminals, providing unique insights on the organization and function of neural projections. Here, we consider what information can be gleaned from axonal recordings made at scales ranging from the summed activity extracted from multi-cell axon projections to single boutons. In particular, we discuss the application of different recently developed multi photon and fiber photometry methods for recording neural activity in axons of rodents. We define experimental difficulties associated with imaging approaches in the axonal compartment and highlight the latest methodological advances for addressing these issues. Finally, we reflect on ways in which new technologies can be used in conjunction with axon calcium imaging to address current questions in neurobiology.
Collapse
Affiliation(s)
| | - Leopoldo Petreanu
- Champalimaud Research, Champalimaud Center for the Unknown, Lisbon, Portugal.
| |
Collapse
|
32
|
Nall RW, Heinsbroek JA, Nentwig TB, Kalivas PW, Bobadilla AC. Circuit selectivity in drug versus natural reward seeking behaviors. J Neurochem 2021; 157:1450-1472. [PMID: 33420731 PMCID: PMC8178159 DOI: 10.1111/jnc.15297] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/16/2020] [Accepted: 01/03/2021] [Indexed: 12/23/2022]
Abstract
Substance use disorder (SUD) is characterized, in part by behavior biased toward drug use and away from natural sources of reward (e.g., social interaction, food, sex). The neurobiological underpinnings of SUDs reveal distinct brain regions where neuronal activity is necessary for the manifestation of SUD-characteristic behaviors. Studies that specifically examine how these regions are involved in behaviors motivated by drug versus natural reward allow determinations of which regions are necessary for regulating seeking of both reward types, and appraisals of novel SUD therapies for off-target effects on behaviors motivated by natural reward. Here, we evaluate studies directly comparing regulatory roles for specific brain regions in drug versus natural reward. While it is clear that many regions drive behaviors motivated by all reward types, based on the literature reviewed we propose a set of interconnected regions that become necessary for behaviors motivated by drug, but not natural rewards. The circuitry is selectively necessary for drug seeking includes an Action/Reward subcircuit, comprising nucleus accumbens, ventral pallidum, and ventral tegmental area, a Prefrontal subcircuit comprising prelimbic, infralimbic, and insular cortices, a Stress subcircuit comprising the central nucleus of the amygdala and the bed nucleus of the stria terminalis, and a Diencephalon circuit including lateral hypothalamus. Evidence was mixed for nucleus accumbens shell, insular cortex, and ventral pallidum. Studies for all other brain nuclei reviewed supported a necessary role in regulating both drug and natural reward seeking. Finally, we discuss emerging strategies to further disambiguate the necessity of brain regions in drug- versus natural reward-associated behaviors.
Collapse
Affiliation(s)
- Rusty W. Nall
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Jasper A. Heinsbroek
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Todd B. Nentwig
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- These authors share senior authorship
| | - Ana-Clara Bobadilla
- School of Pharmacy, University of Wyoming, Laramie, WY, USA
- These authors share senior authorship
| |
Collapse
|
33
|
Jones-Tabah J, Mohammad H, Clarke PBS, Hébert TE. In vivo detection of GPCR-dependent signaling using fiber photometry and FRET-based biosensors. Methods 2021; 203:422-430. [PMID: 34022351 DOI: 10.1016/j.ymeth.2021.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/03/2021] [Accepted: 05/11/2021] [Indexed: 12/27/2022] Open
Abstract
Genetically encoded fluorescent biosensors allow intracellular signaling dynamics to be tracked in live cells and tissues using optical detection. Many such biosensors are based on the principle of Förster resonance energy transfer (FRET), and we have recently developed a simple approach for in vivo detection of FRET-based biosensor signals using fiber photometry. By combining fiber photometry with FRET-based biosensors, we were able to track GPCR-dependent signaling pathways over time, and in response to drug treatments in freely-moving adult rats. Recording from specific neuronal populations, we can quantify intracellular signaling while simultaneously measuring behavioral responses. Our approach, described in detail here, uses adeno-associated viruses infused intracerebrally in order to express genetically-encoded FRET-based biosensors. After several weeks to allow biosensor expression, fiber photometry is used in order to record drug responses in real time from freely-moving adult rats. This methodology would be compatible with other mammalian species and with many biosensors. Hence, it has wide applicability across a spectrum of neuroscience research, ranging from the study of neural circuits and behavior, to preclinical drug development and screening.
Collapse
Affiliation(s)
- Jace Jones-Tabah
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Hanan Mohammad
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Paul B S Clarke
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
34
|
Role of protons in calcium signaling. Biochem J 2021; 478:895-910. [PMID: 33635336 DOI: 10.1042/bcj20200971] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 02/03/2023]
Abstract
Thirty-six years after the publication of the important article by Busa and Nuccitelli on the variability of intracellular pH (pHi) and the interdependence of pHi and intracellular Ca2+ concentration ([Ca2+]i), little research has been carried out on pHi and calcium signaling. Moreover, the results appear to be contradictory. Some authors claim that the increase in [Ca2+]i is due to a reduction in pHi, others that it is caused by an increase in pHi. The reasons for these conflicting results have not yet been discussed and clarified in an exhaustive manner. The idea that variations in pHi are insignificant, because cellular buffers quickly stabilize the pHi, may be a limiting and fundamentally wrong concept. In fact, it has been shown that protons can move and react in the cell before they are neutralized. Variations in pHi have a remarkable impact on [Ca2+]i and hence on some of the basic biochemical mechanisms of calcium signaling. This paper focuses on the possible triggering role of protons during their short cellular cycle and it suggests a new hypothesis for an IP3 proton dependent mechanism of action.
Collapse
|
35
|
Li M, Cabrera-Garcia D, Salling MC, Au E, Yang G, Harrison NL. Alcohol reduces the activity of somatostatin interneurons in the mouse prefrontal cortex: A neural basis for its disinhibitory effect? Neuropharmacology 2021; 188:108501. [PMID: 33636191 DOI: 10.1016/j.neuropharm.2021.108501] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/01/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
The prefrontal cortex (PFC) is involved in executive ("top-down") control of behavior and its function is especially susceptible to the effects of alcohol, leading to behavioral disinhibition that is associated with alterations in decision making, response inhibition, social anxiety and working memory. The circuitry of the PFC involves a complex interplay between pyramidal neurons (PNs) and several subclasses of inhibitory interneurons (INs), including somatostatin (SST)-expressing INs. Using in vivo calcium imaging, we showed that alcohol dose-dependently altered network activity in layers 2/3 of the prelimbic subregion of the mouse PFC. Low doses of alcohol (1 g/kg, intraperitoneal, i.p.) caused moderate activation of SST INs and weak inhibition of PNs. At moderate to high doses, alcohol (2-3 g/kg) strongly inhibited the activity of SST INs in vivo, and this effect may result in disinhibition, as the activity of a subpopulation of PNs was simultaneously enhanced. In contrast, recordings in brain slices using ex vivo electrophysiology revealed no direct effect of alcohol on the excitability of either SST INs or PNs over a range of concentrations (20 and 50 mM) consistent with the blood alcohol levels reached in the in vivo experiments. This dose-dependent effect of alcohol on SST INs in vivo may reveal a neural basis for the disinhibitory effect of alcohol in the PFC mediated by other neurons within or external to the PFC circuitry.
Collapse
Affiliation(s)
- Miao Li
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA
| | - David Cabrera-Garcia
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA
| | - Michael C Salling
- Louisiana State University, Department of Anatomy, New Orleans, LA, 1901 Perdido Street, New Orleans, LA, 70112, USA
| | - Edmund Au
- Columbia University, Department of Pathology & Cell Biology and Rehabilitative Medicine and Regeneration, Columbia Translational Neuroscience Initiative Scholar, 630 West 168th Street, New York, NY, 10032, USA
| | - Guang Yang
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA.
| | - Neil L Harrison
- Columbia University, Department of Anesthesiology, 630 West 168th Street, New York, NY, 10032, USA; Columbia University, Department of Molecular Pharmacology and Therapeutics, 630 West 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
36
|
Bruno CA, O'Brien C, Bryant S, Mejaes JI, Estrin DJ, Pizzano C, Barker DJ. pMAT: An open-source software suite for the analysis of fiber photometry data. Pharmacol Biochem Behav 2021; 201:173093. [PMID: 33385438 PMCID: PMC7853640 DOI: 10.1016/j.pbb.2020.173093] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/17/2020] [Accepted: 12/15/2020] [Indexed: 10/22/2022]
Abstract
The combined development of new technologies for neuronal recordings and the development of novel sensors for recording both cellular activity and neurotransmitter binding has ushered in a new era for the field of neuroscience. Among these new technologies is fiber photometry, a technique wherein an implanted fiber optic is used to record signals from genetically encoded fluorescent sensors in bulk tissue. Fiber photometry has been widely adapted due to its cost-effectiveness, ability to examine the activity of neurons with specific anatomical or genetic identities, and the ability to use these highly modular systems to record from one or more sensors or brain sites in both superficial and deep-brain structures. Despite these many benefits, one major hurdle for laboratories adopting this technique is the steep learning curve associated with the analysis of fiber photometry data. This has been further complicated by a lack of standardization in analysis pipelines. In the present communication, we present pMAT, a 'photometry modular analysis tool' that allows users to accomplish common analysis routines through the use of a graphical user interface. This tool can be deployed in MATLAB and edited by more advanced users, but is also available as an independently deployable, open-source application.
Collapse
Affiliation(s)
- Carissa A Bruno
- Department of Psychology, Rutgers, The State University of New Jersey, United States of America
| | - Chris O'Brien
- Department of Psychology, Rutgers, The State University of New Jersey, United States of America
| | - Svetlana Bryant
- Department of Psychology, Rutgers, The State University of New Jersey, United States of America
| | - Jennifer I Mejaes
- Department of Psychology, Rutgers, The State University of New Jersey, United States of America
| | - David J Estrin
- Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, United States of America
| | - Carina Pizzano
- Department of Psychology, Rutgers, The State University of New Jersey, United States of America
| | - David J Barker
- Department of Psychology, Rutgers, The State University of New Jersey, United States of America.
| |
Collapse
|
37
|
Wang Y, DeMarco EM, Witzel LS, Keighron JD. A selected review of recent advances in the study of neuronal circuits using fiber photometry. Pharmacol Biochem Behav 2021; 201:173113. [PMID: 33444597 DOI: 10.1016/j.pbb.2021.173113] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/17/2020] [Accepted: 01/06/2021] [Indexed: 12/21/2022]
Abstract
To understand the correlation between animal behaviors and the underlying neuronal circuits, it is important to monitor and record neurotransmission in the brain of freely moving animals. With the development of fiber photometry, based on genetically encoded biosensors, and novel electrochemical biosensors, it is possible to measure some key neuronal transmission events specific to cell types or neurotransmitters of interest with high temporospatial resolution. This review discusses the recent advances and achievements of these two techniques in the study of neurotransmission in animal models and how they can be used to complement other techniques in the neuroscientist's toolbox.
Collapse
Affiliation(s)
- Yuanmo Wang
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
| | - Emily M DeMarco
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Program in Neuroscience, University of Maryland, Baltimore, MD 21201, USA
| | - Lisa Sophia Witzel
- Department of Biological and Chemical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA
| | - Jacqueline D Keighron
- Department of Biological and Chemical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA.
| |
Collapse
|
38
|
Melugin PR, Nolan SO, Siciliano CA. Bidirectional causality between addiction and cognitive deficits. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 157:371-407. [PMID: 33648674 PMCID: PMC8566632 DOI: 10.1016/bs.irn.2020.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cognitive deficits are highly comorbid with substance use disorders. Deficits span multiple cognitive domains, are associated with disease severity across substance classes, and persist long after cessation of substance use. Furthermore, recovery of cognitive function during protracted abstinence is highly predictive of treatment adherence, relapse, and overall substance use disorder prognosis, suggesting that addiction may be best characterized as a disease of executive dysfunction. While the association between cognitive deficits and substance use disorders is clear, determining causalities is made difficult by the complex interplay between these variables. Cognitive dysfunction present prior to first drug use can act as a risk factor for substance use initiation, likelihood of pathology, and disease trajectory. At the same time, substance use can directly cause cognitive impairments even in individuals without preexisting deficits. Thus, parsing preexisting risk factors from substance-induced adaptations, and how they may interact, poses significant challenges. Here, focusing on psychostimulants and alcohol, we review evidence from clinical literature implicating cognitive deficits as a risk factor for addiction, a consequence of substance use, and the role the prefrontal cortex plays in these phenomena. We then review corresponding preclinical literature, highlighting the high degree of congruency between animal and human studies, and emphasize the unique opportunity that animal models provide to test causality between cognitive phenotypes and substance use, and to investigate the underlying neurobiology at a cellular and molecular level. Together, we provide an accessible resource for assessing the validity and utility of forward- and reverse-translation between these clinical and preclinical literatures.
Collapse
Affiliation(s)
- Patrick R Melugin
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, United States
| | - Suzanne O Nolan
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, United States
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, United States.
| |
Collapse
|
39
|
Evolution of in vivo dopamine monitoring techniques. Pharmacol Biochem Behav 2020; 200:173078. [PMID: 33278398 DOI: 10.1016/j.pbb.2020.173078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/06/2020] [Accepted: 11/18/2020] [Indexed: 01/01/2023]
Abstract
The brain dopamine system is central to numerous behavioral processes, including movement, learning, and motivation. Accordingly, disruptions of this neural system underlie numerous neurological and psychiatric disorders. Current understanding of how dopamine neurotransmission contributes to behavior and its dysfunction has been driven by technological advancements that permit spatiotemporally-defined measurements of dopaminergic signaling in behaving animals. In this review, we will discuss the evolution of in vivo neural monitoring technologies for measuring dopamine neuron function. We focus on the dopamine system for two reasons: (1) the central role of dopamine neurotransmission in normal behavior and disease, and (2) dopamine neuron measurements have long been at the forefront of in vivo neural monitoring technologies. We will provide a brief overview of standard techniques for monitoring dopamine function, including electrophysiology, microdialysis, and voltammetry. Then, we will discuss recent advancements in optical technologies using genetically-encoded fluorescent proteins (GEFPs), including a critical evaluation of their advantages and limitations.
Collapse
|
40
|
Labouesse MA, Cola RB, Patriarchi T. GPCR-Based Dopamine Sensors-A Detailed Guide to Inform Sensor Choice for In vivo Imaging. Int J Mol Sci 2020; 21:E8048. [PMID: 33126757 PMCID: PMC7672611 DOI: 10.3390/ijms21218048] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/12/2022] Open
Abstract
Understanding how dopamine (DA) encodes behavior depends on technologies that can reliably monitor DA release in freely-behaving animals. Recently, red and green genetically encoded sensors for DA (dLight, GRAB-DA) were developed and now provide the ability to track release dynamics at a subsecond resolution, with submicromolar affinity and high molecular specificity. Combined with rapid developments in in vivo imaging, these sensors have the potential to transform the field of DA sensing and DA-based drug discovery. When implementing these tools in the laboratory, it is important to consider there is not a 'one-size-fits-all' sensor. Sensor properties, most importantly their affinity and dynamic range, must be carefully chosen to match local DA levels. Molecular specificity, sensor kinetics, spectral properties, brightness, sensor scaffold and pharmacology can further influence sensor choice depending on the experimental question. In this review, we use DA as an example; we briefly summarize old and new techniques to monitor DA release, including DA biosensors. We then outline a map of DA heterogeneity across the brain and provide a guide for optimal sensor choice and implementation based on local DA levels and other experimental parameters. Altogether this review should act as a tool to guide DA sensor choice for end-users.
Collapse
Affiliation(s)
- Marie A. Labouesse
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA;
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Reto B. Cola
- Anatomy and Program in Neuroscience, University of Fribourg, 1700 Fribourg, Switzerland;
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
41
|
Shemesh OA, Linghu C, Piatkevich KD, Goodwin D, Celiker OT, Gritton HJ, Romano MF, Gao R, Yu CCJ, Tseng HA, Bensussen S, Narayan S, Yang CT, Freifeld L, Siciliano CA, Gupta I, Wang J, Pak N, Yoon YG, Ullmann JFP, Guner-Ataman B, Noamany H, Sheinkopf ZR, Park WM, Asano S, Keating AE, Trimmer JS, Reimer J, Tolias AS, Bear MF, Tye KM, Han X, Ahrens MB, Boyden ES. Precision Calcium Imaging of Dense Neural Populations via a Cell-Body-Targeted Calcium Indicator. Neuron 2020; 107:470-486.e11. [PMID: 32592656 DOI: 10.1016/j.neuron.2020.05.029] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 05/09/2019] [Accepted: 05/20/2020] [Indexed: 01/11/2023]
Abstract
Methods for one-photon fluorescent imaging of calcium dynamics can capture the activity of hundreds of neurons across large fields of view at a low equipment complexity and cost. In contrast to two-photon methods, however, one-photon methods suffer from higher levels of crosstalk from neuropil, resulting in a decreased signal-to-noise ratio and artifactual correlations of neural activity. We address this problem by engineering cell-body-targeted variants of the fluorescent calcium indicators GCaMP6f and GCaMP7f. We screened fusions of GCaMP to natural, as well as artificial, peptides and identified fusions that localized GCaMP to within 50 μm of the cell body of neurons in mice and larval zebrafish. One-photon imaging of soma-targeted GCaMP in dense neural circuits reported fewer artifactual spikes from neuropil, an increased signal-to-noise ratio, and decreased artifactual correlation across neurons. Thus, soma-targeting of fluorescent calcium indicators facilitates usage of simple, powerful, one-photon methods for imaging neural calcium dynamics.
Collapse
Affiliation(s)
- Or A Shemesh
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Biological Engineering, MIT, Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Changyang Linghu
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
| | - Kiryl D Piatkevich
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Biological Engineering, MIT, Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
| | - Daniel Goodwin
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Orhan Tunc Celiker
- MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
| | - Howard J Gritton
- Boston University, Department of Biomedical Engineering, Boston, MA 02215, USA
| | - Michael F Romano
- Boston University, Department of Biomedical Engineering, Boston, MA 02215, USA
| | - Ruixuan Gao
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Chih-Chieh Jay Yu
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Biological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Hua-An Tseng
- Boston University, Department of Biomedical Engineering, Boston, MA 02215, USA
| | - Seth Bensussen
- Boston University, Department of Biomedical Engineering, Boston, MA 02215, USA
| | - Sujatha Narayan
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Chao-Tsung Yang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Limor Freifeld
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Faculty of Biomedical Engineering, Technion, Haifa, Israel
| | - Cody A Siciliano
- Vanderbilt Center for Addiction Research, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Ishan Gupta
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Biological Engineering, MIT, Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Joyce Wang
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Nikita Pak
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Department of Mechanical Engineering, MIT, Cambridge, MA, USA
| | - Young-Gyu Yoon
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA; School of Electrical Engineering, KAIST Institute for Health Science and Technology, Daejeon, Republic of Korea
| | - Jeremy F P Ullmann
- Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital & Harvard Medical School, Boston, MA, USA
| | - Burcu Guner-Ataman
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Habiba Noamany
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Zoe R Sheinkopf
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Shoh Asano
- Internal Medicine Research Unit, Pfizer, Cambridge, MA, USA
| | - Amy E Keating
- Department of Biological Engineering, MIT, Cambridge, MA, USA; Department of Biology, MIT, Cambridge, MA, USA; Koch Institute, MIT, Cambridge, MA 02139, USA
| | - James S Trimmer
- Department of Physiology and Membrane Biology, University of California, Davis School of Medicine, Davis, CA, USA
| | - Jacob Reimer
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Center for Neuroscience and AI, Baylor College of Medicine, Houston, TX, USA
| | - Andreas S Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Center for Neuroscience and AI, Baylor College of Medicine, Houston, TX, USA; Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
| | - Mark F Bear
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Kay M Tye
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Xue Han
- Boston University, Department of Biomedical Engineering, Boston, MA 02215, USA
| | - Misha B Ahrens
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Edward S Boyden
- The MIT Media Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Biological Engineering, MIT, Cambridge, MA, USA; MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA; MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, USA; Koch Institute, MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
42
|
Siciliano CA, Noamany H, Chang CJ, Brown AR, Chen X, Leible D, Lee JJ, Wang J, Vernon AN, Vander Weele CM, Kimchi EY, Heiman M, Tye KM. A cortical-brainstem circuit predicts and governs compulsive alcohol drinking. Science 2020; 366:1008-1012. [PMID: 31754002 DOI: 10.1126/science.aay1186] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/04/2019] [Indexed: 12/31/2022]
Abstract
What individual differences in neural activity predict the future escalation of alcohol drinking from casual to compulsive? The neurobiological mechanisms that gate the transition from moderate to compulsive drinking remain poorly understood. We longitudinally tracked the development of compulsive drinking across a binge-drinking experience in male mice. Binge drinking unmasked individual differences, revealing latent traits in alcohol consumption and compulsive drinking despite equal prior exposure to alcohol. Distinct neural activity signatures of cortical neurons projecting to the brainstem before binge drinking predicted the ultimate emergence of compulsivity. Mimicry of activity patterns that predicted drinking phenotypes was sufficient to bidirectionally modulate drinking. Our results provide a mechanistic explanation for individual variance in vulnerability to compulsive alcohol drinking.
Collapse
Affiliation(s)
- Cody A Siciliano
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. .,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Habiba Noamany
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chia-Jung Chang
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alex R Brown
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Xinhong Chen
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel Leible
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jennifer J Lee
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joyce Wang
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Amanda N Vernon
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Caitlin M Vander Weele
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eyal Y Kimchi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Myriam Heiman
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kay M Tye
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. .,The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
43
|
Cameron CM, Murugan M, Choi JY, Engel EA, Witten IB. Increased Cocaine Motivation Is Associated with Degraded Spatial and Temporal Representations in IL-NAc Neurons. Neuron 2019; 103:80-91.e7. [PMID: 31101395 DOI: 10.1016/j.neuron.2019.04.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 02/07/2019] [Accepted: 04/05/2019] [Indexed: 01/03/2023]
Abstract
Craving for cocaine progressively increases in cocaine users during drug-free periods, contributing to relapse. The projection from the infralimbic cortex to the nucleus accumbens shell (IL-NAc) is thought to inhibit cocaine seeking. However, it is not known whether and how IL-NAc neurons contribute to the increased motivation associated with a drug-free period. We first performed cellular resolution imaging of IL-NAc neurons in rats during a drug-seeking test. This revealed neurons with spatial selectivity within the cocaine-associated context, a decrease in activity around the time of cocaine seeking, and an inverse relationship between cocaine-seeking activity and subsequent cocaine motivation. All these properties were reduced by a drug-free period. Next, we transiently activated this projection, which resulted in reduced drug seeking, regardless of the drug-free period. Taken together, this suggests that altered IL-NAc activity after a drug-free period may enhance cocaine motivation without fundamentally altering the projection's ability to inhibit drug seeking.
Collapse
Affiliation(s)
- Courtney M Cameron
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Malavika Murugan
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Jung Yoon Choi
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA; Department of Psychology, Princeton University, Princeton, NJ 08544, USA
| | - Esteban A Engel
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA; Department of Psychology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
44
|
Ron D, Weiner J. Special issue on new technologies for alcohol research and treatment. Alcohol 2019; 74:1-2. [PMID: 30409742 DOI: 10.1016/j.alcohol.2018.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 06/11/2018] [Indexed: 11/17/2022]
Affiliation(s)
- Dorit Ron
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143 United States.
| | - Jeff Weiner
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157 United States.
| |
Collapse
|